1
|
Zhang L, Yu Y, Ding K, Ji C, Zhang D, Liang P, Tang BZ, Feng G. Tumor microenvironment ameliorative and adaptive nanoparticles with photothermal-to-photodynamic switch for cancer phototherapy. Biomaterials 2025; 313:122771. [PMID: 39190940 DOI: 10.1016/j.biomaterials.2024.122771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024]
Abstract
The notorious tumor microenvironment (TME) usually becomes more deteriorative during phototherapeutic progress that hampers the antitumor efficacy. To overcome this issue, we herein report the ameliorative and adaptive nanoparticles (TPASIC-PFH@PLGA NPs) that simultaneously reverse hypoxia TME and switch photoactivities from photothermal-dominated state to photodynamic-dominated state to maximize phototherapeutic effect. TPASIC-PFH@PLGA NPs are designed by incorporating oxygen-rich liquid perfluorohexane (PFH) into the intraparticle microenvironment to regulate the intramolecular motions of AIE photosensitizer TPASIC. TPASIC exhibits a unique aggregation-enhanced reactive oxygen species (ROS) generation feature. PFH incorporation affords TPASIC the initially dispersed state, thus promoting active intramolecular motions and photothermal conversion efficiency. While PFH volatilization leads to nanoparticle collapse and the formation of tight TPASIC aggregates with largely enhanced ROS generation efficiency. As a consequence, PFH incorporation not only currently promotes both photothermal and photodynamic efficacies of TPASIC and increases the intratumoral oxygen level, but also enables the smart photothermal-to-photodynamic switch to maximize the phototherapeutic performance. The integration of PFH and AIE photosensitizer eventually delivers more excellent antitumor effect over conventional phototherapeutic agents with fixed photothermal and photodynamic efficacies. This study proposes a new nanoengineering strategy to ameliorate TME and adapt the treatment modality to fit the changed TME for advanced antitumor applications.
Collapse
Affiliation(s)
- Le Zhang
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, School of Materials Science and Engineering, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Yuewen Yu
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, School of Materials Science and Engineering, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Keke Ding
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 Zheshan Road, Wuhu, 241001, China
| | - Chao Ji
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, School of Materials Science and Engineering, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Di Zhang
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, School of Materials Science and Engineering, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Ping Liang
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, School of Materials Science and Engineering, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Ben Zhong Tang
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen City, Guangdong, 518172, China
| | - Guangxue Feng
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, School of Materials Science and Engineering, AIE Institute, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
2
|
Um‐e‐Kalsoom, Wang S, Qu J, Liu L. Innovative optical imaging strategies for monitoring immunotherapy in the tumor microenvironments. Cancer Med 2024; 13:e70155. [PMID: 39387259 PMCID: PMC11465031 DOI: 10.1002/cam4.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a critical role in cancer progression and response to immunotherapy. Immunotherapy targeting the immune system has emerged as a promising treatment modality, but challenges in understanding the TME limit its efficacy. Optical imaging strategies offer noninvasive, real-time insights into the interactions between immune cells and the TME. OBJECTIVE This review assesses the progress of optical imaging technologies in monitoring immunotherapy within the TME and explores their potential applications in clinical trials and personalized cancer treatment. METHODS This is a comprehensive literature review based on the advances in optical imaging modalities including fluorescence imaging (FLI), bioluminescence imaging (BLI), and photoacoustic imaging (PAI). These modalities were analyzed for their capacity to provide high-resolution, real-time imaging of immune cell dynamics, tumor vasculature, and other critical components of the TME. RESULTS Optical imaging techniques have shown significant potential in tracking immune cell infiltration, assessing immune checkpoint inhibitors, and visualizing drug delivery within the TME. Technologies like FLI and BLI are pivotal in tracking immune responses in preclinical models, while PAI provides functional imaging with deeper tissue penetration. The integration of these modalities with immunotherapy holds promise for improving treatment monitoring and outcomes. CONCLUSION Optical imaging is a powerful tool for understanding the complexities of the TME and optimizing immunotherapy. Further advancements in imaging technologies, combined with nanomaterial-based approaches, could pave the way for enhanced diagnostic accuracy and therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Um‐e‐Kalsoom
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| |
Collapse
|
3
|
Liu X, Ullah I, Yuan Y. Tumor Acidity-Triggered Bioorthogonal Reactions for Biomedical Applications. Chembiochem 2024; 25:e202400452. [PMID: 38940000 DOI: 10.1002/cbic.202400452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 06/29/2024]
Abstract
Cancer is one of the most serious threats to human health. Over the past few years, researchers have incrementally uncovered the pivotal role of tumor acidity in tumor formation, development, and treatment. In addition, bioorthogonal reactions have been widely used in tumor diagnosis and therapy, owing to their advantageous characteristics, including small ligand size, biocompatibility, fast reaction kinetics, and high chemospecificity. Consequently, bioorthogonal reactions triggered by tumor acidity have become an emerging strategy in biomedical applications. On this basis, we first elucidate the concept and major strategies of tumor acidity-triggered bioorthogonal reactions. Additionally, we review the progress in biomedical applications, with a particular focus on their importance in disease diagnosis and treatment. Finally, clinical challenges and future trends are also outlooked.
Collapse
Affiliation(s)
- Xiajian Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Ihsan Ullah
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
4
|
Yang Y, Wang N, Yan F, Shi Z, Feng S. Metal-organic frameworks as candidates for tumor sonodynamic therapy: Designable structures for targeted multifunctional transformation. Acta Biomater 2024; 181:67-97. [PMID: 38697383 DOI: 10.1016/j.actbio.2024.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/25/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Sonodynamic therapy (SDT), utilizing ultrasound (US) as the trigger, has gained popularity recently as a therapeutic approach with significant potential for treating various diseases. Metal-organic frameworks (MOFs), characterized by structural flexibility, are prominently emerging in the SDT realm as an innovative type of sonosensitizer, offering functional tunability and biocompatibility. However, due to the inherent limitations of MOFs, such as low reactivity to reactive oxygen species and challenges posed by the complex tumor microenvironment, MOF-based sonosensitizers with singular functions are unable to demonstrate the desired therapeutic efficacy and may pose risks of toxicity, limiting their biological applications to superficial tissues. MOFs generally possess distinctive crystalline structures and properties, and their controlled coordination environments provide a flexible platform for exploring structure-effect relationships and guiding the design and development of MOF-based nanomaterials to unlock their broader potential in biological fields. The primary focus of this paper is to summarize cases involving the modification of different MOF materials and the innovative strategies developed for various complex conditions. The paper outlines the diverse application areas of functionalized MOF-based sonosensitizers in tumor synergistic therapies, highlighting the extensive prospects of SDT. Additionally, challenges confronting SDT are briefly summarized to stimulate increased scientific interest in the practical application of MOFs and the successful clinical translation of SDT. Through these discussions, we strive to foster advancements that lead to early-stage clinical benefits for patients. STATEMENT OF SIGNIFICANCE: 1. An overview for the progresses in SDT explored from a novel and fundamental perspective. 2. Different modification strategies to improve the MOFs-mediated SDT efficacy are provided. 3. Guidelines for the design of multifunctional MOFs-based sonosensitizers are offered. 4. Powerful tumor ablation potential is reflected in SDT-led synergistic therapies. 5. Future challenges in the field of MOFs-based SDT in clinical translation are suggested.
Collapse
Affiliation(s)
- Yilin Yang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Ning Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China.
| | - Zhan Shi
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China.
| | - Shouhua Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| |
Collapse
|
5
|
Zhou Y, Li Q, Wu Y, Zhang W, Ding L, Ji C, Li P, Chen T, Feng L, Tang BZ, Huang X. Synergistic Brilliance: Engineered Bacteria and Nanomedicine Unite in Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313953. [PMID: 38400833 DOI: 10.1002/adma.202313953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Engineered bacteria are widely used in cancer treatment because live facultative/obligate anaerobes can selectively proliferate at tumor sites and reach hypoxic regions, thereby causing nutritional competition, enhancing immune responses, and producing anticancer microbial agents in situ to suppress tumor growth. Despite the unique advantages of bacteria-based cancer biotherapy, the insufficient treatment efficiency limits its application in the complete ablation of malignant tumors. The combination of nanomedicine and engineered bacteria has attracted increasing attention owing to their striking synergistic effects in cancer treatment. Engineered bacteria that function as natural vehicles can effectively deliver nanomedicines to tumor sites. Moreover, bacteria provide an opportunity to enhance nanomedicines by modulating the TME and producing substrates to support nanomedicine-mediated anticancer reactions. Nanomedicine exhibits excellent optical, magnetic, acoustic, and catalytic properties, and plays an important role in promoting bacteria-mediated biotherapies. The synergistic anticancer effects of engineered bacteria and nanomedicines in cancer therapy are comprehensively summarized in this review. Attention is paid not only to the fabrication of nanobiohybrid composites, but also to the interpromotion mechanism between engineered bacteria and nanomedicine in cancer therapy. Additionally, recent advances in engineered bacteria-synergized multimodal cancer therapies are highlighted.
Collapse
Affiliation(s)
- Yaofeng Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Qianying Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Yuhao Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Wan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Lu Ding
- Department of Cardiology, Jiangxi Hypertension Research Institute, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Chenlin Ji
- School of Engineering, Westlake University, Hangzhou, 310030, P. R. China
| | - Ping Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330036, P. R. China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ben Zhong Tang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, P. R. China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| |
Collapse
|
6
|
Lu XX, Xue C, Dong JH, Zhang YZ, Gao F. Nanoplatform-based strategies for enhancing the lethality of current antitumor PDT. J Mater Chem B 2024; 12:3209-3225. [PMID: 38497405 DOI: 10.1039/d4tb00008k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Photodynamic therapy (PDT) exhibits great application prospects in future clinical oncology due to its spatiotemporal controllability and good biosafety. However, the antitumor efficacy of PDT is seriously hindered by many factors, including tumor hypoxia, limited light penetration ability, and strong defense mechanisms of tumors. Considering that it is difficult to completely solve the first two problems, enhancing the lethality of antitumor PDT has become a good idea to extend its clinical application. Herein, we summarize the nanoplatform-involved strategies to effectively amplify the tumoricidal capability of current PDT and then discuss the present bottlenecks and prospects of the nanoplatform-based PDT sensitization strategies in tumor therapy. We hope this review will provide some references for others to design high-performance PDT nanoplatforms for tumor therapy.
Collapse
Affiliation(s)
- Xin-Xin Lu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Chun Xue
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Jian-Hui Dong
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Yi-Zhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| |
Collapse
|
7
|
Liu Y, Wang L, Zhang T, Wang C, Fan Y, Wang C, Song N, Zhou P, Yan CH, Tang Y. Tumor Microenvironment-Regulating Two-Photon Probe Based on Bimetallic Post-Coordinated MOF Facilitating the Dual-Modal and Deep Imaging-Guided Synergistic Therapies. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12289-12301. [PMID: 38418381 DOI: 10.1021/acsami.3c18990] [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: 03/01/2024]
Abstract
The intricate tumor microenvironment (TME) always brings about unsatisfactory therapeutic effects for treatments, although nanomedicines have been demonstrated to be highly beneficial for synergistic therapies to avoid the side effects caused by the complexity and heterogeneity of cancer. Developing nanotheranostics with the functionalities of both synergistic therapies and TME regulation is a good strategy but is still in its infancy. Herein, an "all-in-one" nanoplatform for integrated diagnosis and treatment, namely, Carrier@ICG@DOX@FA (CIDF), is constructed. Benefiting from the bimetallic coordination of Eu3+-HTHA (4,4,4-trifluoro-1-(9-hexylcarbazol-3-yl)-1,3-butanedione) and Fe3+ with the ligands in UiO-67, CIDF can simultaneously achieve two-photon fluorescence imaging, fluorescent lifetime imaging in deep tumors, and regulation of TME. Owing to its porosity, CIDF can encapsulate indocyanine green as photosensitizers and doxorubicin as chemotherapeutic agent, further realizing light-controlled drug release. Moreover, CIDF exhibited good biocompatibility and tumor targeting by coating with folic-acid-modified polymers. Both in vitro and in vivo experiments demonstrate the excellent therapeutic efficacy of CIDF through dual-modal-imaging-guided synergistic photothermal-, photodynamic-, and chemotherapy. CIDF provides a new paradigm for the construction of TME-regulated synergistic nanotheranostics and realizes the complete elimination of tumors without recurrence.
Collapse
Affiliation(s)
- Yanjun Liu
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Lu Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, P. R. China
| | - Tong Zhang
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Chunya Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yifan Fan
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Congcong Wang
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Nan Song
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Ping Zhou
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, P. R. China
| | - Chun-Hua Yan
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yu Tang
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
- State Key Laboratory of Baiyunobo Rare Earth Resource Researches and Comprehensive Utilization, Baotou Research Institute of Rare Earths, Baotou 014030, P. R. China
| |
Collapse
|
8
|
Cai J, Hu G, Hu L, Chen J, Chen D, Liu D, Wang X, Hu B, Li C. A CaCO 3-based nanoplatform with sonodynamic and tumor microenvironment activated for combined in vitro cancer therapy. Transl Oncol 2023; 38:101771. [PMID: 37729741 PMCID: PMC10518365 DOI: 10.1016/j.tranon.2023.101771] [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: 04/12/2023] [Revised: 07/27/2023] [Accepted: 08/28/2023] [Indexed: 09/22/2023] Open
Abstract
INTRODUCTION Sonodynamic therapy (SDT) has potential clinical applications for cancer therapy, and is yet restricted by complex tumor microenvironmental (TME) factors. Thus, the research problem of TME modulation as well as efficient tumor treatment still needs to be clarified. METHOD In this study, a calcium carbonate (CaCO3) nanoplatform was designed for ultrasound (US) and TME response-triggered, which encapsulated Ag2S and loaded with l-Arg, and further wrapped with RBC/Platelet membrane, named as QD@Ca/ML-Arg. RESULTS Non-invasive US-triggered SDT by Ag2S and acidic environment-responsive drug release were achieved. In vitro experiments validated the efficacy of SDT, Ca-ion interference and nitric oxide (NO) gas therapy as combined therapy for cancer treatment. By means of RNA sequencing, the cancer therapeutic mechanism of SDT in redox-related pathways was elucidated. The immunosuppressive TME was simulated with a M2-macrophage/cancer cell co-culture system to confirm the immune activating effect of immunogenic cell death (ICD). CONCLUSION Accordingly, the potential of QD@Ca/ML-Arg-was demonstrated for in vitro TME modulation, cancer therapeutic efficacy and clinical translation.
Collapse
Affiliation(s)
- Jiale Cai
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Guiping Hu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Lihua Hu
- Department of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Dan Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Dan Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Xiaolei Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Boxian Hu
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China
| | - Cheng Li
- Beijing Advanced Innovation Center for Biomedical Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine Beijing, Beihang University, Beijing 100191, China.
| |
Collapse
|
9
|
Liu X, Chu Z, Chen B, Ma Y, Xu L, Qian H, Yu Y. Cancer cell membrane-coated upconversion nanoparticles/Zn xMn 1-xS core-shell nanoparticles for targeted photodynamic and chemodynamic therapy of pancreatic cancer. Mater Today Bio 2023; 22:100765. [PMID: 37636984 PMCID: PMC10457453 DOI: 10.1016/j.mtbio.2023.100765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/29/2023] [Accepted: 08/05/2023] [Indexed: 08/29/2023] Open
Abstract
Oxidative stress induced by reactive oxygen species (ROS) is promising treatment approach for pancreatic ductal adenocarcinoma (PDAC), which is typically insensitive to conventional chemotherapy. In this study, BxPC-3 pancreatic cancer cell membrane-coated upconversion nanoparticles/ZnxMn1-xS core-shell nanoparticles (abbreviated as BUC@ZMS) were developed for tumor-targeted cancer therapy via synergistically oxidative stress and overcoming glutathione (GSH) overexpression. Using a combination of photodynamic therapy (PDT) and chemodynamic therapy (CDT), the BUC@ZMS core-shell nanoparticles were able to elicit the death of pancreatic cancer cells through the high production of ROS. Additionally, the BUC@ZMS core-shell nanoparticles could deplete intracellular GSH and increase the sensitivity of tumor cells to oxidative stress. The in vivo results indicated that BUC@ZMS nanoparticles can accumulate specifically in tumor locations and suppress PDAC without generating obvious toxicity. Thus, it was determined that the as-prepared core-shell nanoparticles would be a viable treatment option for solid malignancies.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
- Department of Gastroenterology, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui, 237000, PR China
| | - Zhaoyou Chu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, PR China
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Benjin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, PR China
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Lingling Xu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Yue Yu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| |
Collapse
|
10
|
Gao W, Shen R. Nanogel enhances the efficacy of MLN8237 in treating hepatocellular carcinoma. J Biomater Appl 2023; 38:527-537. [PMID: 37695622 DOI: 10.1177/08853282231202326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
MLN8237, a specific inhibitor of Aurora-A kinase, is proved to be a potential treatment strategy for hepatocellular carcinoma (HCC). Nanogels improve the efficacy of doxorubicin. Therefore, this study aims to investigate the increase in the effect of nanogels on MLN8237 in inhibiting HCC. Doxorubicin or MLN8237 was used as an anti-tumor drug models which were packaged by organic solvent volatilization method to obtain the doxorubicin-loaded nanogel and the MLN8237-loaded nanogel. Subsequently, CCK8 assay, cell cycle assay, apoptosis assay, real-time PCR, western blotting assay and animal experiments were used to detect the effects of MLN8237 nanogel on the proliferation, cell cycle, apoptosis, tumor growth, mRNA and protein levels of aurora-A and PUMA, and AKT phosphorylation levels in HCC cell lines. The results show that the nanogels can realize pH-regulated hydrophobicity reversal, have certain stability, and can realize lysosomal escape. Moreover, the MLN8237-loaded nanogel has a stronger ability to inhibit HCC cell proliferation, block cell cycle, promote apoptosis and inhibit tumor growth than free MLN8237 by suppressing aurora-A and AKT phosphorylation. In short, nanogel can enhance the efficacy of MLN8237.
Collapse
Affiliation(s)
- Wei Gao
- Department of General Surgery, The Second People's Hospital of Tongxiang, Zhejiang, China
| | - Rongxing Shen
- Department of General Surgery, The Second People's Hospital of Tongxiang, Zhejiang, China
| |
Collapse
|
11
|
Li J, Wang Q, Xia G, Adilijiang N, Li Y, Hou Z, Fan Z, Li J. Recent Advances in Targeted Drug Delivery Strategy for Enhancing Oncotherapy. Pharmaceutics 2023; 15:2233. [PMID: 37765202 PMCID: PMC10534854 DOI: 10.3390/pharmaceutics15092233] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Targeted drug delivery is a precise and effective strategy in oncotherapy that can accurately deliver drugs to tumor cells or tissues to enhance their therapeutic effect and, meanwhile, weaken their undesirable side effects on normal cells or tissues. In this research field, a large number of researchers have achieved significant breakthroughs and advances in oncotherapy. Typically, nanocarriers as a promising drug delivery strategy can effectively deliver drugs to the tumor site through enhanced permeability and retention (EPR) effect-mediated passive targeting and various types of receptor-mediated active targeting, respectively. Herein, we review recent targeted drug delivery strategies and technologies for enhancing oncotherapy. In addition, we also review two mainstream drug delivery strategies, passive and active targeting, based on various nanocarriers for enhancing tumor therapy. Meanwhile, a comparison and combination of passive and active targeting are also carried out. Furthermore, we discuss the associated challenges of passive and active targeted drug delivery strategies and the prospects for further study.
Collapse
Affiliation(s)
- Jianmin Li
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Qingluo Wang
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Guoyu Xia
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Nigela Adilijiang
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Ying Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhenqing Hou
- College of Materials, Xiamen University, Xiamen 361002, China;
| | - Zhongxiong Fan
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Jinyao Li
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| |
Collapse
|
12
|
Yue M, Guo T, Nie DY, Zhu YX, Lin M. Advances of nanotechnology applied to cancer stem cells. World J Stem Cells 2023; 15:514-529. [PMID: 37424953 PMCID: PMC10324502 DOI: 10.4252/wjsc.v15.i6.514] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/01/2023] [Accepted: 04/18/2023] [Indexed: 06/26/2023] Open
Abstract
Cancer stem cells (CSCs) are a small proportion of the cells that exist in cancer tissues. They are considered to be the culprit of tumor genesis, development, drug resistance, metastasis and recurrence because of their self-renewal, proliferation, and differentiation potential. The elimination of CSCs is thus the key to cure cancer, and targeting CSCs provides a new method for tumor treatment. Due to the advantages of controlled sustained release, targeting and high biocompatibility, a variety of nanomaterials are used in the diagnosis and treatments targeting CSCs and promote the recognition and removal of tumor cells and CSCs. This article mainly reviews the research progress of nanotechnology in sorting CSCs and nanodrug delivery systems targeting CSCs. Furthermore, we identify the problems and future research directions of nanotechnology in CSC therapy. We hope that this review will provide guidance for the design of nanotechnology as a drug carrier so that it can be used in clinic for cancer therapy as soon as possible.
Collapse
Affiliation(s)
- Miao Yue
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Ting Guo
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Deng-Yun Nie
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Yin-Xing Zhu
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Mei Lin
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| |
Collapse
|
13
|
Lu J, Zhang A, Zhang F, Linhardt RJ, Zhu Z, Yang Y, Zhang T, Lin Z, Zhang S, Zhao H, Sun P. Ganoderenic acid D-loaded functionalized graphene oxide-based carrier for active targeting therapy of cervical carcinoma. Biomed Pharmacother 2023; 164:114947. [PMID: 37269813 DOI: 10.1016/j.biopha.2023.114947] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/05/2023] Open
Abstract
Ganoderenic acid D (GAD), extracted from the Chinese herb Ganoderma lucidum, was loaded onto a graphene oxide-polyethylene glycol-anti-epidermal growth factor receptor (GO-PEG-EGFR) carrier to develop a targeting antitumor nanocomposite (GO-PEG@GAD). The carrier was fabricated from PEG and anti-EGFR aptamer modified GO. Targeting was mediated by the grafted anti-EGFR aptamer, which targets the membrane of HeLa cells. Physicochemical properties were characterized by transmission electron microscopy, dynamic light scattering, X-ray powder diffraction, and Fourier transform infrared spectroscopy. High loading content (77.3 % ± 1.08 %) and encapsulation efficiency (89.1 % ± 2.11 %) were achieved. Drug release continued for approximately 100 h. The targeting effect both in vitro and in vivo was confirmed by confocal laser scanning microscopy (CLSM) and imaging analysis system. The mass of the subcutaneous implanted tumor was significantly decreased by 27.27 ± 1.23 % after treatment with GO-PEG@GAD compared with the negative control group. Moreover, the in vivo anti-cervical carcinoma activity of this medicine was due to activation of the intrinsic mitochondrial pathway.
Collapse
Affiliation(s)
- Jiahui Lu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 311402 Hangzhou, PR China; College of Food Science and Engineering, Zhejiang University of Technology, 310014 Hangzhou, PR China
| | - Anqiang Zhang
- College of Food Science and Engineering, Zhejiang University of Technology, 310014 Hangzhou, PR China
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 12180 Troy, NY, USA
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 12180 Troy, NY, USA; Departments of Chemistry and Chemical Biology and Biomedical Engineering, Biological Science, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 12180 Troy, NY, USA
| | - Zhihui Zhu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 311402 Hangzhou, PR China
| | - Yanzi Yang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 311402 Hangzhou, PR China
| | - Tinghuang Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 311402 Hangzhou, PR China
| | - Zhibin Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 100191 Beijing, PR China
| | - Su Zhang
- Zhejiang Provincial Rural Industrial Development Co., Ltd, 310000, Hangzhou, PR China
| | - Huajun Zhao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 311402 Hangzhou, PR China.
| | - Peilong Sun
- College of Food Science and Engineering, Zhejiang University of Technology, 310014 Hangzhou, PR China.
| |
Collapse
|
14
|
Xia HY, Li BY, Kankala RK, Chen AZ, Wang SB. Hybrid nanoarchitectonics of molybdenum dioxide (MoO 2) and doxorubicin (DOX) for synergistic chemo-photothermal-based breast carcinoma therapy. Colloids Surf B Biointerfaces 2023; 227:113387. [PMID: 37285669 DOI: 10.1016/j.colsurfb.2023.113387] [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: 02/19/2023] [Revised: 05/10/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Cancer has emerged as one of the severe ailments due to the uncontrolled proliferation rate of cells, accounting for millions of deaths annually. Despite the availability of various treatment strategies, including surgical interventions, radiation, and chemotherapy, tremendous advancements in the past two decades of research have evidenced the generation of different nanotherapeutic designs toward providing synergistic therapy. In this study, we demonstrate the assembly of a versatile nanoplatform based on the hyaluronic acid (HA)-coated molybdenum dioxide (MoO2) assemblies to act against breast carcinoma. The hydrothermal approach-assisted MoO2 constructs are immobilized with doxorubicin (DOX) molecules on the surface. Further, these MoO2-DOX hybrids are encapsulated with the HA polymeric framework. Furthermore, the versatile nanocomposites of HA-coated MoO2-DOX hybrids are systematically characterized using various characterization techniques, and explored biocompatibility in the mouse fibroblasts (L929 cell line), as well as synergistic photothermal (808-nm laser irradiation for 10 min, 1 W/cm2) and chemotherapeutic properties against breast carcinoma (4T1 cells). Finally, the mechanistic views concerning the apoptosis rate are explored using the JC-1 assay to measure the intracellular mitochondrial membrane potential (MMP) levels. In conclusion, these findings indicated excellent photothermal and chemotherapeutic efficacies, exploring the enormous potential of MoO2 composites against breast cancer.
Collapse
Affiliation(s)
- Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, PR China
| | - Bo-Yi Li
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, PR China; College of Chemical Engineering, Huaqiao University, Xiamen 361021, PR China; Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, PR China.
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, PR China; College of Chemical Engineering, Huaqiao University, Xiamen 361021, PR China; Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, PR China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, PR China; Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, PR China.
| |
Collapse
|
15
|
Zhen W, Weichselbaum RR, Lin W. Nanoparticle-Mediated Radiotherapy Remodels the Tumor Microenvironment to Enhance Antitumor Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206370. [PMID: 36524978 PMCID: PMC10213153 DOI: 10.1002/adma.202206370] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/12/2022] [Indexed: 05/26/2023]
Abstract
Radiotherapy (RT) uses ionizing radiation to eradicate localized tumors and, in rare cases, control tumors outside of the irradiated fields via stimulating an antitumor immune response (abscopal effect). However, the therapeutic effect of RT is often limited by inherent physiological barriers of the tumor microenvironment (TME), such as hypoxia, abnormal vasculature, dense extracellular matrix (ECM), and an immunosuppressive TME. Thus, it is critical to develop new RT strategies that can remodel the TME to overcome radio-resistance and immune suppression. In the past decade, high-Z-element nanoparticles have been developed to increase radiotherapeutic indices of localized tumors by reducing X-ray doses and side effects to normal tissues and enhance abscopal effects by activating the TME to elicit systemic antitumor immunity. In this review, the principles of RT and radiosensitization, the mechanisms of radio-resistance and immune suppression, and the use of various nanoparticles to sensitize RT and remodel TMEs for enhanced antitumor efficacy are discussed. The challenges in clinical translation of multifunctional TME-remodeling nanoradiosensitizers are also highlighted.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
16
|
Li D, Ma S, Xu D, Meng X, Lei N, Liu C, Zhao Y, Qi Y, Cheng Z, Wang F. Peptide-functionalized therapeutic nanoplatform for treatment orthotopic triple negative breast cancer and bone metastasis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102669. [PMID: 36933756 DOI: 10.1016/j.nano.2023.102669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/26/2022] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Epidermal Growth Factor Receptor (EGFR) is a promising therapeutic target for triple-negative breast cancer (TNBC). Recently, specific EGFR-targeting peptide GE11-based delivery nano-system shows excellent potential because of its chemical versatility and good targeting ability. However, no further research focusing on the downstream of EGFR after binding with GE11 was explored. Hence, we tailor-designed a self-assembled nanoplatform named GENP using amphiphilic molecule of stearic acid-modified GE11. After loading doxorubicin (DOX), the resulted nanoplatform GENP@DOX demonstrated high loading efficiency and sustainable drug release. Importantly, our findings proved that GENP alone significantly suppressed the proliferation of MDA-MB-231 cells via EGFR-downstream PI3K/AKT signaling pathways, contributing to the synergistic treatment with its DOX release. Further work illustrated remarkable therapeutic efficacy both in orthotopic TNBC and its bone metastasis models with minimal biotoxicity. Together, the results highlight that our GENP-functionalized nanoplatform is a promising strategy for the synergistic therapeutic efficacy targeting EGFR-overexpressed cancer.
Collapse
Affiliation(s)
- Daifeng Li
- Department of Orthopedics, Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Shengnan Ma
- Department of Orthopedics, Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Denghui Xu
- Department of Orthopedics, Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Xiaocao Meng
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ningjing Lei
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Chen Liu
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ying Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yingqiu Qi
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 201203, China; Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| | - Fazhan Wang
- Department of Orthopedics, Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
17
|
Xie L, Zhang C, Liu M, Huang J, Jin X, Zhu C, Lv M, Yang N, Chen S, Shao M, Du X, Feng G. Nucleus-Targeting Manganese Dioxide Nanoparticles Coated with the Human Umbilical Cord Mesenchymal Stem Cell Membrane for Cancer Cell Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10541-10553. [PMID: 36787533 PMCID: PMC9982816 DOI: 10.1021/acsami.3c01176] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/03/2023] [Indexed: 05/29/2023]
Abstract
Recently, development of drug delivery systems for accurate delivery of antitumor drugs to tumor sites to improve their antitumor efficacy has attracted great interest in the area of cancer immunotherapy. In this report, an intelligent biodegradable hollow manganese dioxide (HMnO2) nanoparticle (NP) with a human umbilical cord mesenchymal stem cell (hUC-MSC) membrane coating was designed to exert efficient chemo-immunotherapy for cancer treatment. A TAT peptide-modified membrane structure was constructed for nuclear targeting. Our findings showed that this new nanoreactor inherited the active targeting capability of MSCs and exhibited tumoritropic accumulation significantly at the cancerous parts. Compared with other formulations, intravenous injection of the NPs markedly inhibited tumor growth, relapse, and metastasis. Moreover, we found that the NPs effectively boosted dendritic cell maturation and recruited effector T cells into tumors. Overall, this work demonstrates the great potential of applying MSC membrane-coated manganese dioxide NPs as nucleus-targeting nanocarriers in cancer chemo-immunotherapy.
Collapse
Affiliation(s)
- Lixu Xie
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
- Department
of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Changwen Zhang
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Miao Liu
- Department
of Pediatrics, Qingyun County People’s
Hospital, Dezhou 253700, China
| | - Jianling Huang
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Xiao Jin
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Changjun Zhu
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Minjie Lv
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Ning Yang
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Sixi Chen
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Mingyue Shao
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Xingran Du
- Department
of Infectious Disease, The Second Affiliated
Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| | - Ganzhu Feng
- Department
of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211000, China
| |
Collapse
|
18
|
Cao D, Chen L, Zhang Z, Luo Y, Zhao L, Yuan C, Lu J, Liu X, Li J. Biodegradable nanomaterials for diagnosis and therapy of tumors. J Mater Chem B 2023; 11:1829-1848. [PMID: 36786439 DOI: 10.1039/d2tb02591d] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although degradable nanomaterials have been widely designed and applied for cancer bioimaging and various cancer treatments, few reviews of biodegradable nanomaterials have been reported. Herein, we have summarized the representative research advances of biodegradable nanomaterials with respect to the mechanism of degradation and their application in tumor imaging and therapy. First, four kinds of tumor microenvironment (TME) responsive degradation are presented, including pH, glutathione (GSH), hypoxia and matrix metalloproteinase (MMP) responsive degradation. Second, external stimulation degradation is summarized briefly. Next, we have outlined the applications of nanomaterials in bioimaging. Finally, we have focused on some typical examples of biodegradable nanomaterials in radiotherapy (RT), photothermal therapy (PTT), starvation therapy, photodynamic therapy (PDT), chemotherapy, chemodynamic therapy (CDT), sonodynamic therapy (SDT), gene therapy, immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Dongmiao Cao
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Ziwen Zhang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Yu Luo
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Linjing Zhao
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Chunping Yuan
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Jie Lu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Xijian Liu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
19
|
Ma H, Yu G, Cheng J, Song L, Zhou Z, Zhao Y, Zhao Q, Liu L, Wei X, Yang M. Design of an Injectable Magnetic Hydrogel Based on the Tumor Microenvironment for Multimodal Synergistic Cancer Therapy. Biomacromolecules 2023; 24:868-885. [PMID: 36692905 DOI: 10.1021/acs.biomac.2c01300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Conventional tumor chemotherapy is limited by its low therapeutic efficacy and side effects, which severely hold back its further application. Drug delivery systems (DDSs) based on nanomaterials have attracted wide interest in cancer treatment; especially, the system can realize efficient synergistic therapies. Here, we designed a smart hydrogel drug delivery system with multiple responses to enhance the tumor treatment effect. By cross-linking oxidized hydroxypropyl cellulose with carboxymethyl chitosan, an injectable hydrogel was obtained, into which artesunate (ART), ferroferric oxide (Fe3O4) nanoparticles, and black phosphorus nanosheets (BPs) were preloaded. This DDS has multiple functions including magnetic targeting, pH sensitivity, chemodynamic therapy, and photothermal response. This nanoparticle-composited hydrogel not only preserved excellent rheological properties but also allowed for an accurate stable drug release at tumor sites and synergistic effects of multiple therapies. The in vitro and in vivo experiments revealed that this DDS could efficiently eliminate the HepG2 tumor with good biocompatibility. Taken together, this study clarifies the possible antitumor mechanism of this ART-loaded nanoparticle-composited hydrogel and provides a new strategy for synergistic photothermal-chemo-chemodynamic therapy.
Collapse
Affiliation(s)
- Hai Ma
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Guanghao Yu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Jinlai Cheng
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Lixia Song
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Ziyu Zhou
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Yu Zhao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Qinghe Zhao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Li Liu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Xiaolu Wei
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| | - Miyi Yang
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Dongzhimen Nei Ave., Nanxiaojie 16#, Dongcheng District, Beijing100700, China
| |
Collapse
|
20
|
Liu W, Ma X, Kheyr SM, Dong A, Zhang J. Covalent Organic Frameworks as Nanocarriers for Improved Delivery of Chemotherapeutic Agents. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7215. [PMID: 36295281 PMCID: PMC9611971 DOI: 10.3390/ma15207215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 06/16/2023]
Abstract
Cancer has become one of the main causes of death worldwide. Chemotherapy as one of the main therapy modalities is very unsatisfactory. The various nanocarriers have brought new opportunities for effective tumor treatment. However, most of the current nanocarriers still suffer from low efficiency and confront significant challenges in overcoming multiple biological barriers. Compared with conventional nanocarriers, covalent organic frameworks (COFs) with unique and attractive features exhibited great potential to serve as a promising platform for anticancer drug delivery. In this review, we first summarize the strategies and challenges of nanocarriers for cancer chemotherapy and then highlight the recent advances in COF-based nanocarriers for improved delivery of chemotherapeutic agents. Finally, the challenges remaining for COF-based nanocarriers for clinical applications are outlined.
Collapse
Affiliation(s)
- Weiming Liu
- Key Laboratory of Systems Bioengineering of the Ministry of Education, Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Xinyu Ma
- Key Laboratory of Systems Bioengineering of the Ministry of Education, Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Shuayb Mohamed Kheyr
- Key Laboratory of Systems Bioengineering of the Ministry of Education, Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Anjie Dong
- Key Laboratory of Systems Bioengineering of the Ministry of Education, Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Jianhua Zhang
- Key Laboratory of Systems Bioengineering of the Ministry of Education, Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300350, China
| |
Collapse
|
21
|
Chu S, Shi X, Tian Y, Gao F. pH-Responsive Polymer Nanomaterials for Tumor Therapy. Front Oncol 2022; 12:855019. [PMID: 35392227 PMCID: PMC8980858 DOI: 10.3389/fonc.2022.855019] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
The complexity of the tumor microenvironment presents significant challenges to cancer therapy, while providing opportunities for targeted drug delivery. Using characteristic signals of the tumor microenvironment, various stimuli-responsive drug delivery systems can be constructed for targeted drug delivery to tumor sites. Among these, the pH is frequently utilized, owing to the pH of the tumor microenvironment being lower than that of blood and healthy tissues. pH-responsive polymer carriers can improve the efficiency of drug delivery in vivo, allow targeted drug delivery, and reduce adverse drug reactions, enabling multifunctional and personalized treatment. pH-responsive polymers have gained increasing interest due to their advantageous properties and potential for applicability in tumor therapy. In this review, recent advances in, and common applications of, pH-responsive polymer nanomaterials for drug delivery in cancer therapy are summarized, with a focus on the different types of pH-responsive polymers. Moreover, the challenges and future applications in this field are prospected.
Collapse
Affiliation(s)
- Shunli Chu
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiaolu Shi
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ye Tian
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Fengxiang Gao
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
22
|
Shen J, Chang R, Chang L, Wang Y, Deng K, Wang D, Qin J. Light emitting CMC-CHO based self-healing hydrogel with injectability for in vivo wound repairing applications. Carbohydr Polym 2022; 281:119052. [DOI: 10.1016/j.carbpol.2021.119052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 01/23/2023]
|
23
|
Zhuang Y, Han S, Fang Y, Huang H, Wu J. Multidimensional transitional metal-actuated nanoplatforms for cancer chemodynamic modulation. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214360] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Homotypic Cancer Cell Membranes Camouflaged Nanoparticles for Targeting Drug Delivery and Enhanced Chemo-Photothermal Therapy of Glioma. Pharmaceuticals (Basel) 2022; 15:ph15020157. [PMID: 35215270 PMCID: PMC8879672 DOI: 10.3390/ph15020157] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Glioma is among the deadliest types of brain cancer, for which there currently is no effective treatment. Chemotherapy is mainstay in the treatment of glioma. However, drug tolerance, non-targeting, and poor blood–brain barrier penetrance severely inhibits the efficacy of chemotherapeutics. An improved treatment method is thus urgently needed. Herein, a multifunctional biomimetic nanoplatform was developed by encapsulating graphene quantum dots (GQDs) and doxorubicin (DOX) inside a homotypic cancer cell membrane (CCM) for targeted chemo-photothermal therapy of glioma. The GQDs with stable fluorescence and a superior light-to-heat conversion property were synthesized as photothermal therapeutic agents and co-encapsulated with DOX in CCM. The as-prepared nanoplatform exhibited a high DOX loading efficiency. The cell membrane coating protected drugs from leakage. Upon an external laser stimuli, the membrane could be destroyed, resulting in rapid DOX release. By taking advantage of the homologous targeting of the cancer cell membrane, the GQDs/DOX@CCM were found to actively target tumor cells, resulting in significantly enhanced cellular uptake. Moreover, a superior suppression efficiency of GQDs/DOX@CCM to cancer cells through chemo-photothermal treatment was also observed. The results suggest that this biomimetic nanoplatform holds potential for efficient targeting of drug delivery and synergistic chemo-photothermal therapy of glioma.
Collapse
|
25
|
Chen D, Jiang L, Lei T, Xiao G, Wang Y, Zuo X, Li B, Li L, Wang J. Magnetic CuFe 2O 4 with intrinsic protease-like activity inhibited cancer cell proliferation and migration through mediating intracellular proteins. BIOMATERIALS AND BIOSYSTEMS 2022; 5:100038. [PMID: 36825110 PMCID: PMC9934488 DOI: 10.1016/j.bbiosy.2021.100038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 11/17/2022] Open
Abstract
Protease has been widely used in biological and industrial fields. Developing efficient artificial enzyme mimics remains a major technical challenge due to the high stability of peptide bonds. Nanoenzymes with high stability, high activity and low cost, provided new opportunities to break through natural enzyme inherent limitations. However, compared with many nanomaterials with inherent peroxidase activity, the intrinsic mimic proteases properties of magnetic nanomaterials were seldom explored, let alone the interaction between magnetic nanomaterials and cellular proteins. Herein, we reported for the first time that magnetic CuFe2O4 possesses inherent protease activity to hydrolyze bovine serum albumin (BSA) and casein under physiological conditions, and the CuFe2O4 is more resistant to high temperature than the natural trypsin. It also exhibited significantly higher catalytic efficiency than other copper nanomaterials and can be recycled for many times. Protease participated in pathophysiological processes and all stages of tumor progression. Interesting, CuFe2O4 exhibited anti-proliferative effect on A549, SKOV3, HT-29, BABL-3T3 and HUVEC cells, as well as it was particularly sensitive against SKOV3 cells. CuFe2O4 was about 30 times more effective than conventional chemotherapy drugs oxaliplatin and artesunate against SKOV3 cells. In addition, CuFe2O4 also mediated the expression of intracellular proteins, such as MMP-2, MMP-9, F-actin, and NF-kB, which may be associated with global protein hydrolysis by CuFe2O4, leading to inhibition of cell migration. The merits of the high magnetic properties, good protease-mimic and antitumor activities make CuFe2O4 nanoparticles very prospective candidates for many applications such as proteomics and biotechnology.
Collapse
Affiliation(s)
- Daomei Chen
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
| | - Liang Jiang
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- School of Chemical Sciences & Technology, Yunnan University, Kunming 650091, P R China
| | - Tao Lei
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- School of Chemical Sciences & Technology, Yunnan University, Kunming 650091, P R China
| | - Guo Xiao
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- School of Chemical Sciences & Technology, Yunnan University, Kunming 650091, P R China
| | - Yuanfeng Wang
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- School of Chemical Sciences & Technology, Yunnan University, Kunming 650091, P R China
| | - Xiaoqiong Zuo
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- School of Chemical Sciences & Technology, Yunnan University, Kunming 650091, P R China
| | - Bin Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- Corresponding authors.
| | - Lingli Li
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- School of Chemical Sciences & Technology, Yunnan University, Kunming 650091, P R China
| | - Jiaqiang Wang
- National Center for International Research on Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming 650091, P R China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming 650091, P R China
- School of Chemical Sciences & Technology, Yunnan University, Kunming 650091, P R China
- Corresponding authors.
| |
Collapse
|
26
|
Liu X, Gao P, Shi M, Chen Y, Pan W, Li N, Tang B. An autophagy-inhibitory MOF nanoreactor for tumor-targeted synergistic therapy. Biomater Sci 2022; 10:3088-3091. [DOI: 10.1039/d2bm00579d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An autophagy-inhibitory metal-organic framework (MOF) nanoreactor was developed for tumor-targeted synergistic therapy. The nanoreactor could inhibit autophagy to enhance the glucose oxidase (GOx)-mediated starvation therapy. And the H2O2 generated in...
Collapse
|
27
|
Bai M, Yang M, Gong J, Xu H, Wei Z. Progress and Principle of Drug Nanocrystals for Tumor Targeted Delivery. AAPS PharmSciTech 2021; 23:41. [PMID: 34964079 DOI: 10.1208/s12249-021-02200-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/09/2021] [Indexed: 12/26/2022] Open
Abstract
Drugs are referred to as drug nanocrystals when they exist as nanoscale crystal structures. This kind of nanocarrier has been widely utilized to increase the solubility and absorption for poorly aqueous soluble drugs after oral administration, or prolong the drug circulation when intravenous administration. The systemic cytotoxicity caused by antitumor drugs usually come from the nonspecific drug distribution. To solve the disadvantage of poor targetability, drug nanocrystals for tumor targeted delivery have been developed in recent years. In this review, the targeting mechanisms of various surface modified drug nanocrystals are introduced with the focus on passive targeting, active targeting and stimuli-responsive targeting in details. Function and application of common surface modified materials are also discussed.
Collapse
|
28
|
Ouyang A, Zhao D, Wang X, Zhang W, Jiang T, Li A, Liu W. Covalent RGD-graphene-phthalocyanine nanocomposite for fluorescence imaging-guided dual active/passive tumor-targeted combinatorial phototherapy. J Mater Chem B 2021; 10:306-320. [PMID: 34935023 DOI: 10.1039/d1tb02254g] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Poor tumor selectivity, low stability and quenched fluorescence are the main challenges to be overcome for nanomedicine, and are mainly caused by the dissociation of the nanostructure and aggregation of chromophores in the biological environment. Herein, covalently connected nanoparticles RGD-graphene-phthalocyanine (RGD-GO-SiPc) were constructed based on RGD peptide, silicon phthalocyanine (SiPc) and graphene oxide (GO) via a conjugation reaction for fluorescence imaging-guided cancer-targeted combinatorial phototherapy. The prepared RGD-GO-SiPc exhibited supreme biological stability, high-contrast fluorescence imaging, significantly enhanced NIR absorption, high photothermal conversion efficiency (25.6%), greatly improved cancer-targeting capability, and synergistic photodynamic (PDT) and photothermal therapy (PTT) efficacy along with low toxicity. Both in vitro and in vivo biological studies showed that RGD-GO-SiPc is a kind of promising multifunctional nanomedicine for fluorescence imaging-guided combined photothermal and photodynamic therapy with dual active/passive tumor-targeting properties.
Collapse
Affiliation(s)
- Ancheng Ouyang
- Institute of Crystal Materials, State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Dongmu Zhao
- Institute of Crystal Materials, State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Xianglei Wang
- Institute of Crystal Materials, State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Wei Zhang
- Institute of Crystal Materials, State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| | - Tianyu Jiang
- Helmholtz International Lab for Anti-Infectives, Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao - 266237, P. R. China
| | - Aiying Li
- Helmholtz International Lab for Anti-Infectives, Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao - 266237, P. R. China
| | - Wei Liu
- Institute of Crystal Materials, State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China.
| |
Collapse
|
29
|
Ravi Kiran AVVV, Kusuma Kumari G, Krishnamurthy PT, Khaydarov RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021; 9:7667-7704. [PMID: 34673853 DOI: 10.1039/d1bm01127h] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over recent years, advancements in nanomedicine have allowed new approaches to diagnose and treat tumors. Nano drug delivery systems exploit the enhanced permeability and retention (EPR) effect and enter the tumor tissue's interstitial space. However, tumor barriers play a crucial role, and cause inefficient EPR or the homing effect. Mounting evidence supports the hypothesis that the components of the tumor microenvironment, such as the extracellular matrix, and cellular and physiological components collectively or cooperatively hinder entry and distribution of drugs, and therefore, limit the theragnostic applications of cancer nanomedicine. This abnormal tumor microenvironment plays a pivotal role in cancer nanomedicine and was recently recognized as a promising target for improving nano-drug delivery and their therapeutic outcomes. Strategies like passive or active targeting, stimuli-triggered nanocarriers, and the modulation of immune components have shown promising results in achieving anticancer efficacy. The present review focuses on the tumor microenvironment and nanoparticle-based strategies (polymeric, inorganic and organic nanoparticles) for intruding the tumor barrier and improving therapeutic effects.
Collapse
Affiliation(s)
- Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Renat R Khaydarov
- Institute of Nuclear Physics, Uzbekistan Academy of Sciences, Tashkent, 100047, Uzbekistan.
| |
Collapse
|
30
|
Valenzuela C, Chen C, Sun M, Ye Z, Zhang J. Strategies and applications of covalent organic frameworks as promising nanoplatforms in cancer therapy. J Mater Chem B 2021; 9:3450-3483. [PMID: 33909746 DOI: 10.1039/d1tb00041a] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer nanomedicine is the best option to face the limits of conventional chemotherapy and phototherapy methods, and thus the intensive quest for new nanomaterials to improve therapeutic efficacy and safety is still underway. Owing to their low density, well-defined structures, large surface area, finely tunable pore size, and metal ion free features, covalent organic frameworks (COFs) have been extensively studied in many research fields. The recent great interest in nanoscale COFs to improve the properties of bulk COFs has led to broadening of their applicability in the biomedical field, such as nanocarriers with an outstanding loading capacity and efficient delivery of therapeutic agents, smart theranostic nanoplatforms with excellent stability, high ROS generation, light-to-heat conversion capabilities, and different response and diagnostic characteristics. The COFs and related nanoplatforms with a wide variety of designability and functionalization have opened up a new avenue for exciting opportunities in cancer therapy. Herein we review the state-of-the-art technical and scientific developments in this emerging field, focusing on the overall progress addressed so far in building versatile COF-based nanoplatforms to enhance chemotherapy, photodynamic/photothermal therapy, and combination. Future perspectives for achieving the synergistic effect of cancer elimination and clinical translation are further discussed to motivate future contributions and explore new possibilities.
Collapse
Affiliation(s)
- Cristian Valenzuela
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Chu Chen
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Mengxiao Sun
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Zhanpeng Ye
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Jianhua Zhang
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China. and Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300350, China
| |
Collapse
|
31
|
Liang JL, Luo GF, Chen WH, Zhang XZ. Recent Advances in Engineered Materials for Immunotherapy-Involved Combination Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007630. [PMID: 34050564 DOI: 10.1002/adma.202007630] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/18/2020] [Indexed: 06/12/2023]
Abstract
Immunotherapy that can activate immunity or enhance the immunogenicity of tumors has emerged as one of the most effective methods for cancer therapy. Nevertheless, single-mode immunotherapy is still confronted with several critical challenges, such as the low immune response, the low tumor infiltration, and the complex immunosuppression tumor microenvironment. Recently, the combination of immunotherapy with other therapeutic modalities has emerged as a powerful strategy to augment the therapeutic outcome in fighting against cancer. In this review, recent research advances of the combination of immunotherapy with chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, metabolic therapy, and microwave thermotherapy are summarized. Critical challenges and future research direction of immunotherapy-based cancer therapeutic strategy are also discussed.
Collapse
Affiliation(s)
- Jun-Long Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
32
|
Zhang C, Chen W, Zhang T, Jiang X, Hu Y. Hybrid nanoparticle composites applied to photodynamic therapy: strategies and applications. J Mater Chem B 2021; 8:4726-4737. [PMID: 32104868 DOI: 10.1039/d0tb00093k] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photodynamic therapy (PDT), as a robust strategy, has long been applied to cancer treatment owing to the meaningful breakthroughs and unique advantages, including ignorable invasiveness and spatio-temporal selectivity. Numerous PDT agents, especially hybrid nanoparticle composite (hybrid)-based sensitizers consisting of an organic polymer and inorganic nanoparticles (NPs), feature the synergetic pros of the components, which have unlocked the additional potentials of PDT. Although reviews relating to the applications of hybrids to PDT have been previously reported, most of them only focus on the designs of smart hybrids integrating multimodal imaging-guided multiple treatment modalities. Traditional PDT treatment has several limitations, such as inadequate PDT agents accumulating in cancer tissues, inferior PDT effect due to the devastating cancer hypoxia environment, relevant systemic toxicity in non-intelligent stimulation response treatment systems, and serious dependence of PDT on external light sources. Many strategies have been developed for overcoming these limitations, including improvement of cancer-homing ability by introducing active targeting groups, remodeling of the cancer hypoxia environment through oxygen regulators, intratumor release of ROS through activatable molecules, and replacement of laser light by X-rays or self-luminescence. This review aims to summarize the most recent advances in designing hybrids for improving the therapeutic efficacy of PDT.
Collapse
Affiliation(s)
- Chao Zhang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China. and Shenzhen Research Institute of Nanjing University, Shenzhen, 518057, China
| | - Weizhi Chen
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210093, China.
| | - Taixing Zhang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210093, China.
| | - Yong Hu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China. and Shenzhen Research Institute of Nanjing University, Shenzhen, 518057, China
| |
Collapse
|
33
|
Wang J, Ma P, Kim DH, Liu BF, Demirci U. Towards Microfluidic-Based Exosome Isolation and Detection for Tumor Therapy. NANO TODAY 2021; 37:101066. [PMID: 33777166 PMCID: PMC7990116 DOI: 10.1016/j.nantod.2020.101066] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Exosomes are a class of cell-secreted, nano-sized extracellular vesicles with a bilayer membrane structure of 30-150 nm in diameter. Their discovery and application have brought breakthroughs in numerous areas, such as liquid biopsies, cancer biology, drug delivery, immunotherapy, tissue repair, and cardiovascular diseases. Isolation of exosomes is the first step in exosome-related research and its applications. Standard benchtop exosome separation and sensing techniques are tedious and challenging, as they require large sample volumes, multi-step operations that are complex and time-consuming, requiring cumbersome and expensive instruments. In contrast, microfluidic platforms have the potential to overcome some of these limitations, owing to their high-precision processing, ability to handle liquids at a microscale, and integrability with various functional units, such as mixers, actuators, reactors, separators, and sensors. These platforms can optimize the detection process on a single device, representing a robust and versatile technique for exosome separation and sensing to attain high purity and high recovery rates with a short processing time. Herein, we overview microfluidic strategies for exosome isolation based on their hydrodynamic properties, size filtration, acoustic fields, immunoaffinity, and dielectrophoretic properties. We focus especially on advances in label-free isolation of exosomes with active biological properties and intact morphological structures. Further, we introduce microfluidic techniques for the detection of exosomal proteins and RNAs with high sensitivity, high specificity, and low detection limits. We summarize the biomedical applications of exosome-mediated therapeutic delivery targeting cancer cells. To highlight the advantages of microfluidic platforms, conventional techniques are included for comparison. Future challenges and prospects of microfluidics towards exosome isolation applications are also discussed. Although the use of exosomes in clinical applications still faces biological, technical, regulatory, and market challenges, in the foreseeable future, recent developments in microfluidic technologies are expected to pave the way for tailoring exosome-related applications in precision medicine.
Collapse
Affiliation(s)
- Jie Wang
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Peng Ma
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Daniel H Kim
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Bi-Feng Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| |
Collapse
|
34
|
Cong X, Chen J, Xu R. Tumor-Acidity Responsive Polymeric Nanoparticles for Targeting Delivery of Angiogenesis Inhibitor for Enhanced Antitumor Efficacy With Decreased Toxicity. Front Bioeng Biotechnol 2021; 9:664051. [PMID: 33842451 PMCID: PMC8024478 DOI: 10.3389/fbioe.2021.664051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/01/2021] [Indexed: 11/21/2022] Open
Abstract
Various nanocarriers with tumor targeting ability and improved pharmacokinetic property have been extensively utilized to reduce the toxicity of existing clinical chemotherapeutics. Herein, we showed that by encapsulating angiogenesis inhibitor anlotinib into polymeric nanoparticles, we could significantly decrease its in vivo toxicity. The introduction of pH-responsiveness into the nanocarrier further enhanced its anti-tumor activity. Systemic administration of the anlotinib-loaded nanocarrier into mice bearing A549 and 4T1 subcutaneous tumor received a higher tumor growth suppression and metastasis inhibition without detectable side effects. This strategy offers a promising option to improve the patient compliance of anlotinib.
Collapse
Affiliation(s)
- Xiufeng Cong
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ran Xu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
35
|
Chen D, Li B, Lei T, Na D, Nie M, Yang Y, Congjia, Xie, He Z, Wang J. Selective mediation of ovarian cancer SKOV3 cells death by pristine carbon quantum dots/Cu 2O composite through targeting matrix metalloproteinases, angiogenic cytokines and cytoskeleton. J Nanobiotechnology 2021; 19:68. [PMID: 33663548 PMCID: PMC7934478 DOI: 10.1186/s12951-021-00813-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/22/2021] [Indexed: 01/07/2023] Open
Abstract
It was shown that some nanomaterials may have anticancer properties, but lack of selectivity is one of challenges, let alone selective suppression of cancer growth by regulating the cellular microenvironment. Herein, we demonstrated for the first time that carbon quantum dots/Cu2O composite (CQDs/Cu2O) selectively inhibited ovarian cancer SKOV3 cells by targeting cellular microenvironment, such as matrix metalloproteinases, angiogenic cytokines and cytoskeleton. The result was showed CQDs/Cu2O possessed anticancer properties against SKOV3 cells with IC50 = 0.85 μg mL-1, which was approximately threefold lower than other tested cancer cells and approximately 12-fold lower than normal cells. Compared with popular anticancer drugs, the IC50 of CQDs/Cu2O was approximately 114-fold and 75-fold lower than the IC50 of commercial artesunate (ART) and oxaliplatin (OXA). Furthermore, CQDs/Cu2O possessed the ability to decrease the expression of MMP-2/9 and induced alterations in the cytoskeleton of SKOV3 cells by disruption of F-actin. It also exhibited stronger antiangiogenic effects than commercial antiangiogenic inhibitor (SU5416) through down-regulating the expression of VEGFR2. In addition, CQDs/Cu2O has a vital function on transcriptional regulation of multiple genes in SKOV3 cells, where 495 genes were up-regulated and 756 genes were down-regulated. It is worth noting that CQDs/Cu2O also regulated angiogenesis-related genes in SKOV3 cells, such as Maspin and TSP1 gene, to suppress angiogenesis. Therefore, CQDs/Cu2O selectively mediated of ovarian cancer SKOV3 cells death mainly through decreasing the expression of MMP-2, MMP-9, F-actin, and VEGFR2, meanwhile CQDs/Cu2O caused apoptosis of SKOV3 via S phase cell cycle arrest. These findings reveal a new application for the use of CQDs/Cu2O composite as potential therapeutic interventions in ovarian cancer SKOV3 cells.
Collapse
Affiliation(s)
- Daomei Chen
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China
| | - Bin Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China.
| | - Tao Lei
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China
- School of Chemical Sciences & Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Di Na
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China
- School of Chemical Sciences & Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Minfang Nie
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China
- School of Chemical Sciences & Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Yepeng Yang
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China
- School of Chemical Sciences & Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | | | - Xie
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China
- School of Chemical Sciences & Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Zijuan He
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China
- School of Chemical Sciences & Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Jiaqiang Wang
- National Center for International Research On Photoelectric and Energy Materials, School of Materials and Energy, Yunnan University, Kunming, 650091, People's Republic of China.
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, 650091, People's Republic of China.
- School of Chemical Sciences & Technology, Yunnan University, Kunming, 650091, People's Republic of China.
| |
Collapse
|
36
|
Cook A, Decuzzi P. Harnessing Endogenous Stimuli for Responsive Materials in Theranostics. ACS NANO 2021; 15:2068-2098. [PMID: 33555171 PMCID: PMC7905878 DOI: 10.1021/acsnano.0c09115] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/02/2021] [Indexed: 05/04/2023]
Abstract
Materials that respond to endogenous stimuli are being leveraged to enhance spatiotemporal control in a range of biomedical applications from drug delivery to diagnostic tools. The design of materials that undergo morphological or chemical changes in response to specific biological cues or pathologies will be an important area of research for improving efficacies of existing therapies and imaging agents, while also being promising for developing personalized theranostic systems. Internal stimuli-responsive systems can be engineered across length scales from nanometers to macroscopic and can respond to endogenous signals such as enzymes, pH, glucose, ATP, hypoxia, redox signals, and nucleic acids by incorporating synthetic bio-inspired moieties or natural building blocks. This Review will summarize response mechanisms and fabrication strategies used in internal stimuli-responsive materials with a focus on drug delivery and imaging for a broad range of pathologies, including cancer, diabetes, vascular disorders, inflammation, and microbial infections. We will also discuss observed challenges, future research directions, and clinical translation aspects of these responsive materials.
Collapse
Affiliation(s)
- Alexander
B. Cook
- Laboratory of Nanotechnology
for Precision Medicine, Istituto Italiano
di Tecnologia, Via Morego
30, 16163 Genova, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology
for Precision Medicine, Istituto Italiano
di Tecnologia, Via Morego
30, 16163 Genova, Italy
| |
Collapse
|
37
|
Zhang Y, Han X, Nie G. Responsive and activable nanomedicines for remodeling the tumor microenvironment. Nat Protoc 2021; 16:405-430. [PMID: 33311713 DOI: 10.1038/s41596-020-00421-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/23/2020] [Indexed: 01/19/2023]
Abstract
Here we describe two protocols for the construction of responsive and activable nanomedicines that regulate the tumor microenvironment (TME). The TME is composed of all non-cellular and cellular components surrounding a tumor, including the surrounding blood vessels, immune cells, fibroblasts, signaling molecules, and extracellular matrix and has a crucial role in tumor initiation, growth, and metastasis. Owing to the relatively stable properties of the TME compared to tumor cells, which exhibit frequent genetic mutations and epigenetic changes, therapeutic strategies targeting the TME using multifunctional nanomedicines hold great potential for anti-tumor therapy. By regulating tumor-associated platelets and pancreatic stellate cells (PSCs), the two major players in the TME, we can effectively manipulate the physiological barriers for enhanced drug delivery and significantly improve the tumor penetration and therapeutic efficacy of chemotherapeutics. The preparation and characterization of the multifunctional nanoparticles takes ~10 h for tumor-associated platelet regulation and 16 h for PSC regulation. These nanoformulations can be readily applied to regulate other components in the TME to realize synergistic or additive anti-tumor activity.
Collapse
Affiliation(s)
- Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xuexiang Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
- GBA Research Innovation Institute for Nanotechnology, Guangdong, China.
| |
Collapse
|
38
|
Chen B, Zhang C, Wang W, Chu Z, Zha Z, He X, Zhou W, Liu T, Wang H, Qian H. Ultrastable AgBiS 2 Hollow Nanospheres with Cancer Cell-Specific Cytotoxicity for Multimodal Tumor Therapy. ACS NANO 2020; 14:14919-14928. [PMID: 33137257 DOI: 10.1021/acsnano.0c04370] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Specific cytotoxicity for catalytic nanomedicine triggered by the tumor microenvironment (TME) has attracted increasing interest. In this work, we prepared AgBiS2 hollow nanospheres with narrow bandgaps via rapid precipitation in a weakly polar solvent, which lowered the intrinsic energy gap for the active production of highly reactive hydroxyl radicals (•OH), especially in the TME. The as-prepared AgBiS2 hollow nanospheres exhibited enhanced optical absorption and high photothermal conversion efficiency (44.2%). In addition, the hollow structured AgBiS2 nanospheres were found to have a peroxidase-mimicking feature to induce cancer cell-specific cytotoxicity while exhibiting negligible cytotoxicity toward normal cells, which might be attributed to the efficient production of highly reactive •OH originating from the overexpression H2O2 in the TME caused by surface catalysis. In particular, the cancer cell-specific cytotoxicity of the nanospheres was greatly enhanced both in vitro and in vivo upon irradiation with a near-infrared (NIR) laser (808 nm). The above-mentioned features of the hollow structured AgBiS2 will make it a promising candidate for tumor therapy.
Collapse
Affiliation(s)
- Benjin Chen
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Chenyang Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Wanni Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Zhaoyou Chu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Xiaoyan He
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
- Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Wei Zhou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Tao Liu
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
- Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Hua Wang
- The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P.R. China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
- Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
39
|
Li K, Liu CJ, Zhang XZ. Multifunctional peptides for tumor therapy. Adv Drug Deliv Rev 2020; 160:36-51. [PMID: 33080257 DOI: 10.1016/j.addr.2020.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
Controlled nano-systems for drug delivery are designed to deliver therapeutical drugs to desirable sites on demand. Due to the diverse physiological functions of peptides, it is reasonable to introduce peptides into anti-tumor nano-system. The integration of peptides into nanomaterials has complementary advantages, which not only avoids the rapid degradation of peptides in vivo, but also improves the intelligence and functionality of the nano-system. We summarized the functional peptides with targeting and stimulus-responsive properties, and the present review outlined the most relevant and recent developed peptide-based multifunctional nanomaterials for tumor therapy.
Collapse
|
40
|
Jiang B, Zhou L, Lu J, Wang Y, Liu C, You L, Guo J. Stroma-Targeting Therapy in Pancreatic Cancer: One Coin With Two Sides? Front Oncol 2020; 10:576399. [PMID: 33178608 PMCID: PMC7593693 DOI: 10.3389/fonc.2020.576399] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignancy with one of the worst prognoses worldwide and has an overall 5-year survival rate of only 9%. Although chemotherapy is the recommended treatment for patients with advanced PDAC, its efficacy is not satisfactory. The dense dysplastic stroma of PDAC is a major obstacle to the delivery of chemotherapy drugs and plays an important role in the progression of PDAC. Therefore, stroma-targeting therapy is considered a potential treatment strategy to improve the efficacy of chemotherapy and patient survival. While several preclinical studies have shown encouraging results, the anti-tumor potential of the PDAC stroma has also been revealed, and the extreme depletion might promote tumor progression and undermine patient survival. Therefore, achieving a balance between stromal abundance and depletion might be the further of stroma-targeting therapy. This review summarized the current progress of stroma-targeting therapy in PDAC and discussed the double-edged sword of its therapeutic effects.
Collapse
Affiliation(s)
- Bolun Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yizhi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengxi Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
41
|
Belhadj Z, He B, Fu J, Zhang H, Wang X, Dai W, Zhang Q. Regulating Interactions Between Targeted Nanocarriers and Mononuclear Phagocyte System via an Esomeprazole-Based Preconditioning Strategy. Int J Nanomedicine 2020; 15:6385-6399. [PMID: 32922007 PMCID: PMC7458613 DOI: 10.2147/ijn.s258054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose The mononuclear phagocyte system (MPS) presents a formidable obstacle that hampers the delivery of various nanopreparations to tumors. Therefore, there is an urgent need to improve the off-MPS targeting ability of nanomedicines. In the present study, we present a novel preconditioning strategy to substantially increase the circulation times and tumor targeting of nanoparticles by regulating nanocarrier-MPS interactions. Methods In vitro, the effect of different vacuolar H+-ATPase inhibitors on macrophage uptake of targeted or nontargeted lipid vesicles was evaluated. Specifically, the clinically approved proton-pump inhibitor esomeprazole (ESO) was selected as a preconditioning agent. Then, we further investigated the blocking effect of ESO on the macrophage endocytosis of nanocarriers. In vivo, ESO was first intravenously administered into A549-tumor-bearing nude mice, and 24 h later, the c(RGDm7)-modified vesicles co-loaded with doxorubicin and gefitinib were intravenously injected. Results In vitro, ESO was found to reduce the interactions between macrophages and c(RGDm7)-modified vesicles by interfering with the latter’s lysosomal trafficking. Studies conducted in vivo confirmed that ESO pretreatment greatly decreased the liver and spleen distribution of the targeted vesicles, enhanced their tumor accumulation, and improved the therapeutic outcome of the drug-loaded nanomedicines. Conclusion Our findings indicate that ESO can regulate the nanoparticle-MPS interaction, which provides a feasible option for enhancing the off-MPS targeting of nanomedicines.
Collapse
Affiliation(s)
- Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Jijun Fu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| |
Collapse
|
42
|
Fathi M, Abdolahinia ED, Barar J, Omidi Y. Smart stimuli-responsive biopolymeric nanomedicines for targeted therapy of solid tumors. Nanomedicine (Lond) 2020; 15:2171-2200. [DOI: 10.2217/nnm-2020-0146] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Solid tumors form a permissive microenvironment with irregular features, including high pressured tumor interstitial fluid with acidic pH, co-adaptation of cancer cells with other cells like the immune system cells, abnormal metabolism and anomalous overexpression of various pieces of molecular machinery. The functional expressions of several oncomarkers in different solid tumors have led to the development of targeted drug-delivery systems (DDSs). As a new class of DDSs, stimuli-responsive nanomedicines (SRNMs) have been developed using advanced nanobiomaterials such as biopolymers that show excellent biocompatibility with low inherent immunogenicity. In this review, we aim to overview different types of SRNMs, present deep insights into the stimuli-responsive biopolymers and discuss the most up-to-date progress in the design and development of SRNMs used as advanced DDSs for targeted therapy of cancer.
Collapse
Affiliation(s)
- Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
43
|
Abdou P, Wang Z, Chen Q, Chan A, Zhou DR, Gunadhi V, Gu Z. Advances in engineering local drug delivery systems for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1632. [PMID: 32255276 PMCID: PMC7725287 DOI: 10.1002/wnan.1632] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aims to leverage the immune system to suppress the growth of tumors and to inhibit metastasis. The recent promising clinical outcomes associated with cancer immunotherapy have prompted research and development efforts towards enhancing the efficacy of immune checkpoint blockade, cancer vaccines, cytokine therapy, and adoptive T cell therapy. Advancements in biomaterials, nanomedicine, and micro-/nano-technology have facilitated the development of enhanced local delivery systems for cancer immunotherapy, which can enhance treatment efficacy while minimizing toxicity. Furthermore, locally administered cancer therapies that combine immunotherapy with chemotherapy, radiotherapy, or phototherapy have the potential to achieve synergistic antitumor effects. Herein, the latest studies on local delivery systems for cancer immunotherapy are surveyed, with an emphasis on the therapeutic benefits associated with the design of biomaterials and nanomedicines. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peter Abdou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Road, Suzhou, 215123, Jiangsu, PR China
| | - Amanda Chan
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Daojia R. Zhou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Vivienne Gunadhi
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
44
|
Li S, Zhang W, Xue H, Xing R, Yan X. Tumor microenvironment-oriented adaptive nanodrugs based on peptide self-assembly. Chem Sci 2020; 11:8644-8656. [PMID: 34123123 PMCID: PMC8163399 DOI: 10.1039/d0sc02937h] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The aberrant metabolism of tumor cells creates an inimitable microenvironment featuring acidic pH, high glutathione (GSH) levels, and overexpression of certain enzymes, which benefits the overwhelming progress of a tumor. Peptide self-assembly, emerging as a biofriendly and versatile fabrication strategy, harnesses multiple noncovalent interactions to obtain a variety of nanostructures tailored on demand. Orchestrating the reversible nature of noncovalent interactions and abnormal physiological parameters in the tumor microenvironment enables peptide-based nanodrugs to be targetable or switchable, thereby improving the drugs' bioavailability and optimizing the treatment outcome. This review will focus on peptide-modulated self-assembly of photosensitizers, chemotherapeutic drugs, immunoactive agents for tumor microenvironment-oriented adaptive phototherapy, chemotherapy, immunotherapy and combinatorial therapy. We will emphasize the building block design, the intermolecular interaction principle, adaptive structural transformation in the tumor microenvironment and corresponding therapeutic efficacy, and aim to elucidate the critical role of peptide-modulated, tumor microenvironment-oriented adaptive assemblies in improving the therapeutic index. Challenges and opportunities will be covered as well to advance the development and clinical application of tumor therapies based on peptide self-assembly materials and techniques.
Collapse
Affiliation(s)
- Shukun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China http://www.yan-assembly.org/.,School of Chemical Engineering, University of Chinese Academy of Sciences Beijing 100049 China
| | - Wenjia Zhang
- Department of Radiology, Peking Union Medical College Hospital Beijing 100730 China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital Beijing 100730 China
| | - Ruirui Xing
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China http://www.yan-assembly.org/
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China http://www.yan-assembly.org/.,School of Chemical Engineering, University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
45
|
Zhang W, Yu L, Ji T, Wang C. Tumor Microenvironment-Responsive Peptide-Based Supramolecular Drug Delivery System. Front Chem 2020; 8:549. [PMID: 32775317 PMCID: PMC7388741 DOI: 10.3389/fchem.2020.00549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/28/2020] [Indexed: 12/22/2022] Open
Abstract
Physical and biochemical differences between tumor tissues and normal tissues provide promising triggering factors that can be utilized to engineer stimuli-responsive drug delivery platforms for cancer treatment. Rationally designed peptide-based supramolecular architectures can perform structural conversion by responding to the tumor microenvironment and achieve the controlled release of antitumor drugs. This mini review summarizes recent approaches for designing internal trigger-responsive drug delivery platforms using peptide-based materials. Peptide assemblies that exhibit a stimuli-responsive structural conversion upon acidic pH, high temperature, high oxidative potential, and the overexpressed proteins in tumor tissues are emphatically introduced. We also discuss the challenges of current peptide-based supramolecular delivery platforms against cancer.
Collapse
Affiliation(s)
- Wenbo Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biophysics and Structural Biology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Lanlan Yu
- State Key Laboratory of Medical Molecular Biology, Department of Biophysics and Structural Biology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Tianjiao Ji
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Chenxuan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biophysics and Structural Biology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
46
|
Affiliation(s)
- Chunhui Wang
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| | - Jingxian Yang
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| | - Chunyan Dong
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| | - Shuo Shi
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| |
Collapse
|
47
|
Abstract
Peptides are one of the most important functional motifs for constructing smart drug delivery systems (DDSs). Functional peptides can be conjugated with drugs or carriers via covalent bonds, or assembled into DDSs via supramolecular forces, which enables the DDSs to acquire desired functions such as targeting and/or environmental responsiveness. In this mini review, we first introduce the different types of functional peptides that are commonly used for constructing DDSs, and we highlight representative strategies for designing smart DDSs by using functional peptides in the past few years. We also state the challenges of peptide-based DDSs and come up with prospects.
Collapse
Affiliation(s)
- Zheng Lian
- People's Public Security University of China, Beijing 100038, China
| | | |
Collapse
|
48
|
Su M, Xiao S, Shu M, Lu Y, Zeng Q, Xie J, Jiang Z, Liu J. Enzymatic multifunctional biodegradable polymers for pH- and ROS-responsive anticancer drug delivery. Colloids Surf B Biointerfaces 2020; 193:111067. [PMID: 32388121 DOI: 10.1016/j.colsurfb.2020.111067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 04/11/2020] [Accepted: 04/17/2020] [Indexed: 01/09/2023]
Abstract
A new family of multifunctional biodegradable block copolymers, PEG-poly(ω-pentadecalactone-co-N-methyldiethyleneamine sebacate-co-2,2'-thiodiethylene sebacate) (PEG-PMT), were synthesized via lipase-catalyzed copolymerization procedures. Amphiphilic PEG-PMT copolymers can be readily transformed into stable micellar nanoparticles through self-assembling processes in aqueous medium. The particle sizes increase dramatically after exposure of the particles to the acidic pH and high reactive oxygen species (ROS) conditions in tumor microenvironments, due to protonation of thioether groups and oxidation of amino groups in the PMT micelle cores, respectively. For example, docetaxel (DTX)-loaded PEG-PM-19 % TS micelles were triggered synergistically by acidic pH and ROS stimuli to release over 85 % of the anti-cancer drug. In particular, DTX/PEG-PMT-19 % TS and DTX/PEG-PMT-48 % TS micelles performed better than commercial Duopafei formulation in prohibiting growth of CT-26 tumors xenografed in vivo (70 % of tumor-inhibiting efficiency). Biosafety analysis revealed that DTX-loaded PEG-PMT nanoparticles possessed minimal toxicity towards normal organs, such as liver and kidney. These experimental data demonstrated that the pH- and ROS-responsive PEG-PMT micelles are promising vectors for both delivery of anti-tumor drugs and their controlled release at tumor intracellular sites.
Collapse
Affiliation(s)
- Meifei Su
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Shuting Xiao
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Man Shu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Yao Lu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Qiang Zeng
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Jianhua Xie
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Zhaozhong Jiang
- Department of Biomedical Engineering, Integrated Science and Technology Center, Yale University, 600 West Campus Drive, West Haven, CT, 06516, United States.
| | - Jie Liu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
49
|
Ding B, Zheng P, Ma P, Lin J. Manganese Oxide Nanomaterials: Synthesis, Properties, and Theranostic Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1905823. [PMID: 31990409 DOI: 10.1002/adma.201905823] [Citation(s) in RCA: 253] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/23/2019] [Indexed: 05/20/2023]
Abstract
Despite the comprehensive applications in bioimaging, biosensing, drug/gene delivery, and tumor therapy of manganese oxide nanomaterials (MONs including MnO2 , MnO, Mn2 O3 , Mn3 O4 , and MnOx ) and their derivatives, a review article focusing on MON-based nanoplatforms has not been reported yet. Herein, the representative progresses of MONs on synthesis, heterogene, properties, surface modification, toxicity, imaging, biodetection, and therapy are mainly introduced. First, five kinds of primary synthetic methods of MONs are presented, including thermal decomposition method, exfoliation strategy, permanganates reduction method, adsorption-oxidation method, and hydro/solvothermal. Second, the preparations of hollow MONs and MON-based composite materials are summarized specially. Then, the chemical properties, surface modification, and toxicity of MONs are discussed. Next, the diagnostic applications including imaging and sensing are outlined. Finally, some representative rational designs of MONs in photodynamic therapy, photothermal therapy, chemodynamic therapy, sonodynamic therapy, radiotherapy, magnetic hyperthermia, chemotherapy, gene therapy, starvation therapy, ferroptosis, immunotherapy, and various combination therapy are highlighted.
Collapse
Affiliation(s)
- Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Pan Zheng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
50
|
Pi J, Shen L, Yang E, Shen H, Huang D, Wang R, Hu C, Jin H, Cai H, Cai J, Zeng G, Chen ZW. Macrophage‐Targeted Isoniazid–Selenium Nanoparticles Promote Antimicrobial Immunity and Synergize Bactericidal Destruction of Tuberculosis Bacilli. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201912122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jiang Pi
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
- Department of Microbiology Zhongshan School of Medicine Key Laboratory for Tropical Diseases Control of the Ministry of Education Sun Yat-sen University Guangzhou Guangdong 510080 China
| | - Ling Shen
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Enzhuo Yang
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 China
| | - Dan Huang
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Richard Wang
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Chunmiao Hu
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Hua Jin
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Huaihong Cai
- Department of Chemistry Jinan University Guangzhou Guangdong 510632 China
| | - Jiye Cai
- Department of Chemistry Jinan University Guangzhou Guangdong 510632 China
| | - Gucheng Zeng
- Department of Microbiology Zhongshan School of Medicine Key Laboratory for Tropical Diseases Control of the Ministry of Education Sun Yat-sen University Guangzhou Guangdong 510080 China
| | - Zheng W. Chen
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| |
Collapse
|