1
|
Xuanyuan X, Liu W, Jiang M, Zhang X, Wen B, Zheng R, Yao N, Zhang T, Feng Y, Qiao C, Zhang H, Luo D, Feng S, Li M, Gao J, Lu Z. Harnessing prazosin for tumors: Liposome hybrid nanovesicles activate tumor immunotherapy via autophagy inhibition. Biomaterials 2025; 319:123184. [PMID: 39985978 DOI: 10.1016/j.biomaterials.2025.123184] [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: 12/13/2024] [Revised: 02/01/2025] [Accepted: 02/09/2025] [Indexed: 02/24/2025]
Abstract
Prazosin (Prz), an antagonist of alpha-1 adrenergic receptors, is conventionally employed in the treatment of hypertension. Our study pioneers the exploration of Prz in oncology, examining its impact on cellular autophagy and its potential to trigger antitumor immune responses. We have developed a novel Prz-loaded liposome hybrid nanovesicle (Prz@LINV) system, integrating tumor-derived nanovesicles (TNV) with liposomes (LIP) to facilitate targeted Prz delivery to tumor sites. This formulation enhances Prz bioavailability and markedly inhibits tumor cell autophagy, leading to immunogenic cell death (ICD) and the activation of antitumor immune responses. Furthermore, Prz@LINV modulates dendritic cells (DCs), augmenting their antigen cross-presentation capacity and thereby potentiating antitumor immunity. These effects were validated in a colorectal cancer mouse model, demonstrating the good biocompatibility of Prz@LINV and its significant inhibition in tumor growth, along with the enhancement of antitumor immune responses. Our findings elucidate a novel mechanism by which Prz inhibits autophagy and enhances the antitumor immune response, providing a foundation for the development of innovative immunotherapeutic strategies. The efficacy of Prz@LINV suggests that Prz may emerge as a pivotal component in future immunotherapeutic regimens, offering patients more potent therapeutic options.
Collapse
Affiliation(s)
- Xinyang Xuanyuan
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Wenshang Liu
- Department of Dermatology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Min Jiang
- The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Xin Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - BeiBei Wen
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Rui Zheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Ning Yao
- Department of General Surgery, Joint Support Force 903rd Hospital, Hangzhou, 310013, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Yu Feng
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Chaofeng Qiao
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Huiqi Zhang
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Dong Luo
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Sa Feng
- School of Pharmacy, Henan University, Kaifeng, 475004, China.
| | - Meng Li
- Department of Dermatology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China.
| | - Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
2
|
Song Z, Wang Q, Xiong H, Xiao J, Zhou Z, Li T, Sun Q, Qiu L, Tan Y, Liu X, Jiang H, Han S, Wang X. Bionic gene delivery system activates tumor autophagy and immunosuppressive niche to sensitize anti-PD-1 treatment against STK11-mutated lung adenocarcinoma. J Nanobiotechnology 2025; 23:312. [PMID: 40275340 PMCID: PMC12020135 DOI: 10.1186/s12951-025-03404-z] [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: 11/21/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Clinical data have shown that Serine/Threonine Kinase 11 (STK11) mutation may be associated with an immunosuppressive tumor microenvironment (ITEM) and poor prognosis and failure of anti-PD-1 (αPD1) treatment in non-small cell lung cancer (NSCLC). To explore the potential of restoring STK11 protein in immunotherapy, a bionic gene delivery system was prepared by coating the STK11-encoded DNA-cationic polymer complex core with the tumor cell membrane, termed STK11@PPCM. STK11@PPCM could specifically bind with NSCLC cells and achieve precise delivery of STK11-encoded DNA. The released DNA effectively restored the STK11 protein expression, consequently reactivating autophagy and immunogenic cell death (ICD) in cancer cells. The liberated damage-associated molecular patterns (DAMPs) and autophagosome induced dendritic cells (DCs) maturation, which in turn enhanced CD8 + T cell infiltration, M1 macrophage polarization, and proinflammatory factor expression, thereby reversing the ITEM. Moreover, STK11@PPCM was also found to improve the sensitivity of cancer cells to αPD1 by increasing the expression of PD-L1, which was confirmed in STK11-mutated NSCLC cell xenografted mouse models, constructed by CRISPR-Cas9 knockout technology. This work demonstrated for the first time that restoration of functional STK11 can effectively reverse ITME and boost αPD1 efficacy in NSCLC, offering a new therapeutic approach for STK11-mutated lung adenocarcinoma in clinic.
Collapse
Affiliation(s)
- Zhongquan Song
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qikai Wang
- Health Management Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Hongjie Xiong
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Jiang Xiao
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Zihan Zhou
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Tianxiang Li
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qian Sun
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Liping Qiu
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yue Tan
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| | - Shuhua Han
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China.
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| |
Collapse
|
3
|
Batool A, Kopp I, Kubeil M, Bachmann M, Andrews PC, Stephan H. Targeted bismuth-based materials for cancer. Dalton Trans 2025; 54:5614-5639. [PMID: 40040450 DOI: 10.1039/d5dt00163c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
The use of bismuth and its compounds in biomedicine has developed rapidly in recent years. Due to their unique properties, there are great opportunities for the development of new non-invasive strategies for the early diagnosis and effective treatment of cancers. This perspective highlights key fabrication methods to generate well-defined and clinically relevant bismuth materials of varying characteristics. On the one hand, this opens up a wide range of possibilities for unimodal and multimodal imaging. On the other hand, effective treatment strategies, which are increasingly based on combinatorial therapies, are given a great deal of attention. One of the biggest challenges remains the selective tumour targeting, whether active or passive. Here we present an overview on new developments of bismuth based materials moving forward from a simple enrichment at the tumour site via uptake by the mononuclear phagocytic system (MPS) to a more active tumour specific targeting via covalent modification with tumour-seeking molecules based on either small or antibody-derived molecules.
Collapse
Affiliation(s)
- Amna Batool
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Ina Kopp
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Manja Kubeil
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Philip C Andrews
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
| | - Holger Stephan
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| |
Collapse
|
4
|
Aalhate M, Mahajan S, Dhuri A, Singh PK. Biohybrid nano-platforms manifesting effective cancer therapy: Fabrication, characterization, challenges and clinical perspective. Adv Colloid Interface Sci 2025; 335:103331. [PMID: 39522420 DOI: 10.1016/j.cis.2024.103331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Nanotechnology-based delivery systems have brought a paradigm shift in the management of cancer. However, the main obstacles to nanocarrier-based delivery are their limited circulation duration, excessive immune clearance, inefficiency in interacting effectively in a biological context and overcoming biological barriers. This demands effective engineering of nanocarriers to achieve maximum efficacy. Nanocarriers can be maneuvered with biological components to acquire biological identity for further regulating their biodistribution and cell-to-cell cross-talk. Thus, the integration of synthetic and biological components to deliver therapeutic cargo is called a biohybrid delivery system. These delivery systems possess the advantage of synthetic nanocarriers, such as high drug loading, engineerable surface, reproducibility, adequate communication and immune evasion ability of biological constituents. The biohybrid delivery vectors offer an excellent opportunity to harness the synergistic properties of the best entities of the two worlds for improved therapeutic outputs. The major spotlights of this review are different biological components, synthetic counterparts of biohybrid nanocarriers, recent advances in hybridization techniques, and the design of biohybrid delivery systems for cancer therapy. Moreover, this review provides an overview of biohybrid systems with therapeutic and diagnostic applications. In a nutshell, this article summarizes the advantages and limitations of various biohybrid nano-platforms, their clinical potential and future directions for successful translation in cancer management.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Anish Dhuri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India.
| |
Collapse
|
5
|
Zhao H, Du F, Xiang X, Tang Y, Feng Z, Wang Z, Rong X, Qiu L. Progress in application of nanomedicines for enhancing cancer sono-immunotherapy. ULTRASONICS SONOCHEMISTRY 2024; 111:107105. [PMID: 39427436 PMCID: PMC11533716 DOI: 10.1016/j.ultsonch.2024.107105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/22/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Cancer immunotherapy has significant potential as a cancer treatment since it boosts the immune system and prevents immune escape to get rid of or fight cancers. However, its clinical applicability is still limited because of the low response rate and immune-related side effects. Recently ultrasound has been shown to alter the tumor immune microenvironment, enhance the effectiveness of other antitumor therapies, and cause tumors to become more sensitive to immunotherapy, thus providing new insights into cancer treatment. Nanomedicines are also anticipated to have a positive impact on improving the immunological effects and enhancing ultrasound effect for cancer therapy. Therefore, designing effective nanomedicines enhanced ultrasound effect for augmenting sono-immunotherapy has been a pivot on anticancer therapy. In this review, the immunological impacts of various ultrasound therapeutic modalities, ultrasound parameters, and their underlying mechanisms are discussed. Moreover, we highlight the recent progress of nanomedicines synergistically enhancing sono-immunotherapy. Finally, we put forward opportunities and challenges on sono-immunotherapy.
Collapse
Affiliation(s)
- Hongxin Zhao
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fangxue Du
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xi Xiang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Tang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyan Feng
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyao Wang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiao Rong
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Li Qiu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Wu K, He M, Mao B, Xing Y, Wei S, Jiang D, Wang S, Alkuhali AA, Guo J, Gan Z, Li M, Li X, Chen H. Enhanced delivery of CRISPR/Cas9 system based on biomimetic nanoparticles for hepatitis B virus therapy. J Control Release 2024; 374:293-311. [PMID: 39151831 DOI: 10.1016/j.jconrel.2024.08.019] [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: 11/19/2023] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
The persistent presence of covalently closed circular DNA (cccDNA) in hepatocyte nuclei poses a significant obstacle to achieving a comprehensive cure for hepatitis B virus (HBV). Current applications of CRISPR/Cas9 for targeting and eliminating cccDNA have been confined to in vitro studies due to challenges in stable cccDNA expression in animal models and the limited non-immunogenicity of delivery systems. This study addresses these limitations by introducing a novel non-viral gene delivery system utilizing Gemini Surfactant (GS). The developed system creates stable and targeted CRISPR/Cas9 nanodrugs with a negatively charged surface through modification with red blood cell membranes (RBCM) or hepatocyte membranes (HCM), resulting in GS-pDNA@Cas9-CMs complexes. These GS-pDNA complexes demonstrated complete formation at a 4:1 w/w ratio. The in vitro transfection efficiency of GS-pDNA-HCM reached 54.61%, showing homotypic targeting and excellent safety. Additionally, the study identified the most effective single-guide RNA (sgRNA) from six sequences delivered by GS-pDNA@Cas9-HCM. Using GS-pDNA@Cas9-HCM, a significant reduction of 96.47% in in vitro HBV cccDNA and a 52.34% reduction in in vivo HBV cccDNA were observed, along with a notable decrease in other HBV-related markers. The investigation of GS complex uptake by AML-12 cells under varied time and temperature conditions revealed clathrin-mediated endocytosis (CME) for GS-pDNA and caveolin-mediated endocytosis (CVME) for GS-pDNA-HCM and GS-pDNA-RBCM. In summary, this research presents biomimetic gene-editing nanovectors based on GS (GS-pDNA@Cas9-CMs) and explores their precise and targeted clearance of cccDNA using CRISPR/Cas9, demonstrating good biocompatibility both in vitro and in vivo. This innovative approach provides a promising therapeutic strategy for advancing the cure of HBV.
Collapse
Affiliation(s)
- Kexin Wu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Miao He
- Laboratory Animal Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Binli Mao
- Western (Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing National Biomedicine Industry Base, Chongqing 401329, PR China; Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yangchen Xing
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Shiqi Wei
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Dongjun Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Shunyao Wang
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Asma A Alkuhali
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Xi'an Jiaotong University, Shanxi 710049, PR China
| | - Jinjun Guo
- Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Zongjie Gan
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan University, Sichuan 610041, PR China
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China; Western (Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing National Biomedicine Industry Base, Chongqing 401329, PR China.
| | - Huali Chen
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
7
|
Paengjun NK, Polshettiwar V, Ogawa M. Designed Nanoarchitectures of a BiOBr/BiOI Nanosheet Heterojunction Anchored on Dendritic Fibrous Nanosilica as Visible-Light Responsive Photocatalysts. Inorg Chem 2024; 63:11870-11883. [PMID: 38865140 DOI: 10.1021/acs.inorgchem.4c01756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Heterojunctions, particularly those involving BiOBr/BiOI, have attracted significant attention in the field of photocatalysis due to their remarkable properties. In this study, a unique architecture of BiOBr/BiOI was designed to facilitate the rapid transfer of electrons and holes, effectively mitigating the recombination of electron-hole pairs. Accordingly, the BiOBr/BiOI nanosheet heterojunction was anchored on dendritic fibrous nanosilica (DFNS) by the immobilization of Bi2O3 nanodots in DFNS and the subsequent reaction with HBr and then HI vapors at room temperature. The 4 nm-Bi2O3 nanodots acted as a sacrificial template to form BiOX nanosheets by reaction with HX vapors (X = Br, I). The BiOBr/BiOI nanosheet heterojunction with the lateral size remained in the range of 90 to 110 nm and a thickness of 15 nm formed on DFNS, where the BiOBr:BiOI ratio in the product was controlled by the exposure time to HX vapors. The reaction sequence (HBr → HI vapors) was a key for the formation of BiOBr/BiOI nanosheet heterojunction with controlled composition. When the reaction of Bi2O3 nanodots with HI vapor was performed in the reverse sequence (HI→ HBr), the substitution of I- with Br- occurred to form BiOBr sheets on DFNS. The BiOBr/BiOI nanosheet heterojunction anchored on DFNS was used as a visible-light-driven photocatalyst for the decomposition of benzene in water under solar light, and its activity was superior to that of single BiOX nanosheets on DFNS.
Collapse
Affiliation(s)
- Navarut Kan Paengjun
- School of Energy Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), 555 Moo 1, Pa Yup Nai, Wang Chan, Rayong 21210, Thailand
| | - Vivek Polshettiwar
- Division of Chemical Sciences, Tata Institute of Fundamental Research (TIFR), Homi Bhabha Road, Mumbai 400005, India
| | - Makoto Ogawa
- School of Energy Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), 555 Moo 1, Pa Yup Nai, Wang Chan, Rayong 21210, Thailand
| |
Collapse
|
8
|
Guo Q, Wang S, Xu R, Tang Y, Xia X. Cancer cell membrane-coated nanoparticles: a promising anti-tumor bionic platform. RSC Adv 2024; 14:10608-10637. [PMID: 38567339 PMCID: PMC10985588 DOI: 10.1039/d4ra01026d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Nanoparticle (NP) drug delivery systems have shown promise in tumor therapy. However, limitations such as susceptibility to immune clearance and poor targeting in a complex intercellular environment still exist. Recently, cancer cell membrane-encapsulated nanoparticles (CCM-NPs) constructed using biomimetic nanotechnology have been developed to overcome these problems. Proteins on the membrane surface of cancer cells can provide a wide range of activities for CCM-NPs, including immune escape and homologous cell recognition properties. Meanwhile, the surface of the cancer cell membrane exhibits obvious antigen enrichment, so that CCM-NPs can transmit tumor-specific antigen, activate a downstream immune response, and produce an effective anti-tumor effect. In this review, we first provided an overview of the functions of cancer cell membranes and summarized the preparation techniques and characterization methods of CCM-NPs. Then, we focused on the application of CCM-NPs in tumor therapy. In addition, we summarized the functional modifications of cancer cell membranes and compiled the patent applications related to CCM-NPs in recent years. Finally, we proposed the future challenges and directions of this technology in order to provide guidance for researchers in this field.
Collapse
Affiliation(s)
- Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Rubing Xu
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Yingnan Tang
- School of Pharmacy, Hunan Vocational College of Science and Technology Changsha Hunan 410208 China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| |
Collapse
|
9
|
Lu Y, Fan L, Wang J, Hu M, Wei B, Shi P, Li J, Feng J, Zheng Y. Cancer Cell Membrane-Based Materials for Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306540. [PMID: 37814370 DOI: 10.1002/smll.202306540] [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: 08/01/2023] [Revised: 09/18/2023] [Indexed: 10/11/2023]
Abstract
The nanodelivery system provides a novel direction for disease diagnosis and treatment; however, its delivery effectiveness is restricted by the short biological half-life and inadequate tumor targeting. The immune evasion properties and homologous targeting capabilities of natural cell membranes, particularly those of cancer cell membranes (CCM), have gained significant interest. The integration of CCM and nanoparticles has resulted in the emergence of CCM-based nanoplatforms (CCM-NPs), which have gained significant attention due to their unique properties. CCM-NPs not only prolong the blood circulation time of core nanoparticles, but also direct them for homologous tumor targeting. Herein, the history and development of CCM-NPs as well as how these platforms have been used for biomedical applications are discussed. The application of CCM-NPs for cancer therapy will be described in detail. Translational efforts are currently under way and further research to address key areas of need will ultimately be required to facilitate the successful clinical adoption of CCM-NPs.
Collapse
Affiliation(s)
- Yongping Lu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
- Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Linming Fan
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jun Wang
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Mingxiang Hu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Baogang Wei
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Ping Shi
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Jinyan Feng
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Yu Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
10
|
Xia Z, Mu W, Yuan S, Fu S, Liu Y, Zhang N. Cell Membrane Biomimetic Nano-Delivery Systems for Cancer Therapy. Pharmaceutics 2023; 15:2770. [PMID: 38140108 PMCID: PMC10748133 DOI: 10.3390/pharmaceutics15122770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Nano-delivery systems have demonstrated great promise in the therapy of cancer. However, the therapeutic efficacy of conventional nanomedicines is hindered by the clearance of the blood circulation system and the physiological barriers surrounding the tumor. Inspired by the unique capabilities of cells within the body, such as immune evasion, prolonged circulation, and tumor-targeting, there has been a growing interest in developing cell membrane biomimetic nanomedicine delivery systems. Cell membrane modification on nanoparticle surfaces can prolong circulation time, activate tumor-targeting, and ultimately improve the efficacy of cancer treatment. It shows excellent development potential. This review will focus on the advancements in various cell membrane nano-drug delivery systems for cancer therapy and the obstacles encountered during clinical implementation. It is hoped that such discussions will inspire the development of cell membrane biomimetic nanomedical systems.
Collapse
Affiliation(s)
- Zhenxing Xia
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Weiwei Mu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shijun Yuan
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shunli Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Yongjun Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Na Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| |
Collapse
|
11
|
Jarak I, Isabel Santos A, Helena Pinto A, Domingues C, Silva I, Melo R, Veiga F, Figueiras A. Colorectal cancer cell exosome and cytoplasmic membrane for homotypic delivery of therapeutic molecules. Int J Pharm 2023; 646:123456. [PMID: 37778515 DOI: 10.1016/j.ijpharm.2023.123456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
Colorectal cancer (CRC) is one of the most common causes of death in the world. The multi-drug resistance, especially in metastatic colorectal cancer, drives the development of new strategies that secure a positive outcome and reduce undesirable side effects. Nanotechnology has made an impact in addressing some pharmacokinetic and safety issues related to administration of free therapeutic agents. However, demands of managing complex biointerfacing require equally complex methods for introducing stimuli-responsive or targeting elements. In order to procure a more efficient solution to the overcoming of biological barriers, the physiological functions of cancer cell plasma and exosomal membranes provided the source of highly functionalized coatings. Biomimetic nanovehicles based on colorectal cancer (CRC) membranes imparted enhanced biological compatibility, immune escape and protection to diverse classes of therapeutic molecules. When loaded with therapeutic load or used as a coating for other therapeutic nanovehicles, they provide highly efficient and selective cell targeting and uptake. This review presents a detailed overview of the recent application of homotypic biomimetic nanovehicles in the management of CRC. We also address some of the current possibilities and challenges associated with the CRC membrane biomimetics.
Collapse
Affiliation(s)
- Ivana Jarak
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Porto, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Ana Isabel Santos
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Ana Helena Pinto
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Cátia Domingues
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal; Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
| | - Inês Silva
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Raquel Melo
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Francisco Veiga
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal
| | - Ana Figueiras
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal.
| |
Collapse
|
12
|
Aboeleneen SB, Scully MA, Kramarenko GC, Day ES. Combination cancer imaging and phototherapy mediated by membrane-wrapped nanoparticles. Int J Hyperthermia 2023; 40:2272066. [PMID: 37903544 PMCID: PMC10698846 DOI: 10.1080/02656736.2023.2272066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/11/2023] [Indexed: 11/01/2023] Open
Abstract
Cancer is a devastating health problem with inadequate treatment options. Many conventional treatments for solid-tumor cancers lack tumor specificity, which results in low efficacy and off-target damage to healthy tissues. Nanoparticle (NP)-mediated photothermal therapy (PTT) is a promising minimally invasive treatment for solid-tumor cancers that has entered clinical trials. Traditionally, NPs used for PTT are coated with passivating agents and/or targeting ligands, but alternative coatings are being explored to enhance tumor specific delivery. In particular, cell-derived membranes have emerged as promising coatings that improve the biointerfacing of photoactive NPs, which reduces their immune recognition, prolongs their systemic circulation and increases their tumor accumulation, allowing for more effective PTT. To maximize treatment success, membrane-wrapped nanoparticles (MWNPs) that enable dual tumor imaging and PTT are being explored. These multifunctional theranostic NPs can be used to enhance tumor detection and/or ensure a sufficient quantity of NPs that have arrived in the tumor prior to laser irradiation. This review summarizes the current state-of-the-art in engineering MWNPs for combination cancer imaging and PTT and discusses considerations for the path toward clinical translation.
Collapse
Affiliation(s)
- Sara B. Aboeleneen
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Mackenzie A. Scully
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | | | - Emily S. Day
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
- Department of Materials Science & Engineering, University of Delaware, Newark, DE, USA
- Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA
| |
Collapse
|
13
|
Liu WS, Wu LL, Chen CM, Zheng H, Gao J, Lu ZM, Li M. Lipid-hybrid cell-derived biomimetic functional materials: A state-of-the-art multifunctional weapon against tumors. Mater Today Bio 2023; 22:100751. [PMID: 37636983 PMCID: PMC10448342 DOI: 10.1016/j.mtbio.2023.100751] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Tumors are among the leading causes of death worldwide. Cell-derived biomimetic functional materials have shown great promise in the treatment of tumors. These materials are derived from cell membranes, extracellular vesicles and bacterial outer membrane vesicles and may evade immune recognition, improve drug targeting and activate antitumor immunity. However, their use is limited owing to their low drug-loading capacity and complex preparation methods. Liposomes are artificial bionic membranes that have high drug-loading capacity and can be prepared and modified easily. Although they can overcome the disadvantages of cell-derived biomimetic functional materials, they lack natural active targeting ability. Lipids can be hybridized with cell membranes, extracellular vesicles or bacterial outer membrane vesicles to form lipid-hybrid cell-derived biomimetic functional materials. These materials negate the disadvantages of both liposomes and cell-derived components and represent a promising delivery platform in the treatment of tumors. This review focuses on the design strategies, applications and mechanisms of action of lipid-hybrid cell-derived biomimetic functional materials and summarizes the prospects of their further development and the challenges associated with it.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| | - Li-Li Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Cui-Min Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Hao Zheng
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| |
Collapse
|
14
|
Huang JH, Huang CJ, Yu LN, Guan XL, Liang SW, Li JH, Liang L, Wei MY, Zhang LM. Bioinspired PROTAC-induced macrophage fate determination alleviates atherosclerosis. Acta Pharmacol Sin 2023; 44:1962-1976. [PMID: 37169852 PMCID: PMC10545710 DOI: 10.1038/s41401-023-01088-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/03/2023] [Indexed: 05/13/2023]
Abstract
Atherosclerosis is a major cause of death and disability in cardiovascular disease. Atherosclerosis associated with lipid accumulation and chronic inflammation leads to plaques formation in arterial walls and luminal stenosis in carotid arteries. Current approaches such as surgery or treatment with statins encounter big challenges in curing atherosclerosis plaque. The infiltration of proinflammatory M1 macrophages plays an essential role in the occurrence and development of atherosclerosis plaque. A recent study shows that TRIM24, an E3 ubiquitin ligase of a Trim family protein, acts as a valve to inhibit the polarization of anti-inflammatory M2 macrophages, and elimination of TRIM24 opens an avenue to achieve the M2 polarization. Proteolysis-targeting chimera (PROTAC) technology has emerged as a novel tool for the selective degradation of targeting proteins. But the low bioavailability and cell specificity of PROTAC reagents hinder their applications in treating atherosclerosis plaque. In this study we constructed a type of bioinspired PROTAC by coating the PROTAC degrader (dTRIM24)-loaded PLGA nanoparticles with M2 macrophage membrane (MELT) for atherosclerosis treatment. MELT was characterized by morphology, size, and stability. MELT displayed enhanced specificity to M1 macrophages as well as acidic-responsive release of dTRIM24. After intravenous administration, MELT showed significantly improved accumulation in atherosclerotic plaque of high fat and high cholesterol diet-fed atherosclerotic (ApoE-/-) mice through binding to M1 macrophages and inducing effective and precise TRIM24 degradation, thus resulting in the polarization of M2 macrophages, which led to great reduction of plaque formation. These results suggest that MELT can be considered a potential therapeutic agent for targeting atherosclerotic plaque and alleviating atherosclerosis progression, providing an effective strategy for targeted atherosclerosis therapy.
Collapse
Affiliation(s)
- Jiong-Hua Huang
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Chuang-Jia Huang
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Li-Na Yu
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Department of Preventive Dentistry, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510013, China
| | - Xiao-Ling Guan
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shang-Wen Liang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Jian-Hong Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Lu Liang
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Min-Yan Wei
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Ling-Min Zhang
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
15
|
Wen M, Zhao Y, Qiu P, Ren Q, Tao C, Chen Z, Yu N. Efficient sonodynamic ablation of deep-seated tumors via cancer-cell-membrane camouflaged biocompatible nanosonosensitizers. J Colloid Interface Sci 2023; 644:388-396. [PMID: 37120887 DOI: 10.1016/j.jcis.2023.04.088] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Ultrasound (US)-triggered therapies are promising in cancer treatments, and their effectiveness can be enhanced through the proper camouflage of sonosensitizers. Herein, we have constructed cancer cell membrane (CCM)-camouflaged sonosensitizers for homotypic tumor-targeted sonodynamic therapy (SDT). The camouflaged sonosensitizers have been prepared by encapsulating hemoporfin molecules in poly(lactic acid) polymers (H@PLA) and extruding with CCM from Colon Tumor 26 (CT26) cells, forming the H@PLA@CCM. Under excitation with US, the hemoporfin encapsulated in H@PLA@CCM can convert O2 into cytotoxic 1O2, which exerts an efficient sonodynamic effect. The H@PLA@CCM nanoparticles show enhanced cellular internalization to CT26 cells compared to H@PLA, and they also can be more efficiently engulfed by CT26 cells than by mouse breast cancer cells, due to the homologous targeting ability of CT26 CCM. After the intravenous injection, the blood circulation half-life of H@PLA@CCM is determined to be 3.23 h which is 4.3-time that of H@PLA. With high biosafety, homogeneous targeting ability, and sonodynamic effect, the combination of H@PLA@CCM and US irradiation has induced significant apoptosis and necrosis of tumor cells through the efficient SDT, achieving the strongest inhibition rate of tumors among other groups. This study provides insights into designing efficient and targeted cancer therapies using CCM-camouflaged sonosensitizers.
Collapse
Affiliation(s)
- Mei Wen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Yaoyu Zhao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Pu Qiu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Qian Ren
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Cheng Tao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Zhigang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Nuo Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
16
|
Wang M, Liang Y, Jiang X, Shen J. α-Fe 2O 3@Au-PEG-Ce6-Gd Nanoparticles as Acidic H 2O 2-Driven Oxygenators for Multimodal Imaging and Synergistic Tumor Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:5333-5341. [PMID: 37018043 DOI: 10.1021/acs.langmuir.2c03388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Nanoparticles with visual imaging capabilities and synergistic therapeutics have a bright future in antitumor applications. However, most of the current nanomaterials lack multiple imaging-guided therapeutic capabilities. In this study, a novel enhanced photothermal photodynamic antitumor nanoplatform with photothermal imaging, fluorescence (FL) imaging, and MRI-guided therapeutic capabilities was constructed by grafting gold, dihydroporphyrin Ce6, and Gd onto α-iron trioxide. This antitumor nanoplatform can convert NIR light into local hyperthermia at a temperature of up to 53 °C under NIR light irradiation, while Ce6 can generate singlet oxygen, which further synergizes the tumor-killing effect. At the same time, α-Fe2O3@Au-PEG-Ce6-Gd can also have significant photothermal imaging effect under light irradiation, which can guide to see the temperature change near the tumor tissue. It is worth noting that α-Fe2O3@Au-PEG-Ce6-Gd can have obvious MRI and FL imaging effects after tail vein injection in mice with blood circulation, realizing imaging-guided synergistic antitumor therapy. α-Fe2O3@Au-PEG-Ce6-Gd NPs provide a new solution for tumor imaging and treatment.
Collapse
Affiliation(s)
- Mingqian Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
| | - Ying Liang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
| | - Xuefeng Jiang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, 1 Wenyuan Road, Qixia District, Nanjing 210023, China
- Jiangsu Engineering Research Center of Interfacial Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Qixia District, Nanjing 210023, China
| |
Collapse
|
17
|
Current understanding of passive and active targeting nanomedicines to enhance tumor accumulation. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
18
|
Długosz O, Matyjasik W, Hodacka G, Szostak K, Matysik J, Krawczyk P, Piasek A, Pulit-Prociak J, Banach M. Inorganic Nanomaterials Used in Anti-Cancer Therapies:Further Developments. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13061130. [PMID: 36986024 PMCID: PMC10051539 DOI: 10.3390/nano13061130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 05/14/2023]
Abstract
In this article, we provide an overview of the progress of scientists working to improve the quality of life of cancer patients. Among the known methods, cancer treatment methods focusing on the synergistic action of nanoparticles and nanocomposites have been proposed and described. The application of composite systems will allow precise delivery of therapeutic agents to cancer cells without systemic toxicity. The nanosystems described could be used as a high-efficiency photothermal therapy system by exploiting the properties of the individual nanoparticle components, including their magnetic, photothermal, complex, and bioactive properties. By combining the advantages of the individual components, it is possible to obtain a product that would be effective in cancer treatment. The use of nanomaterials to produce both drug carriers and those active substances with a direct anti-cancer effect has been extensively discussed. In this section, attention is paid to metallic nanoparticles, metal oxides, magnetic nanoparticles, and others. The use of complex compounds in biomedicine is also described. A group of compounds showing significant potential in anti-cancer therapies are natural compounds, which have also been discussed.
Collapse
|
19
|
Lee Y, Kim M, Ha J, Lee M. Brain-targeted exosome-mimetic cell membrane nanovesicles with therapeutic oligonucleotides elicit anti-tumor effects in glioblastoma animal models. Bioeng Transl Med 2023; 8:e10426. [PMID: 36925699 PMCID: PMC10013800 DOI: 10.1002/btm2.10426] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/19/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022] Open
Abstract
The brain-targeted delivery of therapeutic oligonucleotides has been investigated as a new treatment modality for various brain diseases, such as brain tumors. However, delivery efficiency into the brain has been limited due to the blood-brain barrier. In this research, brain-targeted exosome-mimetic cell membrane nanovesicles (CMNVs) were designed to enhance the delivery of therapeutic oligonucleotides into the brain. First, CMNVs were produced by extrusion with isolated C6 cell membrane fragments. Then, CMNVs were decorated with cholesterol-linked T7 peptides as a targeting ligand by hydrophobic interaction, producing T7-CMNV. T7-CMNV was in aqueous solution maintained its nanoparticle size for over 21 days. The targeting and delivery effects of T7-CMNVs were evaluated in an orthotopic glioblastoma animal model. 2'-O-metyl and cholesterol-TEG modified anti-microRNA-21 oligonucleotides (AMO21c) were loaded into T7-CMNVs, and biodistribution experiments indicated that T7-CMNVs delivered AMO21c more efficiently into the brain than CMNVs, scrambled T7-CMNVs, lipofectamine, and naked AMO21c after systemic administration. In addition, AMO21c down-regulated miRNA-21 (miR-21) levels in glioblastoma tissue most efficiently in the T7-CMNVs group. This enhanced suppression of miR-21 resulted in the up-regulation of PDCD4 and PTEN. Eventually, brain tumor size was reduced in the T7-CMNVs group more efficiently than in the other control groups. With stability, low toxicity, and targeting efficiency, T7-CMNVs may be useful to the development of oligonucleotide therapy for brain tumors.
Collapse
Affiliation(s)
- Youngki Lee
- Department of BioengineeringCollege of Engineering, Hanyang UniversitySeoulKorea
| | - Minkyung Kim
- Department of BioengineeringCollege of Engineering, Hanyang UniversitySeoulKorea
| | - Junkyu Ha
- Department of BioengineeringCollege of Engineering, Hanyang UniversitySeoulKorea
| | - Minhyung Lee
- Department of BioengineeringCollege of Engineering, Hanyang UniversitySeoulKorea
| |
Collapse
|
20
|
Ouyang R, Zhang Q, Cao P, Yang Y, Zhao Y, Liu B, Miao Y, Zhou S. Efficient improvement in chemo/photothermal synergistic therapy against lung cancer using Bi@Au nano-acanthospheres. Colloids Surf B Biointerfaces 2023; 222:113116. [PMID: 36603409 DOI: 10.1016/j.colsurfb.2022.113116] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
Novel highly hydrophilic and biocompatible bismuth nanospheres with gold nanoparticles growing outside (Bi@Au nano-acanthospheres, Bi@Au NASs) were synthesized through a simple procedure, which demonstrated to be a promising photothermal agent owing to the ultrahigh photothermal conversion efficiency (η = 46.6 %). The as-prepared Bi@Au NASs showed excellent blood compatibility and fairly low cytotoxicity to human lung cancer A549 cells, as well as efficient photothermal ablation (PTA) therapy induced by a near-infrared laser. Under the 808 nm laser radiation, the tumour temperature could be elevated by ∼25 °C high enough to kill the cancer cells. Moreover, the anticancer drug doxorubicin hydrochloride (DOX) was successfully loaded in Bi@Au NASs with a loading content as high as 16.78 % and released under a pH sensitive release profile, a characteristic beneficial for intravenous delivery of DOX into cancer cells for chemotherapy. The presence of the Bi element enabled Bi@Au NASs to act as a favourable computed tomography (CT) contrast medium for CT imaging-guided tumour treatment. Compared with cancer treatment through either photothermal therapy or chemotherapy, the chemo-photothermal synergistic therapy using Bi@Au NASs as both a photothermal agent and a drug carrier has efficiently enhanced the in vitro and in vivo therapeutic effects in cancer treatment.
Collapse
Affiliation(s)
- Ruizhuo Ouyang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Qiupeng Zhang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Penghui Cao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yang Yang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yuefeng Zhao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yuqing Miao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Shuang Zhou
- Cancer Institute, Tongji University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
21
|
He YC, Hao ZN, Li Z, Gao DW. Nanomedicine-based multimodal therapies: Recent progress and perspectives in colon cancer. World J Gastroenterol 2023; 29:670-681. [PMID: 36742173 PMCID: PMC9896619 DOI: 10.3748/wjg.v29.i4.670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/26/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Colon cancer has attracted much attention due to its annually increasing incidence. Conventional chemotherapeutic drugs are unsatisfactory in clinical application because of their lack of targeting and severe toxic side effects. In the past decade, nanomedicines with multimodal therapeutic strategies have shown potential for colon cancer because of their enhanced permeability and retention, high accumulation at tumor sites, co-loading with different drugs, and comb-ination of various therapies. This review summarizes the advances in research on various nanomedicine-based therapeutic strategies including chemotherapy, radiotherapy, phototherapy (photothermal therapy and photodynamic therapy), chemodynamic therapy, gas therapy, and immunotherapy. Additionally, the therapeutic mechanisms, limitations, improvements, and future of the above therapies are discussed.
Collapse
Affiliation(s)
- Yu-Chu He
- State Key Laboratory of Metastable Materials Science and Technology, Nano-Biotechnology Key Laboratory of Hebei Province, Applying Chemistry Key Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066000, Hebei Province, China
| | - Zi-Ning Hao
- State Key Laboratory of Metastable Materials Science and Technology, Nano-Biotechnology Key Laboratory of Hebei Province, Applying Chemistry Key Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066000, Hebei Province, China
| | - Zhuo Li
- State Key Laboratory of Metastable Materials Science and Technology, Nano-Biotechnology Key Laboratory of Hebei Province, Applying Chemistry Key Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066000, Hebei Province, China
| | - Da-Wei Gao
- State Key Laboratory of Metastable Materials Science and Technology, Nano-Biotechnology Key Laboratory of Hebei Province, Applying Chemistry Key Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066000, Hebei Province, China
| |
Collapse
|
22
|
Wu L, Luo Y, Wang C, Wu S, Zheng Y, Li Z, Cui Z, Liang Y, Zhu S, Shen J, Liu X. Self-Driven Electron Transfer Biomimetic Enzymatic Catalysis of Bismuth-Doped PCN-222 MOF for Rapid Therapy of Bacteria-Infected Wounds. ACS NANO 2023; 17:1448-1463. [PMID: 36622022 DOI: 10.1021/acsnano.2c10203] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In this work, a biomimetic nanozyme catalyst with rapid and efficient self-bacteria-killing and wound-healing performances was synthesized. Through an in situ reduction reaction, a PCN-222 metal organic framework (MOF) was doped with bismuth nanoparticles (Bi NPs) to form Bi-PCN-222, an interfacial Schottky heterojunction biomimetic nanozyme catalyst, which can kill 99.9% of Staphylococcus aureus (S. aureus). The underlying mechanism was that Bi NP doping can endow Bi-PCN-222 MOF with self-driven charge transfer through the Schottky interface and the capability of oxidase-like and peroxidase-like activity, because a large number of free electrons can be captured by surrounding oxygen species to produce radical oxygen species (ROS). Furthermore, once bacteria contact Bi-PCN-222 in a physiological environment, its appropriate redox potential can trigger electron transfer through the electron transport pathway in bacterial membranes and then the interior of the bacteria, which disturbs the bacterial respiration process and subsequent metabolism. Additionally, Bi-PCN-222 can also accelerate tissue regeneration by upregulating fibroblast proliferation and angiogenesis genes (bFGF, VEGF, and HIF-1α), thereby promoting wound healing. This biomimetic enzyme-catalyzed strategy will bring enlightenment to the design of self-bacterial agents for efficient disinfection and tissue reconstruction simultaneously.
Collapse
Affiliation(s)
- Lihua Wu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan430062, People's Republic of China
| | - Yue Luo
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan430062, People's Republic of China
| | - Chaofeng Wang
- School of Health Science and Biomedical Engineering, Hebei University of Technology, Tianjin300401, People's Republic of China
| | - Shuilin Wu
- School of Materials Science and Engineering, Peking University, Beijing100871, People's Republic of China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing100871, People's Republic of China
| | - Zhaoyang Li
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin300072, People's Republic of China
| | - Zhenduo Cui
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin300072, People's Republic of China
| | - Yanqin Liang
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin300072, People's Republic of China
| | - Shengli Zhu
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin300072, People's Republic of China
| | - Jie Shen
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen516473, People's Republic of China
| | - Xiangmei Liu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan430062, People's Republic of China
- School of Health Science and Biomedical Engineering, Hebei University of Technology, Tianjin300401, People's Republic of China
| |
Collapse
|
23
|
Huang F, Zhang Q, Xiao J, Zhang X, Han X, Shi X, Hu J, Li L, Qian X. Cancer Cell Membrane-Coated Gambogic Acid Nanoparticles for Effective Anticancer Vaccination by Activating Dendritic Cells. Int J Nanomedicine 2023; 18:2261-2273. [PMID: 37159807 PMCID: PMC10163893 DOI: 10.2147/ijn.s408521] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/24/2023] [Indexed: 05/11/2023] Open
Abstract
Purpose Recent studies have shown that traditional Chinese medicine (TCM), such as gambogic acid (GA), is involved in the regulation of tumor immune microenvironment and can be combined with other anti-tumor treatment strategies. Here, we used GA as an adjuvant to construct a nano-vaccine to improve the anti-tumor immune response of colorectal cancer (CRC). Materials and Methods We used a previously reported two-step emulsification method to obtain poly (lactic-co-glycolic acid) /GA nanoparticles (PLGA/GA NPs), and then CT26 colon cancer cell membrane (CCM) was used to obtain CCM-PLGA/GA NPs. This novel nano-vaccine, CCM-PLGA/GA NPs, was co-synthesized with GA as an adjuvant and neoantigen provided by CT26 CCM. We further confirmed the stability, tumor targeting, and cytotoxicity of CCM-PLGA/GA NPs. The regulatory effect on the tumor immune microenvironment, the anti-tumor efficacy, and the combined anti-tumor efficacy with anti-PD-1 monoclonal Antibodies (mAbs) of this novel nano-vaccine was also detected in vivo. Results We successfully constructed the CCM-PLGA/GA NPs. In vitro and in vivo tests showed low biological toxicity, as well as the high tumor-targeting ability of the CCM-PLGA/GA NPs. Besides, we revealed a remarkable effect of CCM-PLGA/GA NPs to activate the maturation of dendritic cells (DCs) and the formation of a positive anti-tumor immune microenvironment. Conclusion This novel nano-vaccine constructed with GA as the adjuvant and CCM providing the tumor antigen can not only directly kill tumors by enhancing the ability of GA to target tumors, but also indirectly kill tumors by regulating tumor immune microenvironment, providing a new strategy for immunotherapy of CRC.
Collapse
Affiliation(s)
- Fengli Huang
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Qun Zhang
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing, People’s Republic of China
| | - Jie Xiao
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Xin Zhang
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Xingzhi Han
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Xiao Shi
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Jing Hu
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing, People’s Republic of China
| | - Li Li
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing, People’s Republic of China
| | - Xiaoping Qian
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing, People’s Republic of China
- Correspondence: Xiaoping Qian, Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People’s Republic of China, Tel +86-13951743162, Fax +86-25-68182342, Email
| |
Collapse
|
24
|
Rodrigues CF, Fernandes N, de Melo‐Diogo D, Correia IJ, Moreira AF. Cell-Derived Vesicles for Nanoparticles' Coating: Biomimetic Approaches for Enhanced Blood Circulation and Cancer Therapy. Adv Healthc Mater 2022; 11:e2201214. [PMID: 36121767 PMCID: PMC11481079 DOI: 10.1002/adhm.202201214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/11/2022] [Indexed: 01/28/2023]
Abstract
Cancer nanomedicines are designed to encapsulate different therapeutic agents, prevent their premature release, and deliver them specifically to cancer cells, due to their ability to preferentially accumulate in tumor tissue. However, after intravenous administration, nanoparticles immediately interact with biological components that facilitate their recognition by the immune system, being rapidly removed from circulation. Reports show that less than 1% of the administered nanoparticles effectively reach the tumor site. This suboptimal pharmacokinetic profile is pointed out as one of the main factors for the nanoparticles' suboptimal therapeutic effectiveness and poor translation to the clinic. Therefore, an extended blood circulation time may be crucial to increase the nanoparticles' chances of being accumulated in the tumor and promote a site-specific delivery of therapeutic agents. For that purpose, the understanding of the forces that govern the nanoparticles' interaction with biological components and the impact of the physicochemical properties on the in vivo fate will allow the development of novel and more effective nanomedicines. Therefore, in this review, the nano-bio interactions are summarized. Moreover, the application of cell-derived vesicles for extending the blood circulation time and tumor accumulation is reviewed, focusing on the advantages and shortcomings of each cell source.
Collapse
Affiliation(s)
- Carolina F. Rodrigues
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Natanael Fernandes
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Duarte de Melo‐Diogo
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Ilídio J. Correia
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - André F. Moreira
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
- CPIRN‐UDI/IPG – Center of Potential and Innovation in Natural Resources, Research Unit for Inland DevelopmentInstituto Politécnico da GuardaAvenida Dr. Francisco de Sá CarneiroGuarda6300‐559Portugal
| |
Collapse
|
25
|
Zhang X, Liu B, Yao S, Liu Z, Li J. Multifunctional Bi NSs@BSA Nanoplatform Guided by CT Imaging for Effective Photothermal Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:14355-14363. [PMID: 36350293 DOI: 10.1021/acs.langmuir.2c02474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Photothermal therapy (PTT) has attracted great attention as an anticancer treatment strategy. With the rapid development of nanomedicine, multifunctional inorganic nanophotothermal agents provide a new way to improve the effect of PTT. Herein, bovine serum albumin (BSA)-modified Bi nanosheets (Bi NSs) with good biocompatibility were synthesized by a facile redox and ball milling method and applied as a photothermal agent for the enhancement of PTT. Owing to the strong near-infrared absorption, Bi NSs exhibit high photothermal conversion efficiency (η = 36.17%) under 808 nm laser irradiation and can serve as a nanotherapeutic agent for cancer therapy. In addition, in vitro cell safety analysis also suggests that the toxicity of BSA-modified Bi NSs is negligible. Upon 808 nm irradiation, the uptake ability of tumor cells to Bi NSs@BSA has been improved. Moreover, Bi NSs@BSA also can be used as a good contrast agent for CT imaging and then to observe the distribution of materials in the tumor site. Finally, Bi NSs@BSA-mediated PTT results show a high ablation rate of A549 tumor cells both in vitro and in vivo. All results reveal that Bi NSs@BSA is a promising nanotherapeutic platform for PTT.
Collapse
Affiliation(s)
- Xiaolei Zhang
- School of Material Science and Engineering, University of Jinan, Jinan, China250022
| | - Bin Liu
- School of Material Science and Engineering, University of Jinan, Jinan, China250022
| | | | - Zongming Liu
- School of Material Science and Engineering, University of Jinan, Jinan, China250022
| | - Jinkai Li
- School of Material Science and Engineering, University of Jinan, Jinan, China250022
| |
Collapse
|
26
|
Aboeleneen SB, Scully MA, Harris JC, Sterin EH, Day ES. Membrane-wrapped nanoparticles for photothermal cancer therapy. NANO CONVERGENCE 2022; 9:37. [PMID: 35960404 PMCID: PMC9373884 DOI: 10.1186/s40580-022-00328-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/27/2022] [Indexed: 05/31/2023]
Abstract
Cancer is a global health problem that needs effective treatment strategies. Conventional treatments for solid-tumor cancers are unsatisfactory because they cause unintended harm to healthy tissues and are susceptible to cancer cell resistance. Nanoparticle-mediated photothermal therapy is a minimally invasive treatment for solid-tumor cancers that has immense promise as a standalone therapy or adjuvant to other treatments like chemotherapy, immunotherapy, or radiotherapy. To maximize the success of photothermal therapy, light-responsive nanoparticles can be camouflaged with cell membranes to endow them with unique biointerfacing capabilities that reduce opsonization, prolong systemic circulation, and improve tumor delivery through enhanced passive accumulation or homotypic targeting. This ensures a sufficient dose of photoresponsive nanoparticles arrives at tumor sites to enable their complete thermal ablation. This review summarizes the state-of-the-art in cell membrane camouflaged nanoparticles for photothermal cancer therapy and provides insights to the path forward for clinical translation.
Collapse
Affiliation(s)
| | | | - Jenna C Harris
- Materials Science and Engineering, University of Delaware, Newark, DE, USA
| | - Eric H Sterin
- Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Emily S Day
- Biomedical Engineering, University of Delaware, Newark, DE, USA.
- Materials Science and Engineering, University of Delaware, Newark, DE, USA.
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA.
| |
Collapse
|
27
|
Xu X, Li T, Jin K. Bioinspired and Biomimetic Nanomedicines for Targeted Cancer Therapy. Pharmaceutics 2022; 14:1109. [PMID: 35631695 PMCID: PMC9147382 DOI: 10.3390/pharmaceutics14051109] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Undesirable side effects and multidrug resistance are the major obstacles in conventional chemotherapy towards cancers. Nanomedicines provide alternative strategies for tumor-targeted therapy due to their inherent properties, such as nanoscale size and tunable surface features. However, the applications of nanomedicines are hampered in vivo due to intrinsic disadvantages, such as poor abilities to cross biological barriers and unexpected off-target effects. Fortunately, biomimetic nanomedicines are emerging as promising therapeutics to maximize anti-tumor efficacy with minimal adverse effects due to their good biocompatibility and high accumulation abilities. These bioengineered agents incorporate both the physicochemical properties of diverse functional materials and the advantages of biological materials to achieve desired purposes, such as prolonged circulation time, specific targeting of tumor cells, and immune modulation. Among biological materials, mammalian cells (such as red blood cells, macrophages, monocytes, and neutrophils) and pathogens (such as viruses, bacteria, and fungi) are the functional components most often used to confer synthetic nanoparticles with the complex functionalities necessary for effective nano-biointeractions. In this review, we focus on recent advances in the development of bioinspired and biomimetic nanomedicines (such as mammalian cell-based drug delivery systems and pathogen-based nanoparticles) for targeted cancer therapy. We also discuss the biological influences and limitations of synthetic materials on the therapeutic effects and targeted efficacies of various nanomedicines.
Collapse
Affiliation(s)
- Xiaoqiu Xu
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Li
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ke Jin
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Niculescu AG, Grumezescu AM. Novel Tumor-Targeting Nanoparticles for Cancer Treatment-A Review. Int J Mol Sci 2022; 23:5253. [PMID: 35563645 PMCID: PMC9101878 DOI: 10.3390/ijms23095253] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/28/2022] [Accepted: 05/05/2022] [Indexed: 12/04/2022] Open
Abstract
Being one of the leading causes of death and disability worldwide, cancer represents an ongoing interdisciplinary challenge for the scientific community. As currently used treatments may face limitations in terms of both efficiency and adverse effects, continuous research has been directed towards overcoming existing challenges and finding safer specific alternatives. In particular, increasing interest has been gathered around integrating nanotechnology in cancer management and subsequentially developing various tumor-targeting nanoparticles for cancer applications. In this respect, the present paper briefly describes the most used cancer treatments in clinical practice to set a reference framework for recent research findings, further focusing on the novel developments in the field. More specifically, this review elaborates on the top recent studies concerning various nanomaterials (i.e., carbon-based, metal-based, liposomes, cubosomes, lipid-based, polymer-based, micelles, virus-based, exosomes, and cell membrane-coated nanomaterials) that show promising potential in different cancer applications.
Collapse
Affiliation(s)
- Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania;
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania;
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov No. 3, 050044 Bucharest, Romania
| |
Collapse
|
29
|
Chen G, Li Y, Miao Y, Liu B. Recent developments on bismuth oxyhalide-based functional nanomaterials for biomedical applications. Biomater Sci 2022; 10:5809-5830. [DOI: 10.1039/d2bm01182d] [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
Multifunctional bismuth oxyhalide (BiOX, X = F, Cl, Br, and I) nanomaterials have great potential advantages in medical diagnostic and therapeutic applications. Pure BiOX nanomaterials have some limitations such as...
Collapse
|
30
|
Qiu N, Yang X, Zhang Y, Zhang J, Ji J, Zhang Y, Kong X, Xi Y, Liu D, Ye L, Zhai G. A molybdenum oxide-based degradable nanosheet for combined chemo-photothermal therapy to improve tumor immunosuppression and suppress distant tumors and lung metastases. J Nanobiotechnology 2021; 19:428. [PMID: 34923976 PMCID: PMC8684628 DOI: 10.1186/s12951-021-01162-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/23/2021] [Indexed: 01/12/2023] Open
Abstract
Molybdenum oxide (MoOx) nanosheets have drawn increasing attention for minimally invasive cancer treatments but still face great challenges, including complex modifications and the lack of efficient accumulation in tumor. In this work, a novel multifunctional degradable FA-BSA-PEG/MoOx nanosheet was fabricated (LA-PEG and FA-BSA dual modified MoOx): the synergistic effect of PEG and BSA endows the nanosheet with excellent stability and compatibility; the FA, a targeting ligand, facilitates the accumulation of nanosheets in the tumor. In addition, DTX, a model drug for breast cancer treatment, was loaded (76.49%, 1.5 times the carrier weight) in the nanosheets for in vitro and in vivo antitumor evaluation. The results revealed that the FA-BSA-PEG/MoOx@DTX nanosheets combined photothermal and chemotherapy could not only inhibit the primary tumor growth but also suppress the distant tumor growth (inhibition rate: 51.7%) and lung metastasis (inhibition rate: 93.6%), which is far more effective compared to the commercial Taxotere®. Exploration of the molecular mechanism showed that in vivo immune response induced an increase in positive immune responders, suppressed negative immune suppressors, and established an inflammatory tumor immune environment, which co-contributes towards effective suppression of tumor and lung metastasis. Our experiments demonstrated that this novel multifunctional nanosheet is a promising platform for combined chemo-photothermal therapy. ![]()
Collapse
Affiliation(s)
- Na Qiu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Yanan Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Jicheng Zhang
- Department of Chemistry and Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Yu Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Xinru Kong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Yanwei Xi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Dongzhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan, 250012, People's Republic of China.
| |
Collapse
|
31
|
Guo K, Xiao N, Liu Y, Wang Z, Tóth J, Gyenis J, Thakur VK, Oyane A, Shubhra QT. Engineering polymer nanoparticles using cell membrane coating technology and their application in cancer treatments: Opportunities and challenges. NANO MATERIALS SCIENCE 2021. [DOI: 10.1016/j.nanoms.2021.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
32
|
Han S, Lee Y, Lee M. Biomimetic cell membrane-coated DNA nanoparticles for gene delivery to glioblastoma. J Control Release 2021; 338:22-32. [PMID: 34391836 DOI: 10.1016/j.jconrel.2021.08.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/29/2023]
Abstract
Gene therapy has been introduced as an alternative to radiation and chemical therapy for glioblastoma. Biomimetic nanoparticles coated with cell membranes (CM) have advantages such as high biocompatibility and prolong half-life. To apply CM coated nanoparticles to gene delivery, the polyethylenimine (PEI25k)/plasmid DNA (pDNA) complexes were coated with CM from C6 rat glioblastoma cells. With the CM covering, the PEI25k/pDNA complexes formed stable nanoparticles with negative surface charge. The PEI25k/pDNA/CM nanoparticles had high colloidal stability and could be stored for approximately 20 days without aggregation. The transfection efficiency of the PEI25k/pDNA/CM nanoparticles was higher than that of the PEI25k/pDNA complex in serum-containing medium. This suggests that serum does not interfere with transfection efficiency of the nanoparticles. Moreover, the PEI25k/pDNA/CM nanoparticles had lower toxicity than the PEI25k/DNA complex in vitro and in vivo. The PEI25k/pDNA/CM nanoparticles prepared with CMs of different types of cells were transfected into cells. The results showed that the PEI25k/pDNA/CM nanoparticles with the C6 CM had the highest transfection efficiency to C6 cells, suggesting the homotypic targeting effect. The therapeutic effects of the nanoparticles were evaluated in intracranial C6 transplanted glioblastoma animal models. The PEI25k/pDNA/CM nanoparticles were prepared with herpes simplex virus thymidine kinase plasmid (pHSVtk) and injected into the tumor locally. The results showed that the PEI25k/pHSVtk/CM nanoparticles induced higher HSVtk expression compared with the PEI25k/pHSVtk complex. Furthermore, tumor size was reduced more efficiently by the PEI25k/pHSVtk/CM nanoparticles than by the PEI25k/pHSVtk complex. Overall results indicate that PEI25k/pDNA/CM nanoparticles are suitable for pDNA delivery to glioblastoma.
Collapse
Affiliation(s)
- Sangrok Han
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Youngki Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
33
|
Xia J, Li Z, Xie Z, Zheng M. Near-Infrared absorbing J-Aggregates of boron dipyrromethene for high efficient photothermal therapy. J Colloid Interface Sci 2021; 599:476-483. [PMID: 33962208 DOI: 10.1016/j.jcis.2021.04.086] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/13/2021] [Accepted: 04/18/2021] [Indexed: 10/21/2022]
Abstract
Constructing bioactive materials remains a big challenge through the aggregates of molecules. Herein, a boron dipyrromethene (BODIPY) derivative containing three nitro groups (BDP-(NO2)3) was synthesized, which displays the characteristic of J-aggregate with pronounced red-shifted absorption in nonpolar solvent and aqueous media. The bathochromic shift from 635 to 765 nm facilitates photothermal transition upon the irradiation of near-infrared (NIR) light. Interestingly, BDP-(NO2)3 nanoparticles (NPs) fabricated from BDP-(NO2)3 and poly(oxyethylene)-poly(oxypropylene) copolymer (F-127), still exhibit obvious J-aggregate, which possess the merits of hydrophilicity, NIR absorption, high photothermal conversion efficiency, excellent biosafety, and can behave as unique candidates for photothermal therapy. In vitro and in vivo experiments validate that BDP-(NO2)3 NPs can effectively suppress the proliferation of cancer cells and lead to tumor ablation. This assembly method would be a generic and efficient mode for reasonable design of functional nanomaterials, and could inspire more study on aggregates of organic molecules.
Collapse
Affiliation(s)
- Jinxiu Xia
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun, Jilin 130012, PR China
| | - Zhensheng Li
- Key Laboratory of Chemo/Biosensing and Detection, School of Chemistry and Chemical Engineering, Xuchang University, 88 Bayi Road, Xuchang 461000, PR China
| | - Zhigang Xie
- State Key Laboratory of Polymer Chemistry and Physics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences,5625 Renmin Street, Changchun, Jilin 130022, PR China.
| | - Min Zheng
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun, Jilin 130012, PR China.
| |
Collapse
|