1
|
Cai W, Sun T, Qiu C, Sheng H, Chen R, Xie C, Kou L, Yao Q. Stable triangle: nanomedicine-based synergistic application of phototherapy and immunotherapy for tumor treatment. J Nanobiotechnology 2024; 22:635. [PMID: 39420366 PMCID: PMC11488210 DOI: 10.1186/s12951-024-02925-3] [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: 10/03/2023] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
In recent decades, cancer has posed a challenging obstacle that humans strive to overcome. While phototherapy and immunotherapy are two emerging therapies compared to traditional methods, they each have their advantages and limitations. These limitations include easy metastasis and recurrence, low response rates, and strong side effects. To address these issues, researchers have increasingly focused on combining these two therapies by utilizing a nano-drug delivery system due to its superior targeting effect and high drug loading rate, yielding remarkable results. The combination therapy demonstrates enhanced response efficiency and effectiveness, leading to a preparation that is highly targeted, responsive, and with low recurrence rates. This paper reviews several main mechanisms of anti-tumor effects observed in combination therapy based on the nano-drug delivery system over the last five years. Furthermore, the challenges and future prospects of this combination therapy are also discussed.
Collapse
Affiliation(s)
- Wenjing Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Tuyue Sun
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Huixiang Sheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Congying Xie
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
Alimohammadvand S, Kaveh Zenjanab M, Mashinchian M, Shayegh J, Jahanban-Esfahlan R. Recent advances in biomimetic cell membrane-camouflaged nanoparticles for cancer therapy. Biomed Pharmacother 2024; 177:116951. [PMID: 38901207 DOI: 10.1016/j.biopha.2024.116951] [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: 04/14/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
The emerging strategy of biomimetic nanoparticles (NPs) via cellular membrane camouflage holds great promise in cancer therapy. This scholarly review explores the utilization of cellular membranes derived from diverse cellular entities; blood cells, immune cells, cancer cells, stem cells, and bacterial cells as examples of NP coatings. The camouflaging strategy endows NPs with nuanced tumor-targeting abilities such as self-recognition, homotypic targeting, and long-lasting circulation, thus also improving tumor therapy efficacy overall. The comprehensive examination encompasses a variety of cell membrane camouflaged NPs (CMCNPs), elucidating their underlying targeted therapy mechanisms and delineating diverse strategies for anti-cancer applications. Furthermore, the review systematically presents the synthesis of source materials and methodologies employed in order to construct and characterize these CMCNPs, with a specific emphasis on their use in cancer treatment.
Collapse
Affiliation(s)
- Sajjad Alimohammadvand
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Mashinchian
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Shayegh
- Department of Microbiology, Faculty of Veterinary and Agriculture, Islamic Azad University, Shabestar branch, Shabestar, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Wan Y, Li C, Fu LH, Feng T, Zhang Y, Li Y, Lin J, Huang P, Cui DX. Erythrocyte Membrane Camouflaged Nanotheranostics for Optical Molecular Imaging-Escorted Self-Oxygenation Photodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309026. [PMID: 38477698 DOI: 10.1002/smll.202309026] [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: 10/08/2023] [Revised: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Hypoxic tumor microenvironment (TME) hampers the application of oxygen (O2)-dependent photodynamic therapy (PDT) in solid tumors. To address this problem, a biomimetic nanotheranostics (named MMCC@EM) is developed for optical molecular imaging-escorted self-oxygenation PDT. MMCC@EM is synthesized by encapsulating chlorin e6 (Ce6) and catalase (CAT) in metal-organic framework (MOF) nanoparticles with erythrocyte membrane (EM) camouflage. Based on the biomimetic properties of EM, MMCC@EM efficiently accumulates in tumor tissues. The enriched MMCC@EM achieves TME-activatable drug release, thereby releasing CAT and Ce6, and this process can be monitored through fluorescence (FL) imaging. In addition, endogenous hydrogen peroxide (H2O2) will be decomposed by CAT to produce O2, which can be reflected by the measurement of intratumoral oxygen concentration using photoacoustic (PA) imaging. Such self-oxygenation nanotheranostics effectively mitigate tumor hypoxia and improve the generation of singlet oxygen (1O2). The 1O2 disrupts mitochondrial function and triggers caspase-3-mediated cellular apoptosis. Furthermore, MMCC@EM triggers immunogenic cell death (ICD) effect, leading to an increased infiltration of cytotoxic T lymphocytes (CTLs) into tumor tissues. As a result, MMCC@EM exhibits good therapeutic effects in 4T1-tumor bearing mice under the navigation of FL/PA duplex imaging.
Collapse
Affiliation(s)
- Yilin Wan
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Chunying Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Lian-Hua Fu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Ting Feng
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yifan Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Youyan Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Da-Xiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| |
Collapse
|
4
|
Qureshi SA, Rafiya K, Awasthi S, Jain A, Nadaf A, Hasan N, Kesharwani P, Ahmad FJ. Biomembrane camouflaged nanoparticles: A paradigm shifts in targeted drug delivery system. Colloids Surf B Biointerfaces 2024; 238:113893. [PMID: 38631282 DOI: 10.1016/j.colsurfb.2024.113893] [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: 12/10/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
Targeted drug delivery has emerged as a pivotal approach within precision medicine, aiming to optimize therapeutic efficacy while minimizing systemic side effects. Advanced biomimetic membrane-coated formulations have garnered significant interest from researchers as a promising strategy for targeted drug delivery, site-specific accumulation and heightened therapeutic outcomes. Biomimetic nanotechnology is able to retain the biological properties of the parent cell thus are able to exhibit superior targeting compared to conventional formulations. In this review, we have described different types of cell membrane camouflaged NPs. Mechanism of isolation and coating of the membranes along with the applications of each type of membrane and their mechanism to reach the desired site. Furthermore, a fusion of different membranes in order to prepare hybrid membrane biomimetic NPs which could possess better efficacy is discussed in detail in the review. Later, applications of the hybrid membrane-cloaked NPs along with current development were discussed in detail along with the challenges associated with it. Although membrane-cloaked NPs are currently in the preliminary stage of development, there is a huge potential to explore this biodegradable and biocompatible delivery system.
Collapse
Affiliation(s)
- Saba Asif Qureshi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Km Rafiya
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sakshi Awasthi
- Lloyd Institute of Management and Technology, Greater Noida, India
| | - Abhishek Jain
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Arif Nadaf
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
5
|
Mao M, Wu Y, He Q. Recent advances in targeted drug delivery for the treatment of glioblastoma. NANOSCALE 2024; 16:8689-8707. [PMID: 38606460 DOI: 10.1039/d4nr01056f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is one of the highly malignant brain tumors characterized by significant morbidity and mortality. Despite the recent advancements in the treatment of GBM, major challenges persist in achieving controlled drug delivery to tumors. The management of GBM poses considerable difficulties primarily due to unresolved issues in the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB) and GBM microenvironment. These factors limit the uptake of anti-cancer drugs by the tumor, thus limiting the therapeutic options. Current breakthroughs in nanotechnology provide new prospects concerning unconventional drug delivery approaches for GBM treatment. Specifically, swimming nanorobots show great potential in active targeted delivery, owing to their autonomous propulsion and improved navigation capacities across biological barriers, which further facilitate the development of GBM-targeted strategies. This review presents an overview of technological progress in different drug administration methods for GBM. Additionally, the limitations in clinical translation and future research prospects in this field are also discussed. This review aims to provide a comprehensive guideline for researchers and offer perspectives on further development of new drug delivery therapies to combat GBM.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
6
|
Zhou M, Lin X, Wang L, Yang C, Yu Y, Zhang Q. Preparation and Application of Hemostatic Hydrogels. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309485. [PMID: 38102098 DOI: 10.1002/smll.202309485] [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: 10/19/2023] [Revised: 11/28/2023] [Indexed: 12/17/2023]
Abstract
Hemorrhage remains a critical challenge in various medical settings, necessitating the development of advanced hemostatic materials. Hemostatic hydrogels have emerged as promising solutions to address uncontrolled bleeding due to their unique properties, including biocompatibility, tunable physical characteristics, and exceptional hemostatic capabilities. In this review, a comprehensive overview of the preparation and biomedical applications of hemostatic hydrogels is provided. Particularly, hemostatic hydrogels with various materials and forms are introduced. Additionally, the applications of hemostatic hydrogels in trauma management, surgical procedures, wound care, etc. are summarized. Finally, the limitations and future prospects of hemostatic hydrogels are discussed and evaluated. This review aims to highlight the biomedical applications of hydrogels in hemorrhage management and offer insights into the development of clinically relevant hemostatic materials.
Collapse
Affiliation(s)
- Minyu Zhou
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiang Lin
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Li Wang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Chaoyu Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yunru Yu
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Qingfei Zhang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
7
|
Rodrigues CF, Correia IJ, Moreira AF. Red blood cell membrane-camouflaged gold-core silica shell nanorods for cancer drug delivery and photothermal therapy. Int J Pharm 2024; 655:124007. [PMID: 38493844 DOI: 10.1016/j.ijpharm.2024.124007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Gold core mesoporous silica shell (AuMSS) nanorods are multifunctional nanomedicines that can act simultaneously as photothermal, drug delivery, and bioimaging agents. Nevertheless, it is reported that once administrated, nanoparticles can be coated with blood proteins, forming a protein corona, that directly impacts on nanomedicines' circulation time, biodistribution, and therapeutic performance. Therefore, it become crucial to develop novel alternatives to improve nanoparticles' half-life in the bloodstream. In this work, Polyethylenimine (PEI) and Red blood cells (RBC)-derived membranes were combined for the first time to functionalize AuMSS nanorods and simultaneously load acridine orange (AO). The obtained results revealed that the RBC-derived membranes promoted the neutralization of the AuMSS' surface charge and consequently improved the colloidal stability and biocompatibility of the nanocarriers. Indeed, the in vitro data revealed that PEI/RBC-derived membranes' functionalization also improved the nanoparticles' cellular internalization and was capable of mitigating the hemolytic effects of AuMSS and AuMSS/PEI nanorods. In turn, the combinatorial chemo-photothermal therapy mediated by AuMSS/PEI/RBC_AO nanorods was able to completely eliminate HeLa cells, contrasting with the less efficient standalone therapies. Such data reinforce the potential of AuMSS nanomaterials to act simultaneously as photothermal and chemotherapeutic agents.
Collapse
Affiliation(s)
- Carolina F Rodrigues
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ilídio J Correia
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, 3030-790 Coimbra, Portugal; AEROG-LAETA, Aerospace Sciences Department, Universidade da Beira Interior, Covilhã, Portugal.
| | - André F Moreira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CPIRN-UDI/IPG - Centro de Potencial e Inovação em Recursos Naturais, Unidade de Investigação para o Desenvolvimento do Interior do Instituto Politécnico da Guarda, Avenida Dr. Francisco de Sá Carneiro, No. 50, 6300-559 Guarda, Portugal.
| |
Collapse
|
8
|
Zhang D, Chen Y, Hao M, Xia Y. Putting Hybrid Nanomaterials to Work for Biomedical Applications. Angew Chem Int Ed Engl 2024; 63:e202319567. [PMID: 38429227 PMCID: PMC11478030 DOI: 10.1002/anie.202319567] [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: 12/18/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/03/2024]
Abstract
Hybrid nanomaterials have found use in many biomedical applications. This article provides a comprehensive review of the principles, techniques, and recent advancements in the design and fabrication of hybrid nanomaterials for biomedicine. We begin with an introduction to the general concept of material hybridization, followed by a discussion of how this approach leads to materials with additional functionality and enhanced performance. We then highlight hybrid nanomaterials in the forms of nanostructures, nanocomposites, metal-organic frameworks, and biohybrids, including their fabrication methods. We also showcase the use of hybrid nanomaterials to advance biomedical engineering in the context of nanomedicine, regenerative medicine, diagnostics, theranostics, and biomanufacturing. Finally, we offer perspectives on challenges and opportunities.
Collapse
Affiliation(s)
- Dong Zhang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | - Yidan Chen
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332 (USA)
| | - Min Hao
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA); School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332 (USA)
| |
Collapse
|
9
|
Liu Z, Liu X, Zhang W, Gao R, Wei H, Yu CY. Current advances in modulating tumor hypoxia for enhanced therapeutic efficacy. Acta Biomater 2024; 176:1-27. [PMID: 38232912 DOI: 10.1016/j.actbio.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Hypoxia is a common feature of most solid tumors, which promotes the proliferation, invasion, metastasis, and therapeutic resistance of tumors. Researchers have been developing advanced strategies and nanoplatforms to modulate tumor hypoxia to enhance therapeutic effects. A timely review of this rapidly developing research topic is therefore highly desirable. For this purpose, this review first introduces the impact of hypoxia on tumor development and therapeutic resistance in detail. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are also systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We provide a detailed discussion of the rationale and research progress of these strategies. Through a review of current trends, it is hoped that this comprehensive overview can provide new prospects for clinical application in tumor treatment. STATEMENT OF SIGNIFICANCE: As a common feature of most solid tumors, hypoxia significantly promotes tumor progression. Advanced nanoplatforms have been developed to modulate tumor hypoxia to enhanced therapeutic effects. In this review, we first introduce the impact of hypoxia on tumor progression. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We discuss the rationale and research progress of the above strategies in detail, and finally introduce future challenges for treatment of hypoxic tumors. By reviewing the current trends, this comprehensive overview can provide new prospects for clinical translatable tumor therapy.
Collapse
Affiliation(s)
- Zihan Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xinping Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Wei Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ruijie Gao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
10
|
Li S, Meng X, Peng B, Huang J, Liu J, Xiao H, Ma L, Liu Y, Tang J. Cell membrane-based biomimetic technology for cancer phototherapy: Mechanisms, recent advances and perspectives. Acta Biomater 2024; 174:26-48. [PMID: 38008198 DOI: 10.1016/j.actbio.2023.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/04/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Despite significant advances in medical technology and antitumour treatments, the diagnosis and treatment of tumours have undergone remarkable transformations. Noninvasive phototherapy methods, such as photodynamic therapy (PDT) and photothermal therapy (PTT), have gained significant interest in antitumour medicine. However, traditional photosensitisers or photothermal agents face challenges like immune system recognition, rapid clearance from the bloodstream, limited tumour accumulation, and phototoxicity concerns. Researchers combine photosensitisers or photothermal agents with natural cell membranes to overcome these obstacles to create a nano biomimetic therapeutic platform. When used to coat nanoparticles, red blood cells, platelets, cancer cells, macrophages, lymphocytes, and bacterial outer membranes could provide prolonged circulation, tumour targeting, immune stimulation, or antigenicity. This article covers the principles of cellular membrane biomimetic nanotechnology and phototherapy, along with recent advancements in applying nano biomimetic technology to PDT, PTT, PCT, and combined diagnosis and treatment. Furthermore, the challenges and issues of using nano biomimetic nanoparticles in phototherapy are discussed. STATEMENT OF SIGNIFICANCE: Currently, there has been significant progress in the field of cell membrane biomimetic technology. Researchers are exploring its potential application in tumor diagnosis and treatment through phototherapy. Scholars have conducted extensive research on combining cell membrane technology and phototherapy in anticancer diagnosis and treatment. This review aims to highlight the mechanisms of phototherapy and the latest advancements in single phototherapy (PTT, PDT) and combination phototherapy (PCT, PRT, and PIT), as well as diagnostic approaches. The review provides an overview of various cell membrane technologies, including RBC membranes, platelet membranes, macrophage cell membranes, tumour cell membranes, bacterial membranes, hybrid membranes, and their potential for anticancer applications under phototherapy. Lastly, the review discusses the challenges and future directions in this field.
Collapse
Affiliation(s)
- Songtao Li
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xiangrui Meng
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Bo Peng
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Ju Huang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jingwen Liu
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Hang Xiao
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, PR China
| | - Li Ma
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, PR China
| | - Yiyao Liu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, PR China.
| | - Jianyuan Tang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
11
|
Peng C, Xu Y, Wu J, Wu D, Zhou L, Xia X. TME-Related Biomimetic Strategies Against Cancer. Int J Nanomedicine 2024; 19:109-135. [PMID: 38192633 PMCID: PMC10773252 DOI: 10.2147/ijn.s441135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in various stages of tumor generation, metastasis, and evasion of immune monitoring and treatment. TME targeted therapy is based on TME components, related pathways or active molecules as therapeutic targets. Therefore, TME targeted therapy based on environmental differences between TME and normal cells has been widely studied. Biomimetic nanocarriers with low clearance, low immunogenicity, and high targeting have enormous potential in tumor treatment. This review introduces the composition and characteristics of TME, including cancer‑associated fibroblasts (CAFs), extracellular matrix (ECM), tumor blood vessels, non-tumor cells, and the latest research progress of biomimetic nanoparticles (NPs) based on TME. It also discusses the opportunities and challenges of clinical transformation of biomimetic nanoparticles.
Collapse
Affiliation(s)
- Cheng Peng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Yilin Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Jing Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Donghai Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| |
Collapse
|
12
|
Lu J, Yu J, Xie W, Gao X, Guo Z, Jin Z, Li Y, Fahad A, Pambe NU, Che S, Wei Y, Zhao L. Physical Dissolution Combined with Photodynamic Depletion: A Two-Pronged Nanoapproach for Deoxygenation-Driven and Hypoxia-Activated Prodrug Therapy. ACS APPLIED BIO MATERIALS 2023; 6:3902-3911. [PMID: 37644623 DOI: 10.1021/acsabm.3c00566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Hypoxia may enhance the chemoresistance of cancer cells and can significantly compromise the effectiveness of chemotherapy. Many efforts have been made to relieve or reverse hypoxia by introducing more oxygen into the tumor microenvironment (TME). Acting in a diametrically opposite way, in the current study, a novel nanocarrier was designed to further exhaust the oxygen level of the hypoxic TME. By creating such an oxygen depleted TME, the hypoxia-selective cytotoxin can work effectively, and oxygen exhaustion triggered chemotherapy can be achieved. Herein, deoxygenation agent, FDA-approved perfluorocarbon (PFC) and photosensitizer indocyanine green (ICG) for oxygen depletion, along with the hypoxia-activating drug tirapazamine (TPZ), were coincorporated within the poly(lactic-co-glycolic acid) (PLGA) nanoemulsion (ICG/TPZ@PPs) for the treatment of hypoxic tumors. Following hypoxia amplifying through physical oxygen dissolution and photodynamic depletion in tumors, hypoxic chemotherapy could be effectively activated to improve multitreatment synergy. After achieving local tumor enrichment, PFC-mediated oxygen dissolution combined with further ICG-mediated photodynamic therapy (PDT) under near-infrared (NIR) laser irradiation could induce enhanced hypoxia, which would activate the antitumor activity of codelivered TPZ to synergize cytotoxicity. Remarkably, in vivo experimental results exhibited that deoxygenated ICG/TPZ@PPs-based photothermal therapy (PTT), PDT, and hypoxia activated chemotherapy have an excellent synergistic ablation of tumors without obvious side effects, and therefore, a broad prospect of application of this nanocarrier could be expected.
Collapse
Affiliation(s)
- Jingsong Lu
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jing Yu
- Research Center of Magnetic and Electronic Materials, College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wensheng Xie
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaohan Gao
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100084, China
| | - Zhenhu Guo
- Institute of Process Engineering Chinese Academy of Sciences, State Key Laboratories of Biochemical Engineering, Beijing 100190, China
| | - Zeping Jin
- Key Laboratory of Advanced Materials, Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Ying Li
- Key Laboratory of Advanced Materials, Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Abdul Fahad
- Key Laboratory of Advanced Materials, Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Neema Ufurahi Pambe
- Key Laboratory of Advanced Materials, Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Shenglei Che
- Research Center of Magnetic and Electronic Materials, College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yen Wei
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- Key Laboratory of Advanced Materials, Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
13
|
Abstract
Ischemia or hypoxia can lead to pathological changes in the metabolism and function of tissues and then lead to various diseases. Timely and effective blood resuscitation or improvement of hypoxia is very important for the treatment of diseases. However, there is a need to develop stable, nontoxic, and immunologically inert oxygen carriers due to limitations such as blood shortages, different blood types, and the risk of transmitting infections. With the development of various technologies, oxygen carriers based on hemoglobin and perfluorocarbon have been widely studied in recent years. This paper reviews the development and application of hemoglobin and perfluorocarbon oxygen carriers. The design of oxygen carriers was analyzed, and their application as blood substitutes or oxygen carriers in various hypoxic diseases was discussed. Finally, the characteristics and future research of ideal oxygen carriers were prospected to provide reference for follow-up research.
Collapse
Affiliation(s)
- Qingsong Ye
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Deyuan Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Kaiyuan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| |
Collapse
|
14
|
Zhang C, Hu X, Jin L, Lin L, Lin H, Yang Z, Huang W. Strategic Design of Conquering Hypoxia in Tumor for Advanced Photodynamic Therapy. Adv Healthc Mater 2023; 12:e2300530. [PMID: 37186515 DOI: 10.1002/adhm.202300530] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/08/2023] [Indexed: 05/17/2023]
Abstract
Photodynamic therapy (PDT), with its advantages of high targeting, minimally invasive, and low toxicity side effects, has been widely used in the clinical therapy of various tumors, especially superficial tumors. However, the tumor microenvironment (TME) presents hypoxia due to the low oxygen (O2 ) supply caused by abnormal vascularization in neoplastic tissues and high O2 consumption induced by the rapid proliferation of tumor cells. The efficacy of oxygen-consumping PDT can be hampered by a hypoxic TME. To address this problem, researchers have been developing advanced nanoplatforms and strategies to enhance the therapeutic effect of PDT in tumor treatment. This review summarizes recent advanced PDT therapeutic strategies to against the hypoxic TME, thus enhancing PDT efficacy, including increasing O2 content in TME through delivering O2 to the tumors and in situ generations of O2 ; decreasing the O2 consumption during PDT by design of type I photosensitizers. Moreover, recent synergistically combined therapy of PDT and other therapeutic methods such as chemotherapy, photothermal therapy, immunotherapy, and gas therapy is accounted for by addressing the challenging problems of mono PDT in hypoxic environments, including tumor resistance, proliferation, and metastasis. Finally, perspectives of the opportunities and challenges of PDT in future clinical research and translations are provided.
Collapse
Affiliation(s)
- Cheng Zhang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Xiaoming Hu
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
- Jiangxi Key Laboratory of Nanobiomaterials, School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, P. R. China
| | - Long Jin
- Department of Pathology, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, P. R. China
| | - Lisheng Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Hongxin Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Zhen Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
| | - Wei Huang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
- Frontiers Science Center for Flexible Electronics (FSCFE), MIIT Key Laboratory of Flexible Electronics (KLoFE) Northwestern Polytechnical University Xi'an, Xi'an, 710072, P. R. China
| |
Collapse
|
15
|
Chen M, Leng Y, He C, Li X, Zhao L, Qu Y, Wu Y. Red blood cells: a potential delivery system. J Nanobiotechnology 2023; 21:288. [PMID: 37608283 PMCID: PMC10464085 DOI: 10.1186/s12951-023-02060-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023] Open
Abstract
Red blood cells (RBCs) are the most abundant cells in the body, possessing unique biological and physical properties. RBCs have demonstrated outstanding potential as delivery vehicles due to their low immunogenicity, long-circulating cycle, and immune characteristics, exhibiting delivery abilities. There have been several developments in understanding the delivery system of RBCs and their derivatives, and they have been applied in various aspects of biomedicine. This article compared the various physiological and physical characteristics of RBCs, analyzed their potential advantages in delivery systems, and summarized their existing practices in biomedicine.
Collapse
Affiliation(s)
- Mengran Chen
- Department of Hematology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yamei Leng
- Department of Hematology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Chuan He
- Guang'an People's Hospital, Guang'an, 638001, Sichuan, People's Republic of China
| | - Xuefeng Li
- Department of Hematology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lei Zhao
- Department of Hematology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Ying Qu
- Department of Hematology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Yu Wu
- Department of Hematology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
16
|
Chen X, Song P, Li W, Wang J, Gui T, Zhang W, Ge F, Zhu L. A pH-responsive polymer-coated CaO 2as oxygen-generating nanoparticle in situfor enhanced chemo-photodynamic synergistic therapy against tumors. NANOTECHNOLOGY 2023; 34:455101. [PMID: 37544302 DOI: 10.1088/1361-6528/aced9c] [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: 03/22/2023] [Accepted: 08/06/2023] [Indexed: 08/08/2023]
Abstract
Photodynamic therapy (PDT) has emerged as an efficient strategy for tumor treatment. However, Insufficient amounts of inherent hypoxia and intrinsic hydrogen peroxide (H2O2) in the tumor microenvironment severely constrained PDT, as oxygen is the critical substrate for photosensitivity reaction. Here, a pH-responsive H2O2and O2self-supplying hybrid nanoparticle was designed. Through, the calcium peroxide (CaO2) as carriers loading a chemotherapeutic drug a photosensitizer 5,10,15,20-tetrakis(4-aminophenyl) porphyrin (TAPP) and doxorubicin (DOX), was covered with polyacrylic acid (PAA) to build up a feature material DOX-TAPP-CaO2@OA@PAA (denoted as DTCOP) through the reverse microemulsion method. In the acidic tumor microenvironment conditions exposing the water-sensitive CaO2nanocore to generate hydrogen peroxide (H2O2) and O2, the self-supplied O2alleviates hypoxia to enhance the PDT, and releasing DOX and TAPP. Synthetic characterization shows that the succeeded synthesized Nanocarriers could effectively carry DOX and TAPP to the tumor site and release O2at the low pH of TME. And the experimental results demonstrated that this interpose exogenous oxygen strategy is efficient at inhibition of tumor growth bothin vitroandin vivo. The nanocomposite exhibits excellent biocompatibility and the ability to inhibit tumor growth and has significant potential for the treatment of hypoxic tumors.
Collapse
Affiliation(s)
- Xiaolu Chen
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| | - Ping Song
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| | - Wanzhen Li
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| | - Jun Wang
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| | - Ting Gui
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| | - Weiwei Zhang
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| | - Fei Ge
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| | - Longbao Zhu
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, People's Republic of China
| |
Collapse
|
17
|
Duan H, Wang L, Wang S, He Y. Surface modification potentials of cell membrane-based materials for targeted therapies: a chemotherapy-focused review. Nanomedicine (Lond) 2023; 18:1281-1303. [PMID: 37753724 DOI: 10.2217/nnm-2023-0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023] Open
Abstract
Nanotechnology has significant potential for cancer management at all stages, including prevention, diagnosis and treatment. In therapeutic applications, nanoparticles (NPs) have biological stability, targeting and body-clearance issues. To overcome these difficulties, biomimetic or cell membrane-coating methods using immune cell membranes are advised. Macrophage or neutrophil cell membrane-coated NPs may impede cancer progression in malignant tissue. Immune cell surface proteins and their capacity to maintain activity after membrane extraction and NP coating determine NP functioning. Immune cell surface proteins may offer NPs higher cellular interactions, blood circulation, antigen recognition for targeting, progressive drug release and reduced in vivo toxicity. This article examines nano-based systems with immune cell membranes, their surface modification potential, and their application in cancer treatment.
Collapse
Affiliation(s)
- Hongliang Duan
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun, 130000, China
| | - LiJuan Wang
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, 130000, China
| | - Sen Wang
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun, 130000, China
| | - Yangfang He
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, 130000, China
| |
Collapse
|
18
|
Huang S, Song C, Miao J, Zhu X, Jia Y, Liu Y, Fu D, Li B, Miao M, Duan S, Zhang Z, Hu Y. Red Blood Cell Membrane-coated Functionalized Au Nanocage as a Biomimetic Platform for Improved MicroRNA Delivery in Hepatocellular Carcinoma. Int J Pharm 2023:123044. [PMID: 37178790 DOI: 10.1016/j.ijpharm.2023.123044] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Dysregulation of microRNAs (miRNAs) expression is closely related to cancers and managing miRNA expression holds great promise for cancer therapy. However, their wide clinical application has been hampered by their poor stability, short half-life and non-specific biodistribution in vivo. Herein, a novel biomimetic platform designated as RHAuNCs-miRNA for improved miRNA delivery was prepared through wrapping miRNA-loaded functionalized Au nanocages (AuNCs) with red blood cell (RBC) membrane. RHAuNCs-miRNA not only successfully loaded miRNAs but also effectively protected them from enzymatic degradation. With good stability, RHAuNCs-miRNA had the characteristics of photothermal conversion and sustained release. Cellular uptake of RHAuNCs-miRNA by SMMC-7721 cells was in a time-dependent manner via clathrin- and caveolin-mediated endocytosis. The uptake of RHAuNCs-miRNAs was affected by cell types and improved by mild near infrared (NIR) laser irradiation. More importantly, RHAuNCs-miRNA exhibited a prolonged circulation time without the occurrence of accelerated blood clearance (ABC) in vivo, resulting in efficient delivery to tumor tissues. This study may demonstrate the great potential of RHAuNCs-miRNA for improved miRNAs delivery.
Collapse
Affiliation(s)
- Shengnan Huang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China; School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Chengzhi Song
- Center for Quantitative Biology, Peking University, Beijing 100871, PR China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Xiali Zhu
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Yongyan Jia
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Yafei Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Dongjun Fu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Benyi Li
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, United State
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Shaofeng Duan
- School of Pharmacy, Henan University, Henan International Joint Laboratory of Chinese Medicine Efficacy, Kaifeng, Henan Province 475004, PR China.
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.
| | - Yurong Hu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.
| |
Collapse
|
19
|
Wang X, Sun Y, Wangpraseurt D. Engineered photoresponsive biohybrids for tumor therapy. SMART MEDICINE 2023; 2:e20220041. [PMID: 39188274 PMCID: PMC11235730 DOI: 10.1002/smmd.20220041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/30/2023] [Indexed: 08/28/2024]
Abstract
Engineered biohybrids have recently emerged as innovative biomimetic platforms for cancer therapeutic applications. Particularly, engineered photoresponsive biohybrids hold tremendous potential against tumors due to their intriguing biomimetic properties, photoresponsive ability, and enhanced biotherapeutic functions. In this review, the design principles of engineered photoresponsive biohybrids and their latest progresses for tumor therapy are summarized. Representative engineered photoresponsive biohybrids are highlighted including biomolecules-associated, cell membrane-based, eukaryotic cell-based, bacteria-based, and algae-based photoresponsive biohybrids. Representative tumor therapeutic modalities of the engineered photoresponsive biohybrids are presented, including photothermal therapy, photodynamic therapy, synergistic therapy, and tumor therapy combined with tissue regeneration. Moreover, the challenges and future perspectives of these photoresponsive biohybrids for clinical practice are discussed.
Collapse
Affiliation(s)
- Xiaocheng Wang
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Yazhi Sun
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Daniel Wangpraseurt
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
- Scripps Institution of OceanographyUniversity of California San DiegoSan DiegoCaliforniaUSA
| |
Collapse
|
20
|
Kolarikova M, Hosikova B, Dilenko H, Barton-Tomankova K, Valkova L, Bajgar R, Malina L, Kolarova H. Photodynamic therapy: Innovative approaches for antibacterial and anticancer treatments. Med Res Rev 2023. [PMID: 36757198 DOI: 10.1002/med.21935] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 02/10/2023]
Abstract
Photodynamic therapy is an alternative treatment mainly for cancer but also for bacterial infections. This treatment dates back to 1900 when a German medical school graduate Oscar Raab found a photodynamic effect while doing research for his doctoral dissertation with Professor Hermann von Tappeiner. Unexpectedly, Raab revealed that the toxicity of acridine on paramecium depends on the intensity of light in his laboratory. Photodynamic therapy is therefore based on the administration of a photosensitizer with subsequent light irradiation within the absorption maxima of this substance followed by reactive oxygen species formation and finally cell death. Although this treatment is not a novelty, there is an endeavor for various modifications to the therapy. For example, selectivity and efficiency of the photosensitizer, as well as irradiation with various types of light sources are still being modified to improve final results of the photodynamic therapy. The main aim of this review is to summarize anticancer and antibacterial modifications, namely various compounds, approaches, and techniques, to enhance the effectiveness of photodynamic therapy.
Collapse
Affiliation(s)
- Marketa Kolarikova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Barbora Hosikova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hanna Dilenko
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Katerina Barton-Tomankova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Lucie Valkova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Robert Bajgar
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Lukas Malina
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hana Kolarova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
21
|
Biomimetic selenium nanosystems for infectious wound healing. ENGINEERED REGENERATION 2023. [DOI: 10.1016/j.engreg.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
22
|
Imran M, Gowd V, Saha P, Rashid S, Ahmad Chaudhary A, Mohamed MYA, Alawam AS, Khan R. Biologically inspired stealth - Camouflaged strategies in nanotechnology for the improved therapies in various diseases. Int J Pharm 2023; 631:122407. [PMID: 36402290 DOI: 10.1016/j.ijpharm.2022.122407] [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: 07/19/2022] [Revised: 10/27/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022]
Abstract
Nanotechnology has received increasing attention in the past decade and it's being used as a model for developing better treatments for a variety of diseases. Despite the fact that nanotechnology-based therapy has greatly improved treatment regimens, it still faces challenges such as inadequate circulation, insufficient accumulation at the target region, and undesired toxicity. In this regard, scientists are working on producing cell-membrane camouflaged nanoparticles as a biomimetic technique for modifying the surface of existing nanoparticles to produce significant therapeutic benefits following imparting myriad of desired functionalities. Membranes originating from erythrocytes, white blood cells, cancer cells, stem cells, platelets, or bacterial cells have been used to coat nanoparticle surfaces and create biologically inspired camouflaged nanoparticles. These biomemitic delivery systems have been proven to have potential applications in diagnosing and treating vaiorus diseases, including drug administration, immunisation, immunological regulation, and detoxification. From its inception to the present, we provide a complete description of this advanced technique for functionalizing nanoparticle surfaces. The method of making these membrane coated nanoparticles as well as their characterisation have been thoroughly discussed. Following that, we focused on the diversity of cell membranes derived from distinct cells in the evolution of nanoparticles, emphasising how these biologically inspired stealth - camouflaged techniques have led to increased therapeutic efficacy in a variety of disease states.
Collapse
Affiliation(s)
- Mohammad Imran
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India
| | - Vemana Gowd
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India
| | - Puspita Saha
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh 11623, Saudi Arabia
| | - Marwa Yousry A Mohamed
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh 11623, Saudi Arabia
| | - Abdullah S Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh 11623, Saudi Arabia
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India.
| |
Collapse
|
23
|
Tumor-Targeted Erythrocyte Membrane Nanoparticles for Theranostics of Triple-Negative Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15020350. [PMID: 36839675 PMCID: PMC9966336 DOI: 10.3390/pharmaceutics15020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/24/2023] Open
Abstract
Triple-negative breast cancer (TNBC) cells do not contain various receptors for targeted treatment, a reason behind the poor prognosis of this disease. In this study, biocompatible theranostic erythrocyte-derived nanoparticles (EDNs) were developed and evaluated for effective early diagnosis and treatment of TNBC. The anti-cancer drug, doxorubicin (DOX), was encapsulated into the EDNs and diagnostic quantum dots (QDs) were incorporated into the lipid bilayers of EDNs for tumor bio-imaging. Then, anti-epidermal growth factor receptor (EGFR) antibody molecules were conjugated to the surface of EDNs for TNBC targeting (iEDNs). According to the confocal microscopic analyses and biodistribution assay, iEDNs showed a higher accumulation in EGFR-positive MDA-MB-231 cancers in vitro as well as in vivo, compared to untargeted EDNs. iEDNs containing doxorubicin (iEDNs-DOX) showed a stronger inhibition of target tumor growth than untargeted ones. The resulting anti-EGFR iEDNs exhibited strong biocompatibility, prolonged blood circulation, and efficient targeting of TNBC in mice. Therefore, iEDNs may be used as potential TNBC-targeted co-delivery systems for therapeutics and diagnostics.
Collapse
|
24
|
Huis in ‘t Veld RV, Heuts J, Ma S, Cruz LJ, Ossendorp FA, Jager MJ. Current Challenges and Opportunities of Photodynamic Therapy against Cancer. Pharmaceutics 2023; 15:pharmaceutics15020330. [PMID: 36839652 PMCID: PMC9965442 DOI: 10.3390/pharmaceutics15020330] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Photodynamic therapy (PDT) is an established, minimally invasive treatment for specific types of cancer. During PDT, reactive oxygen species (ROS) are generated that ultimately induce cell death and disruption of the tumor area. Moreover, PDT can result in damage to the tumor vasculature and induce the release and/or exposure of damage-associated molecular patterns (DAMPs) that may initiate an antitumor immune response. However, there are currently several challenges of PDT that limit its widespread application for certain indications in the clinic. METHODS A literature study was conducted to comprehensively discuss these challenges and to identify opportunities for improvement. RESULTS The most notable challenges of PDT and opportunities to improve them have been identified and discussed. CONCLUSIONS The recent efforts to improve the current challenges of PDT are promising, most notably those that focus on enhancing immune responses initiated by the treatment. The application of these improvements has the potential to enhance the antitumor efficacy of PDT, thereby broadening its potential application in the clinic.
Collapse
Affiliation(s)
- Ruben V. Huis in ‘t Veld
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
- Department of Radiology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
- Correspondence:
| | - Jeroen Heuts
- Department of Immunology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Sen Ma
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Luis J. Cruz
- Department of Radiology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Ferry A. Ossendorp
- Department of Immunology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
25
|
Das CGA, Kumar VG, Dhas TS, Karthick V, Kumar CMV. Nanomaterials in anticancer applications and their mechanism of action - A review. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 47:102613. [PMID: 36252911 DOI: 10.1016/j.nano.2022.102613] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The current challenges in cancer treatment using conventional therapies have made the emergence of nanotechnology with more advancements. The exponential growth of nanoscience has drawn to develop nanomaterials (NMs) with therapeutic activities. NMs have enormous potential in cancer treatment by altering the drug toxicity profile. Nanoparticles (NPs) with enhanced surface characteristics can diffuse more easily inside tumor cells, thus delivering an optimal concentration of drugs at tumor site while reducing the toxicity. Cancer cells can be targeted with greater affinity by utilizing NMs with tumor specific constituents. Furthermore, it bypasses the bottlenecks of indiscriminate biodistribution of the antitumor agent and high administration dosage. Here, we focus on the recent advances on the use of various nanomaterials for cancer treatment, including targeting cancer cell surfaces, tumor microenvironment (TME), organelles, and their mechanism of action. The paradigm shift in cancer management is achieved through the implementation of anticancer drug delivery using nano routes.
Collapse
Affiliation(s)
- C G Anjali Das
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - V Ganesh Kumar
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - T Stalin Dhas
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - V Karthick
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - C M Vineeth Kumar
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| |
Collapse
|
26
|
Construction of a 980 nm laser-activated Pt(II) metallacycle nanosystem for efficient and safe photo-induced bacteria sterilization. Sci China Chem 2022. [DOI: 10.1007/s11426-022-1440-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Lai C, Luo B, Shen J, Shao J. Biomedical engineered nanomaterials to alleviate tumor hypoxia for enhanced photodynamic therapy. Pharmacol Res 2022; 186:106551. [PMID: 36370918 DOI: 10.1016/j.phrs.2022.106551] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Photodynamic therapy (PDT), as a highly selective, widely applicable, and non-invasive therapeutic modality that is an alternative to radiotherapy and chemotherapy, is extensively applied to cancer therapy. Practically, the efficiency of PDT is severely hindered by the existence of hypoxia in tumor tissue. Hypoxia is a typical hallmark of malignant solid tumors, which remains an essential impediment to many current treatments, thereby leading to poor clinical prognosis after therapy. To address this issue, studies have been focused on modulating tumor hypoxia to augment the therapeutic efficacy. Although nanomaterials to relieve tumor hypoxia for enhanced PDT have been demonstrated in many research articles, a systematical summary of the role of nanomaterials in alleviating tumor hypoxia is scarce. In this review, we introduced the mechanism of PDT, and the involved therapeutic modality of PDT for ablation of tumor cells was specifically summarized. Moreover, current advances in nanomaterials-mediated tumor oxygenation via oxygen-carrying or oxygen-generation tactics to alleviate tumor hypoxia are emphasized. Based on these considerable summaries and analyses, we proposed some feasible perspectives on nanoparticle-based tumor oxygenation to ameliorate the therapeutic outcomes, which may provide some detailed information in designing new oxygenation nanomaterials in this burgeneous field.
Collapse
Affiliation(s)
- Chunmei Lai
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Bangyue Luo
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jiangwen Shen
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jingwei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China; College of Materials and Chemical Engineering, Minjiang University, Fuzhou 350108, China.
| |
Collapse
|
28
|
Zheng BD, Xiao MT. Red blood cell membrane nanoparticles for tumor phototherapy. Colloids Surf B Biointerfaces 2022; 220:112895. [PMID: 36242941 DOI: 10.1016/j.colsurfb.2022.112895] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Non-invasive phototherapy includes photodynamic therapy (PDT) and photothermal therapy (PTT), and has garnered special interest in anti-tumor therapy. However, traditional photosensitizers or photothermal agents are faced with major challenges, including easy recognition by immune system, rapid clearance from blood circulation, and low accumulation in target sites. Combining the characteristics of natural cell membrane with the characteristics of photosensitizer or photothermal agent is an important technology to achieve the ideal therapeutic effect of cancer. Red cell membrane (RBMs) coated can disguise phototherapy agents as endogenous substances, thus constructing a new nano bionic therapeutic platform, resisting blood clearance and prolonging circulation time. At present, a variety of phototherapy agents based on Nano-RBMs have been isolated or designed. In this review, firstly, the basic principles of Nano-RBMs and phototherapy are expounded respectively. Then, the latest progress of Nano-RBMs for PDT, PTT and PDT/PTT applications in recent five years has been introduced respectively. Finally, the problems and challenges of Nano-RBMs in the field of phototherapy are put forward.
Collapse
Affiliation(s)
- Bing-De Zheng
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Mei-Tian Xiao
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| |
Collapse
|
29
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [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
|
30
|
Meng D, Yang S, Yang Y, Zhang L, Cui L. Synergistic chemotherapy and phototherapy based on red blood cell biomimetic nanomaterials. J Control Release 2022; 352:146-162. [PMID: 36252749 DOI: 10.1016/j.jconrel.2022.10.019] [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: 07/22/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Novel drug delivery systems (DDSs) have become the mainstay of research in targeted cancer therapy. By combining different therapeutic strategies, potential DDSs and synergistic treatment approaches are needed to effectively deal with evolving drug resistance and the adverse effects of cancer. Nowadays, developing and optimizing human cell-based DDSs has become a new research strategy. Among them, red blood cells can be used as DDSs as they significantly enhance the pharmacokinetics of the transported drug cargo. Phototherapy, as a novel adjuvant in cancer treatment, can be divided into photodynamic therapy and photothermal therapy. Phototherapy using erythropoietic nanocarriers to mimic the unique properties of erythrocytes and overcome the limitations of existing DDSs shows excellent prospects in clinical settings. This review provides an overview of the development of photosensitizers and research on bio-nano-delivery systems based on erythrocytes and erythrocyte membranes that are used in achieving synergistic outcomes during phototherapy/chemotherapy.
Collapse
Affiliation(s)
- Di Meng
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China
| | - Shuoye Yang
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China.
| | - Yanan Yang
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China
| | - Lu Zhang
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China
| | - Lan Cui
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China
| |
Collapse
|
31
|
Li P, Wang D, Hu J, Yang X. The role of imaging in targeted delivery of nanomedicine for cancer therapy. Adv Drug Deliv Rev 2022; 189:114447. [PMID: 35863515 DOI: 10.1016/j.addr.2022.114447] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/27/2022] [Accepted: 07/06/2022] [Indexed: 01/24/2023]
Abstract
Nanomedicines overcome the pharmacokinetic limitations of traditional drug formulations and have promising prospect in cancer treatment. However, nanomedicine delivery in vivo is still facing challenges from the complex physiological environment. For the purpose of effective tumor therapy, they should be designed to guarantee the five features principle, including long blood circulation, efficient tumor accumulation, deep matrix penetration, enhanced cell internalization and accurate drug release. To ensure the excellent performance of the designed nanomedicine, it would be better to monitor the drug delivery process as well as the therapeutic effects by real-time imaging. In this review, we summarize strategies in developing nanomedicines for efficiently meeting the five features of drug delivery, and the role of several imaging modalities (fluorescent imaging (FL), magnetic resonance imaging (MRI), computed tomography (CT), photoacoustic imaging (PAI), positron emission tomography (PET), and electron microscopy) in tracing drug delivery and therapeutic effect in vivo based on five features principle.
Collapse
Affiliation(s)
- Puze Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dongdong Wang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
32
|
Wang S, Wang Y, Jin K, Zhang B, Peng S, Nayak AK, Pang Z. Recent advances in erythrocyte membrane-camouflaged nanoparticles for the delivery of anti-cancer therapeutics. Expert Opin Drug Deliv 2022; 19:965-984. [PMID: 35917435 DOI: 10.1080/17425247.2022.2108786] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Red blood cell (or erythrocyte) membrane-camouflaged nanoparticles (RBC-NPs) not only have a superior circulation life and do not induce accelerated blood clearance, but also possess special functions, which offers great potential in cancer therapy. AREAS COVERED This review focuses on the recent advances of RBC-NPs for delivering various agents to treat cancers in light of their vital role in improving drug delivery. Meanwhile, the construction and in vivo behavior of RBC-NPs are discussed to provide an in-depth understanding of the basis of RBC-NPs for improved cancer drug delivery. EXPERT OPINION Although RBC-NPs are quite prospective in delivering anti-cancer therapeutics, they are still in their infancy stage and many challenges need to be overcome for successful translation into the clinic. The preparation and modification of RBC membranes, the optimization of coating methods, the scale-up production and the quality control of RBC-NPs, and the drug loading and release should be carefully considered in the clinical translation of RBC-NPs for cancer therapy.
Collapse
Affiliation(s)
- Siyu Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| | - Yiwei Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Kai Jin
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong 519000, China
| | - Amit Kumar Nayak
- Department of Pharmaceutics, Seemanta Institute of Pharmaceutical Sciences, Mayurbhanj-757086, Odisha, India
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| |
Collapse
|
33
|
Red Blood Cell Inspired Strategies for Drug Delivery: Emerging Concepts and New Advances. Pharm Res 2022; 39:2673-2698. [PMID: 35794397 DOI: 10.1007/s11095-022-03328-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/29/2022] [Indexed: 12/09/2022]
Abstract
In the past five decades, red blood cells (RBCs) have been extensively explored as drug delivery systems due to their distinguishing potential in modulating the pharmacokinetic, pharmacodynamics, and biological activity of carried payloads. The extensive interests in RBC-mediated drug delivery technologies are in part derived from RBCs' unique biological features such as long circulation time, wide access to many tissues in the body, and low immunogenicity. Owing to these outstanding properties, a large body of efforts have led to the development of various RBC-inspired strategies to enable precise drug delivery with enhanced therapeutic efficacy and reduced off-target toxicity. In this review, we discuss emerging concepts and new advances in such RBC-inspired strategies, including native RBCs, ghost RBCs, RBC-mimetic nanoparticles, and RBC-derived extracellular vesicles, for drug delivery.
Collapse
|
34
|
Shi J, Nie W, Zhao X, Yang X, Cheng H, Zhou T, Zhang Y, Zhang K, Liu J. An Intracellular Self-Assembly-Driven Uninterrupted ROS Generator Augments 5-Aminolevulinic-Acid-Based Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201049. [PMID: 35488781 DOI: 10.1002/adma.202201049] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/25/2022] [Indexed: 06/14/2023]
Abstract
Free radical therapy based on 5-aminolevulinic acid (ALA, a precursor of the photosensitizer protoporphyrin IX (PpIX)) has been approved by the US Food and Drug Administration for clinical tumor treatment. However, PpIX can be quickly converted into photoinactive heme, leading to unexpectedly paused production of free radicals and severely hindering its therapeutic benefits. Here, inspired by the natural biotransformation of ALA (ALA-PpIX-heme), an uninterrupted reactive oxygen species generator (URG) that converts useless heme to peroxidase mimics via intracellular self-assembly is developed. The URG is prepared by enwrapping ALA-loaded polyamide-amine dendrimers in red blood cell membrane vesicles with a further surface modification of G-quadruplex-structured AS1411. The URGs realize "1 O2 -•OH" uninterrupted generation through "recycling waste" in two steps: i) PpIX generates 1 O2 under laser irradiation; and ii) the photoinactive metabolite heme self-assembled with AS1411 to catalyze H2 O2 conversion into •OH. Interestingly, the specific generation of 1 O2 in mitochondria and •OH in nuclei further augments the free-radical-induced damage. It is demonstrated that URG can continuously produce free radicals for 6 h postirradiation, and shows 3.3-times more than that of the nonassembly group, achieving nearly 80% regression of tumors in vivo.
Collapse
Affiliation(s)
- Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Science and Technology, Department of Henan Province, Zhengzhou, 450001, China
| | - Weimin Nie
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiu Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinyuan Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hui Cheng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Tonghai Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yun Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Science and Technology, Department of Henan Province, Zhengzhou, 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Science and Technology, Department of Henan Province, Zhengzhou, 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Science and Technology, Department of Henan Province, Zhengzhou, 450001, China
| |
Collapse
|
35
|
Chen J, Chen D, Chen J, Shen T, Jin T, Zeng B, Li L, Yang C, Mu Z, Deng H, Cai X. An all-in-one CO gas therapy-based hydrogel dressing with sustained insulin release, anti-oxidative stress, antibacterial, and anti-inflammatory capabilities for infected diabetic wounds. Acta Biomater 2022; 146:49-65. [PMID: 35500813 DOI: 10.1016/j.actbio.2022.04.043] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 12/16/2022]
Abstract
To effectively treat diabetic wounds, the development of versatile medical dressings that can long-term regulate blood glucose and highly effective anti-oxidative stress, antibacterial and anti-inflammatory are critical. Here, an all-in-one CO gas-therapy-based versatile hydrogel dressing (ICOQF) was developed via the dynamic Schiff base reaction between the amino groups on quaternized chitosan (QCS) and the aldehyde groups on benzaldehyde-terminated F108 (F108-CHO) micelles. CORM-401 (an oxidant-sensitive CO-releasing molecules) was encapsulated in the hydrophobic core of F108-CHO micelles and insulin was loaded in the three-dimensional network structure of ICOQF. The dynamic Schiff base bonds not only endowed ICOQF with good tissue adhesion, injectability and self-healing, but also gave it sustained and controllable insulin release ability. In addition, ICOQF could quickly generate CO in inflamed wound tissue by consuming reactive oxygen species. The generated CO could effectively anti-oxidative stress by activating the expression of heme oxygenase; antibacterial by inducing the rupture of bacterial cell membranes and mitochondrial dysfunction and inhibiting the synthesis of adenosine triphosphate; and anti-inflammatory by inhibiting the proliferation of activated macrophages and promoting the polarization of the M1 phenotype to the M2 phenotype. Due to these outstanding properties, ICOQF significantly promoted the healing of STZ-induced MRSA-infected diabetic wounds accompanied by good biocompatibility. This study clearly shows that ICOQF is a versatile hydrogel dressing with great application potential for the management of diabetic wounds. STATEMENT OF SIGNIFICANCE: The development of some versatile hydrogel dressings that can not only provide a prolonged and controlled insulin release property but also utilize a non-antibiotic treatment modality for highly effective antibacterial, anti-inflammatory, and anti-oxidative stress effects is vital for the successful treatment of diabetic wounds. Herein, we developed an all-in-one CO gas-therapy-based versatile hydrogel dressing (ICOQF) with sustained and controllable insulin release abilities. Moreover, ICOQF could not only quickly release CO in the inflamed wound tissue by consumption of reactive oxygen species but also utilize the generated CO to highly effectively anti-oxidative stress, antibacterial, and anti-inflammatory. ICOQF therapy substantially promoted the healing of STZ-induced MRSA-infected diabetic wounds. Overall, this work provides a multifunctional hydrogel dressing for the management of diabetic wounds.
Collapse
|
36
|
Choi MJ, Choi KC, Lee DH, Jeong HY, Kang SJ, Kim MW, Jeong IH, You YM, Lee JS, Lee YK, Im CS, Park YS. EGF Receptor-Targeting Cancer Therapy Using CD47-Engineered Cell-Derived Nanoplatforms. Nanotechnol Sci Appl 2022; 15:17-31. [PMID: 35818431 PMCID: PMC9270928 DOI: 10.2147/nsa.s352038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Avoiding phagocytic cells and reducing off-target toxicity are the primary hurdles in the clinical application of nanoparticles containing therapeutics. For overcoming these errors, in this study, nanoparticles expressing CD47 proteins inhibiting the phagocytic attack of immune cells were prepared and then evaluated as an anti-cancer drug delivery vehicle. Methods The CD47+ cell-derived nanoparticles (CDNs) were prepared from the plasma membranes of human embryonic kidney cells transfected with a plasmid encoding CD47. And the doxorubicin (DOX) was loaded into the CDNs, and anti-EGF receptor (EGFR) antibodies were conjugated to the surface of the CDNs to target tumors overexpressing EGFR. Results The CD47+iCDNs-DOX was successfully synthesized having a stable structure. The CD47+CDNs were taken up less by RAW264.7 macrophages compared to control CDNs. Anti-EGFR CD47+CDNs (iCDNs) selectively recognized EGFR-positive MDA-MB-231 cells in vitro and accumulated more effectively in the target tumor xenografts in mice. Moreover, iCDNs encapsulating doxorubicin (iCDNs-DOX) exhibited the highest suppression of tumor growth in mice, presumably due to the enhanced DOX delivery to tumor tissues, compared to non-targeting CDNs or CD47- iCDNs. Discussion These results suggest that the clinical application of biocompatible cell membrane-derived nanocarriers could be facilitated by functionalization with macrophage-avoiding CD47 and tumor-targeting antibodies.
Collapse
Affiliation(s)
- Moon Jung Choi
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Kang Chan Choi
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Do Hyun Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Hwa Yeon Jeong
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Seong Jae Kang
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Min Woo Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - In Ho Jeong
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Young Myoung You
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Jin Suk Lee
- Department of Anatomy, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Yeon Kyung Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Chan Su Im
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
- Correspondence: Chan Su Im; Yong Serk Park, Department of Biomedical Laboratory Science, Yonsei University, Wonju, Gangwon, 220-710, Republic of Korea, Email ;
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
37
|
Wang H, Liu H, Guo Y, Zai W, Li X, Xiong W, Zhao X, Yao Y, Hu Y, Zou Z, Wu J. Photosynthetic microorganisms coupled photodynamic therapy for enhanced antitumor immune effect. Bioact Mater 2022; 12:97-106. [PMID: 35087966 PMCID: PMC8777206 DOI: 10.1016/j.bioactmat.2021.10.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 02/09/2023] Open
Abstract
The ideal photodynamic therapy (PDT) should effectively remove the primary tumor, and produce a stronger immune memory effect to inhibit the tumor recurrence and tumor metastasis. However, limited by the hypoxic and immunosuppressive microenvironment, the PDT efficiency is apparently low. Here, Chlorella (Chl.) is exploited to enhance local effect by producing oxygen to reverse hypoxia, and release adjuvants to reverse immunosuppressive microenvironment to enhance abscopal effect afterwards. Results from different animal models indicated that Chl. could enhance local effect and PDT related immune response. Ultimately, Chl. coupled PDT elicited anti-tumor effects toward established primary tumors (inhibition rate: 90%) and abscopal tumors (75%), controlled the challenged tumors (100%) and alleviated metastatic tumors (90%). This Chl. coupled PDT strategy can also produce a stronger anti-tumor immune memory effect. Overall, this Chl. coupled PDT strategy generates enhanced local tumor killing, boosts PDT-induced immune responses and promotes anti-tumor immune memory effect, which may be a great progress for realizing systemic effect of PDT. Chlorella can act as oxygen supplier and release adjuvants under light irradiation to enhance photodynamic therapy (PDT). The dual characteristics of Chlorella strengthen the occurrence of effective anti-tumor immune responses. Enhanced local and abscopal anti-tumor effect can be achieved by Chlorella with good biocompatibility.
Collapse
|
38
|
Malhotra S, Dumoga S, Singh N. Red blood cells membrane-derived nanoparticles: Applications and key challenges in their clinical translation. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1776. [PMID: 35106966 DOI: 10.1002/wnan.1776] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/16/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022]
Abstract
Cellular membrane-derived nanoparticles, particularly of red blood cells (RBCs), represent an emerging class of drug delivery systems. The lack of nucleus and organelles in these cells makes them easy to process and empty out intracellular contents. The empty vesicle membranes can then be either used as a coating on nanoparticles or can be reassembled into a nanovesicle. Engineered RBCs membrane has unique ability to retain its lipid bilayer architecture with host's proteins during top-down approach, thus allowing it to form stable nanoformulations mimicking RBCs stealth properties. In addition, its core-shell structure allows loading of different drug molecules, and its surface chemistry can be manipulated by facile conjugation with ligands on the shell. The remarkable ability of RBCs membrane to fuse with membranes of other cells enables the formation of hybrid nanovesicles. In this review, we highlight the biomedical applications of such vesicles and discuss the potential challenges related to its clinical translation. Although nano-RBCs retain much of the host's proteins, which may give an edge over synthetic nanoparticles in terms of lower immunogenicity, its production at industrial level is more challenging. This review gives the critical analysis of barriers involved in the translation of RBCs-derived nanoparticles from preclinical to clinical level. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Sahil Malhotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Shweta Dumoga
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India.,Biomedical Engineering unit, All India Institute of Medical Sciences New Delhi, New Delhi, India
| |
Collapse
|
39
|
Guo Y, Li W, Liu S, Jing D, Wang Y, Feng Q, Zhang K, Xu J. Construction of nanocarriers based on endogenous cell membrane and its application in nanomedicine. CHINESE J CHEM 2022. [DOI: 10.1002/cjoc.202100946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yingshu Guo
- Shandong Provincial Key Laboratory of Molecular Engineering School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250353 China
| | - Wenxin Li
- School of Chemistry and Chemical Engineering Linyi University Linyi 276005 China
| | - Shiwei Liu
- Shandong Provincial Key Laboratory of Molecular Engineering School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250353 China
| | - Dan Jing
- Shandong Provincial Key Laboratory of Molecular Engineering School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250353 China
| | - Yifan Wang
- Shandong Provincial Key Laboratory of Molecular Engineering School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250353 China
| | - Qingfang Feng
- Shandong Provincial Key Laboratory of Molecular Engineering School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250353 China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Jing‐Juan Xu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Centre of Chemistry for Life Sciences Nanjing University, 163 Xianlin Road Nanjing 210023 China
| |
Collapse
|
40
|
Wu N, Fan CH, Yeh CK. Ultrasound-activated nanomaterials for sonodynamic cancer theranostics. Drug Discov Today 2022; 27:1590-1603. [PMID: 35247594 DOI: 10.1016/j.drudis.2022.02.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/10/2022] [Accepted: 02/28/2022] [Indexed: 12/28/2022]
Abstract
Despite intensive efforts to develop diagnostic and therapeutic tools, the successful treatment of cancer is still hampered by the obscure boundary between cancerous cells and normal cells, recurrence of the cancer, and the development of drug resistance during chemotherapy. In recent years, sonodynamic therapy (SDT), employing therapeutic ultrasound with sonosensitizers, has attracted attention as a potentially promising approach for cancer therapy. This review describes the current understanding of the mechanisms and the preclinical and clinical efficacy of SDT-based applications in tumors, providing an insight into the therapeutic potential offered by SDT. The limitations and future directions of this emerging treatment are also discussed.
Collapse
Affiliation(s)
- Nan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
41
|
Rehman MU, Khan A, Imtiyaz Z, Ali S, Makeen HA, Rashid S, Arafah A. Current Nano-therapeutic Approaches Ameliorating Inflammation in Cancer Progression. Semin Cancer Biol 2022; 86:886-908. [DOI: 10.1016/j.semcancer.2022.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/22/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
|
42
|
Mei H, Cai S, Huang D, Gao H, Cao J, He B. Carrier-free nanodrugs with efficient drug delivery and release for cancer therapy: From intrinsic physicochemical properties to external modification. Bioact Mater 2022; 8:220-240. [PMID: 34541398 PMCID: PMC8424425 DOI: 10.1016/j.bioactmat.2021.06.035] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
The considerable development of carrier-free nanodrugs has been achieved due to their high drug-loading capability, simple preparation method, and offering "all-in-one" functional platform features. However, the native defects of carrier-free nanodrugs limit their delivery and release behavior throughout the in vivo journey, which significantly compromise the therapeutic efficacy and hinder their further development in cancer treatment. In this review, we summarized and discussed the recent strategies to enhance drug delivery and release of carrier-free nanodrugs for improved cancer therapy, including optimizing the intrinsic physicochemical properties and external modification. Finally, the corresponding challenges that carrier-free nanodrugs faced are discussed and the future perspectives for its application are presented. We hope this review will provide constructive information for the rational design of more effective carrier-free nanodrugs to advance therapeutic treatment.
Collapse
Affiliation(s)
- Heng Mei
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Shengsheng Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Dennis Huang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78731, USA
| | - Huile Gao
- West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Jun Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
43
|
Zeng Y, Li S, Zhang S, Wang L, Yuan H, Hu F. Cell membrane coated-nanoparticles for cancer immunotherapy. Acta Pharm Sin B 2022; 12:3233-3254. [PMID: 35967284 PMCID: PMC9366230 DOI: 10.1016/j.apsb.2022.02.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/14/2021] [Accepted: 02/19/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy can effectively inhibit cancer progression by activating the autoimmune system, with low toxicity and high effectiveness. Some of cancer immunotherapy had positive effects on clinical cancer treatment. However, cancer immunotherapy is still restricted by cancer heterogeneity, immune cell disability, tumor immunosuppressive microenvironment and systemic immune toxicity. Cell membrane-coated nanoparticles (CMCNs) inherit abundant source cell-relevant functions, including “self” markers, cross-talking with the immune system, biological targeting, and homing to specific regions. These enable them to possess preferred characteristics, including better biological compatibility, weak immunogenicity, immune escaping, a prolonged circulation, and tumor targeting. Therefore, they are applied to precisely deliver drugs and promote the effect of cancer immunotherapy. In the review, we summarize the latest researches of biomimetic CMCNs for cancer immunotherapy, outline the existing specific cancer immune therapies, explore the unique functions and molecular mechanisms of various cell membrane-coated nanoparticles, and analyze the challenges which CMCNs face in clinical translation.
Collapse
|
44
|
Recent Advances in Strategies for Addressing Hypoxia in Tumor Photodynamic Therapy. Biomolecules 2022; 12:biom12010081. [PMID: 35053229 PMCID: PMC8774200 DOI: 10.3390/biom12010081] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 01/10/2023] Open
Abstract
Photodynamic therapy (PDT) is a treatment modality that uses light to target tumors and minimize damage to normal tissues. It offers advantages including high spatiotemporal selectivity, low side effects, and maximal preservation of tissue functions. However, the PDT efficiency is severely impeded by the hypoxic feature of tumors. Moreover, hypoxia may promote tumor metastasis and tumor resistance to multiple therapies. Therefore, addressing tumor hypoxia to improve PDT efficacy has been the focus of antitumor treatment, and research on this theme is continuously emerging. In this review, we summarize state-of-the-art advances in strategies for overcoming hypoxia in tumor PDTs, categorizing them into oxygen-independent phototherapy, oxygen-economizing PDT, and oxygen-supplementing PDT. Moreover, we highlight strategies possessing intriguing advantages such as exceedingly high PDT efficiency and high novelty, analyze the strengths and shortcomings of different methods, and envision the opportunities and challenges for future research.
Collapse
|
45
|
Khatoon N, Zhang Z, Zhou C, Chu M. Macrophage membrane coated nanoparticles: a biomimetic approach for enhanced and targeted delivery. Biomater Sci 2022; 10:1193-1208. [DOI: 10.1039/d1bm01664d] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The enhanced and targeted drug delivery with low systemic toxicity and subsequent release of drugs is the major concern among researchers and pharmaceutics. Inspite of greater advancement and discoveries in...
Collapse
|
46
|
Guo M, Xia C, Wu Y, Zhou N, Chen Z, Li W. Research Progress on Cell Membrane-Coated Biomimetic Delivery Systems. Front Bioeng Biotechnol 2021; 9:772522. [PMID: 34869288 PMCID: PMC8636778 DOI: 10.3389/fbioe.2021.772522] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/29/2021] [Indexed: 01/12/2023] Open
Abstract
Cell membrane-coated biomimetic nanoplatforms have many inherent properties, such as bio-interfacing abilities, self-identification, and signal transduction, which enable the biomimetic delivery system to escape immune clearance and opsonization. This can also maximize the drug delivery efficiency of synthetic nanoparticles (NPs) and functional cell membranes. As a new type of delivery system, cell membrane-coated biomimetic delivery systems have broadened the prospects for biomedical applications. In this review, we summarize research progress on cell membrane biomimetic technology from three aspects, including sources of membrane, modifications, and applications, then analyze their limitations and propose future research directions.
Collapse
Affiliation(s)
- Mengyu Guo
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenjie Xia
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nong Zhou
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area's Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Zhipeng Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weidong Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
47
|
Wan Y, Fu LH, Li C, Lin J, Huang P. Conquering the Hypoxia Limitation for Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103978. [PMID: 34580926 DOI: 10.1002/adma.202103978] [Citation(s) in RCA: 252] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/11/2021] [Indexed: 06/13/2023]
Abstract
Photodynamic therapy (PDT) has aroused great research interest in recent years owing to its high spatiotemporal selectivity, minimal invasiveness, and low systemic toxicity. However, due to the hypoxic nature characteristic of many solid tumors, PDT is frequently limited in therapeutic effect. Moreover, the consumption of O2 during PDT may further aggravate the tumor hypoxic condition, which promotes tumor proliferation, metastasis, and invasion resulting in poor prognosis of treatment. Therefore, numerous efforts have been made to increase the O2 content in tumor with the goal of enhancing PDT efficacy. Herein, these strategies developed in past decade are comprehensively reviewed to alleviate tumor hypoxia, including 1) delivering exogenous O2 to tumor directly, 2) generating O2 in situ, 3) reducing tumor cellular O2 consumption by inhibiting respiration, 4) regulating the TME, (e.g., normalizing tumor vasculature or disrupting tumor extracellular matrix), and 5) inhibiting the hypoxia-inducible factor 1 (HIF-1) signaling pathway to relieve tumor hypoxia. Additionally, the O2 -independent Type-I PDT is also discussed as an alternative strategy. By reviewing recent progress, it is hoped that this review will provide innovative perspectives in new nanomaterials designed to combat hypoxia and avoid the associated limitation of PDT.
Collapse
Affiliation(s)
- Yilin Wan
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Lian-Hua Fu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Chunying Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
48
|
Chugh V, Vijaya Krishna K, Pandit A. Cell Membrane-Coated Mimics: A Methodological Approach for Fabrication, Characterization for Therapeutic Applications, and Challenges for Clinical Translation. ACS NANO 2021; 15:17080-17123. [PMID: 34699181 PMCID: PMC8613911 DOI: 10.1021/acsnano.1c03800] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/13/2021] [Indexed: 05/04/2023]
Abstract
Cell membrane-coated (CMC) mimics are micro/nanosystems that combine an isolated cell membrane and a template of choice to mimic the functions of a cell. The design exploits its physicochemical and biological properties for therapeutic applications. The mimics demonstrate excellent biological compatibility, enhanced biointerfacing capabilities, physical, chemical, and biological tunability, ability to retain cellular properties, immune escape, prolonged circulation time, and protect the encapsulated drug from degradation and active targeting. These properties and the ease of adapting them for personalized clinical medicine have generated a significant research interest over the past decade. This review presents a detailed overview of the recent advances in the development of cell membrane-coated (CMC) mimics. The primary focus is to collate and discuss components, fabrication methodologies, and the significance of physiochemical and biological characterization techniques for validating a CMC mimic. We present a critical analysis of the two main components of CMC mimics: the template and the cell membrane and mapped their use in therapeutic scenarios. In addition, we have emphasized on the challenges associated with CMC mimics in their clinical translation. Overall, this review is an up to date toolbox that researchers can benefit from while designing and characterizing CMC mimics.
Collapse
Affiliation(s)
| | | | - Abhay Pandit
- CÚRAM, SFI Research
Centre for Medical Devices, National University
of Ireland Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
49
|
Zhou Z, Jiang N, Chen J, Zheng C, Guo Y, Ye R, Qi R, Shen J. Selectively down-regulated PD-L1 by albumin-phenformin nanoparticles mediated mitochondrial dysfunction to stimulate tumor-specific immunological response for enhanced mild-temperature photothermal efficacy. J Nanobiotechnology 2021; 19:375. [PMID: 34794446 PMCID: PMC8600872 DOI: 10.1186/s12951-021-01124-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
Background Mild-temperature photothermal therapy (mild-PTT) has emerged as a highly promising antitumor strategy by triggering immunogenic cell death (ICD) to elicit both innate and adaptive immune responses for tumor control. However, mild-PTT still leads to the risk of tumor recurrence or metastasis because it could hardly completely eradicate tumors due to its impaired immunological efficacy owing to the enhanced PD-L1 expression in tumor cells after treatment. Results In this study, we described a hydrogen peroxide (H2O2) responsive manganese dioxide mineralized albumin nanocomposite loading with mitochondria function inhibitor phenformin (PM) and near-infrared photothermal dye indocyanine green (ICG) by modified two-step biomineralization method. In combination with ICG induced mild-PTT and PM mediated mitochondria dysfunction, PD-L1 expression was obviously down-regulated and the generated immunological responses was able to effectively attack the remaining tumor cells. Meanwhile, the risk of tumor metastasis was effectively inhibited by reducing the expression of tumor invasion-related signal molecules (TGF-β and vimentin) after combining treatment. Conclusion Such a strategy offers novel insight into the development of nanomedicine for mild-PTT as well as cancer immunotherapy, which can provide protection against tumor relapse post elimination of their initial and metastatic tumors. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01124-8. Over-expression of PD-L1 after mild-photothermal therapy significantly limited its efficacy. Phenformin could effectively downregulate PD-L1 expression and inhibit tumor metastasis through AMPK activation. Hydrogen peroxide responsive manganese dioxide mineralized albumin nanocomplex co-loading with phenformin and ICG named ICG@PM@NP was constructed by modified two-step biomineralization method. ICG@PM@NP could enhance T cell infiltration and antitumor metastasis in vivo. ICG@PM@NP mediated mild-photothermal therapy could make up the defects of conventional mild-photothermal therapy in lacking the anti-metastasis ability and inducing enhanced PD-L1 expression.
Collapse
Affiliation(s)
- Zaigang Zhou
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ning Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jiashe Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Chunjuan Zheng
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yuanyuan Guo
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ruirong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Ruogu Qi
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jianliang Shen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China. .,Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China. .,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
50
|
Xiong W, Qi L, Si D, Jiang X, Liu Y, Zheng C, Li Y, Shen J, Zhou Z. Effective tumor vessel barrier disruption mediated by perfluoro-N-(4-methylcyclohexyl) piperidine nanoparticles to enhance the efficacy of photodynamic therapy. NANOSCALE 2021; 13:13473-13486. [PMID: 34477752 DOI: 10.1039/d1nr02880d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Currently, limited tumor drug permeation, poor oxygen perfusion and immunosuppressive microenvironments are the most important bottlenecks that significantly reduce the efficacy of photodynamic therapy (PDT). The main cause of these major bottlenecks is the platelet activation maintained abnormal tumor vessel barriers. Thus, platelet inhibition may present a new way to most effectively enhance the efficacy of PDT. However, to the best of our knowledge, few studies have validated the effectiveness of such a way in enhancing the efficacy of PDT both in vivo and in vitro. In this study, perfluoro-N-(4-methylcyclohexyl) piperidine-loaded albumin (PMP@Alb) nanoparticles were discovered, which possess excellent platelet inhibition ability. After PMP@Alb treatment, remarkably enhanced intra-tumoral drug accumulation, oxygen perfusion and T cell infiltration could be observed owing to the disrupted tumor vessel barriers. Besides, the effect of ICG@Lip mediated PDT was significantly amplified by PMP@Alb nanoparticles. It was demonstrated that PMP@Alb could be used as a useful tool to improve the efficacy of existing PDT by disrupting tumor vessel barriers through effective platelet inhibition.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | | | | | | | | | | | | | | | | |
Collapse
|