1
|
Amirian H, Franco FB, Dabiri B, Alessandrino F. Urologic Imaging of the Bladder: Cancers and Mimics. Urol Clin North Am 2025; 52:111-124. [PMID: 39537297 DOI: 10.1016/j.ucl.2024.07.011] [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: 11/16/2024]
Abstract
Bladder cancer (BC) represents a significant global health challenge with notable incidence and mortality rates. Despite treatment advancements, its management remains complex, requiring a multidisciplinary approach. Imaging techniques play a pivotal role in diagnosis, staging, and treatment planning by aiding lesion localization, differentiation, and assessment of tumor extent. Primary modalities like computed tomography and MRI offer detailed anatomic insights. Imaging provides valuable insights into tumor biology, vascular patterns, and molecular profiles, enabling personalized medicine strategies to optimize therapeutic efficacy and minimize adverse effects, crucial for improving BC management and prognosis.
Collapse
Affiliation(s)
- Haleh Amirian
- Department of Surgical Oncology, University of Miami, 1150 Northwest 14th Street #511, Miami, FL 33136, USA
| | - Felipe B Franco
- Department of Body Imaging, Radiology Associates of South Florida, 8900 North Kendall Drive, Miami, FL 33176, USA.
| | - Borna Dabiri
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Francesco Alessandrino
- Division of Abdominal Imaging, Department of Radiology, University of Miami, 1150 Northwest 14th Street #511, Miami, FL 33136, USA
| |
Collapse
|
2
|
Butt A, Bach H. Advancements in nanotechnology for diagnostics: a literature review, part II: advanced techniques in nuclear and optical imaging. Nanomedicine (Lond) 2024:1-24. [PMID: 39670826 DOI: 10.1080/17435889.2024.2439778] [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: 09/08/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024] Open
Abstract
Modern molecular imaging routes, such as nuclear imaging and optical imaging, derive significant advantages from nanoparticles, where multimodality use and multipurpose are key benefits. Nanoparticles also showcase benefits over traditional imaging agents in nuclear and optical imaging, including improved resolution, penetration, and specificity. The goal of this literature review was to explore recent advancements in nanomaterials within these molecular imaging techniques to expand on the current state of nanomedicine in these modalities. This review derives findings from relevant reviews, original research papers, in-human clinical trials, and patents in the literature. Au- and Fe oxide-based nanosystems are just as ubiquitous within more modern modalities due to their multimodal diagnostic and therapeutic potential. It is also repeatedly highlighted in the literature, patents, and clinical trials that the use of nanoparticles, specifically in multimodal imaging techniques and theranostics, present innovative methods in recent years, enabling researchers and clinicians to overcome the limitations of unimodal imaging modalities and further advancing accuracy in the diagnosis and treatment of important pathologies, particularly cancer. Overall, nanoparticle-based imaging represents a transformative approach in advanced imaging modalities, offering new approaches to limitations of conventional agents currently being applied in clinical settings.
Collapse
Affiliation(s)
- Ahmad Butt
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Horacio Bach
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Division of Infectious Diseases, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Guo Y, Qian R, Li Z, Lv T, Yang C, Li W, Pan T, Hou X, Wang Z. Tumor-derived nanovesicles enhance cancer synergistic chemo-immunotherapy by promoting cGAS/STING pathway activation and immunogenetic cell death. Life Sci 2024; 348:122687. [PMID: 38718856 DOI: 10.1016/j.lfs.2024.122687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
AIMS Checkpoint blockade immunotherapy is a promising therapeutic modality that has revolutionized cancer treatment; however, the therapy is only effective on a fraction of patients due to the tumor environment. In tumor immunotherapy, the cGAS-STING pathway is a crucial intracellular immune response pathway. Therefore, this study aimed to develop an immunotherapy strategy based on the cGAS-STING pathway. MATERIALS AND METHODS The physicochemical properties of the nanoparticles EM@REV@DOX were characterized by TEM, DLS, and WB. Subcutaneous LLC xenograft tumors were used to determine the biodistribution, antitumor efficacy, and immune response. Blood samples and tissues of interest were harvested for hematological analysis and H&E staining. SIGNIFICANCE Overall, our designed nanovesicles provide a new perspective on tumor immunotherapy by ICD and cGAS-STING pathway, promoting DCs maturation, macrophage polarization, and activating T cells, offering a meaningful strategy for accelerating the clinical development of immunotherapy. KEY FINDINGS EM@REV@DOX accumulated in the tumor site through EPR and homing targeting effect to release REV and DOX, resulting in DNA damage and finally activating the cGAS-STING pathway, thereby promoting DCs maturation, macrophage polarization, and activating T cells. Additionally, EM@REV@DOX increased the production of pro-inflammatory cytokines (e.g., TNF-α and IFN-β). As a result, EM@REV@DOX was effective in treating tumor-bearing mice and prolonged their lifespans. When combined with αPD-L1, EM@REV@DOX significantly inhibited distant tumor growth, extended the survival of mice, and prevented long-term postoperative tumor metastasis, exhibiting great potential in antitumor immunotherapy.
Collapse
Affiliation(s)
- Yawen Guo
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Ruijie Qian
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, PR China
| | - Zijie Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Tingting Lv
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Chunwang Yang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Wen Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Teng Pan
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xiaoming Hou
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Zhiyu Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China.
| |
Collapse
|
4
|
Hou Y, Zhu C, Ban G, Shen Z, Liang Y, Chen K, Wang C, Shi H. Advancements and Challenges in the Application of Metal-Organic Framework (MOF) Nanocomposites for Tumor Diagnosis and Treatment. Int J Nanomedicine 2024; 19:6295-6317. [PMID: 38919774 PMCID: PMC11198007 DOI: 10.2147/ijn.s463144] [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: 02/06/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Nanoscale metal-organic frameworks (MOFs) offer high biocompatibility, nanomaterial permeability, substantial specific surface area, and well-defined pores. These properties make MOFs valuable in biomedical applications, including biological targeting and drug delivery. They also play a critical role in tumor diagnosis and treatment, including tumor cell targeting, identification, imaging, and therapeutic methods such as drug delivery, photothermal effects, photodynamic therapy, and immunogenic cell death. The diversity of MOFs with different metal centers, organics, and surface modifications underscores their multifaceted contributions to tumor research and treatment. This review is a summary of these roles and mechanisms. The final section of this review summarizes the current state of the field and discusses prospects that may bring MOFs closer to pharmaceutical applications.
Collapse
Affiliation(s)
- Yingze Hou
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
- Clinical Medical College, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Can Zhu
- Department of Urology, The Second Clinical Medical College of Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Ge Ban
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Zhean Shen
- Heart Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| | - Yingbing Liang
- Department of Chemistry and Biotechnology, Graduate School of Engineering Tottori University Koyama-Minami 4-101, Tottori, 680-8552, Japan
| | - Kun Chen
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Chenbo Wang
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Heng Shi
- Heart Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| |
Collapse
|
5
|
Zhao D, Li Z, Ji DK, Xia Q. Recent Progress of Multifunctional Molecular Probes for Triple-Negative Breast Cancer Theranostics. Pharmaceutics 2024; 16:803. [PMID: 38931924 PMCID: PMC11207493 DOI: 10.3390/pharmaceutics16060803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer (BC) poses a significant threat to women's health, with triple-negative breast cancer (TNBC) representing one of the most challenging and aggressive subtypes due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Traditional TNBC treatments often encounter issues such as low drug efficiency, limited tumor enrichment, and substantial side effects. Therefore, it is crucial to explore novel diagnostic and treatment systems for TNBC. Multifunctional molecular probes (MMPs), which integrate target recognition as well as diagnostic and therapeutic functions, introduce advanced molecular tools for TNBC theranostics. Using an MMP system, molecular drugs can be precisely delivered to the tumor site through a targeted ligand. Real-time dynamic monitoring of drug release achieved using imaging technology allows for the evaluation of drug enrichment at the tumor site. This approach enables accurate drug release, thereby improving the therapeutic effect. Therefore, this review summarizes the recent advancements in MMPs for TNBC theranostics, encompassing the design and synthesis of MMPs as well as their applications in the field of TNBC theranostics.
Collapse
Affiliation(s)
- Deyi Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (D.Z.); (Z.L.)
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhe Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (D.Z.); (Z.L.)
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ding-Kun Ji
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qian Xia
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
6
|
Hassibian S, Taghdisi SM, Jamshidi Z, Samie A, Alinezhad Nameghi M, Shayan M, Farrokhi N, Alibolandi M, Ramezani M, Dehnavi SM, Abnous K. Surface modification of hollow gold nanoparticles conducted by incorporating cancer cell membrane and AS1411 aptamer, aiming to achieve a dual-targeted therapy for colorectal cancer. Int J Pharm 2024; 655:124036. [PMID: 38522491 DOI: 10.1016/j.ijpharm.2024.124036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Due to its inherent membrane structure, a nanostructure enveloped by an active cell membrane possesses distinctive characteristics such as prolonged presence in the bloodstream, precise identification capabilities, and evasion of immune responses. This research involved the production of biomimetic nanoparticles, specifically hollow gold nanoparticles (HGNPs) loaded with methotrexate (MTX), which were further coated with cancer cell membrane. These nanoparticles were then adorned with AS1411 aptamer to serve as a targeting agent (Apt-CCM-HG@MTX). The nanoplatform demonstrated precise targeting towards cancer cells due to its dual-targeting characteristic (AS1411 aptamer and C26 cancer cell membrane), exhibiting uniformity in distribution. It also displayed a desirable response to photothermal stimulation, controlled release of drugs, and exceptional properties for fluorescence imaging. The system was composed of spherical HGNPs measuring 51.33 ± 5.70 nm in diameter, which were effectively loaded with MTX using a physical absorption method. The encapsulation efficiency achieved was recorded at 79.54 %, while the loading efficiency reached 38.21 %. The targeted formulation demonstrated a noteworthy mortality of approximately 45 % in the nucleolin positive cell line, C26, as determined by in vitro cytotoxicity assays. As a result of the functionalization process applied to the homologous binding adhesion molecules found in cancer cell membranes and targeting ability of AS1411 aptamer, Apt-CCM-HG@MTX demonstrated a substantial enhancement in targeting tumors and facilitating cellular uptake during in vivo experiments. Furthermore, under NIR radiation the photothermal effect exhibited by Apt-CCM-HG@MTX in the tumor area was notably robust due to the distinctive attributes of HGNPs. The conclusions obtained from this study have the potential to assist in adopting a bioinspired strategy that will significantly improve the effective management of MTX and therapy for individuals with colorectal cancer.
Collapse
Affiliation(s)
- Sepideh Hassibian
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, P.O. Box 19839-69411, Tehran, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Jamshidi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Samie
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mersedeh Shayan
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Naser Farrokhi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, P.O. Box 19839-69411, Tehran, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohsen Dehnavi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, P.O. Box 19839-69411, Tehran, Iran.
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Yuan M, Han Z, Li Y, Zhan X, Sun Y, He B, Liang Y, Luo K, Li F. A pH-responsive nanoplatform with dual-modality imaging for enhanced cancer phototherapy and diagnosis of lung metastasis. J Nanobiotechnology 2024; 22:180. [PMID: 38622591 PMCID: PMC11017640 DOI: 10.1186/s12951-024-02431-6] [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: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024] Open
Abstract
To address the limitations of traditional photothermal therapy (PTT)/ photodynamic therapy (PDT) and real-time cancer metastasis detection, a pH-responsive nanoplatform (NP) with dual-modality imaging capability was rationally designed. Herein, 1 H,1 H-undecafluorohexylamine (PFC), served as both an oxygen carrier and a 19F magnetic resonance imaging (MRI) probe, and photosensitizer indocyanine green (ICG) were grafted onto the pH-responsive peptide hexahistidine (H6) to form H6-PFC-ICG (HPI). Subsequently, the heat shock protein 90 inhibitor, gambogic acid (GA), was incorporated into hyaluronic acid (HA) modified HPI (HHPI), yielding the ultimate HHPI@GA NPs. Upon self-assembly, HHPI@GA NPs passively accumulated in tumor tissues, facilitating oxygen release and HA-mediated cell uptake. Once phagocytosed by lysosomes, protonation of H6 was triggered due to the low pH, resulting in the release of GA. With near-infrared laser irradiation, GA-mediated decreased HSP90 expression and PFC-mediated increased ROS generation amplified the PTT/PDT effect of HHPI@GA, leading to excellent in vitro and in vivo anticancer efficacies. Additionally, the fluorescence and 19F MRI dual-imaging capabilities of HHPI@GA NPs enabled effective real-time primary cancer and lung metastasis monitoring. This work offers a novel approach for enhanced cancer phototherapy, as well as precise cancer diagnosis.
Collapse
Affiliation(s)
- Mujie Yuan
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Zeyu Han
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xin Zhan
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yong Sun
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Yan Liang
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China.
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fan Li
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China.
| |
Collapse
|
8
|
Guo Q, Wang S, Xu R, Tang Y, Xia X. Cancer cell membrane-coated nanoparticles: a promising anti-tumor bionic platform. RSC Adv 2024; 14:10608-10637. [PMID: 38567339 PMCID: PMC10985588 DOI: 10.1039/d4ra01026d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Nanoparticle (NP) drug delivery systems have shown promise in tumor therapy. However, limitations such as susceptibility to immune clearance and poor targeting in a complex intercellular environment still exist. Recently, cancer cell membrane-encapsulated nanoparticles (CCM-NPs) constructed using biomimetic nanotechnology have been developed to overcome these problems. Proteins on the membrane surface of cancer cells can provide a wide range of activities for CCM-NPs, including immune escape and homologous cell recognition properties. Meanwhile, the surface of the cancer cell membrane exhibits obvious antigen enrichment, so that CCM-NPs can transmit tumor-specific antigen, activate a downstream immune response, and produce an effective anti-tumor effect. In this review, we first provided an overview of the functions of cancer cell membranes and summarized the preparation techniques and characterization methods of CCM-NPs. Then, we focused on the application of CCM-NPs in tumor therapy. In addition, we summarized the functional modifications of cancer cell membranes and compiled the patent applications related to CCM-NPs in recent years. Finally, we proposed the future challenges and directions of this technology in order to provide guidance for researchers in this field.
Collapse
Affiliation(s)
- Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Rubing Xu
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Yingnan Tang
- School of Pharmacy, Hunan Vocational College of Science and Technology Changsha Hunan 410208 China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| |
Collapse
|
9
|
Liu X, Chu Z, Chen B, Ma Y, Xu L, Qian H, Yu Y. Cancer cell membrane-coated upconversion nanoparticles/Zn xMn 1-xS core-shell nanoparticles for targeted photodynamic and chemodynamic therapy of pancreatic cancer. Mater Today Bio 2023; 22:100765. [PMID: 37636984 PMCID: PMC10457453 DOI: 10.1016/j.mtbio.2023.100765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/29/2023] [Accepted: 08/05/2023] [Indexed: 08/29/2023] Open
Abstract
Oxidative stress induced by reactive oxygen species (ROS) is promising treatment approach for pancreatic ductal adenocarcinoma (PDAC), which is typically insensitive to conventional chemotherapy. In this study, BxPC-3 pancreatic cancer cell membrane-coated upconversion nanoparticles/ZnxMn1-xS core-shell nanoparticles (abbreviated as BUC@ZMS) were developed for tumor-targeted cancer therapy via synergistically oxidative stress and overcoming glutathione (GSH) overexpression. Using a combination of photodynamic therapy (PDT) and chemodynamic therapy (CDT), the BUC@ZMS core-shell nanoparticles were able to elicit the death of pancreatic cancer cells through the high production of ROS. Additionally, the BUC@ZMS core-shell nanoparticles could deplete intracellular GSH and increase the sensitivity of tumor cells to oxidative stress. The in vivo results indicated that BUC@ZMS nanoparticles can accumulate specifically in tumor locations and suppress PDAC without generating obvious toxicity. Thus, it was determined that the as-prepared core-shell nanoparticles would be a viable treatment option for solid malignancies.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
- Department of Gastroenterology, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui, 237000, PR China
| | - Zhaoyou Chu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, PR China
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Benjin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, PR China
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Lingling Xu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Yue Yu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| |
Collapse
|
10
|
Zhou Z, Zhang S, Xue N. Research progress of cancer cell membrane coated nanoparticles for the diagnosis and therapy of breast cancer. Front Oncol 2023; 13:1270407. [PMID: 37781205 PMCID: PMC10539574 DOI: 10.3389/fonc.2023.1270407] [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: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Nanoparticles (NPs) disguised in the cell membrane are a new type of biomimetic platform. Due to their ability to simulate the unique biological functions of membrane-derived cells, they have become one of the hotspots of research at home and abroad. The tumor-specific antigen antibody carried by breast cancer cell membranes can modify nanoparticles to have homologous tumor targeting. Therefore, nanoparticles wrapped in cancer cell membranes have been widely used in research on the diagnosis and treatment of breast cancer. This article reviews the current situation, prospects, advantages and limitations of nanoparticles modified by cancer cell membranes in the treatment and diagnosis of breast cancer.
Collapse
Affiliation(s)
| | - Shengmin Zhang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Nianyu Xue
- Department of Ultrasound Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
11
|
Li W, Cheng J, He F, Zhang P, Zhang N, Wang J, Song Q, Hou Y, Gan Z. Cell membrane-based nanomaterials for theranostics of central nervous system diseases. J Nanobiotechnology 2023; 21:276. [PMID: 37596631 PMCID: PMC10439658 DOI: 10.1186/s12951-023-02004-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/13/2023] [Indexed: 08/20/2023] Open
Abstract
Central nervous system (CNS) diseases have been widely acknowledged as one of the major healthy concerns globally, which lead to serious impacts on human health. There will be about 135 million CNS diseases cases worldwide by mid-century, and CNS diseases will become the second leading cause of death after the cardiovascular disease by 2040. Most CNS diseases lack of effective diagnostic and therapeutic strategies with one of the reasons that the biological barrier extremely hampers the delivery of theranostic agents. In recent years, nanotechnology-based drug delivery is a quite promising way for CNS diseases due to excellent properties. Among them, cell membrane-based nanomaterials with natural bio-surface, high biocompatibility and biosafety, are of great significance in both the diagnosis and treatment of different CNS diseases. In this review, the state of art of the fabrication of cell membranes-based nanomaterials is introduced. The characteristics of different CNS diseases, and the application of cell membranes-based nanomaterials in the theranostics are summarized. In addition, the future prospects and limitations of cell membrane nanotechnology are anticipated. Through summarizing the state of art of the fabrication, giving examples of CNS diseases, and highlighting the applications in theranostics, the current review provides designing methods and ideas for subsequent cell membrane nanomaterials.
Collapse
Affiliation(s)
- Wenyue Li
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Junwei Cheng
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fangfei He
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Peisen Zhang
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ni Zhang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Jian Wang
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China.
| | - Qiliang Song
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang, 262700, China.
| | - Yi Hou
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhihua Gan
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
12
|
Diao L, Liu M. Rethinking Antigen Source: Cancer Vaccines Based on Whole Tumor Cell/tissue Lysate or Whole Tumor Cell. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300121. [PMID: 37254712 PMCID: PMC10401146 DOI: 10.1002/advs.202300121] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/29/2023] [Indexed: 06/01/2023]
Abstract
Cancer immunotherapies have improved human health, and one among the important technologies for cancer immunotherapy is cancer vaccine. Antigens are the most important components in cancer vaccines. Generally, antigens in cancer vaccines can be divided into two categories: pre-defined antigens and unidentified antigens. Although, cancer vaccines loaded with predefined antigens are commonly used, cancer vaccine loaded with mixed unidentified antigens, especially whole cancer cells or cancer cell lysates, is a very promising approach, and such vaccine can obviate some limitations in cancer vaccines. Their advantages include, but are not limited to, the inclusion of pan-spectra (all or most kinds of) antigens, inducing pan-clones specific T cells, and overcoming the heterogeneity of cancer cells. In this review, the recent advances in cancer vaccines based on whole-tumor antigens, either based on whole cancer cells or whole cancer cell lysates, are summarized. In terms of whole cancer cell lysates, the focus is on applying whole water-soluble cell lysates as antigens. Recently, utilizing the whole cancer cell lysates as antigens in cancer vaccines has become feasible. Considering that pre-determined antigen-based cancer vaccines (mainly peptide-based or mRNA-based) have various limitations, developing cancer vaccines based on whole-tumor antigens is a promising alternative.
Collapse
Affiliation(s)
- Lu Diao
- Department of PharmaceuticsCollege of Pharmaceutical Sciences, Soochow University199 of Ren ai RoadSuzhouJiangsu215123P. R. China
- Kunshan Hospital of Traditional Chinese MedicineKunshanJiangsu215300P. R. China
- Suzhou Ersheng Biopharmaceutical Co., Ltd.Suzhou215123P. R. China
| | - Mi Liu
- Department of PharmaceuticsCollege of Pharmaceutical Sciences, Soochow University199 of Ren ai RoadSuzhouJiangsu215123P. R. China
- Kunshan Hospital of Traditional Chinese MedicineKunshanJiangsu215300P. R. China
- Suzhou Ersheng Biopharmaceutical Co., Ltd.Suzhou215123P. R. China
| |
Collapse
|
13
|
Chen B, Xiao L, Wang W, Xu L, Jiang Y, Zhang G, Liu L, Li X, Yu Y, Qian H. Bi 2-xMn xO 3 Nanospheres Engaged Radiotherapy with Amplifying DNA Damage. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37410709 DOI: 10.1021/acsami.3c06838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Radiotherapy efficacy was greatly limited by hypoxia and overexpression of glutathione (GSH) in the tumor microenvironment (TME), which maintained the immunosuppressive microenvironment and promoted DNA repair. In this work, 4T1 cell membrane-coated Bi2-xMnxO3 nanospheres have been achieved via a facile protocol, which showed enhanced therapeutic efficacy for a combination of radiotherapy and immunotherapy. Bi2-xMnxO3 nanospheres showed appreciable performance in generating O2 in situ and depleting GSH to amplify DNA damage and remodel the tumor immunosuppressive microenvironment, thus enhancing radiotherapy efficacy. Cancer cell membrane-coated Bi2-xMnxO3 nanospheres (T@BM) prolonged blood circulation time and enriched the accumulation of the materials in the tumor. Meanwhile, the released Mn2+ could activate STING pathway-induced immunotherapy, resulting in the immune infiltration of CD8+ T cells on in situ mammary tumors and the inhibition of pulmonary nodules. As a result, approximately 1.9-fold recruitment of CD8+ T cells and 4.0-fold transformation of mature DC cells were observed compared with the phosphate-buffered saline (PBS) group on mammary tumors (in situ). In particular, the number of pulmonary nodules significantly decreased and the proliferation of pulmonary metastatic lesions was substantially inhibited, which provided a longer survival period. Therefore, T@BM exhibited great potential for the treatment of 4T1 tumors in situ and lung metastasis.
Collapse
Affiliation(s)
- Benjin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Liang Xiao
- Department of Radiology, Research Center of Clinical Medical Imaging, Anhui Province Clinical Image Quality Control Center, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Wanni Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, Anhui, P. R. China
- Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei 230012, Anhui, P. R. China
| | - Lingling Xu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, Anhui, P. R. China
- Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei 230012, Anhui, P. R. China
| | - Yechun Jiang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Guoqiang Zhang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Lin Liu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Xiaohu Li
- Department of Radiology, Research Center of Clinical Medical Imaging, Anhui Province Clinical Image Quality Control Center, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Yongqiang Yu
- Department of Radiology, Research Center of Clinical Medical Imaging, Anhui Province Clinical Image Quality Control Center, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, Anhui, P. R. China
- Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei 230012, Anhui, P. R. China
| |
Collapse
|
14
|
Desai N, Rana D, Pande S, Salave S, Giri J, Benival D, Kommineni N. "Bioinspired" Membrane-Coated Nanosystems in Cancer Theranostics: A Comprehensive Review. Pharmaceutics 2023; 15:1677. [PMID: 37376125 DOI: 10.3390/pharmaceutics15061677] [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: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Achieving precise cancer theranostics necessitates the rational design of smart nanosystems that ensure high biological safety and minimize non-specific interactions with normal tissues. In this regard, "bioinspired" membrane-coated nanosystems have emerged as a promising approach, providing a versatile platform for the development of next-generation smart nanosystems. This review article presents an in-depth investigation into the potential of these nanosystems for targeted cancer theranostics, encompassing key aspects such as cell membrane sources, isolation techniques, nanoparticle core selection, approaches for coating nanoparticle cores with the cell membrane, and characterization methods. Moreover, this review underscores strategies employed to enhance the multi-functionality of these nanosystems, including lipid insertion, membrane hybridization, metabolic engineering, and genetic modification. Additionally, the applications of these bioinspired nanosystems in cancer diagnosis and therapeutics are discussed, along with the recent advances in this field. Through a comprehensive exploration of membrane-coated nanosystems, this review provides valuable insights into their potential for precise cancer theranostics.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Shreya Pande
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | | |
Collapse
|
15
|
Ahmadi M, Emzhik M, Mosayebnia M. Nanoparticles labeled with gamma-emitting radioisotopes: an attractive approach for in vivo tracking using SPECT imaging. Drug Deliv Transl Res 2023; 13:1546-1583. [PMID: 36811810 DOI: 10.1007/s13346-023-01291-1] [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] [Accepted: 01/03/2023] [Indexed: 02/24/2023]
Abstract
Providing accurate molecular imaging of the body and biological process is critical for diagnosing disease and personalizing treatment with the minimum side effects. Recently, diagnostic radiopharmaceuticals have gained more attention in precise molecular imaging due to their high sensitivity and appropriate tissue penetration depth. The fate of these radiopharmaceuticals throughout the body can be traced using nuclear imaging systems, including single-photon emission computed tomography (SPECT) and positron emission tomography (PET) modalities. In this regard, nanoparticles are attractive platforms for delivering radionuclides into targets because they can directly interfere with the cell membranes and subcellular organelles. Moreover, applying radiolabeled nanomaterials can decrease their toxicity concerns because radiopharmaceuticals are usually administrated at low doses. Therefore, incorporating gamma-emitting radionuclides into nanomaterials can provide imaging probes with valuable additional properties compared to the other carriers. Herein, we aim to review (1) the gamma-emitting radionuclides used for labeling different nanomaterials, (2) the approaches and conditions adopted for their radiolabeling, and (3) their application. This study can help researchers to compare different radiolabeling methods in terms of stability and efficiency and choose the best way for each nanosystem.
Collapse
Affiliation(s)
- Mahnaz Ahmadi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Emzhik
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Mosayebnia
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Niayesh Junction, Vali-E-Asr Ave, Tehran, 14155-6153, Iran.
| |
Collapse
|
16
|
Xu D, Li C, Li W, Lin B, Lv R. Recent advances in lanthanide-doped up-conversion probes for theranostics. Front Chem 2023; 11:1036715. [PMID: 36846851 PMCID: PMC9949555 DOI: 10.3389/fchem.2023.1036715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Up-conversion (or anti-Stokes) luminescence refers to the phenomenon whereby materials emit high energy, short-wavelength light upon excitation at longer wavelengths. Lanthanide-doped up-conversion nanoparticles (Ln-UCNPs) are widely used in biomedicine due to their excellent physical and chemical properties such as high penetration depth, low damage threshold and light conversion ability. Here, the latest developments in the synthesis and application of Ln-UCNPs are reviewed. First, methods used to synthesize Ln-UCNPs are introduced, and four strategies for enhancing up-conversion luminescence are analyzed, followed by an overview of the applications in phototherapy, bioimaging and biosensing. Finally, the challenges and future prospects of Ln-UCNPs are summarized.
Collapse
Affiliation(s)
| | | | | | - Bi Lin
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, China
| | | |
Collapse
|
17
|
Mettenbrink EM, Yang W, Wilhelm S. Bioimaging with Upconversion Nanoparticles. ADVANCED PHOTONICS RESEARCH 2022; 3:2200098. [PMID: 36686152 PMCID: PMC9858112 DOI: 10.1002/adpr.202200098] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Bioimaging enables the spatiotemporal visualization of biological processes at various scales empowered by a range of different imaging modalities and contrast agents. Upconversion nanoparticles (UCNPs) represent a distinct type of such contrast agents with the potential to transform bioimaging due to their unique optical properties and functional design flexibilities. This review explores and discusses the opportunities, challenges, and limitations that UCNPs exhibit as bioimaging probes and highlights applications with spatial dimensions ranging from the single nanoparticle level to cellular, tissue, and whole animal imaging. We further summarized recent advancements in bioimaging applications enabled by UCNPs, including super-resolution techniques and multimodal imaging methods, and provide a perspective on the future potential of UCNP-based technologies in bioimaging research and clinical translation. This review may provide a valuable resource for researchers interested in exploring and applying UCNP-based bioimaging technologies.
Collapse
Affiliation(s)
- Evan M. Mettenbrink
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), University of Oklahoma, Norman, Oklahoma, 73019, USA
| |
Collapse
|
18
|
Gerelkhuu Z, Lee YI, Yoon TH. Upconversion Nanomaterials in Bioimaging and Biosensor Applications and Their Biological Response. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3470. [PMID: 36234598 PMCID: PMC9565472 DOI: 10.3390/nano12193470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/16/2022] [Accepted: 09/30/2022] [Indexed: 06/16/2023]
Abstract
In recent decades, upconversion nanomaterials (UCNMs) have attracted considerable research interest because of their unique optical properties, such as large anti-Stokes shifts, sharp emissions, non-photobleaching, and long lifetime. These unique properties make them ideal candidates for unified applications in biomedical fields, including drug delivery, bioimaging, biosensing, and photodynamic therapy for specific cancers. This review describes the general mechanisms of upconversion, synthesis methods, and potential applications in biology and their biological responses. Additionally, the biological toxicity of UCNMs is explained and summarized with the associated intracellular association mechanisms. Finally, the prospects and future challenges of UCNMs at the clinical level in biological applications are described, along with a summary of opportunity for biological as well as clinical applications of UCNMs.
Collapse
Affiliation(s)
- Zayakhuu Gerelkhuu
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
| | - Yong-Ill Lee
- Department of Materials Convergence and System Engineering, Changwon National University, Changwon 51140, Korea
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City 71408, Vietnam
| | - Tae Hyun Yoon
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
19
|
Su J, Yao Z, Chen Z, Zhou S, Wang Z, Xia H, Liu S, Wu Y. TfR Aptamer Enhanced Blood-Brain Barrier Penetration of Biomimetic Nanocomplexes for Intracellular Transglutaminase 2 Imaging and Silencing in Glioma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203448. [PMID: 35980938 DOI: 10.1002/smll.202203448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Engineering a versatile nanocomplex integrating effective penetration of the blood-brain barrier (BBB), accurate diagnosis, and boosting therapy has always been an intractable challenge in glioblastoma multiforme (GBM). Herein, biomimetic nanocomplexes (TMPsM) for single intracellular transglutaminase 2 (TG2)-triggered self-assembly imaging and RNAi therapy for GBM are subtly developed. To prove the concept, transferrin receptor (TfR) aptamer-modified brain metastatic tumor cell membrane is prepared as the shell for dual BBB targeting capability and prolonged blood retention time. Upon targeting entering into GBM, hollow MnO2 is decomposed to release KKGKGQQ-tetraphenylethene (Pep-TPE) and siRNA. Owing to TG2 dependence, the non-emissive Pep-TPE would be self-aggregated to induce the emission turn-on in GBM that contain overexpressed TG2. The resulting aggregation-induced emission fluorescence imaging with a high signal-to-noise ratio can achieve the precise localization of the tumor and dynamic detection of TG2 activity, thereby allowing the GBM accurate diagnosis. Notably, the TG2 can be silenced by the released siRNA to cause cell apoptosis and increase chemotherapeutic sensitivity, ultimately realizing excellent antitumor efficacy. In vitro and in vivo results demonstrate that the as-prepared TMPsM indeed possess superior BBB penetration, precise diagnosis, and effective therapy of GBM. The proposed strategy may pioneer a new path for the theranostics of brain tumors.
Collapse
Affiliation(s)
- Juan Su
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Zhipeng Yao
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- The Translational Research Institute for Neurological Disorders, Department of Neurosurgery of Wannan Medical College, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Zixuan Chen
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Sisi Zhou
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Zhi Wang
- Testing and Certification, Wuxi Institute of Inspection, Wuxi, 214125, China
| | - Hongping Xia
- The Translational Research Institute for Neurological Disorders, Department of Neurosurgery of Wannan Medical College, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Yafeng Wu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| |
Collapse
|
20
|
Wu M, Chen T, Wang L, Akakuru OU, Ma X, Xu J, Hu J, Chen J, Fang Q, Wu A, Li Q. The strategy of precise targeting and in situ oxygenating for enhanced triple-negative breast cancer chemophototherapy. NANOSCALE 2022; 14:8349-8361. [PMID: 35635070 DOI: 10.1039/d2nr00985d] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The absence of effective therapeutic targets and tumor hypoxia are the main causes of failure in the treatment of triple-negative breast cancer (TNBC). Biomimetic nanotechnology and tumor microenvironment (TME) responsiveness bring hope and opportunity to address this problem. Here, we develop a core membrane nanoplatform (HM/D-I-BL) using hollow mesoporous manganese dioxide (HM) coated with a biomimetic cancer cell membrane for enhanced chemotherapy/phototherapy via the strategy of precise drug delivery and hypoxia amelioration. Cancer cell membrane modification endows HM/D-I-BL with excellent homologous targeting and immune escape performance. Cellular uptake and fluorescence imaging studies confirmed that HM/D-I-BL can be accurately delivered to tumor sites. HM/D-I-BL also features efficient in situ O2 generation in tumors upon laser irradiation, and subsequently enhanced chemotherapy/phototherapy, pointing to its usefulness as a TME-responsive nanozyme to alleviate tumor hypoxia in the presence of H2O2. In addition, HM/D-I-BL showed good fluorescence and magnetic resonance imaging performances, which offers a reliable multimodal image-guided combination tumor therapy for precision theranostics in the future. In general, this intelligent biomimetic nanoplatform with its homotypic tumor targeting, in situ alleviation of tumor hypoxia and synergetic chemophototherapy would open up a new dimension for the precision treatment of TNBC.
Collapse
Affiliation(s)
- Manxiang Wu
- Department of Radiology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315100, China.
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 Zhongguan West Road, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Tianxiang Chen
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 Zhongguan West Road, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Lianfu Wang
- Department of Radiology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315100, China.
| | - Ozioma Udochukwu Akakuru
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 Zhongguan West Road, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Xuehua Ma
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 Zhongguan West Road, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Jinshan Xu
- Department of Radiology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315100, China.
| | - Jiapeng Hu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 Zhongguan West Road, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jia Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qianlan Fang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 Zhongguan West Road, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 Zhongguan West Road, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Qiang Li
- Department of Radiology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315100, China.
| |
Collapse
|
21
|
Qian R, Wang K, Guo Y, Li H, Zhu Z, Huang X, Gong C, Gao Y, Guo R, Yang B, Wang C, Jiang D, Lan X, An R, Gao Z. Minimizing adverse effects of Cerenkov radiation induced photodynamic therapy with transformable photosensitizer-loaded nanovesicles. J Nanobiotechnology 2022; 20:203. [PMID: 35477389 PMCID: PMC9044600 DOI: 10.1186/s12951-022-01401-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/28/2022] [Indexed: 01/12/2023] Open
Abstract
Background Photodynamic therapy (PDT) is a promising antitumor strategy with fewer adverse effects and higher selectivity than conventional therapies. Recently, a series of reports have suggested that PDT induced by Cerenkov radiation (CR) (CR-PDT) has deeper tissue penetration than traditional PDT; however, the strategy of coupling radionuclides with photosensitizers may cause severe side effects. Methods We designed tumor-targeting nanoparticles (131I-EM@ALA) by loading 5-aminolevulinic acid (ALA) into an 131I-labeled exosome mimetic (EM) to achieve combined antitumor therapy. In addition to playing a radiotherapeutic role, 131I served as an internal light source for the Cerenkov radiation (CR). Results The drug-loaded nanoparticles effectively targeted tumors as confirmed by confocal imaging, flow cytometry, and small animal fluorescence imaging. In vitro and in vivo experiments demonstrated that 131I-EM@ALA produced a promising antitumor effect through the synergy of radiotherapy and CR-PDT. The nanoparticles killed tumor cells by inducing DNA damage and activating the lysosome-mitochondrial pathways. No obvious abnormalities in the hematology analyses, blood biochemistry, or histological examinations were observed during the treatment. Conclusions We successfully engineered a nanocarrier coloaded with the radionuclide 131I and a photosensitizer precursor for combined radiotherapy and PDT for the treatment of breast cancer. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01401-0.
Collapse
Affiliation(s)
- Ruijie Qian
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Kun Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yawen Guo
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaojuan Huang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Department of Nuclear Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Chengpeng Gong
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Rong Guo
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Biao Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chenyang Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Rui An
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China. .,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Zairong Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China. .,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
22
|
Ansari AA, Parchur AK, Chen G. Surface modified lanthanide upconversion nanoparticles for drug delivery, cellular uptake mechanism, and current challenges in NIR-driven therapies. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214423] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Zhu L, Zhong Y, Wu S, Yan M, Cao Y, Mou N, Wang G, Sun D, Wu W. Cell membrane camouflaged biomimetic nanoparticles: Focusing on tumor theranostics. Mater Today Bio 2022; 14:100228. [PMID: 35265826 PMCID: PMC8898969 DOI: 10.1016/j.mtbio.2022.100228] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/19/2022] [Accepted: 02/26/2022] [Indexed: 12/16/2022] Open
Abstract
Nanoparticles (NPs) modified by cell membranes represent an emerging biomimetic platform that can mimic the innate biological functions resulting from the various cell membranes in biological systems. researchers focus on constructing the cell membrane camouflaged NPs using a wide variety of cells, such as red blood cell membranes (RBC), macrophages and cancer cells. Cell membrane camouflaged NPs (CMNPs) inherit the composition of cell membranes, including specific receptors, antigens, proteins, for target delivering to the tumor, escaping immune from clearance, and prolonging the blood circulation time, etc. Combining cell membrane-derived biological functions and the NP cores acted cargo carriers to encapsulate the imaging agents, CMNPs are widely developed to apply in tumor imaging techniques, including computed tomography (CT), magnetic resonance imaging (MRI), fluorescence imaging (FL) and photoacoustic imaging (PA). Herein, in this review, we systematically summarize the superior functions of various CMNPs in tumor imaging, especially highlighting the advanced applications in different imaging techniques, which is to provide the theoretical supports for the development of precise guided imaging and tumor treatment.
Collapse
Affiliation(s)
- Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
| | - Shuai Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
| | - Meng Yan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
| | - Yu Cao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
| | - Nianlian Mou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
| | - Da Sun
- Institute of Life Sciences & Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, People's Republic of China
- Institute of Life Sciences & Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou, 325035, China
| |
Collapse
|
24
|
Arnett LP, Liu J, Zhang Y, Cho H, Lu E, Closson T, Allo B, Winnik MA. Biotinylated Lipid-Coated NaLnF 4 Nanoparticles: Demonstrating the Use of Lanthanide Nanoparticle-Based Reporters in Suspension and Imaging Mass Cytometry. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:2525-2537. [PMID: 35167296 DOI: 10.1021/acs.langmuir.1c03002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Lanthanide nanoparticles (LnNPs) have the potential to be used as high-sensitivity mass tag reporters in mass cytometry immunoassays. For this application, however, the LnNPs must be made colloidally stable in aqueous buffers, demonstrate minimal non-specific binding to cells, and have functional groups to attach antibodies or other targeting agents. One possible approach to address these requirements is by using lipid coating to modify the surface of the LnNPs. In this work, 39 nm diameter NaYF4:Yb, Er NPs (LnNPs) were coated with a lipid formulation consisting of egg sphingomyelin, 1,2-dioleoyl-sn-glycero-3-phosphocholine, 1,2-dioleoyl-3-trimethylammonium propane, cholesterol-(polyethylene glycol-600), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[biotinyl(polyethylene glycol)-2000]. The resulting biotinylated lipid-coated LnNPs were characterized by dynamic light scattering to determine the hydrodynamic size and stability in phosphate buffered saline, and the composition of the lipid coatings was quantified by liquid chromatography-tandem mass spectrometry. The specific and non-specific binding of the biotinylated lipid-coated LnNPs to a model system of functionalized polystyrene microbeads were then tested by both suspension and imaging mass cytometry. We found that targeted binding with minimal non-specific binding can be achieved with the lipid-coated LnNPs and that the lipid composition of the coating has an impact on the performance of the LnNPs as mass cytometry reporters. These results additionally establish the importance of quantifying the composition of lipid-coated nanomaterials to optimize them more effectively for their desired application.
Collapse
Affiliation(s)
- Loryn P Arnett
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 1H6, Canada
| | - Jieyi Liu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E2, Canada
| | - Yefeng Zhang
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 1H6, Canada
| | - Hyungjun Cho
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 1H6, Canada
| | - Elsa Lu
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 1H6, Canada
| | - Taunia Closson
- Fluidigm Canada Inc., 1380 Rodick Road, Markham, Ontario L3R 4G5, Canada
| | - Bedilu Allo
- Fluidigm Canada Inc., 1380 Rodick Road, Markham, Ontario L3R 4G5, Canada
| | - Mitchell A Winnik
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 1H6, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E2, Canada
| |
Collapse
|
25
|
Engineered lanthanide-doped upconversion nanoparticles for biosensing and bioimaging application. Mikrochim Acta 2022; 189:109. [PMID: 35175435 DOI: 10.1007/s00604-022-05180-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/07/2022] [Indexed: 01/26/2023]
Abstract
Various fluctuations of intracellular ions, biomolecules, and other conditions in the physiological environment play crucial roles in fundamental biological processes. These factors are of great importance for analysis in biomedical detection. Nevertheless, developments of the simple, rapid, and accurate proof for specific detection still encounter major challenges. Upconversion nanoparticles (UCNPs), which could absorb multiple low-energy near-infrared light (NIR) photon excitation and emits high-energy photons caused by anti-Stokes shift, show unique upconversion luminescence (UCL) properties, for example, sharp emission band, high physicochemical stability like near-zero photobleaching, photo blinking in biological tissues, and long luminescence lifetime. Furthermore, the NIR used for the light source to excite UCNPs enable lower photo-damage effect and deeper penetration of tissue, and in the meantime, it can avoid the auto-fluorescence and light scattering from biological tissue interference. Thus, the lanthanide-doped UCNP-based functional platform with controlled structure, crystalline phase, size, and multicolor emission has become an appropriate nanomaterial for bioapplications such as biosensing, bioimaging, drug release, and therapies. In this review, the recent progress about synthesis and biomedical applications of UCNPs related to sensing and bioimaging is summarized. Firstly, the different luminescence mechanisms of the upconversion process are presented. Secondly, four of the most common methods for synthesizing UCNPs are compared as well as the advantages and disadvantages of these synthetic routes. Meanwhile, the surface modification of lanthanide-doped UCNPs was introduced to pave the way for their biochemistry applications. Next, this review detailed the biological applications of lanthanide-doped UCNPs, particularly in bioimaging, including UCL and multi-modal imaging and biosensing (monitoring intracellular ions and biomolecules). Finally, the challenges and future perspectives in materials science and biomedical fields of UCNPs are concluded: the low quantum yield of the upconversion process should be considered when they are executed as imaging contrast agents. And the biosafety of lanthanide-doped UCNPs needs to be evaluated.
Collapse
|
26
|
Wei X, Zhao H, Huang G, Liu J, He W, Huang Q. ES-MION-Based Dual-Modality PET/MRI Probes for Acidic Tumor Microenvironment Imaging. ACS OMEGA 2022; 7:3442-3451. [PMID: 35128253 PMCID: PMC8811892 DOI: 10.1021/acsomega.1c05815] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/31/2021] [Indexed: 06/14/2023]
Abstract
Among all characteristics of the tumor microenvironment (TME), which are caused by abnormal proliferation of solid tumors, extracellular acidity is an important indicator for malignancy grading. pH-low insertion peptides (pHLIPs) are adopted to discern the acidic TME. To date, different imaging agents including fluorescent, positron emission tomography (PET), single photon emission computed tomography (SPECT), and magnetic resonance (MR) contrast agents with pHLIPs to target the acidic TME have been used to image various tumor models successfully. In this article, a PET/MRI dual-modality probe, based on extremely small magnetic iron oxide nanoparticles (ES-MIONs) with pHLIPs as a targeting unit, was proposed for the first time. In the phantom study, the probe showed relatively high r 1 relaxivity (r 1 = 1.03 mM-1 s-1), indicating that it could be used as a T1-weighted MR contrast agent. The 68Ga-radiolabeled probe was further studied in vitro and in vivo to evaluate pHLIP targeting efficacy and feasibility for PET/MRI. PET with intratumoral injection and T1-weighted MRI with intravenous injection both showed pHLIP-specific delivery of the probe. Therefore, we successfully designed and developed a radiolabeled ES-MION-based dual-modality PET/MRI agent to target the acidic tumor microenvironment. Although the accumulation of the probe in tumors with intravenous injection was not high enough to exhibit signals in the PET imaging study, our study still provides further insights into the ES-MION-based PET/MRI strategy.
Collapse
Affiliation(s)
- Xiuyan Wei
- Medical
Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haitao Zhao
- Department
of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji
Hospital, School of Medicine, Shanghai Jiao
Tong University, Shanghai 200127, China
| | - Gang Huang
- Shanghai
Key Laboratory of Molecular Imaging, Shanghai
University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianhua Liu
- Medical
Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weina He
- Medical
Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qingqing Huang
- Shanghai
Key Laboratory of Molecular Imaging, Shanghai
University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
27
|
Huang C, Guan X, Lin H, Liang L, Miao Y, Wu Y, Bao H, Wu X, Shen A, Wei M, Huang J. Efficient Photoacoustic Imaging With Biomimetic Mesoporous Silica-Based Nanoparticles. Front Bioeng Biotechnol 2021; 9:762956. [PMID: 34917596 PMCID: PMC8669651 DOI: 10.3389/fbioe.2021.762956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022] Open
Abstract
Indocyanine green (ICG), a near-infrared (NIR) fluorescent dye approved by the Food and Drug Administration (FDA), has been extensively used as a photoacoustic (PA) probe for PA imaging. However, its practical application is limited by poor photostability in water, rapid body clearance, and non-specificity. Herein, we fabricated a novel biomimetic nanoprobe by coating ICG-loaded mesoporous silica nanoparticles with the cancer cell membrane (namely, CMI) for PA imaging. This probe exhibited good dispersion, large loading efficiency, good biocompatibility, and homologous targeting ability to Hela cells in vitro. Furthermore, the in vivo and ex vivo PA imaging on Hela tumor-bearing nude mice demonstrated that CMI could accumulate in tumor tissue and display a superior PA imaging efficacy compared with free ICG. All these results demonstrated that CMI might be a promising contrast agent for PA imaging of cervical carcinoma.
Collapse
Affiliation(s)
- Chuangjia Huang
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoling Guan
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hui Lin
- Department of Oncology, Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, China
| | - Lu Liang
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yingling Miao
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yueheng Wu
- School of Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, China
| | - Huiqiong Bao
- School of Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, China
| | - Xiaodan Wu
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ao Shen
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Minyan Wei
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jionghua Huang
- Department of Cardiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
28
|
Radionuclide-Based Imaging of Breast Cancer: State of the Art. Cancers (Basel) 2021; 13:cancers13215459. [PMID: 34771622 PMCID: PMC8582396 DOI: 10.3390/cancers13215459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Breast cancer is one of the most commonly diagnosed malignant tumors, possessing high incidence and mortality rates that threaten women’s health. Thus, early and effective breast cancer diagnosis is crucial for enhancing the survival rate. Radionuclide molecular imaging displays its advantages for detecting breast cancer from a functional perspective. Noninvasive visualization of biological processes with radionuclide-labeled small metabolic compounds helps elucidate the metabolic state of breast cancer, while radionuclide-labeled ligands/antibodies for receptor-targeted radionuclide molecular imaging is sensitive and specific for visualization of the overexpressed molecular markers in breast cancer. This review focuses on the most recent developments of novel radiotracers as promising tools for early breast cancer diagnosis. Abstract Breast cancer is a malignant tumor that can affect women worldwide and endanger their health and wellbeing. Early detection of breast cancer can significantly improve the prognosis and survival rate of patients, but with traditional anatomical imagine methods, it is difficult to detect lesions before morphological changes occur. Radionuclide-based molecular imaging based on positron emission tomography (PET) and single-photon emission computed tomography (SPECT) displays its advantages for detecting breast cancer from a functional perspective. Radionuclide labeling of small metabolic compounds can be used for imaging biological processes, while radionuclide labeling of ligands/antibodies can be used for imaging receptors. Noninvasive visualization of biological processes helps elucidate the metabolic state of breast cancer, while receptor-targeted radionuclide molecular imaging is sensitive and specific for visualization of the overexpressed molecular markers in breast cancer, contributing to early diagnosis and better management of cancer patients. The rapid development of radionuclide probes aids the diagnosis of breast cancer in various aspects. These probes target metabolism, amino acid transporters, cell proliferation, hypoxia, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), gastrin-releasing peptide receptor (GRPR) and so on. This article provides an overview of the development of radionuclide molecular imaging techniques present in preclinical or clinical studies, which are used as tools for early breast cancer diagnosis.
Collapse
|
29
|
Jia L, Zhang P, Sun H, Dai Y, Liang S, Bai X, Feng L. Optimization of Nanoparticles for Smart Drug Delivery: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2790. [PMID: 34835553 PMCID: PMC8622036 DOI: 10.3390/nano11112790] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/16/2022]
Abstract
Nanoparticle delivery systems have good application prospects in the treatment of various diseases, especially in cancer treatment. The effect of drug delivery is regulated by the properties of nanoparticles. There have been many studies focusing on optimizing the structure of nanoparticles in recent years, and a series of achievements have been made. This review summarizes the optimization strategies of nanoparticles from three aspects-improving biocompatibility, increasing the targeting efficiency of nanoparticles, and improving the drug loading rate of nanoparticles-aiming to provide some theoretical reference for the subsequent drug delivery of nanoparticles.
Collapse
Affiliation(s)
- Lina Jia
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China; (L.J.); (P.Z.); (H.S.); (Y.D.); (S.L.)
| | - Peng Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China; (L.J.); (P.Z.); (H.S.); (Y.D.); (S.L.)
| | - Hongyan Sun
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China; (L.J.); (P.Z.); (H.S.); (Y.D.); (S.L.)
| | - Yuguo Dai
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China; (L.J.); (P.Z.); (H.S.); (Y.D.); (S.L.)
| | - Shuzhang Liang
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China; (L.J.); (P.Z.); (H.S.); (Y.D.); (S.L.)
| | - Xue Bai
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China; (L.J.); (P.Z.); (H.S.); (Y.D.); (S.L.)
| | - Lin Feng
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China; (L.J.); (P.Z.); (H.S.); (Y.D.); (S.L.)
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
30
|
Ansari AA, Parchur AK, Thorat ND, Chen G. New advances in pre-clinical diagnostic imaging perspectives of functionalized upconversion nanoparticle-based nanomedicine. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213971] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
31
|
Liu Z, Liang G, Zhan W. In situ Activatable Peptide-based Nanoprobes for Tumor Imaging. Chem Res Chin Univ 2021. [DOI: 10.1007/s40242-021-1181-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Ansari AA, Thakur VK, Chen G. Functionalized upconversion nanoparticles: New strategy towards FRET-based luminescence bio-sensing. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213821] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
33
|
Cell membrane cloaked nanomedicines for bio-imaging and immunotherapy of cancer: Improved pharmacokinetics, cell internalization and anticancer efficacy. J Control Release 2021; 335:130-157. [PMID: 34015400 DOI: 10.1016/j.jconrel.2021.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/13/2023]
Abstract
Despite enormous advancements in the field of oncology, the innocuous and effectual treatment of various types of malignancies remained a colossal challenge. The conventional modalities such as chemotherapy, radiotherapy, and surgery have been remained the most viable options for cancer treatment, but lacking of target-specificity, optimum safety and efficacy, and pharmacokinetic disparities are their impliable shortcomings. Though, in recent decades, numerous encroachments in the field of onco-targeted drug delivery have been adapted but several limitations (i.e., short plasma half-life, early clearance by reticuloendothelial system, immunogenicity, inadequate internalization and localization into the onco-tissues, chemoresistance, and deficient therapeutic efficacy) associated with these onco-targeted delivery systems limits their clinical viability. To abolish the aforementioned inadequacies, a promising approach has been emerged in which stealthing of synthetic nanocarriers has been attained by cloaking them into the natural cell membranes. These biomimetic nanomedicines not only retain characteristics features of the synthetic nanocarriers but also inherit the cell-membrane intrinsic functionalities. In this review, we have summarized preparation methods, mechanism of cloaking, and pharmaceutical and therapeutic superiority of cell-membrane camouflaged nanomedicines in improving the bio-imaging and immunotherapy against various types of malignancies. These pliable adaptations have revolutionized the current drug delivery strategies by optimizing the plasma circulation time, improving the permeation into the cancerous microenvironment, escaping the immune evasion and rapid clearance from the systemic circulation, minimizing the immunogenicity, and enabling the cell-cell communication via cell membrane markers of biomimetic nanomedicines. Moreover, the preeminence of cell-membrane cloaked nanomedicines in improving the bio-imaging and theranostic applications, alone or in combination with phototherapy or radiotherapy, have also been pondered.
Collapse
|
34
|
Chen X, Liu B, Tong R, Zhan L, Yin X, Luo X, Huang Y, Zhang J, He W, Wang Y. Orchestration of biomimetic membrane coating and nanotherapeutics in personalized anticancer therapy. Biomater Sci 2021; 9:590-625. [PMID: 33305765 DOI: 10.1039/d0bm01617a] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nanoparticle-based therapeutic and detectable modalities can augment anticancer efficiency, holding potential in capable target and suppressive metastases post administration. However, the individual discrepancies of the current "one-size-fits-all" strategies for anticancer nanotherapeutics have heralded the need for "personalized therapy". Benefiting from the special inherency of various cells, diverse cell membrane-coated nanoparticles (CMCNs) were established on a patient-by-patient basis, which would facilitate the personalized treatment of individual cancer patients. CMCNs in a complex microenvironment can evade the immune system and target homologous tumors with a suppressed immune response, as well as a prolonged circulation time, consequently increasing the drug accumulation at the tumor site and anticancer therapeutic efficacy. This review focuses on the emerging strategies and advances of CMCNs to synergistically integrate the merit of source cells with nanoparticulate delivery systems for the orchestration of personalized anticancer nanotherapeutics, thus discussing their rationalities in facilitating chemotherapy, imaging, immunotherapy, phototherapy, radiotherapy, sonodynamic, magnetocaloric, chemodynamic and gene therapy. Furthermore, the mechanism, challenges and opportunities of CMCNs in personalized anticancer therapy were highlighted to further boost cooperation from different fields, including materials science, chemistry, medicine, pharmacy and biology for the lab-to-clinic translation of CMCNs combined with the individual advantages of source cells and nanotherapeutics.
Collapse
Affiliation(s)
- Xuerui Chen
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Bingbing Liu
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Rongliang Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lin Zhan
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xuelian Yin
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xin Luo
- Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanan Huang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Wen He
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanli Wang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
35
|
Oladipo AO, Unuofin JO, Iku SII, Nkambule TTI, Mamba BB, Msagati TAM. Bimetallic Au@Pd nanodendrite system incorporating multimodal intracellular imaging for improved doxorubicin antitumor efficiency. Int J Pharm 2021; 602:120661. [PMID: 33933638 DOI: 10.1016/j.ijpharm.2021.120661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/19/2022]
Abstract
The sufficient accumulation of drugs is crucial for efficient treatment in a complex tumor microenvironment. Drug delivery systems (DDS) with high surface area and selective cytotoxicity present a novel approach to mitigate insufficient drug loading for improved therapeutic response. Herein, a doxorubicin-conjugated bimetallic gold-core palladium-shell nanocarrier with multiple dense arrays of branches (Au@PdNDs.PEG/DOX) was characterized and its efficacy against breast adenocarcinoma (MCF-7) and lung adenocarcinoma (A549) cells were evaluated. Enhanced darkfield and hyperspectral imaging (HSI) microscopy were used to study the intracellular uptake and accumulation of the DOX-loaded nanodendrites A fascinating data from a 3D-CytoViva fluorescence imaging technique provided information about the dynamics of localization and distribution of the nanocarrier. In vitro cytotoxicity assays indicated that Au@PdNDs.PEG/DOX inhibited the proliferative effects of MCF-7 cells at equivalent IC50 dosage compared to DOX alone. The nanocarrier triggered higher induction of apoptosis proved by a time-dependent phosphatidylserine V release, cell cycle arrest, and flow cytometry analysis. Moreover, the cell cycle phase proportion increase suggests that the enhanced apoptotic effect induced by Au@PdNDs.PEG/DOX was via a G2/M phase arrest. Thus, this study demonstrated the potential of dendritic nanoparticles to improve DOX therapeutic efficiency and plasmonic-mediated intracellular imaging as a suitable theranostic platform for deployment in nanomedicine.
Collapse
Affiliation(s)
- Adewale O Oladipo
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa.
| | - Jeremiah O Unuofin
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Solange I I Iku
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Thabo T I Nkambule
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Bhekie B Mamba
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Titus A M Msagati
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa.
| |
Collapse
|
36
|
Fang H, Gai Y, Wang S, Liu Q, Zhang X, Ye M, Tan J, Long Y, Wang K, Zhang Y, Lan X. Biomimetic oxygen delivery nanoparticles for enhancing photodynamic therapy in triple-negative breast cancer. J Nanobiotechnology 2021; 19:81. [PMID: 33743740 PMCID: PMC7981819 DOI: 10.1186/s12951-021-00827-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/09/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a kind of aggressive breast cancer with a high rate of metastasis, poor overall survival time, and a low response to targeted therapies. To improve the therapeutic efficacy and overcome the drug resistance of TNBC treatments, here we developed the cancer cell membrane-coated oxygen delivery nanoprobe, CCm-HSA-ICG-PFTBA, which can improve the hypoxia at tumor sites and enhance the therapeutic efficacy of the photodynamic therapy (PDT), resulting in relieving the tumor growth in TNBC xenografts. RESULTS The size of the CCm-HSA-ICG-PFTBA was 131.3 ± 1.08 nm. The in vitro 1O2 and ROS concentrations of the CCm-HSA-ICG-PFTBA group were both significantly higher than those of the other groups (P < 0.001). In vivo fluorescence imaging revealed that the best time window was at 24 h post-injection of the CCm-HSA-ICG-PFTBA. Both in vivo 18F-FMISO PET imaging and ex vivo immunofluorescence staining results exhibited that the tumor hypoxia was significantly improved at 24 h post-injection of the CCm-HSA-ICG-PFTBA. For in vivo PDT treatment, the tumor volume and weight of the CCm-HSA-ICG-PFTBA with NIR group were both the smallest among all the groups and significantly decreased compared to the untreated group (P < 0.01). No obvious biotoxicity was observed by the injection of CCm-HSA-ICG-PFTBA till 14 days. CONCLUSIONS By using the high oxygen solubility of perfluorocarbon (PFC) and the homologous targeting ability of cancer cell membranes, CCm-HSA-ICG-PFTBA can target tumor tissues, mitigate the hypoxia of the tumor microenvironment, and enhance the PDT efficacy in TNBC xenografts. Furthermore, the HSA, ICG, and PFC are all FDA-approved materials, which render the nanoparticles highly biocompatible and enhance the potential for clinical translation in the treatment of TNBC patients.
Collapse
Affiliation(s)
- Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Sheng Wang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Min Ye
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jianling Tan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Kuanyin Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
37
|
Kostiv U, Kučka J, Lobaz V, Kotov N, Janoušková O, Šlouf M, Krajnik B, Podhorodecki A, Francová P, Šefc L, Jirák D, Horák D. Highly colloidally stable trimodal 125I-radiolabeled PEG-neridronate-coated upconversion/magnetic bioimaging nanoprobes. Sci Rep 2020; 10:20016. [PMID: 33208804 PMCID: PMC7675969 DOI: 10.1038/s41598-020-77112-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
"All-in-one" multifunctional nanomaterials, which can be visualized simultaneously by several imaging techniques, are required for the efficient diagnosis and treatment of many serious diseases. This report addresses the design and synthesis of upconversion magnetic NaGdF4:Yb3+/Er3+(Tm3+) nanoparticles by an oleic acid-stabilized high-temperature coprecipitation of lanthanide precursors in octadec-1-ene. The nanoparticles, which emit visible or UV light under near-infrared (NIR) irradiation, were modified by in-house synthesized PEG-neridronate to facilitate their dispersibility and colloidal stability in water and bioanalytically relevant phosphate buffered saline (PBS). The cytotoxicity of the nanoparticles was determined using HeLa cells and human fibroblasts (HF). Subsequently, the particles were modified by Bolton-Hunter-neridronate and radiolabeled by 125I to monitor their biodistribution in mice using single-photon emission computed tomography (SPECT). The upconversion and the paramagnetic properties of the NaGdF4:Yb3+/Er3+(Tm3+)@PEG nanoparticles were evaluated by photoluminescence, magnetic resonance (MR) relaxometry, and magnetic resonance imaging (MRI) with 1 T and 4.7 T preclinical scanners. MRI data were obtained on phantoms with different particle concentrations and during pilot long-time in vivo observations of a mouse model. The biological and physicochemical properties of the NaGdF4:Yb3+/Er3+(Tm3+)@PEG nanoparticles make them promising as a trimodal optical/MRI/SPECT bioimaging and theranostic nanoprobe for experimental medicine.
Collapse
Affiliation(s)
- Uliana Kostiv
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Jan Kučka
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Volodymyr Lobaz
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Nikolay Kotov
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Miroslav Šlouf
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Bartosz Krajnik
- Department of Experimental Physics, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Artur Podhorodecki
- Department of Experimental Physics, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Pavla Francová
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, 120 00, Prague 2, Czech Republic
| | - Luděk Šefc
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, 120 00, Prague 2, Czech Republic
| | - Daniel Jirák
- Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 140 21, Prague 4, Czech Republic
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovská 1, 120 00, Prague 2, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic.
| |
Collapse
|
38
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
39
|
Huang Y, Li Q, Yang Q, Huang Z, Gao H, Xu Y, Liao L. WITHDRAWN: Analysis of Risk Factors for Prognosis and Infection of Child with Refractory Epilepsy Via Artificial Intelligence Neural Network Image Information. Neurosci Lett 2020:S0304-3940(20)30468-7. [PMID: 32585258 DOI: 10.1016/j.neulet.2020.135198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/14/2020] [Accepted: 06/21/2020] [Indexed: 01/08/2023]
Abstract
This article has been withdrawn at the request of the Editor-in-Chief. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Yueyan Huang
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, China
| | - Qingfeng Li
- Department of Radiology, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, China.
| | - Qian Yang
- Center for Diagnosis and Research of Pathological Diseases, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, China
| | - Zhijing Huang
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, China
| | - Hongbo Gao
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, China
| | - Yunan Xu
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, China
| | - Lianghua Liao
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, China
| |
Collapse
|