1
|
Xu G, Zhang Q, Cheng R, Qu J, Li W. Survival strategies of cancer cells: the role of macropinocytosis in nutrient acquisition, metabolic reprogramming, and therapeutic targeting. Autophagy 2025; 21:693-718. [PMID: 39817564 PMCID: PMC11925119 DOI: 10.1080/15548627.2025.2452149] [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: 10/09/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Macropinocytosis is a nonselective form of endocytosis that allows cancer cells to largely take up the extracellular fluid and its contents, including nutrients, growth factors, etc. We first elaborate meticulously on the process of macropinocytosis. Only by thoroughly understanding this entire process can we devise targeted strategies against it. We then focus on the central role of the MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) in regulating macropinocytosis, highlighting its significance as a key signaling hub where various pathways converge to control nutrient uptake and metabolic processes. The article covers a comprehensive analysis of the literature on the molecular mechanisms governing macropinocytosis, including the initiation, maturation, and recycling of macropinosomes, with an emphasis on how these processes are hijacked by cancer cells to sustain their growth. Key discussions include the potential therapeutic strategies targeting macropinocytosis, such as enhancing drug delivery via this pathway, inhibiting macropinocytosis to starve cancer cells, blocking the degradation and recycling of macropinosomes, and inducing methuosis - a form of cell death triggered by excessive macropinocytosis. Targeting macropinocytosis represents a novel and innovative approach that could significantly advance the treatment of cancers that rely on this pathway for survival. Through continuous research and innovation, we look forward to developing more effective and safer anti-cancer therapies that will bring new hope to patients.Abbreviation: AMPK: AMP-activated protein kinase; ASOs: antisense oligonucleotides; CAD: carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase; DC: dendritic cell; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; ERBB2: erb-b2 receptor tyrosine kinase 2; ESCRT: endosomal sorting complex required for transport; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; GRB2: growth factor receptor bound protein 2; LPP: lipopolyplex; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; NSCLC: non-small cell lung cancer; PADC: pancreatic ductal adenocarcinoma; PDPK1: 3-phosphoinositide dependent protein kinase 1; PI3K: phosphoinositide 3-kinase; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns(3,4,5)P3: phosphatidylinositol-(3,4,5)-trisphosphate; PtdIns(4,5)P2: phosphatidylinositol-(4,5)-bisphosphate; PTT: photothermal therapies; RAC1: Rac family small GTPase 1; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; RTKs: receptor tyrosine kinases; SREBF: sterol regulatory element binding transcription factor; TFEB: transcription factor EB; TNBC: triple-negative breast cancer; TSC2: TSC complex subunit 2; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Guoshuai Xu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Qinghong Zhang
- Emergency Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renjia Cheng
- Department of Intensive Care Medicine, The General Hospital of the Northern Theater Command of the People’s Liberation Army of China, Shenyang, Liaoning, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Wenqiang Li
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
2
|
Vozgirdaite D, Allard-Vannier E, Velge-Roussel F, Douez E, Jolivet L, Boursin F, Chourpa I, Aubrey N, Hervé-Aubert K. Metformin-encapsulating immunoliposomes conjugated with anti-TROP 2 antibody fragments for the active targeting of triple-negative breast cancer. NANOSCALE 2025; 17:4058-4072. [PMID: 39775761 DOI: 10.1039/d4nr03224a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Trophoblast cell-surface antigen 2 (TROP 2) has re-emerged as a promising biomarker in triple-negative breast cancer (TNBC), with high overexpression in many TNBC cases. However, despite its potential and approval as an antibody-drug-conjugate for TNBC treatment, TROP 2-targeted delivery systems are currently underexplored. Therefore, this study was aimed at exploiting the potential of TROP 2 targeting by encapsulating metformin (Met), an antidiabetic drug associated with tumor growth inhibitory properties, inside liposomes decorated with TROP 2-targeting single-chain variable fragments (scFvs). The optimization of scFv grafting resulted in Met-immunoliposomes with an average diameter of less than 200 nm, low polydispersity index (∼0.1), negative surface charge (<-10 mV), high Met drug loading (>150 mg g-1), and high affinity towards TROP 2 binding. Furthermore, Met-immunoliposomes were reproducible, and the scFv conjugation was stable in the presence of serum for five days. Their cellular uptake increased 4 folds in two-dimensional and 9 folds in three-dimensional TNBC models owing to the high affinity towards TROP 2 binding. Finally, it was observed that the therapeutic effect of Met in suppressing cancer cell growth and proliferation was superior when using anti-TROP 2 scFv-grafted Met-immunoliposomes, which completely stopped the spheroid growth and inhibited the expression of adenosine triphosphate. This study is one of the first reports to explore the combination of nanoparticle-based drug delivery systems to target the TROP 2 protein in TNBC, and to the best of our knowledge, this is the first report to specifically combine the use of scFvs with TROP 2 targeting to deliver therapeutics for TNBC treatment.
Collapse
Affiliation(s)
- Daiva Vozgirdaite
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
| | | | | | - Emmanuel Douez
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
- Pharmacy Department, University Hospital Center of Tours, 37200 Tours, France
| | - Louis Jolivet
- ISP UMR 1282, INRAE, BioMAP team, University of Tours, 37200 Tours, France
| | - Fanny Boursin
- ISP UMR 1282, INRAE, BioMAP team, University of Tours, 37200 Tours, France
| | - Igor Chourpa
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
| | - Nicolas Aubrey
- ISP UMR 1282, INRAE, BioMAP team, University of Tours, 37200 Tours, France
| | - Katel Hervé-Aubert
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
| |
Collapse
|
3
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
4
|
Hameedat F, Mendes BB, Conniot J, Di Filippo LD, Chorilli M, Schroeder A, Conde J, Sousa F. Engineering nanomaterials for glioblastoma nanovaccination. NATURE REVIEWS MATERIALS 2024; 9:628-642. [DOI: 10.1038/s41578-024-00684-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 01/03/2025]
|
5
|
Abdelmessih R, Xu J, Hung FR, Auguste DT. Integration of an LPAR1 Antagonist into Liposomes Enhances Their Internalization and Tumor Accumulation in an Animal Model of Human Metastatic Breast Cancer. Mol Pharm 2023; 20:5500-5514. [PMID: 37844135 PMCID: PMC10631474 DOI: 10.1021/acs.molpharmaceut.3c00348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
Lysophosphatidic acid receptor 1 (LPAR1) is elevated in breast cancer. The deregulation of LPAR1, including the function and level of expression, is linked to cancer initiation, progression, and metastasis. LPAR1 antagonists, AM095 or Ki16425, may be effective therapeutic molecules, yet their limited water solubility hinders in vivo delivery. In this study, we report on the synthesis of two liposomal formulations incorporating AM095 or Ki16425, embedded within the lipid bilayer, as targeted nanocarriers for metastatic breast cancer (MBC). The data show that the Ki16425 liposomal formulation exhibited a 50% increase in internalization by MBC mouse epithelial cells (4T1) and a 100% increase in tumor accumulation in a mouse model of MBC compared with that of a blank liposomal formulation (control). At the same time, normal mouse epithelial cells (EpH-4Ev) internalized the Ki16425 liposomal formulation 25% lesser than the control formulation. Molecular dynamics simulations show that the integration of AM095 or Ki16425 modified the physical and mechanical properties of the lipid bilayer, making it more flexible in these liposomal formulations compared with liposomes without drug. The incorporation of an LPAR1 antagonist within a liposomal drug delivery system represents a viable therapeutic approach for targeting the LPA-LPAR1 axis, which may hinder the progression of MBC.
Collapse
Affiliation(s)
- Rudolf
G. Abdelmessih
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Jiaming Xu
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Francisco R. Hung
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Debra T. Auguste
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
6
|
Martins C, Sarmento B. Multi-ligand functionalized blood-to-tumor sequential targeting strategies in the field of glioblastoma nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1893. [PMID: 37186374 DOI: 10.1002/wnan.1893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 05/17/2023]
Abstract
Glioblastoma (GBM) is an unmet clinical need characterized by a standard of care (SOC) 5-year survival rate of only 5%, and a treatment mostly palliative. Significant hurdles in GBM therapies include an effective penetration of therapeutics through the brain protective barrier, namely the blood-brain barrier (BBB), and a successful therapeutic delivery to brain-invading tumor cells post-BBB crossing. These hurdles, along with the poor prognosis and critical heterogeneity of the disease, have shifted attention to treatment modalities with capacity to precisely and sequentially target (i) BBB cells, inducing blood-to-brain transport, and (ii) GBM cells, leading to a higher therapeutic accumulation at the tumor site. This sequential targeting allows therapeutic molecules to reach the brain parenchyma and compromise molecular processes that support tumor cell invasion. Besides improving formulation and pharmacokinetics constraints of drugs, nanomedicines offer the possibility of being surface functionalized with multiple possibilities of targeting ligands, while delivering the desired therapeutic cargos to the biological sites of interest. Targeting ligands exploit the site-specific expression or overexpression of specific molecules on BBB and GBM cells, triggering brain plus tumor transport. Since the efficacy of single-ligand functionalized nanomedicines is limited due to the GBM anatomical site (brain) and disease complexity, this review presents an overview of multi-ligand functionalized, BBB and GBM sequentially- and dual-targeted nanomedicines reported in literature over the last 10 years. The role of the BBB in GBM progression, treatment options, and the multiple possibilities of currently available targeting ligands will be summarized. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- IUCS-CESPU, Gandra, Portugal
| |
Collapse
|
7
|
Sanati M, Afshari AR, Aminyavari S, Kesharwani P, Jamialahmadi T, Sahebkar A. RGD-engineered nanoparticles as an innovative drug delivery system in cancer therapy. J Drug Deliv Sci Technol 2023; 84:104562. [DOI: 10.1016/j.jddst.2023.104562] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
|
8
|
Di Filippo LD, de Carvalho SG, Duarte JL, Luiz MT, Paes Dutra JA, de Paula GA, Chorilli M, Conde J. A receptor-mediated landscape of druggable and targeted nanomaterials for gliomas. Mater Today Bio 2023; 20:100671. [PMID: 37273792 PMCID: PMC10238751 DOI: 10.1016/j.mtbio.2023.100671] [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: 02/02/2023] [Revised: 04/13/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Gliomas are the most common type of brain cancer, and among them, glioblastoma multiforme (GBM) is the most prevalent (about 60% of cases) and the most aggressive type of primary brain tumor. The treatment of GBM is a major challenge due to the pathophysiological characteristics of the disease, such as the presence of the blood-brain barrier (BBB), which prevents and regulates the passage of substances from the bloodstream to the brain parenchyma, making many of the chemotherapeutics currently available not able to reach the brain in therapeutic concentrations, accumulating in non-target organs, and causing considerable adverse effects for the patient. In this scenario, nanocarriers emerge as tools capable of improving the brain bioavailability of chemotherapeutics, in addition to improving their biodistribution and enhancing their uptake in GBM cells. This is possible due to its nanometric size and surface modification strategies, which can actively target nanocarriers to elements overexpressed by GBM cells (such as transmembrane receptors) related to aggressive development, drug resistance, and poor prognosis. In this review, an overview of the most frequently overexpressed receptors in GBM cells and possible approaches to chemotherapeutic delivery and active targeting using nanocarriers will be presented.
Collapse
Affiliation(s)
| | | | - Jonatas Lobato Duarte
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Marcela Tavares Luiz
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | | | - Geanne Aparecida de Paula
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
9
|
Gareev K, Tagaeva R, Bobkov D, Yudintceva N, Goncharova D, Combs SE, Ten A, Samochernych K, Shevtsov M. Passing of Nanocarriers across the Histohematic Barriers: Current Approaches for Tumor Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1140. [PMID: 37049234 PMCID: PMC10096980 DOI: 10.3390/nano13071140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
Over the past several decades, nanocarriers have demonstrated diagnostic and therapeutic (i.e., theranostic) potencies in translational oncology, and some agents have been further translated into clinical trials. However, the practical application of nanoparticle-based medicine in living organisms is limited by physiological barriers (blood-tissue barriers), which significantly hampers the transport of nanoparticles from the blood into the tumor tissue. This review focuses on several approaches that facilitate the translocation of nanoparticles across blood-tissue barriers (BTBs) to efficiently accumulate in the tumor. To overcome the challenge of BTBs, several methods have been proposed, including the functionalization of particle surfaces with cell-penetrating peptides (e.g., TAT, SynB1, penetratin, R8, RGD, angiopep-2), which increases the passing of particles across tissue barriers. Another promising strategy could be based either on the application of various chemical agents (e.g., efflux pump inhibitors, disruptors of tight junctions, etc.) or physical methods (e.g., magnetic field, electroporation, photoacoustic cavitation, etc.), which have been shown to further increase the permeability of barriers.
Collapse
Affiliation(s)
- Kamil Gareev
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia
| | - Ruslana Tagaeva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Danila Bobkov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Daria Goncharova
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
| | - Artem Ten
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Konstantin Samochernych
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| |
Collapse
|
10
|
Na Y, Zhang N, Zhong X, Gu J, Yan C, Yin S, Lei X, Zhao J, Geng F. Polylactic-co-glycolic acid-based nanoparticles modified with peptides and other linkers cross the blood-brain barrier for targeted drug delivery. Nanomedicine (Lond) 2023; 18:125-143. [PMID: 36916394 DOI: 10.2217/nnm-2022-0287] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Because of the blood-brain barrier, only a limited fraction of drugs can penetrate the brain. As a result, there is a need to take larger doses of the drug, which may result in numerous undesirable side effects. Over the past few decades, a plethora of research has been conducted to address this issue. In recent years, the field of nanomedicine research has reported promising findings. Currently, numerous types of polylactic-co-glycolic acid-based drug-delivery systems are being studied, and great progress has been made in the modification of their surfaces with a variety of ligands. In this review, the authors highlight the preparation of polylactic-co-glycolic acid-based nanoparticles and single- and dual-targeted peptide modifications for site-specific drug delivery into the brain.
Collapse
Affiliation(s)
- Yue Na
- Key Laboratory of Photochemistry Biomaterials & Energy Storage Materials of Heilongjiang Province, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang, 150025, China
| | - Ning Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China.,Wuxi Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, 214071, China
| | - Xinyu Zhong
- Key Laboratory of Photochemistry Biomaterials & Energy Storage Materials of Heilongjiang Province, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang, 150025, China
| | - Jinlian Gu
- Key Laboratory of Photochemistry Biomaterials & Energy Storage Materials of Heilongjiang Province, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang, 150025, China
| | - Chang Yan
- Key Laboratory of Photochemistry Biomaterials & Energy Storage Materials of Heilongjiang Province, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang, 150025, China
| | - Shun Yin
- Key Laboratory of Photochemistry Biomaterials & Energy Storage Materials of Heilongjiang Province, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang, 150025, China
| | - Xia Lei
- Wuxi Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, 214071, China
| | - Jihui Zhao
- College of Pharmacy, Hunan University of Medicine, Huaihua, Hunan, 418000, China
| | - Fang Geng
- Key Laboratory of Photochemistry Biomaterials & Energy Storage Materials of Heilongjiang Province, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang, 150025, China
| |
Collapse
|
11
|
Fuloria S, Subramaniyan V, Gupta G, Sekar M, Meenakshi DU, Sathasivam K, Sudhakar K, Alharbi KS, Almutairi SS, Almalki WH, Fuloria NK. Detection of Circulating Tumor Cells and Epithelial Progenitor Cells: A Comprehensive Study. J Environ Pathol Toxicol Oncol 2023; 42:1-29. [PMID: 37017676 DOI: 10.1615/jenvironpatholtoxicoloncol.2022044456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Technological advancement to enhance tumor cells (TC) has allowed discovery of various cellular bio-markers: cancer stem cells (CSC), circulating tumor cells (CTC), and endothelial progenitor cells (EPC). These are responsible for resistance, metastasis, and premetastatic conditions of cancer. Detection of CSC, CTC, and EPC assists in early diagnosis, recurrence prediction, and treatment efficacy. This review describes various methods to detect TC subpopulations such as in vivo assays (sphere-forming, serial dilution, and serial transplantation), in vitro assays (colony-forming cells, microsphere, side-population, surface antigen staining, aldehyde dehydrogenase activity, and Paul Karl Horan label-retaining cells, surface markers, nonenriched and enriched detection), reporter systems, and other analytical methods (flow cytometry, fluorescence microscopy/spectroscopy, etc.). The detailed information on methods to detect CSC, CTC, and EPC in this review will assist investigators in successful prognosis, diagnosis, and cancer treatment with greater ease.
Collapse
Affiliation(s)
- Shivkanya Fuloria
- Faculty of Pharmacy /Centre of Excellence for Biomaterials Engineering, AIMST University, Kedah 08100, Malaysia
| | - Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Bandar Saujana Putra, 42610 Jenjarom Selangor, Malaysia
| | - Gaurav Gupta
- Department of Pharmacology, Suresh GyanVihar University, Jagatpura, Jaipur, India; Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical Sciences, Saveetha University, Chennai, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Perak, Malaysia
| | | | | | - Kalvatala Sudhakar
- School of Pharmaceutical Sciences (LIT-Pharmacy), Lovely Professional University, Jalandhar 144411, India
| | - Khalid Saad Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | | | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Neeraj Kumar Fuloria
- Faculty of Pharmacy/Centre of Excellence for Biomaterials Engineering, AIMST University, Kedah 08100, Malaysia
| |
Collapse
|
12
|
Alhaj-Suliman SO, Wafa EI, Salem AK. Engineering nanosystems to overcome barriers to cancer diagnosis and treatment. Adv Drug Deliv Rev 2022; 189:114482. [PMID: 35944587 DOI: 10.1016/j.addr.2022.114482] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 01/24/2023]
Abstract
Over the past two decades, multidisciplinary investigations into the development of nanoparticles for medical applications have continually increased. However, nanoparticles are still subject to biological barriers and biodistribution challenges, which limit their overall clinical potential. This has motivated the implementation of innovational modifications to a range of nanoparticle formulations designed for cancer imaging and/or cancer treatment to overcome specific barriers and shift the accumulation of payloads toward the diseased tissues. In recent years, novel technological and chemical approaches have been employed to modify or functionalize the surface of nanoparticles or manipulate the characteristics of nanoparticles. Combining these approaches with the identification of critical biomarkers provides new strategies for enhancing nanoparticle specificity for both cancer diagnostic and therapeutic applications. This review discusses the most recent advances in the design and engineering of nanoparticles as well as future directions for developing the next generation of nanomedicines.
Collapse
Affiliation(s)
- Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, United States.
| |
Collapse
|
13
|
Cong VT, Houng JL, Kavallaris M, Chen X, Tilley RD, Gooding JJ. How can we use the endocytosis pathways to design nanoparticle drug-delivery vehicles to target cancer cells over healthy cells? Chem Soc Rev 2022; 51:7531-7559. [PMID: 35938511 DOI: 10.1039/d1cs00707f] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Targeted drug delivery in cancer typically focuses on maximising the endocytosis of drugs into the diseased cells. However, there has been less focus on exploiting the differences in the endocytosis pathways of cancer cells versus non-cancer cells. An understanding of the endocytosis pathways in both cancer and non-cancer cells allows for the design of nanoparticles to deliver drugs to cancer cells whilst restricting healthy cells from taking up anticancer drugs, thus efficiently killing the cancer cells. Herein we compare the differences in the endocytosis pathways of cancer and healthy cells. Second, we highlight the importance of the physicochemical properties of nanoparticles (size, shape, stiffness, and surface chemistry) on cellular uptake and how they can be adjusted to selectively target the dominated endocytosis pathway of cancer cells over healthy cells and to deliver anticancer drug to the target cells. The review generates new thought in the design of cancer-selective nanoparticles based on the endocytosis pathways.
Collapse
Affiliation(s)
- Vu Thanh Cong
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jacinta L Houng
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maria Kavallaris
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.,Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia.,School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, China
| | - Richard D Tilley
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| | - J Justin Gooding
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
14
|
Madani F, Esnaashari SS, Webster TJ, Khosravani M, Adabi M. Polymeric nanoparticles for drug delivery in glioblastoma: State of the art and future perspectives. J Control Release 2022; 349:649-661. [PMID: 35878729 DOI: 10.1016/j.jconrel.2022.07.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is an aggressive, fatal and malignant primary brain tumor. Despite the current standard treatment for glioblastoma patients including neurosurgical resection, followed by concomitant radiation and chemotherapy, the median survival rate is only about 15 months. An unresolved challenge for current therapies is related to getting drugs through the blood-brain barrier (BBB), which hinders many chemotherapeutic agents from reaching tumors cells. Although a large amount of research has been done to circumvent the BBB and deliver drugs to the brain, with nanoparticles (NPs) taking the lead, the challenge is still high. In this regard, the BBB and how to transfer drug pathways through the BBB, especially using NPs, are introduced here. Afterwards, the latest advances in drug delivery, co-drug delivery, and combination modalities are described specifically for GBM treatments using natural and synthetic polymeric NPs and adjuvant therapies including hyperthermia, photodynamic therapy and also ketogenic regimens. In addition, receptor-mediated endocytosis agents that exist in endothelial capillary cells of the brain are explained. Lastly, future directions to finally deliver drugs through the BBB for GBM treatment are emphasized. It is the hope that this review can provide a number of practical pathways for the future development of BBB permeable nanochemotherapeutics against GBM.
Collapse
Affiliation(s)
- Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Sara Esnaashari
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, China
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Vallet-Regí M, Schüth F, Lozano D, Colilla M, Manzano M. Engineering mesoporous silica nanoparticles for drug delivery: where are we after two decades? Chem Soc Rev 2022; 51:5365-5451. [PMID: 35642539 PMCID: PMC9252171 DOI: 10.1039/d1cs00659b] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The present review details a chronological description of the events that took place during the development of mesoporous materials, their different synthetic routes and their use as drug delivery systems. The outstanding textural properties of these materials quickly inspired their translation to the nanoscale dimension leading to mesoporous silica nanoparticles (MSNs). The different aspects of introducing pharmaceutical agents into the pores of these nanocarriers, together with their possible biodistribution and clearance routes, would be described here. The development of smart nanocarriers that are able to release a high local concentration of the therapeutic cargo on-demand after the application of certain stimuli would be reviewed here, together with their ability to deliver the therapeutic cargo to precise locations in the body. The huge progress in the design and development of MSNs for biomedical applications, including the potential treatment of different diseases, during the last 20 years will be collated here, together with the required work that still needs to be done to achieve the clinical translation of these materials. This review was conceived to stand out from past reports since it aims to tell the story of the development of mesoporous materials and their use as drug delivery systems by some of the story makers, who could be considered to be among the pioneers in this area.
Collapse
Affiliation(s)
- María Vallet-Regí
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Ferdi Schüth
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Daniel Lozano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Montserrat Colilla
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Miguel Manzano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
16
|
Taghipour YD, Zarebkohan A, Salehi R, Rahimi F, Torchilin VP, Hamblin MR, Seifalian A. An update on dual targeting strategy for cancer treatment. J Control Release 2022; 349:67-96. [PMID: 35779656 DOI: 10.1016/j.jconrel.2022.06.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 12/18/2022]
Abstract
The key issue in the treatment of solid tumors is the lack of efficient strategies for the targeted delivery and accumulation of therapeutic cargoes in the tumor microenvironment (TME). Targeting approaches are designed for more efficient delivery of therapeutic agents to cancer cells while minimizing drug toxicity to normal cells and off-targeting effects, while maximizing the eradication of cancer cells. The highly complicated interrelationship between the physicochemical properties of nanoparticles, and the physiological and pathological barriers that are required to cross, dictates the need for the success of targeting strategies. Dual targeting is an approach that uses both purely biological strategies and physicochemical responsive smart delivery strategies to increase the accumulation of nanoparticles within the TME and improve targeting efficiency towards cancer cells. In both approaches, either one single ligand is used for targeting a single receptor on different cells, or two different ligands for targeting two different receptors on the same or different cells. Smart delivery strategies are able to respond to triggers that are typical of specific disease sites, such as pH, certain specific enzymes, or redox conditions. These strategies are expected to lead to more precise targeting and better accumulation of nano-therapeutics. This review describes the classification and principles of dual targeting approaches and critically reviews the efficiency of dual targeting strategies, and the rationale behind the choice of ligands. We focus on new approaches for smart drug delivery in which synthetic and/or biological moieties are attached to nanoparticles by TME-specific responsive linkers and advanced camouflaged nanoparticles.
Collapse
Affiliation(s)
- Yasamin Davatgaran Taghipour
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fariborz Rahimi
- Department of Electrical Engineering, University of Bonab, Bonab, Iran
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine and Department of Chemical Engineering, Northeastern University, Boston, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, South Africa
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
17
|
Zhang W, Chen X, Ding D, Zhang G, Zhu Z, Yang X, Li M, Liang L, Shi X, Wang T, Gao R. Real-time in vivo imaging reveals specific nanoparticle target binding in a syngeneic glioma mouse model. NANOSCALE 2022; 14:5678-5688. [PMID: 35195122 DOI: 10.1039/d1nr07591h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanomaterial-based drug delivery is a promising strategy for glioma treatment. However, the detailed dynamics of nanoparticles in solid glioma are still a mystery, including their intratumoral infiltration depth, penetration, retention time, and distribution. Revealing these processes in detail requires repeated intravital imaging of the corresponding brain tumor regions over time during glioma growth. Hereby, we established a syngeneic orthotopic cerebral glioma mouse model by combining the chronic cranial window and two-photon microscopy. Thus, we were able to investigate the dynamics of the nanoparticles during long-term glioma growth. Three hours after the intravenous (i.v.) injection of integrin αVβ3 binding conjugated silicon nanoparticles (SNPs-PEG-RGD-FITC), green nanoparticles had already infiltrated the brain glioma, and then more nanoparticles penetrated into the solid brain tumor and were retained for at least 8 days. However, the amount of control SNPs-PEG-FITC that infiltrated into the solid brain tumor was very low. Moreover, we found that SNPs-PEG-RGD-FITC were not only located in the tumor border but could also infiltrate into the core region of the solid tumor. In vitro assay also confirmed the high binding affinity between GL-261-Tdtomato cells and SNPs-PEG-RGD-FITC. Our results indicate that SNPs-PEG-RGD-FITC has high penetration and retention in a solid glioma and our model provides novel ideas for the investigation of nanoparticle dynamics in brain tumors.
Collapse
Affiliation(s)
- Wenlong Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - Xiangyu Chen
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, 130015, China
| | - Dong Ding
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Guoxin Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - Ziwei Zhu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - XingJiu Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - Mengyuan Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - Lijun Liang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, China
| | - Xudong Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - Tie Wang
- Life and Health Research Institute, Tianjin University of Technology, Tianjin, 300072, China
| | - Ran Gao
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| |
Collapse
|
18
|
Recent advances in the therapeutic strategies of glioblastoma multiforme. Neuroscience 2022; 491:240-270. [PMID: 35395355 DOI: 10.1016/j.neuroscience.2022.03.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most common, most formidable, and deadliest malignant types of primary astrocytoma with a poor prognosis. At present, the standard of care includes surgical tumor resection, followed by radiation therapy concomitant with chemotherapy and temozolomide. New developments and significant advances in the treatment of GBM have been achieved in recent decades. However, despite the advances, recurrence is often inevitable, and the survival of patients remains low. Various factors contribute to the difficulty in identifying an effective therapeutic option, among which are tumor complexity, the presence of the blood-brain barrier (BBB), and the presence of GBM cancer stem cells, prompting the need for improving existing treatment approaches and investigating new treatment alternatives for ameliorating the treatment strategies of GBM. In this review, we outline some of the most recent literature on the various available treatment options such as surgery, radiotherapy, cytotoxic chemotherapy, gene therapy, immunotherapy, phototherapy, nanotherapy, and tumor treating fields in the treatment of GBM, and we list some of the potential future directions of GBM. The reviewed studies confirm that GBM is a sophisticated disease with several challenges for scientists to address. Hence, more studies and a multimodal therapeutic approach are crucial to yield an effective cure and prolong the survival of GBM patients.
Collapse
|
19
|
Li Y, Shen C, Liang X, Deng K, Zeng Z, Xu X. Dynamic-Responsive Virus-Mimetic Nanocapsules Facilitate Protein Drug Penetration and Extracellular-Specific Unpacking for Antitumor Treatment. Biomater Sci 2022; 10:3447-3453. [DOI: 10.1039/d2bm00500j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Protein-based drugs have been demonstrating great potential on the treatment of various diseases, but most of them encounter many difficulties in clinical trials or uses, such as instability, low bioavailability,...
Collapse
|
20
|
Ribovski L, Hamelmann NM, Paulusse JMJ. Polymeric Nanoparticles Properties and Brain Delivery. Pharmaceutics 2021; 13:2045. [PMID: 34959326 PMCID: PMC8705716 DOI: 10.3390/pharmaceutics13122045] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 01/04/2023] Open
Abstract
Safe and reliable entry to the brain is essential for successful diagnosis and treatment of diseases, but it still poses major challenges. As a result, many therapeutic approaches to treating disorders associated with the central nervous system (CNS) still only show limited success. Nano-sized systems are being explored as drug carriers and show great improvements in the delivery of many therapeutics. The systemic delivery of nanoparticles (NPs) or nanocarriers (NCs) to the brain involves reaching the neurovascular unit (NVU), being transported across the blood-brain barrier, (BBB) and accumulating in the brain. Each of these steps can benefit from specifically controlled properties of NPs. Here, we discuss how brain delivery by NPs can benefit from careful design of the NP properties. Properties such as size, charge, shape, and ligand functionalization are commonly addressed in the literature; however, properties such as ligand density, linker length, avidity, protein corona, and stiffness are insufficiently discussed. This is unfortunate since they present great value against multiple barriers encountered by the NPs before reaching the brain, particularly the BBB. We further highlight important examples utilizing targeting ligands and how functionalization parameters, e.g., ligand density and ligand properties, can affect the success of the nano-based delivery system.
Collapse
Affiliation(s)
| | | | - Jos M. J. Paulusse
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands; (L.R.); (N.M.H.)
| |
Collapse
|
21
|
Li W, Little N, Park J, Foster CA, Chen J, Lu J. Tumor-Associated Fibroblast-Targeting Nanoparticles for Enhancing Solid Tumor Therapy: Progress and Challenges. Mol Pharm 2021; 18:2889-2905. [PMID: 34260250 DOI: 10.1021/acs.molpharmaceut.1c00455] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Even though nanoparticle drug delivery systems (nanoDDSs) have improved antitumor efficacy by delivering more drugs to tumor sites compared to free and unencapsulated therapeutics, achieving satisfactory distribution and penetration of nanoDDSs inside solid tumors, especially in stromal fibrous tumors, remains challenging. As one of the most common stromal cells in solid tumors, tumor-associated fibroblasts (TAFs) not only promote tumor growth and metastasis but also reduce the drug delivery efficiency of nanoparticles through the tumor's inherent physical and physiological barriers. Thus, TAFs have been emerging as attractive targets, and TAF-targeting nanotherapeutics have been extensively explored to enhance the tumor delivery efficiency and efficacy of various anticancer agents. The purpose of this Review is to opportunely summarize the underlying mechanisms of TAFs on obstructing nanoparticle-mediated drug delivery into tumors and discuss the current advances of a plethora of nanotherapeutic approaches for effectively targeting TAFs.
Collapse
Affiliation(s)
- Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Nicholas Little
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jonghan Park
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Cole Alexander Foster
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jiawei Chen
- Michigan Institute for Clinical & Health Research, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States.,BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States.,NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, Arizona 85721, United States.,Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
22
|
Sub‐50 nm Supramolecular Nanohybrids with Active Targeting Corona for Image‐Guided Solid Tumor Treatment and Metastasis Inhibition. ADVANCED FUNCTIONAL MATERIALS 2021. [DOI: 10.1002/adfm.202103272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
23
|
Liu M, Ma W, Zhao D, Li J, Li Q, Liu Y, Hao L, Lin Y. Enhanced Penetrability of a Tetrahedral Framework Nucleic Acid by Modification with iRGD for DOX-Targeted Delivery to Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:25825-25835. [PMID: 34038071 DOI: 10.1021/acsami.1c07297] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Poor penetrability and nonselective distribution of chemotherapeutic drugs are the main obstacles for chemotherapy for triple-negative breast cancer (TNBC). In our work, we developed a DNA-based drug delivery system to surmount these barriers. In addition, a tetrahedral framework nucleic acid (tFNA) was employed to load doxorubicin (DOX) with iRGD decoration to form a novel nanoparticle (tFNA/DOX@iRGD). The RGD sequence and the CendR motif in iRGD are used in tumor targeting and tissue penetration, respectively. Based on the sustained serum stability and pH-sensitive release behavior of DOX, tFNA/DOX@iRGD exhibited superiority for biomedical application. Moreover, tFNA/DOX@iRGD showed excellent deep penetration and drug accumulation in three-dimensional (3D) multicellular tumor spheroids compared to DOX and tFNA/DOX. Additionally, the therapeutic effect was verified in a 4T1 subcutaneous tumor model, and the complexes displayed a superior antitumor and antiangiogenic efficiency with fewer collateral damages. Therefore, these findings suggested that tFNA/DOX@iRGD might be a more effective pattern for drug delivery and TNBC therapy.
Collapse
Affiliation(s)
- Mengting Liu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Wenjuan Ma
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Dan Zhao
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Jiajie Li
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Qirong Li
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Yuhao Liu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Liying Hao
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,College of Biomedical Engineering, Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
24
|
Wiwatchaitawee K, Quarterman JC, Geary SM, Salem AK. Enhancement of Therapies for Glioblastoma (GBM) Using Nanoparticle-based Delivery Systems. AAPS PharmSciTech 2021; 22:71. [PMID: 33575970 DOI: 10.1208/s12249-021-01928-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of malignant brain tumor. Current FDA-approved treatments include surgical resection, radiation, and chemotherapy, while hyperthermia, immunotherapy, and most relevantly, nanoparticle (NP)-mediated delivery systems or combinations thereof have shown promise in preclinical studies. Drug-carrying NPs are a promising approach to brain delivery as a result of their potential to facilitate the crossing of the blood-brain barrier (BBB) via two main types of transcytosis mechanisms: adsorptive-mediated transcytosis (AMT) and receptor-mediated transcytosis (RMT). Their ability to accumulate in the brain can thus provide local sustained release of tumoricidal drugs at or near the site of GBM tumors. NP-based drug delivery has the potential to significantly reduce drug-related toxicity, increase specificity, and consequently improve the lifespan and quality of life of patients with GBM. Due to significant advances in the understanding of the molecular etiology and pathology of GBM, the efficacy of drugs loaded into vectors targeting this disease has increased in both preclinical and clinical settings. Multitargeting NPs, such as those incorporating multiple specific targeting ligands, are an innovative technology that can lead to decreased off-target effects while simultaneously having increased accumulation and action specifically at the tumor site. Targeting ligands can include antibodies, or fragments thereof, and peptides or small molecules, which can result in a more controlled drug delivery system compared to conventional drug treatments. This review focuses on GBM treatment strategies, summarizing current options and providing a detailed account of preclinical findings with prospective NP-based approaches aimed at improving tumor targeting and enhancing therapeutic outcomes for GBM patients.
Collapse
|
25
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
26
|
Ogawa K, Kato N, Kawakami S. Recent Strategies for Targeted Brain Drug Delivery. Chem Pharm Bull (Tokyo) 2021; 68:567-582. [PMID: 32611994 DOI: 10.1248/cpb.c20-00041] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Because the brain is the most important human organ, many brain disorders can cause severe symptoms. For example, glioma, one type of brain tumor, is progressive and lethal, while neurodegenerative diseases cause severe disability. Nevertheless, medical treatment for brain diseases remains unsatisfactory, and therefore innovative therapies are desired. However, the development of therapies to treat some cerebral diseases is difficult because the blood-brain barrier (BBB) or blood-brain tumor barrier prevents drugs from entering the brain. Hence, drug delivery system (DDS) strategies are required to deliver therapeutic agents to the brain. Recently, brain-targeted DDS have been developed, which increases the quality of therapy for cerebral disorders. This review gives an overview of recent brain-targeting DDS strategies. First, it describes strategies to cross the BBB. This includes BBB-crossing ligand modification or temporal BBB permeabilization. Strategies to avoid the BBB using local administration are also summarized. Intrabrain drug distribution is a crucial factor that directly determines the therapeutic effect, and thus it is important to evaluate drug distribution using optimal methods. We introduce some methods for evaluating drug distribution in the brain. Finally, applications of brain-targeted DDS for the treatment of brain tumors, Alzheimer's disease, Parkinson's disease, and stroke are explained.
Collapse
Affiliation(s)
- Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Naoya Kato
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| |
Collapse
|
27
|
Wu QL, Xu HL, Xiong C, Lan QH, Fang ML, Cai JH, Li H, Zhu ST, Xu JH, Tao FY, Lu CT, Zhao YZ, Chen B. c(RGDyk)-modified nanoparticles encapsulating quantum dots as a stable fluorescence probe for imaging-guided surgical resection of glioma under the auxiliary UTMD. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 48:143-158. [PMID: 32207347 DOI: 10.1080/21691401.2019.1699821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Surgical resection remains the preferred approach for some patients with glioblastoma (GBM), and eradication of the residual tumour niche after surgical resection is very helpful for prolonging patient survival. However, complete surgical resection of invasive GBM is difficult because of its ambiguous boundary. Herein, a novel targeting material, c(RGDyk)-poloxamer-188, was synthesized by modifying carboxyl-terminated poloxamer-188 with a glioma-targeting cyclopeptide, c(RGDyk). Quantum dots (QDs) as fluorescent probe were encapsulated into the self-assembled c(RGDyk)-poloxamer-188 polymer nanoparticles (NPs) to construct glioma-targeted QDs-c(RGDyk)NP for imaging-guided surgical resection of GBM. QDs-c(RGDyk)NP exhibited a moderate hydrodynamic diameter of 212.4 nm, a negative zeta potential of -10.1 mV and good stability. QDs-c(RGDyk)NP exhibited significantly lower toxicity against PC12 and C6 cells and HUVECs than free QDs. Moreover, in vitro cellular uptake experiments demonstrated that QDs-c(RGDyk)NP specifically targeted C6 cells, making them display strong fluorescence. Combined with ultrasound-targeted microbubble destruction (UTMD), QDs-c(RGDyk)NP specifically accumulated in glioma tissue in orthotropic tumour rats after intravenous administration, evidenced by ex vivo NIR fluorescence imaging of bulk brain and glioma tissue sections. Furthermore, fluorescence imaging with QDs-c(RGDyk)NP guided accurate surgical resection of glioma. Finally, the safety of QDs-c(RGDyk)NP was verified using pathological HE staining. In conclusion, QDs-c(RGDyk)NP may be a potential imaging probe for imaging-guided surgery.
Collapse
Affiliation(s)
- Qi-Long Wu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - He-Lin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Cui Xiong
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Qing-Hua Lan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Ming-Ling Fang
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Jin-Hua Cai
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Hui Li
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Shu-Ting Zhu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Jing-Hong Xu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Fang-Yi Tao
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Cui-Tao Lu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Ying-Zheng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Bin Chen
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| |
Collapse
|
28
|
Bi Q, Song X, Hu A, Luo T, Jin R, Ai H, Nie Y. Magnetofection: Magic magnetic nanoparticles for efficient gene delivery. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.07.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Van Zundert I, Fortuni B, Rocha S. From 2D to 3D Cancer Cell Models-The Enigmas of Drug Delivery Research. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2236. [PMID: 33187231 PMCID: PMC7696259 DOI: 10.3390/nano10112236] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/30/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023]
Abstract
Over the past decades, research has made impressive breakthroughs towards drug delivery systems, resulting in a wide range of multifunctional engineered nanoparticles with biomedical applications such as cancer therapy. Despite these significant advances, well-designed nanoparticles rarely reach the clinical stage. Promising results obtained in standard 2D cell culture systems often turn into disappointing outcomes in in vivo models. Although the overall majority of in vitro nanoparticle research is still performed on 2D monolayer cultures, more and more researchers started acknowledging the importance of using 3D cell culture systems, as better models for mimicking the in vivo tumor physiology. In this review, we provide a comprehensive overview of the 3D cancer cell models currently available. We highlight their potential as a platform for drug delivery studies and pinpoint the challenges associated with their use. We discuss in which way each 3D model mimics the in vivo tumor physiology, how they can or have been used in nanomedicine research and to what extent the results obtained so far affect the progress of nanomedicine development. It is of note that the global scientific output associated with 3D models is limited, showing that the use of these systems in nanomedicine investigation is still highly challenging.
Collapse
Affiliation(s)
| | - Beatrice Fortuni
- Molecular Imaging and Photonics, Chemistry Department, KU Leuven, Celestijnenlaan 200F, 3001 Heverlee, Belgium;
| | - Susana Rocha
- Molecular Imaging and Photonics, Chemistry Department, KU Leuven, Celestijnenlaan 200F, 3001 Heverlee, Belgium;
| |
Collapse
|
30
|
Forest CR, Silva CAC, Thordarson P. Dual‐peptide functionalized nanoparticles for therapeutic use. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Chelsea R. Forest
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of New South Wales Sydney New South Wales Australia
| | - Caitlin A. C. Silva
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of New South Wales Sydney New South Wales Australia
| | - Pall Thordarson
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of New South Wales Sydney New South Wales Australia
| |
Collapse
|
31
|
Campos EVR, Pereira AES, de Oliveira JL, Carvalho LB, Guilger-Casagrande M, de Lima R, Fraceto LF. How can nanotechnology help to combat COVID-19? Opportunities and urgent need. J Nanobiotechnology 2020; 18:125. [PMID: 32891146 PMCID: PMC7474329 DOI: 10.1186/s12951-020-00685-4] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
Incidents of viral outbreaks have increased at an alarming rate over the past decades. The most recent human coronavirus known as COVID-19 (SARS-CoV-2) has already spread around the world and shown R0 values from 2.2 to 2.68. However, the ratio between mortality and number of infections seems to be lower in this case in comparison to other human coronaviruses (such as severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV)). These outbreaks have tested the limits of healthcare systems and have posed serious questions about management using conventional therapies and diagnostic tools. In this regard, the use of nanotechnology offers new opportunities for the development of novel strategies in terms of prevention, diagnosis and treatment of COVID-19 and other viral infections. In this review, we discuss the use of nanotechnology for COVID-19 virus management by the development of nano-based materials, such as disinfectants, personal protective equipment, diagnostic systems and nanocarrier systems, for treatments and vaccine development, as well as the challenges and drawbacks that need addressing.
Collapse
Affiliation(s)
- Estefânia V R Campos
- Human and Natural Sciences Center, Federal University of ABC. Av. dos Estados, 5001. Bl. A, T3 Lab. 503-3. Bangú, Santo André, SP, Brazil
| | - Anderson E S Pereira
- São Paulo State University-UNESP, Institute of Science and Technology, Sorocaba, SP, Brazil
| | | | | | | | - Renata de Lima
- Universidade de Sorocaba, Rodovia Raposo Tavares km 92,5, Sorocaba, São Paulo, Brazil.
| | | |
Collapse
|
32
|
Choi J, Kim G, Cho SB, Im HJ. Radiosensitizing high-Z metal nanoparticles for enhanced radiotherapy of glioblastoma multiforme. J Nanobiotechnology 2020; 18:122. [PMID: 32883290 PMCID: PMC7470617 DOI: 10.1186/s12951-020-00684-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an essential step during the treatment of glioblastoma multiforme (GBM), one of the most lethal malignancies. The survival in patients with GBM was improved by the current standard of care for GBM established in 2005 but has stagnated since then. Since GBM is a radioresistant malignancy and the most of GBM recurrences occur in the radiotherapy field, increasing the effectiveness of radiotherapy using high-Z metal nanoparticles (NPs) has recently attracted attention. This review summarizes the progress in radiotherapy approaches for the current treatment of GBM, the physical and biological mechanisms of radiosensitization through high-Z metal NPs, and the results of studies on radiosensitization in the in vitro and in vivo GBM models using high-Z metal NPs to date.
Collapse
Affiliation(s)
- Jinyeong Choi
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Gaeun Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Su Bin Cho
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hyung-Jun Im
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Del Valle AC, Su CK, Sun YC, Huang YF. NIR-cleavable drug adducts of gold nanostars for overcoming multidrug-resistant tumors. Biomater Sci 2020; 8:1934-1950. [PMID: 32039412 DOI: 10.1039/c9bm01813a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
An aptamer-conjugated gold nanostar (dsDDA-AuNS) has been developed for targeting nucleolin present in both tumor cells and tumor vasculature for conducting a drug-resistant cancer therapy. AuNS with its strong absorption in the near-infrared (NIR) region was assembled with a layer of the anti-nucleolin aptamer AS1411. An anticancer drug, namely doxorubicin (DOX), was specifically conjugated on deoxyguanosine residues employing heat and acid labile methylene linkages. In response to NIR irradiation, dsDDA-AuNS allowed on-demand therapeutics. AS1411 played an active role in drug cargo-nucleus interactions, enhancing drug accumulation in the nuclei of drug-resistant breast cancer cells. The intravenous injection of dsDDA-AuNS allowed higher drug accumulation in drug-resistant tumors over naked drugs, leading to greater therapeutic efficacy even at a 54-fold less equivalent drug dose. The in vivo triggered release of DOX from dsDDA-AuNS was achieved by NIR irradiation, resulting in simultaneous photothermal and chemotherapeutic actions, yielding superior tumor growth inhibition than those obtained from either type of monotherapy for overcoming drug resistance in cancers.
Collapse
Affiliation(s)
- Andrea C Del Valle
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China.
| | - Cheng-Kuan Su
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 20224, Taiwan, Republic of China
| | - Yuh-Chang Sun
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China. and Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China
| | - Yu-Fen Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China. and Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China
| |
Collapse
|
34
|
Chen D, Qu X, Shao J, Wang W, Dong X. Anti-vascular nano agents: a promising approach for cancer treatment. J Mater Chem B 2020; 8:2990-3004. [PMID: 32211649 DOI: 10.1039/c9tb02957e] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Anti-vascular agents (AVAs) are a class of promising therapeutic agents with tumor vasculature targeting properties, which can be divided into two types: anti-angiogenic agents (AAAs, inhibit angiogenesis factors) and vascular disrupting agents (VDAs, disrupt established tumor vasculature). AVAs exhibit an enhanced anti-cancer effect by cutting off the oxygen and nutrition supplement channels of tumors. However, the intrinsic drawbacks, such as poor hydrophilicity, undesirable membrane permeability and inferior tumor targeting ability, discount their anti-vascular efficacy. Fortunately, the development of nanotechnology has brought an opportunity for efficient delivery of AVAs to tumour sites with great therapeutic efficacy. The works summarized in this review will provide an understanding of recent advances of anti-vascular nano agents (AVNAs) with a goal to define the mechanism of anti-vascular-based cancer therapy and discuss the challenges and opportunities of AVNAs for clinical translation.
Collapse
Affiliation(s)
- Dapeng Chen
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211800, China.
| | | | | | | | | |
Collapse
|
35
|
Ullah I, Chung K, Bae S, Li Y, Kim C, Choi B, Nam HY, Kim SH, Yun CO, Lee KY, Kumar P, Lee SK. Nose-to-Brain Delivery of Cancer-Targeting Paclitaxel-Loaded Nanoparticles Potentiates Antitumor Effects in Malignant Glioblastoma. Mol Pharm 2020; 17:1193-1204. [PMID: 31944768 DOI: 10.1021/acs.molpharmaceut.9b01215] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive tumor with no curative treatment. The tumor recurrence after resection often requires chemotherapy or radiation to delay the infiltration of tumor remnants. Intracerebral chemotherapies are preferentially being used to prevent tumor regrowth, but treatments remain unsuccessful because of the poor drug distribution in the brain. In this study, we investigated the therapeutic efficacy of cancer-targeting arginyl-glycyl-aspartic tripeptide (RGD) conjugated paclitaxel (PTX)-loaded nanoparticles (NPs) against GBM by nose-to-brain delivery. Our results demonstrated that RGD-modified PTX-loaded NPs showed cancer-specific delivery and enhanced anticancer effects in vivo. The intranasal (IN) inoculation of RGD-PTX-loaded NPs effectively controls the tumor burden (75 ± 12% reduction) by inducing apoptosis and/or inhibiting cancer cell proliferation without affecting the G0 stage of normal brain cells. Our data provide therapeutic evidence supporting the use of intranasally delivered cancer-targeted PTX-loaded NPs for GBM therapy.
Collapse
Affiliation(s)
- Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea.,Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven 06510, United States
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea
| | - Sumin Bae
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea
| | - Yan Li
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea.,National Cancer Center, Gyeonggi-do, Goyang 10408, Korea
| | - Chunggu Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea
| | - Boyoung Choi
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea.,Samyang Biopharmaceuticals Co., Seoul 13488, Korea
| | | | - Sun Hwa Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Chae-Ok Yun
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea
| | - Kuen Yong Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven 06510, United States
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
36
|
Darrigues E, Nima ZA, Griffin RJ, Anderson JM, Biris AS, Rodriguez A. 3D cultures for modeling nanomaterial-based photothermal therapy. NANOSCALE HORIZONS 2020; 5:400-430. [PMID: 32118219 DOI: 10.1039/c9nh00628a] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Photothermal therapy (PTT) is one of the most promising techniques for cancer tumor ablation. Nanoparticles are increasingly being investigated for use with PTT and can serve as theranostic agents. Based on the ability of near-infrared nano-photo-absorbers to generate heat under laser irradiation, PTT could prove advantageous in certain situations over more classical cancer therapies. To analyze the efficacy of nanoparticle-based PTT, preclinical in vitro studies typically use 2D cultures, but this method cannot completely mimic the complex tumor organization, bioactivity, and physiology that all control the complex penetration depth, biodistribution, and tissue diffusion parameters of nanomaterials in vivo. To fill this knowledge gap, 3D culture systems have been explored for PTT analysis. These models provide more realistic microenvironments that allow spatiotemporal oxygen gradients and cancer cell adaptations to be considered. This review highlights the work that has been done to advance 3D models for cancer microenvironment modeling, specifically in the context of advanced, functionalized nanoparticle-directed PTT.
Collapse
Affiliation(s)
- Emilie Darrigues
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S University Avenue, Little Rock, AR 72204, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Israel LL, Galstyan A, Holler E, Ljubimova JY. Magnetic iron oxide nanoparticles for imaging, targeting and treatment of primary and metastatic tumors of the brain. J Control Release 2020; 320:45-62. [PMID: 31923537 DOI: 10.1016/j.jconrel.2020.01.009] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/21/2022]
Abstract
Magnetic nanoparticles in general, and iron oxide nanoparticles in particular, have been studied extensively during the past 20 years for numerous biomedical applications. The main applications of these nanoparticles are in magnetic resonance imaging (MRI), magnetic targeting, gene and drug delivery, magnetic hyperthermia for tumor treatment, and manipulation of the immune system by macrophage polarization for cancer treatment. Recently, considerable attention has been paid to magnetic particle imaging (MPI) because of its better sensitivity compared to MRI. In recent years, MRI and MPI have been combined as a dual or multimodal imaging method to enhance the signal in the brain for the early detection and treatment of brain pathologies. Because magnetic and iron oxide nanoparticles are so diverse and can be used in multiple applications such as imaging or therapy, they have attractive features for brain delivery. However, the greatest limitations for the use of MRI/MPI for imaging and treatment are in brain delivery, with one of these limitations being the brain-blood barrier (BBB). This review addresses the current status, chemical compositions, advantages and disadvantages, toxicity and most importantly the future directions for the delivery of iron oxide based substances across the blood-brain barrier for targeting, imaging and therapy of primary and metastatic tumors of the brain.
Collapse
Affiliation(s)
- Liron L Israel
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Anna Galstyan
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Eggehard Holler
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Julia Y Ljubimova
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA.
| |
Collapse
|
38
|
Chung K, Ullah I, Kim N, Lim J, Shin J, Lee SC, Jeon S, Kim SH, Kumar P, Lee SK. Intranasal delivery of cancer-targeting doxorubicin-loaded PLGA nanoparticles arrests glioblastoma growth. J Drug Target 2020; 28:617-626. [PMID: 31852284 DOI: 10.1080/1061186x.2019.1706095] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of brain tumour and treatment is very challenging. Despite the recent advances in drug delivery systems, various approaches that allow sufficient deposition of anti-cancer drugs within the brain remain unsuccessful due to limited drug delivery throughout the brain. In this study, we utilised an intranasal (IN) approach to allow delivery of anti-cancer drug, encapsulated in PLGA nanoparticles (NPs). PLGA NPs were modified with the RGD ligand to enable Avβ3 expressing tumour-specific delivery. IN delivery of RGD-conjugated-doxorubicin (DOX)-loaded-PLGA-nanoparticles (RGD-DOX-NP) showed cancer-specific delivery of NP and inhibition of brain tumour growth compared to the free-DOX or non-modified DOX-NP in the C6-implanted GBM model. Further, IN treatment with RGD-DOX-NP induces apoptosis in the tumour region without affecting normal brain cells. Our study provides therapeutic evidence to treat GBM using a non-invasive IN approach, which may further be translated to other brain-associated diseases.
Collapse
Affiliation(s)
- Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea.,Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Nahyeon Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea.,Samsung Bioepis, Incheon, Korea
| | - Jaeyeoung Lim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea.,Celltrion, Incheon, Korea
| | - Jungah Shin
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea.,Chong Kun Dang Pharmaceutics, Seoul, Korea
| | - Sangah C Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea.,Department of Health Services, Policy, and Practice, Brown University, Providence, RI, USA
| | - Sangmin Jeon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea
| |
Collapse
|
39
|
Luo Y, Yang H, Zhou YF, Hu B. Dual and multi-targeted nanoparticles for site-specific brain drug delivery. J Control Release 2019; 317:195-215. [PMID: 31794799 DOI: 10.1016/j.jconrel.2019.11.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/26/2022]
Abstract
In recent years, nanomedicines have emerged as a promising method for central nervous system drug delivery, enabling the drugs to overcome the blood-brain barrier and accumulate preferentially in the brain. Despite the current success of brain-targeted nanomedicines, limitations still exist in terms of the targeting specificity. Based on the molecular mechanism, the exact cell populations and subcellular organelles where the injury occurs and the drugs take effect have been increasingly accepted as a more specific target for the next generation of nanomedicines. Dual and multi-targeted nanoparticles integrate different targeting functionalities and have provided a paradigm for precisely delivering the drug to the pathological site inside the brain. The targeting process often involves the sequential or synchronized navigation of the targeting moieties, which allows highly controlled drug delivery compared to conventional targeting strategies. Herein, we focus on the up-to-date design of pathological site-specific nanoparticles for brain drug delivery, highlighting the dual and multi-targeting strategies that were employed and their impact on improving targeting specificity and therapeutic effects. Furthermore, the background discussion of the basic properties of a brain-targeted nanoparticle and the common lesion features classified by neurological pathology are systematically summarized.
Collapse
Affiliation(s)
- Yan Luo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hang Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
40
|
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults, associated with a high mortality rate and a survival of between 12 and 15 months after diagnosis. Due to current treatment limitations involving surgery, radiotherapy and chemotherapy with temozolamide, there is a high rate of treatment failure and recurrence. To try to overcome these limitations nanotechnology has emerged as a novel alternative. Lipid, polymeric, silica and magnetic nanoparticles, among others, are being developed to improve GBM treatment and diagnosis. These nanoformulations have many advantages, including lower toxicity, biocompatibility and the ability to be directed toward the tumor. This article reviews the progress that have been made and the large variety of nanoparticles currently under study for GBM.
Collapse
|
41
|
Zhang X, Xi Z, Machuki JO, Luo J, Yang D, Li J, Cai W, Yang Y, Zhang L, Tian J, Guo K, Yu Y, Gao F. Gold Cube-in-Cube Based Oxygen Nanogenerator: A Theranostic Nanoplatform for Modulating Tumor Microenvironment for Precise Chemo-Phototherapy and Multimodal Imaging. ACS NANO 2019; 13:5306-5325. [PMID: 31018094 DOI: 10.1021/acsnano.8b09786] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Engineering a versatile oncotherapy nanoplatform integrating both diagnostic and therapeutic functions has always been an intractable challenge in targeted cancer treatment. Herein, to actualize the theme of precise medicine, a nanoplatform is developed by anchoring Mn-Cdots to doxorubicin (DOX)-loaded mesoporous silica-coated gold cube-in-cubes core/shell nanocomposites and further conjugating them to a Arg-Gly-Asp (RGD) peptide (denoted as RGD-CCmMC/DOX) to achieve an active-targeting effect. Under 635 nm irradiation, the nanoplatform acts as oxygen nanogenerator that produces O2 in situ and amplifies the content of singlet oxygen (1O2) in the hypoxic tumor microenvironment (TME), which has been demonstrated to attenuate tumor hypoxia and synchronously enhance photodynamic efficacy. Moreover, the gold cube-in-cube core in this work has been proven as a photothermal agent for hyperthermia, which exhibits a favorable photothermal effect with a 65.6% calculated photothermal conversion efficiency under 808 nm irradiation. In addition, the nanoplatform achieves heat- and pH-sensitive drug release with precise control to specific-tumor sites, executing combined chemo-phototherapy functions. Besides, it functions as a multimodal bioimaging agent of photothermal, fluorescence, and magnetic resonance imaging for the accurate diagnosis and guidance of therapy. As validated by in vivo and in vitro assays, the TME-responsive nanoplatform is highly biocompatible and effectively obliterates 4T1 tumor xenografts on nude mice after triple-synergetic treatment. This work presents a rational design of versatile nanoplatforms, which modulate the TME to enable high therapeutic performance and multiplexed imaging, which provides an innovative paradigm for targeted tumor therapy.
Collapse
Affiliation(s)
- Xing Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
- Institute of Orthopedics, Department of Orthopedics , Affiliated Hospital of Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Zhongqian Xi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Jeremiah Ong'achwa Machuki
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Jianjun Luo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
- Institute of Orthopedics, Department of Orthopedics , Affiliated Hospital of Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Dongzhi Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Jingjing Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Weibing Cai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Yun Yang
- Nanomaterials and Chemistry Key Laboratory , Wenzhou University , Wenzhou , Zhejiang 325027 , PR China
| | - Lijie Zhang
- Nanomaterials and Chemistry Key Laboratory , Wenzhou University , Wenzhou , Zhejiang 325027 , PR China
| | - Jiangwei Tian
- School of Traditional Chinese Pharmacy , China Pharmaceutical University , Nanjing , Jiangsu 211198 , PR China
| | - Kaijin Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
- Institute of Orthopedics, Department of Orthopedics , Affiliated Hospital of Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu 221002 , PR China
| |
Collapse
|
42
|
Castillo RR, Lozano D, González B, Manzano M, Izquierdo-Barba I, Vallet-Regí M. Advances in mesoporous silica nanoparticles for targeted stimuli-responsive drug delivery: an update. Expert Opin Drug Deliv 2019; 16:415-439. [PMID: 30897978 PMCID: PMC6667337 DOI: 10.1080/17425247.2019.1598375] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Mesoporous silica nanoparticles (MSNs) are outstanding nanoplatforms for drug delivery. Herein, the most recent advances to turn MSN-based carriers into minimal side effect drug delivery agents are covered. AREAS COVERED This review summarizes the scientific advances dealing with MSNs for targeted and stimuli-responsive drug delivery since 2015. Delivery aspects to diseased tissues together with approaches to obtain smart MSNs able to respond to internal or external stimuli and their applications are here described. Special emphasis is done on the combination of two or more stimuli on the same nanoplatform and on combined drug therapy. EXPERT OPINION The use of MSNs in nanomedicine is a promising research field because they are outstanding platforms for treating different pathologies. This is possible thanks to their structural, chemical, physical and biological properties. However, there are certain issues that should be overcome to improve the suitability of MSNs for clinical applications. All materials must be properly characterized prior to their in vivo evaluation; furthermore, preclinical in vivo studies need to be standardized to demonstrate the MSNs clinical translation potential.
Collapse
Affiliation(s)
- Rafael R. Castillo
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Daniel Lozano
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Miguel Manzano
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| |
Collapse
|
43
|
Zhang C, Gu Z, Shen L, Liu X, Lin H. In vivo Evaluation and Alzheimer’s Disease Treatment Outcome of siRNA Loaded Dual Targeting Drug Delivery System. Curr Pharm Biotechnol 2019; 20:56-62. [PMID: 30727887 DOI: 10.2174/1389201020666190204141046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/23/2018] [Accepted: 01/23/2019] [Indexed: 01/02/2023]
Abstract
Background:
To deliver drugs to treat Alzheimer’s Disease (AD), nanoparticles should
firstly penetrate through blood brain barrier, and then target neurons.
Methods:
Recently, we developed an Apo A-I and NL4 dual modified nanoparticle (ANNP) to deliver
beta-amyloid converting enzyme 1 (BACE1) siRNA. Although promising in vitro results were obtained,
the in vivo performance was not clear. Therefore, in this study, we further evaluated the in vivo
neuroprotective effect and toxicity of the ANNP/siRNA. The ANNP/siRNA was 80.6 nm with good
stability when incubated with serum. In vivo, the treatment with ANNP/siRNA significantly improves
the spatial learning and memory of APP/PS1 double transgenic mice, as determined by mean escape
latency, times of crossing the platform area during the 60 s swimming and the percentage of the distance
in the target quadrant.
Results and Conclusion:
After the treatment, BACE1 RNA level of ANNP/siRNA group was greatly
reduced, which contributed a good AD treatment outcome. Finally, after repeated administration, the
ANNP/siRNA did not lead to significant change as observed by HE staining of main organs, suggesting
the good biocompatibility of ANNP/siRNA. These results demonstrated that the ANNP was a good
candidate for AD targeting siRNA delivery.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Zhichun Gu
- Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Long Shen
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Xianyan Liu
- Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Houwen Lin
- Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| |
Collapse
|
44
|
Immobilized Peptide on the Surface of Poly l-DOPA/Silica for Targeted Delivery of 5-Fluorouracil to Breast Tumor. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09834-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
45
|
Tomitaka A, Kaushik A, Kevadiya BD, Mukadam I, Gendelman HE, Khalili K, Liu G, Nair M. Surface-engineered multimodal magnetic nanoparticles to manage CNS diseases. Drug Discov Today 2019; 24:873-882. [PMID: 30660756 PMCID: PMC7405805 DOI: 10.1016/j.drudis.2019.01.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/26/2018] [Accepted: 01/11/2019] [Indexed: 01/21/2023]
Abstract
Advanced central nervous system (CNS) therapies exhibited high efficacy but complete treatment of CNS diseases remains challenging owing to limited delivery of therapeutic agents to the brain. Multifunctional magnetic nanoparticles are investigated not only for site-specific drug delivery but also for theranostic applications aiming for an effective CNS therapy. Recently, surface engineering of magnetic nanoparticles was recognized as a crucial area of research to achieve precise therapy and imaging at molecular and cellular levels. This review reports state-of-the-art advancement in the development of surface-engineered magnetic nanoparticles targeting CNS diagnostics and therapies. The challenges and future prospects of magnetic theranostics are also discussed by considering the translation from bench to bedside. Successful translation of magnetic theranostics to the clinical setting will enable precise and efficient diagnostics and therapy to manage CNS diseases.
Collapse
Affiliation(s)
- Asahi Tomitaka
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Ajeet Kaushik
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmacology and Experimental Neuroscience, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kamel Khalili
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
46
|
Guo P, Yang J, Liu D, Huang L, Fell G, Huang J, Moses MA, Auguste DT. Dual complementary liposomes inhibit triple-negative breast tumor progression and metastasis. SCIENCE ADVANCES 2019; 5:eaav5010. [PMID: 30906868 PMCID: PMC6426465 DOI: 10.1126/sciadv.aav5010] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/31/2019] [Indexed: 05/10/2023]
Abstract
Distinguishing malignant cells from non-neoplastic ones is a major challenge in triple-negative breast cancer (TNBC) treatment. Here, we developed a complementary targeting strategy that uses precisely matched, multivalent ligand-receptor interactions to recognize and target TNBC tumors at the primary site and metastatic lesions. We screened a panel of cancer cell surface markers and identified intercellular adhesion molecule-1 (ICAM1) and epithelial growth factor receptor (EGFR) as optimal candidates for TNBC complementary targeting. We engineered a dual complementary liposome (DCL) that precisely complements the molecular ratio and organization of ICAM1 and EGFR specific to TNBC cell surfaces. Our in vitro mechanistic studies demonstrated that DCLs, compared to single-targeting liposomes, exhibited increased binding, enhanced internalization, and decreased receptor signaling. DCLs consistently exhibited substantially increased tumor targeting activity and antitumor efficacy in orthotopic and lung metastasis models, indicating that DCLs are a platform technology for the design of personalized nanomedicines for TNBC.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Jiang Yang
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Daxing Liu
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Lan Huang
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Gillian Fell
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jing Huang
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Marsha A. Moses
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Debra T. Auguste
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| |
Collapse
|
47
|
Fu Q, Zhao Y, Yang Z, Yue Q, Xiao W, Chen Y, Yang Y, Guo L, Wu Y. Liposomes actively recognizing the glucose transporter GLUT1and integrin αvβ3for dual-targeting of glioma. Arch Pharm (Weinheim) 2019; 352:e1800219. [PMID: 30609116 DOI: 10.1002/ardp.201800219] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 11/05/2018] [Accepted: 11/11/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Qiuyi Fu
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Yi Zhao
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Zhongzhen Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Qiming Yue
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Wenjiao Xiao
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Yang Chen
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Yang Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Li Guo
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| | - Yong Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy; Sichuan University; Chengdu P. R. China
| |
Collapse
|
48
|
Li H, Li K, Zeng Q, Zeng Y, Chen D, Pang L, Chen X, Zhan Y. Novel vinyl-modified RGD conjugated silica nanoparticles based on photo click chemistry for in vivo prostate cancer targeted fluorescence imaging. RSC Adv 2019; 9:25318-25325. [PMID: 35530054 PMCID: PMC9070015 DOI: 10.1039/c9ra04513a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 08/02/2019] [Indexed: 11/21/2022] Open
Abstract
Molecular imaging is a powerful tool for non-invasive visualization of tumors that plays an important role in their diagnosis and treatment. The specificity of molecular imaging probes for cancer cells is important for accurate tumor visualization, with antibody and polypeptide nanoprobe conjugates having often been used as targeting agents for tumor detection. However, many traditional chemical conjugation methods employ complex conjugation reactions that result in poor efficiency and poor bioactivity. Herein, we describe the use of photo click methodology for the rapid synthesis of nanoprobes comprised of silica nanoparticles functionalized with RGD targeting units (SiO2@T1-RGDk NPs) (∼80 nm) for in vivo prostate cancer fluorescent imaging applications. These SiO2@T1-RGDk NPs exhibit a maximum absorption wavelength of 380 nm in their UV absorption spectra with a maximum fluorescence emission wavelength of 550 nm. Confocal immunofluorescent imaging reveal that SiO2@T1-RGDk NPs exhibit excellent targeting ability for visualizing cancer cells, with in vivo fluorescence imaging intensity in a subcutaneous tumor model of prostate cancer reaching a maxima after 4 h. Biosafety assessments showed that SiO2@T1-RGDk NPs demonstrate no obvious toxicity in mice, thus demonstrated that these novel NPs may prove to be promising fluorescent imaging agents for the accurate detection and treatment of tumors. Photo click chemistry has been used to prepare RGD conjugated silica nanoprobe (SiO2@T1-RGDk NPs) that exhibits excellent tumor targeting ability and negligible toxicity which enables them to be used for the diagnosis and treatment of cancer.![]()
Collapse
Affiliation(s)
- Hanrui Li
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education
- School of Life Science and Technology
- Xidian University
- Xi'an
- China
| | - Ke Li
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease
- Shaanxi Key Laboratory of Brain Disorders
- Institute of Basic and Translational Medicine
- Xi'an Medical University
- Xi'an
| | - Qi Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education
- School of Life Science and Technology
- Xidian University
- Xi'an
- China
| | - Yun Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education
- School of Life Science and Technology
- Xidian University
- Xi'an
- China
| | - Dan Chen
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education
- School of Life Science and Technology
- Xidian University
- Xi'an
- China
| | - Liaojun Pang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education
- School of Life Science and Technology
- Xidian University
- Xi'an
- China
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education
- School of Life Science and Technology
- Xidian University
- Xi'an
- China
| | - Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education
- School of Life Science and Technology
- Xidian University
- Xi'an
- China
| |
Collapse
|
49
|
Kaur A, Shimoni O, Wallach M. Novel screening test for celiac disease using peptide functionalised gold nanoparticles. World J Gastroenterol 2018; 24:5379-5390. [PMID: 30598582 PMCID: PMC6305529 DOI: 10.3748/wjg.v24.i47.5379] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/01/2018] [Accepted: 10/05/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To develop a screening test for celiac disease based on the coating of gold nanoparticles with a peptide sequence derived from gliadin, the protein that triggers celiac disease.
METHODS 20 nm gold nanoparticles were first coated with NeutrAvidin. A long chain Polyethylene glycol (PEG) linker containing Maleimide at the Ω-end and Biotin group at the α-end was used to ensure peptide coating to the gold nanoparticles. The maleimide group with the thiol (-SH) side chain reacted with the cysteine amino acid in the peptide sequence and the biotinylated and PEGylated peptide was added to the NeutrAvidin coated gold nanoparticles. The peptide coated gold nanoparticles were then converted into a serological assay. We used the peptide functionalised gold nanoparticle-based assay on thirty patient serum samples in a blinded assessment and compared our results with the previously run serological and pathological tests on these patients.
RESULTS A stable colloidal suspension of peptide coated gold nanoparticles was obtained without any aggregation. An absorbance peak shift as well as color change was caused by the aggregation of gold nanoparticles following the addition of anti-gliadin antibody to peptide coated nanoparticles at levels associated with celiac disease. The developed assay has been shown to detect anti-gliadin antibody not only in quantitatively spiked samples but also in a small-scale study on real non-hemolytic celiac disease patient’s samples.
CONCLUSION The study demonstrates the potential of gold nanoparticle-peptide based approach to be adapted for developing a screening assay for celiac disease diagnosis. The assay could be a part of an exclusion based diagnostic strategy and prove particularly useful for testing high celiac disease risk populations.
Collapse
Affiliation(s)
- Anantdeep Kaur
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney 2007, Australia
| | - Olga Shimoni
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney 2007, Australia
| | - Michael Wallach
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney 2007, Australia
| |
Collapse
|
50
|
Zhu X, Zhou H, Liu Y, Wen Y, Wei C, Yu Q, Liu J. Transferrin/aptamer conjugated mesoporous ruthenium nanosystem for redox-controlled and targeted chemo-photodynamic therapy of glioma. Acta Biomater 2018; 82:143-157. [PMID: 30316026 DOI: 10.1016/j.actbio.2018.10.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/27/2018] [Accepted: 10/09/2018] [Indexed: 01/10/2023]
Abstract
The blood-brain barrier (BBB) and low targeting are major obstacles for the treatment of gliomas. Accordingly, overcoming the BBB and enhancing the targeting of drugs to the glioma area are key to achieving a good therapeutic effect. Here, we have developed the mesoporous ruthenium nanosystem RBT@MRN-SS-Tf/Apt with dual targeting function. Transferrin (Tf) and aptamer AS1411 (Apt) are grafted on the surfaces of mesoporous ruthenium nanoparticles (MRN) with high loading capacity. This is achieved via redox-cleavable disulfide bonds, serving as both a capping agent and a targeting ligand, enabling the effective penetration of the blood-brain barrier and targeting the glioma. In addition, RBT@MRN-SS-Tf/Apt can specifically kill glioma cells in vitro and in vivo. Moreover, anti-tumor drugs [Ru(bpy)2(tip)]2+ (RBT) will produce reactive oxygen species and induce apoptosis of tumor cells under laser irradiation, providing photodynamic therapy (PDT) for the treatment of gliomas, and further prolonging the median survival period. The study shows that this chemical photodynamic therapy nanosystem can be used as an efficient and powerful synergistic system for the treatment of brain tumors and other brain diseases of the central nervous system. STATEMENT OF SIGNIFICANCE: In order to overcome the blood-brain barrier and low targeting, and enhance the anti-glioma activities of nanodrugs. We have developed RBT@MRN-SS-Tf/Apt with dual targeting function. It is achieved release drug via redox-cleavable disulfide bonds, and enable the effective penetration of the blood-brain barrier and targeting the glioma. Moreover, anti-tumor drugs RBT will produce reactive oxygen species and induce apoptosis of tumor cells under laser irradiation, providing photodynamic therapy (PDT) for the treatment of gliomas, and further prolonging the median survival period. Therefore, this chemical photodynamic therapy nanosystem can be used as an efficient and powerful synergistic system for the treatment of brain tumors and other brain diseases of the central nervous system.
Collapse
Affiliation(s)
- Xufeng Zhu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Hui Zhou
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yanan Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yayu Wen
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Chunfang Wei
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Qianqian Yu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Jie Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| |
Collapse
|