1
|
Lopes-Nunes J, Oliveira PA, Cruz C. Nanotherapy for human papillomavirus-associated cancers: breakthroughs and challenges. Trends Pharmacol Sci 2024; 45:781-797. [PMID: 39181737 DOI: 10.1016/j.tips.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024]
Abstract
Human papillomaviruses (HPVs) are well-known causative agents of several cancers, yet selective therapies remain under investigation. Nanoparticles, for instance, are emerging as promising solutions to enhance the delivery and efficacy of therapeutic approaches. Despite the increasing number of nanotherapies offering advantages over current treatments, only one has advanced to clinical trials. This review highlights recent advances in nanotherapies for HPV-associated cancers, focusing on the delivery of small molecules, gene-targeted therapies, and vaccines. Some of the challenges faced in nanotherapies translation for clinical application are discussed, emphasizing the most used preclinical models that fail to accurately predict human responses, thereby hindering proper evaluation of nanotherapies. Additionally, we explore and discuss alternative promising new preclinical models that could pave the way for more effective nanotherapeutic evaluations.
Collapse
Affiliation(s)
- Jéssica Lopes-Nunes
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Paula A Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Inov4Agro, Institute for Innovation, Capacity Building, and Sustainability of Agri-food Production, Vila Real, Portugal
| | - Carla Cruz
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; Departamento de Química, Universidade da Beira Interior, Rua Marquês de Ávila e Bolama, 6201-001, Covilhã, Portugal.
| |
Collapse
|
2
|
Zhang J, Zhou J, Tang L, Ma J, Wang Y, Yang H, Wang X, Fan W. Custom-Design of Multi-Stimuli-Responsive Degradable Silica Nanoparticles for Advanced Cancer-Specific Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400353. [PMID: 38651235 DOI: 10.1002/smll.202400353] [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: 01/15/2024] [Revised: 03/24/2024] [Indexed: 04/25/2024]
Abstract
Chemotherapy is crucial in oncology for combating malignant tumors but often encounters obatacles such as severe adverse effects, drug resistance, and biocompatibility issues. The advantages of degradable silica nanoparticles in tumor diagnosis and treatment lie in their ability to target drug delivery, minimizing toxicity to normal tissues while enhancing therapeutic efficacy. Moreover, their responsiveness to both endogenous and exogenous stimuli opens up new possibilities for integrating multiple treatment modalities. This review scrutinizes the burgeoning utility of degradable silica nanoparticles in combination with chemotherapy and other treatment modalities. Commencing the elucidation of degradable silica synthesis and degradation mechanisms, emphasis is placed on the responsiveness of these materials to endogenous (e.g., pH, redox reactions, hypoxia, and enzymes) and exogenous stimuli (e.g., light and high-intensity focused ultrasound). Moreover, this exploration delves into strategies harnessing degradable silica nanoparticles in chemotherapy alone, coupled with radiotherapy, photothermal therapy, photodynamic therapy, gas therapy, immunotherapy, starvation therapy, and chemodynamic therapy, elucidating multimodal synergies. Concluding with an assessment of advances, challenges, and constraints in oncology, despite hurdles, future investigations are anticipated to augment the role of degradable silica in cancer therapy. These insights can serve as a compass for devising more efficacious combined tumor treatment strategies.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, P. R. China
| | - Jiani Zhou
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | | | - Jiayi Ma
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Ying Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Xiaoxiao Wang
- Biochemical Engineering Research Center, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243032, P. R. China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
3
|
Zhang J, Li Y, Guo S, Zhang W, Fang B, Wang S. Moving beyond traditional therapies: the role of nanomedicines in lung cancer. Front Pharmacol 2024; 15:1363346. [PMID: 38389925 PMCID: PMC10883231 DOI: 10.3389/fphar.2024.1363346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Amidst a global rise in lung cancer occurrences, conventional therapies continue to pose substantial side effects and possess notable toxicities while lacking specificity. Counteracting this, the incorporation of nanomedicines can notably enhance drug delivery at tumor sites, extend a drug's half-life and mitigate inadvertent toxic and adverse impacts on healthy tissues, substantially influencing lung cancer's early detection and targeted therapy. Numerous studies signal that while the nano-characteristics of lung cancer nanomedicines play a pivotal role, further interplay with immune, photothermal, and genetic factors exist. This review posits that the progression towards multimodal combination therapies could potentially establish an efficacious platform for multimodal targeted lung cancer treatments. Current nanomedicines split into active and passive targeting. Active therapies focus on a single target, often with unsatisfactory results. Yet, developing combination systems targeting multiple sites could chart new paths in lung cancer therapy. Conversely, low drug delivery rates limit passive therapies. Utilizing the EPR effect to bind specific ligands on nanoparticles to tumor cell receptors might create a new regime combining active-passive targeting, potentially elevating the nanomedicines' concentration at target sites. This review collates recent advancements through the lens of nanomedicine's attributes for lung cancer therapeutics, the novel carrier classifications, targeted therapeutic modalities and their mechanisms, proposing that the emergence of multi-target nanocomposite therapeutics, combined active-passive targeting therapies and multimodal combined treatments will pioneer novel approaches and tools for future lung cancer clinical therapies.
Collapse
Affiliation(s)
- Jingjing Zhang
- Medical College of Qingdao Binhai University, Qingdao, China
- The Affiliated Hospital of Qindao Binhai University (Qingdao Military-Cvil Integration Hospital), Qingdao, China
| | - Yanzhi Li
- Medical College of Qingdao Binhai University, Qingdao, China
| | - Sa Guo
- Ethnic Medicine Academic Heritage Innovation Research Center, Meishan Traditional Chinese Medicine Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weifen Zhang
- Medical College, Weifang University, Weifang, China
| | - Bing Fang
- The Affiliated Hospital of Qindao Binhai University (Qingdao Military-Cvil Integration Hospital), Qingdao, China
| | - Shaohui Wang
- Ethnic Medicine Academic Heritage Innovation Research Center, Meishan Traditional Chinese Medicine Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Wu Y, Tian J, Yang J, Peng Q, Wu Z, Liu R, Luo M, Qiu Y, Cao R. Bufotalin-loaded biomimetic Prussian blue nanoparticles for colorectal cancer chemo-photothermal ferroptosis therapy. Nanomedicine (Lond) 2024; 19:109-125. [PMID: 38197393 DOI: 10.2217/nnm-2023-0293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
Purpose: We constructed biomimetic nanoparticles with biocompatible, tumor-targeting, laser-responsive properties for ferroptosis-induced colorectal cancer chemo-photothermal therapy, with the aim to realize double-hit ferroptosis treatment for colorectal cancer. Methods: The nanoparticles were prepared by first loading the chemotherapy drug bufotalin (CS-5) with Prussian blue (PB), then combining a hybridized erythrocyte-tumor membrane (M) with PB@CS-5 to produce PB@CS-5@M. The chemo-photothermal therapy efficiency of PB@CS-5@M was tested by in vitro and in vivo experiments. Results and conclusion: The combined PB and CS-5 act as promising ferroptosis inducers to enhance ferroptosis efficacy. The hyperthermia induced by laser stimulation can trigger PB to release CS-5 and iron and ferrous ions, which further promotes ferroptosis.
Collapse
Affiliation(s)
- Yi Wu
- The First Affiliated Hospital of Hunan Normal University, Changsha, 410013, China
- Immunodiagnostic Reagents Engineering Research Center of Hunan Province, Hunan Normal University, Changsha, 410013, China
| | - Jiahui Tian
- The First Affiliated Hospital of Hunan Normal University, Changsha, 410013, China
- Department of Medical Laboratory, School of Medicine, Hunan Normal University, Changsha, 410013, China
- Immunodiagnostic Reagents Engineering Research Center of Hunan Province, Hunan Normal University, Changsha, 410013, China
| | - Jialu Yang
- The First Affiliated Hospital of Hunan Normal University, Changsha, 410013, China
| | - Qian Peng
- Department of Medical Laboratory, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | | | - Rushi Liu
- Department of Medical Laboratory, School of Medicine, Hunan Normal University, Changsha, 410013, China
- Immunodiagnostic Reagents Engineering Research Center of Hunan Province, Hunan Normal University, Changsha, 410013, China
| | - Mengjie Luo
- Shenzhen Yantian District People's Hospital, Shenzhen, 518081, China
| | - Yilan Qiu
- Immunodiagnostic Reagents Engineering Research Center of Hunan Province, Hunan Normal University, Changsha, 410013, China
- School of Life Science, Hunan Normal University, Changsha, 410013, China
| | - Ruiyun Cao
- Wujin Hospital of Traditional Chinese Medicine, Changzhou, 213161, China
| |
Collapse
|
5
|
Xia Z, Mu W, Yuan S, Fu S, Liu Y, Zhang N. Cell Membrane Biomimetic Nano-Delivery Systems for Cancer Therapy. Pharmaceutics 2023; 15:2770. [PMID: 38140108 PMCID: PMC10748133 DOI: 10.3390/pharmaceutics15122770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Nano-delivery systems have demonstrated great promise in the therapy of cancer. However, the therapeutic efficacy of conventional nanomedicines is hindered by the clearance of the blood circulation system and the physiological barriers surrounding the tumor. Inspired by the unique capabilities of cells within the body, such as immune evasion, prolonged circulation, and tumor-targeting, there has been a growing interest in developing cell membrane biomimetic nanomedicine delivery systems. Cell membrane modification on nanoparticle surfaces can prolong circulation time, activate tumor-targeting, and ultimately improve the efficacy of cancer treatment. It shows excellent development potential. This review will focus on the advancements in various cell membrane nano-drug delivery systems for cancer therapy and the obstacles encountered during clinical implementation. It is hoped that such discussions will inspire the development of cell membrane biomimetic nanomedical systems.
Collapse
Affiliation(s)
- Zhenxing Xia
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Weiwei Mu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shijun Yuan
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shunli Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Yongjun Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Na Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| |
Collapse
|
6
|
Xu J, Karra V, Large DE, Auguste DT, Hung FR. Understanding the Mechanical Properties of Ultradeformable Liposomes Using Molecular Dynamics Simulations. J Phys Chem B 2023; 127:9496-9512. [PMID: 37879075 PMCID: PMC10641833 DOI: 10.1021/acs.jpcb.3c04386] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/19/2023] [Accepted: 10/04/2023] [Indexed: 10/27/2023]
Abstract
Improving drug delivery efficiency to solid tumor sites is a central challenge in anticancer therapeutic research. Our previous experimental study (Guo et al., Nat. Commun. 2018, 9, 130) showed that soft, elastic liposomes had increased uptake and accumulation in cancer cells and tumors in vitro and in vivo respectively, relative to rigid particles. As a first step toward understanding how liposomes' molecular structure and composition modulates their elasticity, we performed all-atom and coarse-grained classical molecular dynamics (MD) simulations of lipid bilayers formed by mixing a long-tailed unsaturated phospholipid with a short-tailed saturated lipid with the same headgroup. The former types of phospholipids considered were 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and 1,2-dipalmitoleoyl-sn-glycero-3-phosphocholine (termed here DPMPC). The shorter saturated lipids examined were 1,2-diheptanoyl-sn-glycero-3-phosphocholine (DHPC), 1,2-didecanoyl-sn-glycero-3-phosphocholine (DDPC), 1,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC), and 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC). Several lipid concentrations and surface tensions were considered. Our results show that DOPC or DPMPC systems having 25-35 mol % of the shortest lipids DHPC or DDPC are the least rigid, having area compressibility moduli KA that are ∼10% smaller than the values observed in pure DOPC or DPMPC bilayers. These results agree with experimental measurements of the stretching modulus and lysis tension in liposomes with the same compositions. These mixed systems also have lower areas per lipid and form more uneven x-y interfaces with water, the tails of both primary and secondary lipids are more disordered, and the terminal methyl groups in the tails of the long lipid DOPC or DPMPC wriggle more in the vertical direction, compared to pure DOPC or DPMPC bilayers or their mixtures with the longer saturated lipid DLPC or DMPC. These observations confirm our hypothesis that adding increasing concentrations of the short unsaturated lipid DHPC or DDPC to DOPC or DPMPC bilayers alters lipid packing and thus makes the resulting liposomes more elastic and less rigid. No formation of lipid nanodomains was noted in our simulations, and no clear trends were observed in the lateral diffusivities of the lipids as the concentration, type of secondary lipid, and surface tension were varied.
Collapse
Affiliation(s)
- Jiaming Xu
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Vyshnavi Karra
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Danielle E. Large
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Debra T. Auguste
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Francisco R. Hung
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
7
|
Bajwa DE, Salvanou EA, Theodosiou M, Koutsikou TS, Efthimiadou EK, Bouziotis P, Liolios C. Radiolabeled iron oxide nanoparticles functionalized with PSMA/BN ligands for dual-targeting of prostate cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1184309. [PMID: 39380961 PMCID: PMC11460297 DOI: 10.3389/fnume.2023.1184309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2024]
Abstract
Introduction Prostate cancer (PCa) is the second most frequent cancer diagnosis in men and the fifth leading cause of death worldwide. Prostate Specific Membrane Antigen (PSMA) and Gastrin Releasing Peptide (GRP) receptors are overexpressed in PCa. In this study, we have developed iron oxide nanoparticles (IONs) functionalized with the Prostate Specific Membrane Antigen (PSMA) and Gastrin Releasing Peptide (GRP) ligands for dual targeting of Prostate cancer. Methods IONs were developed with a thin silica layer on their surface with MPTES (carrying -SH groups, IONs-SH), and they were coupled either with a pharmacophore targeting PSMA (IONs-PSMA) or with bombesin peptide (IONs-BN), targeting GRP receptors, or with both (IONs-PSMA/BN). The functionalized IONs were characterized for their size, zeta potential, and efficiency of functionalization using dynamic light scattering (DLS) and Fourier-Transform Infrared Spectroscopy (FT-IR). All the aforementioned types of IONs were radiolabeled directly with Technetium-99m (99mTc) and evaluated for their radiolabeling efficiency, stability, and binding ability on two different PCa cell lines (PC3 and LNCaP). Results and Discussion The MTT assay demonstrated low toxicity of the IONs against PC3 and LNCaP cells, while the performed wound-healing assay further proved that these nanostructures did not affect cellular growth mechanisms. The observed hemolysis ratio after co-incubation with red blood cells was extremely low. Furthermore, the 99mTc-radiolabeled IONs showed good stability in human serum, DTPA, and histidine, and high specific binding rates in cancer cells, supporting their future utilization as potential diagnostic tools for PCa with Single Photon Emission Computed Tomography (SPECT) imaging.
Collapse
Affiliation(s)
- Danae Efremia Bajwa
- Radiochemical Studies Laboratory, Energy & Safety, Institute of Nuclear & Radiological Sciences & Technology (INRASTES), National Centre for Scientific Research (NCSR) “Demokritos”, Athens, Greece
| | - Evangelia-Alexandra Salvanou
- Radiochemical Studies Laboratory, Energy & Safety, Institute of Nuclear & Radiological Sciences & Technology (INRASTES), National Centre for Scientific Research (NCSR) “Demokritos”, Athens, Greece
| | - Maria Theodosiou
- Laboratory of Inorganic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora S. Koutsikou
- Radiochemical Studies Laboratory, Energy & Safety, Institute of Nuclear & Radiological Sciences & Technology (INRASTES), National Centre for Scientific Research (NCSR) “Demokritos”, Athens, Greece
- Laboratory of Inorganic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni K. Efthimiadou
- Laboratory of Inorganic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Penelope Bouziotis
- Radiochemical Studies Laboratory, Energy & Safety, Institute of Nuclear & Radiological Sciences & Technology (INRASTES), National Centre for Scientific Research (NCSR) “Demokritos”, Athens, Greece
| | - Christos Liolios
- Radiochemical Studies Laboratory, Energy & Safety, Institute of Nuclear & Radiological Sciences & Technology (INRASTES), National Centre for Scientific Research (NCSR) “Demokritos”, Athens, Greece
- Research Laboratory, Institute of Pharmaceutical Research & Technology (IFET) (Pallini), Athens, Greece
| |
Collapse
|
8
|
Yao J, Zhang A, Qiu Y, Li Z, Wu X, Li Z, Wu A, Yang F. Navigating zinc-involved nanomedicine in oncotherapy. NANOSCALE 2023; 15:4261-4276. [PMID: 36756840 DOI: 10.1039/d2nr06857e] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Zinc (Zn), extolled as "the flower of life" in modern medicine, has been extensively highlighted with its physiological functions to maintain growth, development, and metabolism homeostasis. Driven by the substantial advancement of nanotechnology and oncology, Zn-involved nanomedicines integrating the intrinsic bioactivity of Zn species and the physiochemical attributes of Zn-composed nanosystems have blazed a highly efficient and relatively biosafe antineoplastic path. In this review, we aim to highlight and discuss the recent representative modalities of emerging Zn-involved oncology nanomedicine, mainly emphasizing the rational design, biological effect and biosafety, and therapeutic strategies. In addition, we provide the underlying critical obstacles and future perspectives of Zn-involved oncology nanomedicines, primarily focusing on the chances and challenges of clinical translation. Furthermore, we hope the review can give rise to opportunities within oncology nanomedicine and other biomedical fields, promoting the prosperity and progress of the "Zincic Age".
Collapse
Affiliation(s)
- Junlie Yao
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Aoran Zhang
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, P. R. China
| | - Yue Qiu
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Zihou Li
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Xiaoxia Wu
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Zhouhua Li
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Aiguo Wu
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, P. R. China
| | - Fang Yang
- Ningbo Cixi Institute of BioMedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, P. R. China
| |
Collapse
|
9
|
Xi H, Xu B, Fang A, Li X, Huang Z, Qin S, Xiao W, Li G, Tian M, Fan N, Song X. A cascade-responsive nanoplatform with tumor cell-specific drug burst release for chemotherapy. Acta Biomater 2023; 162:120-134. [PMID: 36828165 DOI: 10.1016/j.actbio.2023.02.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/22/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023]
Abstract
Most of the nanomedicines can reduce the side effects of anti-tumor chemical drugs but do not have good enough therapeutic efficacy, largely due to the sustained drug release profile. It might be a promising alternative strategy to develop a cascade-responsive nanoplatform against tumor with the burst release of chemotherapeutics based on the highly efficient tumor cell targeting delivery. In this work, we constructed innovative nanoparticles (PMP/WPH-NPs) consisting of two functional polymers. PMP contained the MMP-2 enzyme sensitive linker and disulfide bond, which could respond to the tumor-overexpressing enzyme MMP-2 and high-level glutathione. While WPH promoted tumor penetration and acid-responsive drug release by modifying cellular penetrating peptides and polymerizing L-histidine. PMP/WPH-NPs exhibited outstanding features including longer blood circulation time, promoted tumor-specific accumulation, enhanced tumor penetration and efficient escape from lysosomes. Subsequently, the model drug paclitaxel (PTX), widely used in the tumor chemotherapy, was encapsulated into PMP/WPH-NPs via an emulsion solvent evaporation method. Within a short period of time, PTX-PMP/WPH-NP in simulated tumor cellular microenvironment could release 8 times more PTX than that in the physiological environment, demonstrating a good potential in tumor cell-specific burst drug release. In addition, PTX-PMP/WPH-NPs exhibited stronger anti-tumor activity than PTX in vitro and in vivo, which also had good biocompatibility according to the hemolysis assay and H&E staining. In summary, our work has succeeded in designing an original polymeric nanoplatform for programmed burst drug release based on the tailored tumor targeting delivery system. This new approach would facilitate the clinical translation of more anti-tumor nanomedicines. STATEMENT OF SIGNIFICANCE: Biomaterials responsive to the tumor-specific stimulus has conventionally used in the targeted-delivery of anti-tumor drugs. However, the levels of common stimulus are not uniformly distributed and not high enough to effectively trigger drug release. In an effort to achieve a better specific drug release and promote the chemotherapeutic efficacy, we constructed a cascade responsive nanoplatform with tumor cell-specific drug burst release profile. The tailored biomaterial could overcome the bio-barriers in vivo and succeeded in the programmed burst drug release based on the tumor cell-specific delivery of chemotherapeutics.
Collapse
Affiliation(s)
- He Xi
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Xu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Aiping Fang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuan Li
- Department of Gastroenterology, Hospital of Chengdu Office of People's Government of Tibetan autonomous Region, Sichuan, China
| | - Zhiying Huang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shugang Qin
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen Xiao
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guohong Li
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Miaomiao Tian
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Na Fan
- Sichuan University West China Hospital
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Abbasi A, Rahbar Saadat T, Rahbar Saadat Y. Microbial exopolysaccharides-β-glucans-as promising postbiotic candidates in vaccine adjuvants. Int J Biol Macromol 2022; 223:346-361. [PMID: 36347372 DOI: 10.1016/j.ijbiomac.2022.11.003] [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: 09/29/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/08/2022]
Abstract
The urgent task of creating new, enhanced adjuvants is closely related to our comprehension of their mechanisms of action. A few adjuvants have shown sufficient efficacy and low toxicity to be allowed for use in human vaccines, despite the fact that they have a long history and an important function. Adjuvants have long been used without a clear understanding of how precisely they augment the immune response. The rational production of stronger and safer adjuvants has been impeded by this lack of information, which necessitates more mechanistic research to support the development of vaccines. Carbohydrate structures-polygalactans, fructans, β-D-glucans, α-D-glucans, D-galactose, and D-glucose-are desirable candidates for the creation of vaccine adjuvants and immunomodulators because they serve important functions in nature and are often biocompatible, safe, and well tolerated. In this review, we have discussed recent advances in microbial-derived carbohydrate-based adjuvants, their immunostimulatory activity, and the implications of this for vaccine development, along with the critical view on the microbial sources, chemical composition, and biosynthetic pathways.
Collapse
Affiliation(s)
- Amin Abbasi
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Yalda Rahbar Saadat
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Li M, Wang J, Guo P, Jin L, Tan X, Zhang Z, Zhanghuang C, Mi T, Liu J, Wang Z, Wu X, Wei G, He D. Exosome mimetics derived from bone marrow mesenchymal stem cells ablate neuroblastoma tumor in vitro and in vivo. BIOMATERIALS ADVANCES 2022; 142:213161. [PMID: 36308859 DOI: 10.1016/j.bioadv.2022.213161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/26/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
PURPOSE To develop exosome-mimetics derived from bone marrow mesenchymal stem cells (EM) as a novel nanoscale drug delivery system(nanoDDS) with improved tumor targeting activity, therapeutic effect, and biosafety, and to evaluate the therapeutic effect of doxorubicin loaded EM (EM-Dox) on neuroblastoma (NB) in vitro and in vivo. METHODS EM was prepared by serial extrusion of bone marrow mesenchymal stem cells (BMSCs), ammonium sulfate gradient method was used to promote the active loading of doxorubicin, and EM-Dox was obtained after removal of free doxorubicin by dialysis. The obtained EM and EM-Dox were characterized by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), Western Blot assay(WB), and the yield of exosomes and EM was further compared. Confocal fluorescent microscopy was used to verify the uptake of EM-Dox and free doxorubicin (Free-Dox) by NB cells. CCK-8 assay, cell cycle assay, and cell apoptosis assay were used to evaluate the antitumor effect of EM-Dox on NB cells in vitro. In addition, the targeted therapeutic effect and biosafety of EM-Dox against NB were evaluated in tumor-bearing nude mice. RESULTS TEM, NTA, and WB verified that both EM and EM-Dox feature highly similar morphology, size and marker protein expression in comparison with naturally occurred exosomes, but the particle size of EM-Dox increased slightly after loading doxorubicin. The protein yield and particle yield of EM-Dox were 16.8 and 26.3-folds higher than those of exosomes, respectively. Confocal fluorescent microscopy showed that EM and doxorubicin had a definite co-localization. EM-Dox was readily internalized in two well-established human NB cell lines. The intracellular content of doxorubicin in cells treated with EM-Dox was significantly higher than that treated with Free-Dox. CCK-8 assay and flow cytometry confirmed that EM-Dox could inhibit NB cell proliferation, induce G2/M phase cell cycle arrest, and promote NB cell apoptosis in vitro. In vivo bioluminescence imaging results demonstrated that EM-Dox effectively targets NB tumors in vivo. Compared with Free-Dox, EM-Dox had a significantly increased inhibitory effect against NB tumor proliferation and progression in vivo, without inducing any myocardial injury. CONCLUSIONS EM-Dox showed significantly increased anti-tumor activity in comparison with free doxorubicin in vitro and in vivo, and scalable EMs may represent a new class of NanoDDS that can potentially replace naturally occurred exosomes in preclinical or clinical translations.
Collapse
Affiliation(s)
- Mujie Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jinkui Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Peng Guo
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, PR China
| | - Liming Jin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaojun Tan
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Zhaoxia Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Chenghao Zhanghuang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Tao Mi
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jiayan Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Zhang Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xin Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
12
|
Cholujova D, Koklesova L, Lukacova Bujnakova Z, Dutkova E, Valuskova Z, Beblava P, Matisova A, Sedlak J, Jakubikova J. In vitro and ex vivo anti-myeloma effects of nanocomposite As 4S 4/ZnS/Fe 3O 4. Sci Rep 2022; 12:17961. [PMID: 36289430 PMCID: PMC9606304 DOI: 10.1038/s41598-022-22672-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/18/2022] [Indexed: 02/06/2023] Open
Abstract
Nanoparticles in medicine can integrate actively targeted imaging agents and drug delivery vehicles, and combining multiple types of therapeutics in a single particle has numerous advantages, especially in multiple myeloma. MM is an incurable hematological disorder characterized by clonal proliferation of plasma cells in the bone marrow. In this study, we evaluated the anti-myeloma activity of 3 nanocomposites (3NPs): As4S4/ZnS/Fe3O4 (1:4:1), As4S4/ZnS/Fe3O4 with folic acid (FA), and As4S4/ZnS/Fe3O4 with FA and albumin with reduced survival MM cell lines and primary MM samples by each of 3NP. Cytotoxic effects of 3NPs were associated with caspase- and mitochondria-dependent apoptosis induction and reduced c-Myc expression. Modulation of cell cycle regulators, such as p-ATM/ATM and p-ATR/ATR, and increases in p-Chk2, cyclin B1, and histones were accompanied by G2/M arrest triggered by 3NPs. In addition, 3NPs activated several myeloma-related signaling, including JNK1/2/3, ERK1/2 and mTOR. To overcome BM microenvironment-mediated drug resistance, nanocomposites retained its anti-MM activity in the presence of stroma. 3NPs significantly decreased the stem cell-like side population in MM cells, even in the context of stroma. We observed strong synergistic effects of 3NPs combined with lenalidomide, pomalidomide, or melphalan, suggesting the potential of these combinations for future clinical studies.
Collapse
Affiliation(s)
- Danka Cholujova
- grid.420087.90000 0001 2106 1943Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505 Slovakia ,grid.419303.c0000 0001 2180 9405Centre for Advanced Materials Application, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84511 Slovakia
| | - Lenka Koklesova
- grid.420087.90000 0001 2106 1943Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505 Slovakia ,grid.7634.60000000109409708Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Slovakia
| | - Zdenka Lukacova Bujnakova
- grid.419303.c0000 0001 2180 9405Department of Mechanochemistry, Institute of Geotechnics, Slovak Academy of Sciences, Watsonova 45, Košice, 04001 Slovakia
| | - Erika Dutkova
- grid.419303.c0000 0001 2180 9405Department of Mechanochemistry, Institute of Geotechnics, Slovak Academy of Sciences, Watsonova 45, Košice, 04001 Slovakia
| | - Zuzana Valuskova
- grid.420087.90000 0001 2106 1943Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505 Slovakia
| | - Patricia Beblava
- grid.420087.90000 0001 2106 1943Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505 Slovakia
| | - Anna Matisova
- grid.420087.90000 0001 2106 1943Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505 Slovakia
| | - Jan Sedlak
- grid.420087.90000 0001 2106 1943Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505 Slovakia
| | - Jana Jakubikova
- grid.420087.90000 0001 2106 1943Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505 Slovakia ,grid.419303.c0000 0001 2180 9405Centre for Advanced Materials Application, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84511 Slovakia
| |
Collapse
|
13
|
Multi-functionalized single-walled carbon nanotubes as delivery carriers: promote the targeting uptake and antitumor efficacy of doxorubicin. J INCL PHENOM MACRO 2022. [DOI: 10.1007/s10847-022-01163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
14
|
Guan Q, Zhou LL, Dong YB. Metalated covalent organic frameworks: from synthetic strategies to diverse applications. Chem Soc Rev 2022; 51:6307-6416. [PMID: 35766373 DOI: 10.1039/d1cs00983d] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Covalent organic frameworks (COFs) are a class of organic crystalline porous materials discovered in the early 21st century that have become an attractive class of emerging materials due to their high crystallinity, intrinsic porosity, structural regularity, diverse functionality, design flexibility, and outstanding stability. However, many chemical and physical properties strongly depend on the presence of metal ions in materials for advanced applications, but metal-free COFs do not have these properties and are therefore excluded from such applications. Metalated COFs formed by combining COFs with metal ions, while retaining the advantages of COFs, have additional intriguing properties and applications, and have attracted considerable attention over the past decade. This review presents all aspects of metalated COFs, from synthetic strategies to various applications, in the hope of promoting the continued development of this young field.
Collapse
Affiliation(s)
- Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China.
| | - Le-Le Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China.
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China.
| |
Collapse
|
15
|
Nteli P, Bajwa DE, Politakis D, Michalopoulos C, Kefala-Narin A, Efstathopoulos EP, Gazouli M. Nanomedicine approaches for treatment of hematologic and oncologic malignancies. World J Clin Oncol 2022; 13:553-566. [PMID: 36157164 PMCID: PMC9346428 DOI: 10.5306/wjco.v13.i7.553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/10/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of death worldwide. Nowadays, the therapies are inadequate and spur demand for improved technologies. Rapid growth in nanotechnology and novel nanomedicine products represents an opportunity to achieve sophisticated targeting strategies and multi-functionality. Nanomedicine is increasingly used to develop new cancer diagnosis and treatment methods since this technology can modulate the biodistribution and the target site accumulation of chemotherapeutic drugs, thereby reducing their toxicity. Cancer nanotechnology and cancer immunotherapy are two parallel themes that have emerged over the last few decades while searching for a cure for cancer. Immunotherapy is revolutionizing cancer treatment, as it can achieve unprecedented responses in advanced-stage patients, including complete cures and long-term survival. A deeper understanding of the human immune system allows the establishment of combination regimens in which immunotherapy is combined with other treatment modalities (as in the case of the nanodrug Ferumoxytol). Furthermore, the combination of gene therapy approaches with nanotechnology that aims to silence or express cancer-relevant genes via one-time treatment is gradually progressing from bench to bedside. The most common example includes lipid-based nanoparticles that target VEGF-Α and KRAS pathways. This review focuses on nanoparticle-based platforms utilized in recent advances aiming to increase the efficacy of currently available cancer therapies. The insights provided and the evidence obtained in this paper indicate a bright future ahead for immuno-oncology applications of engineering nanomedicines.
Collapse
Affiliation(s)
- Polyxeni Nteli
- Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Danae Efremia Bajwa
- Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Dimitrios Politakis
- Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Charalampos Michalopoulos
- Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Anastasia Kefala-Narin
- Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Efstathios P Efstathopoulos
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, General University Hospital Attikon, Athens12462, Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| |
Collapse
|
16
|
Nanomaterial-Based Drug Delivery System Targeting Lymph Nodes. Pharmaceutics 2022; 14:pharmaceutics14071372. [PMID: 35890268 PMCID: PMC9325242 DOI: 10.3390/pharmaceutics14071372] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/28/2022] [Accepted: 06/22/2022] [Indexed: 02/06/2023] Open
Abstract
The lymphatic system plays an indispensable role in humoral balance, lipid metabolism, and immune regulation. The lymph nodes (LNs) are known as the primary sites of tumor metastasis and the metastatic LNs largely affected the prognosis of the patiens. A well-designed lymphatic-targeted system favors disease treatment as well as vaccination efficacy. In recent years, development of nanotechnologies and emerging biomaterials have gained increasing attention in developing lymph-node-targeted drug-delivery systems. By mimicking the endogenous macromolecules or lipid conjugates, lymph-node-targeted nanocarries hold potential for disease diagnosis and tumor therapy. This review gives an introduction to the physiological functions of LNs and the roles of LNs in diseases, followed by a review of typical lymph-node-targeted nanomaterial-based drug-delivery systems (e.g., liposomes, micelles, inorganic nanomaterials, hydrogel, and nanocapsules). Future perspectives and conclusions concerned with lymph-node-targeted drug-delivery systems are also provided.
Collapse
|
17
|
Song Q, Yu H, Han J, Qiang Lv JL, Yang H. Exosomes in urological diseases - Biological functions and clinical applications. Cancer Lett 2022; 544:215809. [PMID: 35777716 DOI: 10.1016/j.canlet.2022.215809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/02/2022]
Abstract
Exosomes are extracellular vesicles with a variety of biological functions that exist in various biological body fluids and exert their functions through proteins, nucleic acids, lipids, and metabolites. Recent discoveries have revealed the functional and biomarker roles of miRNAs in urological diseases, including benign diseases and malignancies. Exosomes have several uses in the diagnosis, treatment, and monitoring of urological diseases, especially cancer. Proteins and nucleic acids can be used as alternative biomarkers for detecting urological diseases. Additionally, exosomes can be detected in most body fluids, thereby avoiding pathogenesis. More importantly, for urological tumors, exosomes display a higher sensitivity than circulating tumor cells and tumor-derived DNA in body fluid biopsies because of their low immunogenicity and high stability. These advantages have made it a research hotspot in recent years. In this review, we focus on the biological characteristics and functions of exosomes and summarize their advantages and the latest progress in the diagnosis and treatment of urological diseases.
Collapse
Affiliation(s)
- Qiang Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China
| | - Hao Yu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China
| | - Jie Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China
| | - Jiancheng Lv Qiang Lv
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China.
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China.
| |
Collapse
|
18
|
Charó N, Jerez H, Tatti S, Romero EL, Schattner M. The Anti-Inflammatory Effect of Nanoarchaeosomes on Human Endothelial Cells. Pharmaceutics 2022; 14:pharmaceutics14040736. [PMID: 35456570 PMCID: PMC9027062 DOI: 10.3390/pharmaceutics14040736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 01/14/2023] Open
Abstract
Archaebacterias are considered a unique source of novel biomaterials of interest for nanomedicine. In this perspective, the effects of nanoarchaeosomes (ARC), which are nanovesicles prepared from polar lipids extracted from the extreme halophilic Halorubrum tebenquinchense, on human umbilical vein endothelial cells (HUVEC) were investigated in physiological and under inflammatory static conditions. Upon incubation, ARC (170 nm mean size, −41 mV ζ) did not affect viability, cell proliferation, and expression of intercellular adhesion molecule-1 (ICAM-1) and E-selectin under basal conditions, but reduced expression of both molecules and secretion of IL-6 induced by lypopolysaccharide (LPS), Pam3CSK4 or Escherichia coli. Such effects were not observed with TNF-α or IL-1β stimulation. Interestingly, ARC significantly decreased basal levels of von Willebrand factor (vWF) and levels induced by all stimuli. None of these parameters was altered by liposomes of hydrogenated phosphatidylcholine and cholesterol of comparable size and concentration. Only ARC were endocytosed by HUVEC and reduced mRNA expression of ICAM-1 and vWF via NF-ĸB and ERK1/2 in LPS-stimulated cells. This is the first report of the anti-inflammatory effect of ARC on endothelial cells and our data suggest that its future use in vascular disease may hopefully be of particular interest.
Collapse
Affiliation(s)
- Nancy Charó
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina;
| | - Horacio Jerez
- Center for Research and Development in Nanomedicines (CIDEN), National University of Quilmes, Roque Saenz Peña, Bernal 1876, Argentina;
| | - Silvio Tatti
- Department of Obstetrics and Gynecology, Clinical Hospital, Av. Córdoba 2351, Buenos Aires 1120, Argentina;
| | - Eder Lilia Romero
- Center for Research and Development in Nanomedicines (CIDEN), National University of Quilmes, Roque Saenz Peña, Bernal 1876, Argentina;
- Correspondence: (E.L.R.); (M.S.)
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina;
- Correspondence: (E.L.R.); (M.S.)
| |
Collapse
|
19
|
Maldonado-Valderrama J, Yang Y, Jiménez-Guerra M, del Castillo-Santaella T, Ramos J, Martín-Molina A. Complexation of DNA with Thermoresponsive Charged Microgels: Role of Swelling State and Electrostatics. Gels 2022; 8:184. [PMID: 35323297 PMCID: PMC8955517 DOI: 10.3390/gels8030184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/16/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
Micro- and nanogels are being increasingly used to encapsulate bioactive compounds. Their soft structure allows large loading capacity while their stimuli responsiveness makes them extremely versatile. In this work, the complexation of DNA with thermoresponsive microgels is presented. To this end, PEGylated charged microgels based on poly-N-isopropylacrylamide have been synthesized, allowing one to explore the electrostatics of the complexation. Cationic microgels complexate spontaneously by electrostatic attraction to oppositely charged DNA as demonstrated by electrophoretic mobility of the complexes. Then, Langmuir monolayers reveal an increased interaction of DNA with swollen microgels (20 °C). Anionic microgels require the presence of multivalent cations (Ca2+) to promote the complexation, overcoming the electrostatic repulsion with negatively charged DNA. Then again, Langmuir monolayers evidence their complexation at the surface. However, the presence of Ca2+ seems to induce profound changes in the interaction and surface conformation of anionic microgels. These alterations are further explored by measuring adsorbed films with the pendant drop technique. Conformational changes induced by Ca2+ on the structure of the microgel can ultimately affect the complexation with DNA and should be considered in the design. The combination of microstructural and surface properties for microgels offers a new perspective into complexation of DNA with soft particles with biomedical applications.
Collapse
Affiliation(s)
- Julia Maldonado-Valderrama
- Departamento de Física Aplicada, Universidad de Granada, Campus de Fuentenueva sn, 18071 Granada, Granada, Spain; (J.M.-V.); (Y.Y.); (M.J.-G.); (T.d.C.-S.)
- Excellence Research Unit “Modelling Nature” (MNat), Universidad de Granada, 18071 Granada, Granada, Spain
| | - Yan Yang
- Departamento de Física Aplicada, Universidad de Granada, Campus de Fuentenueva sn, 18071 Granada, Granada, Spain; (J.M.-V.); (Y.Y.); (M.J.-G.); (T.d.C.-S.)
| | - Maykel Jiménez-Guerra
- Departamento de Física Aplicada, Universidad de Granada, Campus de Fuentenueva sn, 18071 Granada, Granada, Spain; (J.M.-V.); (Y.Y.); (M.J.-G.); (T.d.C.-S.)
| | - Teresa del Castillo-Santaella
- Departamento de Física Aplicada, Universidad de Granada, Campus de Fuentenueva sn, 18071 Granada, Granada, Spain; (J.M.-V.); (Y.Y.); (M.J.-G.); (T.d.C.-S.)
| | - José Ramos
- IQLIT Emulsiones Poliméricas S.L.U., Autovía Tarragona-Salou Km 3,8., 43110 La Canonja, Tarragona, Spain;
| | - Alberto Martín-Molina
- Departamento de Física Aplicada, Universidad de Granada, Campus de Fuentenueva sn, 18071 Granada, Granada, Spain; (J.M.-V.); (Y.Y.); (M.J.-G.); (T.d.C.-S.)
- Instituto Carlos I de Física Teórica y Computacional, Universdad de Granada, 18071 Granada, Granada, Spain
| |
Collapse
|
20
|
Bariwal J, Ma H, Altenberg GA, Liang H. Nanodiscs: a versatile nanocarrier platform for cancer diagnosis and treatment. Chem Soc Rev 2022; 51:1702-1728. [PMID: 35156110 DOI: 10.1039/d1cs01074c] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer therapy is a significant challenge due to insufficient drug delivery to the cancer cells and non-selective killing of healthy cells by most chemotherapy agents. Nano-formulations have shown great promise for targeted drug delivery with improved efficiency. The shape and size of nanocarriers significantly affect their transport inside the body and internalization into the cancer cells. Non-spherical nanoparticles have shown prolonged blood circulation half-lives and higher cellular internalization frequency than spherical ones. Nanodiscs are desirable nano-formulations that demonstrate enhanced anisotropic character and versatile functionalization potential. Here, we review the recent development of theranostic nanodiscs for cancer mitigation ranging from traditional lipid nanodiscs encased by membrane scaffold proteins to newer nanodiscs where either the membrane scaffold proteins or the lipid bilayers themselves are replaced with their synthetic analogues. We first discuss early cancer detection enabled by nanodiscs. We then explain different strategies that have been explored to carry a wide range of payloads for chemotherapy, cancer gene therapy, and cancer vaccines. Finally, we discuss recent progress on organic-inorganic hybrid nanodiscs and polymer nanodiscs that have the potential to overcome the inherent instability problem of lipid nanodiscs.
Collapse
Affiliation(s)
- Jitender Bariwal
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Hairong Ma
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Hongjun Liang
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
21
|
Li SS, Zhang CM, Wu JD, Liu C, Liu ZP. A branched small molecule-drug conjugate nanomedicine strategy for the targeted HCC chemotherapy. Eur J Med Chem 2022; 228:114037. [PMID: 34883290 DOI: 10.1016/j.ejmech.2021.114037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Off-target toxicity is one of the main challenges faced by anticancer chemotherapeutics. For tumor targeted and precision chemotherapy, we take the advantages of the ligand directed tumor active targeting of small molecule drug conjugates (SMDCs) and the passive tumor targeting of nanoparticles via the enhanced penetration and retention (EPR) effects, put forward a branched small molecule drug conjugate (BSMDC) nanomedicine design concept. In a proof of concept, we used pentaerythritol as the branched moiety, galactosamine (GalN) as the hepatocellular carcinoma (HCC) directing ligands, PTX as a payload, and a stearoyl moiety as the amphiphilic property adjusting group, designed and synthesized BSMDC 1 and prepared its NPs. In cellular level, the BSMDC 1 NPs targeted asialoglycoprotein receptor (ASGPR)-overexpressing HepG2 cells, were effectively taken up in the cells and released in tumor microenvironments, inhibited the HepG2 cell proliferation, arrested HepG2 cell in G2/M phase and induced tumor cell apoptosis. In HepG2 xenograft nude mice, the BSMDC 1 NPs were high specific to target the tumor and demonstrated a higher antitumor efficiency than BSMDC 1, having no apparent influences on mice body weights and major organs, supporting our BSMDC nanomedicine design concept. Therefore, this new strategy may find applications for cancer targeted and precision chemotherapy.
Collapse
Affiliation(s)
- Sha-Sha Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Cheng-Mei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Jing-De Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Chao Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| | - Zhao-Peng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
22
|
Lafuente-Gómez N, Latorre A, Milán-Rois P, Rodriguez Diaz C, Somoza Á. Stimuli-responsive nanomaterials for cancer treatment: boundaries, opportunities and applications. Chem Commun (Camb) 2021; 57:13662-13677. [PMID: 34874370 DOI: 10.1039/d1cc05056g] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Small molecule drugs, including most chemotherapies, are rapidly degraded and/or eliminated from the body, which is why high doses of these drugs are necessary, potentially producing toxic effects. Several types of nanoparticles loaded with anti-cancer drugs have been designed to overcome the disadvantages of conventional therapies. Modified nanoparticles can circulate for a long time, thus improving the solubility and biodistribution of drugs. Furthermore, they also allow the controlled release of the payload once its target tissue has been reached. These mechanisms can reduce the exposure of healthy tissues to chemotherapeutics, since the drugs are only released in the presence of specific tumour stimuli. Overall, these properties can improve the effectiveness of treatments while reducing undesirable side effects. In this article, we review the recent advances in stimuli-responsive albumin, gold and magnetic nanostructures for controlled anti-cancer drug delivery. These nanostructures were designed to release drugs in response to different internal and external stimuli of the cellular environment, including pH, redox, light and magnetic fields. We also describe various examples of applications of these nanomaterials. Overall, we shed light on the properties, potential clinical translation and limitations of stimuli-responsive nanoparticles for cancer treatment.
Collapse
Affiliation(s)
- Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Ana Latorre
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Paula Milán-Rois
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Ciro Rodriguez Diaz
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain. .,Unidad Asociada al Centro Nacional de Biotecnología (CSIC), 28049 Madrid, Spain
| |
Collapse
|
23
|
Guan Q, Zhou LL, Dong YB. Ferroptosis in cancer therapeutics: a materials chemistry perspective. J Mater Chem B 2021; 9:8906-8936. [PMID: 34505861 DOI: 10.1039/d1tb01654g] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Ferroptosis, distinct from apoptosis, is a regulated form of cell death caused by lipid peroxidation that has attracted extensive research interest since it was first defined in 2012. Over the past five years, an increasing number of studies have revealed the close relationship between ferroptosis and materials chemistry, in particular nanobiotechnology, and have concluded that nanotechnology-triggered ferroptosis is an efficient and promising antitumor strategy that provides an alternative therapeutic approach, especially for apoptosis-resistant tumors. In this review, we summarize recent advances in ferroptosis-induced tumor therapy at the intersection of materials chemistry, redox biology, and tumor biology. The biological features and molecular mechanisms of ferroptosis are first outlined, followed by a summary of the feasible strategies to induce ferroptosis using nanomaterials and the applications of ferroptosis in combined tumor therapy. Finally, the existing challenges and future development directions in this emerging field are discussed, with the aim of promoting the progress of ferroptosis-based oncotherapy in materials science and nanoscience and enriching the antitumor arsenal.
Collapse
Affiliation(s)
- Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China.
| | - Le-Le Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China.
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
24
|
Tang L, He S, Yin Y, Liu H, Hu J, Cheng J, Wang W. Combination of Nanomaterials in Cell-Based Drug Delivery Systems for Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13111888. [PMID: 34834304 PMCID: PMC8621332 DOI: 10.3390/pharmaceutics13111888] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cell-based drug delivery systems have shown tremendous advantages in cancer treatment due to their distinctive properties. For instance, delivery of therapeutics using tumor-tropic cells like neutrophils, lymphocytes and mesenchymal stem cells can achieve specific tumor targeting due to the "Trojan Horse" effect. Other circulatory cells like erythrocytes and platelets can greatly improve the circulation time of nanoparticles due to their innate long circulation property. Adipocytes, especially cancer-associated adipocytes, play key roles in tumor development and metabolism, therefore, adipocytes are regarded as promising bio-derived nanoplatforms for anticancer targeted drug delivery. Nanomaterials are important participants in cell-based drug delivery because of their unique physicochemical characteristics. Therefore, the integration of various nanomaterials with different cell types will endow the constructed delivery systems with many attractive properties due to the merits of both. In this review, a number of strategies based on nanomaterial-involved cell-mediated drug delivery systems for cancer treatment will be summarized. This review discusses how nanomaterials can be a benefit to cell-based therapies and how cell-derived carriers overcome the limitations of nanomaterials, which highlights recent advancements and specific biomedical applications based on nanomaterial-mediated, cell-based drug delivery systems.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Jingyi Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 211166, China
- Correspondence: (J.C.); (W.W.)
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
- Correspondence: (J.C.); (W.W.)
| |
Collapse
|
25
|
Guo P, Busatto S, Huang J, Morad G, Moses MA. A facile magnetic extrusion method for preparing endosome-derived vesicles for cancer drug delivery. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2008326. [PMID: 34924915 PMCID: PMC8680268 DOI: 10.1002/adfm.202008326] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Indexed: 06/14/2023]
Abstract
To date, the scaled-up manufacturing and efficient drug loading of exosomes are two existing challenges limiting the clinical translation of exosome-based drug delivery. Herein, we developed a facile magnetic extrusion method for preparing endosome-derived vesicles, also known as exosome mimetics (EMs), which share the same biological origin and similar morphology, composition, and biofunctions with native exosomes. The high yield and consistency of this magnetic extrusion method help to overcome the manufacturing bottleneck in exosome research. Moreover, the proposed standardized multi-step method readily facilitates the ammonium sulfate gradient approach to actively load chemodrugs such as doxorubicin into EMs. The engineered EMs developed and tested here exhibit comparable drug delivery properties as do native exosomes and potently inhibit tumor growth by delivering doxorubicin in an orthotopic breast tumor model. These findings demonstrate that EMs can be prepared in a facile and scaled-up manner as a promising biological nanomedicine for cancer drug delivery.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Sara Busatto
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Jing Huang
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Golnaz Morad
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
- Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, United States
| | - Marsha A. Moses
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
26
|
Busatto S, Morad G, Guo P, Moses MA. The role of extracellular vesicles in the physiological and pathological regulation of the blood-brain barrier. FASEB Bioadv 2021; 3:665-675. [PMID: 34485835 PMCID: PMC8409556 DOI: 10.1096/fba.2021-00045] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are a subclass of biological nanoparticles secreted by most cell types. Once secreted, EVs can travel long distances to deliver their content to target cells thereby playing a key role in cell-to-cell communication and supporting both physiological and pathological processes. In recent years, the functional versatility of EVs has come to be more widely appreciated. Their heterogeneous structure encloses solubilized bioactive cargoes including proteins and nucleic acids. EVs mirror the secreting cell in composition therefore representing a novel source of diagnostic and prognostic biomarkers. Moreover, due to their unique structure, EVs constitute a promising class of biocompatible nanovehicles for drug delivery as well. Importantly, and of burgeoning interest, is the fact that EVs have the intrinsic ability to breach biological barriers including the complex blood-brain barrier (BBB), whose restrictive nature represents a significant therapeutic challenge. EVs have been shown to contribute to the progression of a variety of brain diseases including metastatic brain cancer, neurodegenerative diseases, and acute pathologies including infections and ischemia. In this review, the role of EVs in the maintenance and regulation of the BBB under normal physiological and pathologic conditions are discussed. Applications of EVs as therapeutic and diagnostic tools in the treatment of diseases that affect the central nervous system are presented as are limitations hindering their broad translation and potential solutions to resolve them.
Collapse
Affiliation(s)
- Sara Busatto
- Vascular Biology ProgramBoston Children's HospitalBostonMAUSA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Golnaz Morad
- Department of Surgical OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Peng Guo
- Vascular Biology ProgramBoston Children's HospitalBostonMAUSA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Marsha A. Moses
- Vascular Biology ProgramBoston Children's HospitalBostonMAUSA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
27
|
Mochizuki C, Nakamura J, Nakamura M. Development of Non-Porous Silica Nanoparticles towards Cancer Photo-Theranostics. Biomedicines 2021; 9:73. [PMID: 33451074 PMCID: PMC7828543 DOI: 10.3390/biomedicines9010073] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/09/2021] [Indexed: 02/07/2023] Open
Abstract
Nanoparticles have demonstrated several advantages for biomedical applications, including for the development of multifunctional agents as innovative medicine. Silica nanoparticles hold a special position among the various types of functional nanoparticles, due to their unique structural and functional properties. The recent development of silica nanoparticles has led to a new trend in light-based nanomedicines. The application of light provides many advantages for in vivo imaging and therapy of certain diseases, including cancer. Mesoporous and non-porous silica nanoparticles have high potential for light-based nanomedicine. Each silica nanoparticle has a unique structure, which incorporates various functions to utilize optical properties. Such advantages enable silica nanoparticles to perform powerful and advanced optical imaging, from the in vivo level to the nano and micro levels, using not only visible light but also near-infrared light. Furthermore, applications such as photodynamic therapy, in which a lesion site is specifically irradiated with light to treat it, have also been advancing. Silica nanoparticles have shown the potential to play important roles in the integration of light-based diagnostics and therapeutics, termed "photo-theranostics". Here, we review the recent development and progress of non-porous silica nanoparticles toward cancer "photo-theranostics".
Collapse
Affiliation(s)
- Chihiro Mochizuki
- Department of Organ Anatomy & Nanomedicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan; (C.M.); (J.N.)
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Junna Nakamura
- Department of Organ Anatomy & Nanomedicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan; (C.M.); (J.N.)
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Michihiro Nakamura
- Department of Organ Anatomy & Nanomedicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan; (C.M.); (J.N.)
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
28
|
Wang G, Zhang Z, Wang R, Zhu Z. A Review on Heat Transfer of Nanofluids by Applied Electric Field or Magnetic Field. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2386. [PMID: 33260487 PMCID: PMC7760193 DOI: 10.3390/nano10122386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 12/27/2022]
Abstract
Nanofluids are considered to be a next-generation heat transfer medium due to their excellent thermal performance. To investigate the effect of electric fields and magnetic fields on heat transfer of nanofluids, this paper analyzes the mechanism of thermal conductivity enhancement of nanofluids, the chaotic convection and the heat transfer enhancement of nanofluids in the presence of an applied electric field or magnetic field through the method of literature review. The studies we searched showed that applied electric field and magnetic field can significantly affect the heat transfer performance of nanofluids, although there are still many different opinions about the effect and mechanism of heat transfer. In a word, this review is supposed to be useful for the researchers who want to understand the research state of heat transfer of nanofluids.
Collapse
Affiliation(s)
| | | | - Ruijin Wang
- School of Mechanical Engineering, Hangzhou Dianzi University, Hangzou 310000, China; (G.W.); (Z.Z.)
| | - Zefei Zhu
- School of Mechanical Engineering, Hangzhou Dianzi University, Hangzou 310000, China; (G.W.); (Z.Z.)
| |
Collapse
|
29
|
Mukherjee K. The 'What, Why, Where and How' of Delivering a Drug. Trends Pharmacol Sci 2020; 41:679-680. [PMID: 32946771 PMCID: PMC7497775 DOI: 10.1016/j.tips.2020.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Kusumika Mukherjee
- Trends in Pharmacological Sciences, Cell Press, 50 Hampshire Street, Cambridge, MA 02139, USA.
| |
Collapse
|
30
|
Cerqueira SR, Ayad NG, Lee JK. Neuroinflammation Treatment via Targeted Delivery of Nanoparticles. Front Cell Neurosci 2020; 14:576037. [PMID: 33192321 PMCID: PMC7555434 DOI: 10.3389/fncel.2020.576037] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
The lack of effective treatments for most neurological diseases has prompted the search for novel therapeutic options. Interestingly, neuroinflammation is emerging as a common feature to target in most CNS pathologies. Recent studies suggest that targeted delivery of small molecules to reduce neuroinflammation can be beneficial. However, suboptimal drug delivery to the CNS is a major barrier to modulate inflammation because neurotherapeutic compounds are currently being delivered systemically without spatial or temporal control. Emerging nanomaterial technologies are providing promising and superior tools to effectively access neuropathological tissue in a controlled manner. Here we highlight recent advances in nanomaterial technologies for drug delivery to the CNS. We propose that state-of-the-art nanoparticle drug delivery platforms can significantly impact local CNS bioavailability of pharmacological compounds and treat neurological diseases.
Collapse
Affiliation(s)
- Susana R Cerqueira
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States.,The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Nagi G Ayad
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States.,The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Jae K Lee
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States.,The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|