1
|
Lei K, Zhou L, Dan M, Yang F, Jian T, Xin J, Yu Z, Wang Y. Trojan Horse Delivery Strategies of Natural Medicine Monomers: Challenges and Limitations in Improving Brain Targeting. Pharmaceutics 2025; 17:280. [PMID: 40142943 PMCID: PMC11945504 DOI: 10.3390/pharmaceutics17030280] [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: 01/23/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Central nervous system (CNS) diseases, such as brain tumors, Alzheimer's disease, and Parkinson's disease, significantly impact patients' quality of life and impose substantial economic burdens on society. The blood-brain barrier (BBB) limits the effective delivery of most therapeutic drugs, especially natural products, despite their potential therapeutic effects. The Trojan Horse strategy, using nanotechnology to disguise drugs as "cargo", enables them to bypass the BBB, enhancing targeting and therapeutic efficacy. This review explores the applications of natural products in the treatment of CNS diseases, discusses the challenges posed by the BBB, and analyzes the advantages and limitations of the Trojan Horse strategy. Despite the existing technical challenges, future research is expected to enhance the application of natural drugs in CNS treatment by integrating nanotechnology, improving delivery mechanisms, and optimizing targeting characteristics.
Collapse
Affiliation(s)
- Kelu Lei
- Department of Pharmacy, Ya’an People’s Hospital-West China Ya’an Hospital, Sichuan University, Ya’an 625000, China; (K.L.); (M.D.); (F.Y.); (T.J.); (J.X.)
| | - Lanyu Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Min Dan
- Department of Pharmacy, Ya’an People’s Hospital-West China Ya’an Hospital, Sichuan University, Ya’an 625000, China; (K.L.); (M.D.); (F.Y.); (T.J.); (J.X.)
| | - Fei Yang
- Department of Pharmacy, Ya’an People’s Hospital-West China Ya’an Hospital, Sichuan University, Ya’an 625000, China; (K.L.); (M.D.); (F.Y.); (T.J.); (J.X.)
| | - Tiantian Jian
- Department of Pharmacy, Ya’an People’s Hospital-West China Ya’an Hospital, Sichuan University, Ya’an 625000, China; (K.L.); (M.D.); (F.Y.); (T.J.); (J.X.)
| | - Juan Xin
- Department of Pharmacy, Ya’an People’s Hospital-West China Ya’an Hospital, Sichuan University, Ya’an 625000, China; (K.L.); (M.D.); (F.Y.); (T.J.); (J.X.)
| | - Zhigang Yu
- Department of Pharmacy, Ya’an People’s Hospital-West China Ya’an Hospital, Sichuan University, Ya’an 625000, China; (K.L.); (M.D.); (F.Y.); (T.J.); (J.X.)
| | - Yue Wang
- Department of Pharmacy, Ya’an People’s Hospital-West China Ya’an Hospital, Sichuan University, Ya’an 625000, China; (K.L.); (M.D.); (F.Y.); (T.J.); (J.X.)
| |
Collapse
|
2
|
Kim K, Chejara MR, Yoon B, Park MH. Gold nanorod-based smart platform for efficient cellular uptake and combination therapy. RSC Adv 2024; 14:27385-27393. [PMID: 39205927 PMCID: PMC11350633 DOI: 10.1039/d4ra06051b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Gold nanorods (GNRs) have received much attention as potential drug-delivery vehicles because of their various advantages such as good biocompatibility, passive targeting, responsiveness to stimuli, and easy post-functionalization by surface modification. However, the drug structure might be changed for loading into GNRs, making it difficult to load various drugs, and the space to contain drugs is small, making it difficult to deliver sufficient drugs required for treatment compared with other porous materials. Herein, we report an amphiphilic polymer-coated GNR platform for chemo- and photothermal combination therapy. Amphiphilic polymers comprise hydrophobic alkyl chains for drug encapsulation, polyethylene glycol for biocompatibility, and folic acid for cancer targeting. GNRs generate heat energy under near-infrared light irradiation, promoting controlled drug release, and inducing cellular uptake by deforming the cell membrane. On-demand release behavior was traced with Nile red, and targeting and delivery efficiency were confirmed with paclitaxel through cellular experiments. This GNR-based platform enables combination therapy with passive and active targeting to enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Kibeom Kim
- Department of Chemistry and Life Science, Sahmyook University Seoul 01795 South Korea
| | - Mamta Ramgopal Chejara
- Department of Chemistry and Life Science, Sahmyook University Seoul 01795 South Korea
- Department of Convergence Science, Sahmyook University Seoul 01795 South Korea
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University Seoul 01795 South Korea
| | - Been Yoon
- Department of Convergence Science, Sahmyook University Seoul 01795 South Korea
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University Seoul 01795 South Korea
| | - Myoung-Hwan Park
- Department of Chemistry and Life Science, Sahmyook University Seoul 01795 South Korea
- Department of Convergence Science, Sahmyook University Seoul 01795 South Korea
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University Seoul 01795 South Korea
| |
Collapse
|
3
|
Zheng J, Jiang X, Li Y, Gao J. Inorganic nanoparticle-integrated mesenchymal stem cells: A potential biological agent for multifaceted applications. MedComm (Beijing) 2023; 4:e313. [PMID: 37533768 PMCID: PMC10390757 DOI: 10.1002/mco2.313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 08/04/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based therapies are flourishing. MSCs could be used as potential therapeutic agents for regenerative medicine due to their own repair function. Meanwhile, the natural predisposition toward inflammation or injury sites makes them promising carriers for targeted drug delivery. Inorganic nanoparticles (INPs) are greatly favored for their unique properties and potential applications in biomedical fields. Current research has integrated INPs with MSCs to enhance their regenerative or antitumor functions. This model also allows the in vivo fate tracking of MSCs in multiple imaging modalities, as many INPs are also excellent contrast agents. Thus, INP-integrated MSCs would be a multifunctional biologic agent with great potential. In this review, the current roles performed by the integration of INPs with MSCs, including (i) enhancing their repair and regeneration capacity via the improvement of migration, survival, paracrine, or differentiation properties, (ii) empowering tumor-killing ability through agent loaded or hyperthermia, and (iii) conferring traceability are summarized. An introduction of INP-integrated MSCs for simultaneous treatment and tracking is also included. The promising applications of INP-integrated MSCs in future treatments are emphasized and the challenges to their clinical translation are discussed.
Collapse
Affiliation(s)
- Juan‐Juan Zheng
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Xin‐Chi Jiang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Yao‐Sheng Li
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Jian‐Qing Gao
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Hangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
4
|
Wang X, Sun Y, Wangpraseurt D. Engineered photoresponsive biohybrids for tumor therapy. SMART MEDICINE 2023; 2:e20220041. [PMID: 39188274 PMCID: PMC11235730 DOI: 10.1002/smmd.20220041] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/30/2023] [Indexed: 08/28/2024]
Abstract
Engineered biohybrids have recently emerged as innovative biomimetic platforms for cancer therapeutic applications. Particularly, engineered photoresponsive biohybrids hold tremendous potential against tumors due to their intriguing biomimetic properties, photoresponsive ability, and enhanced biotherapeutic functions. In this review, the design principles of engineered photoresponsive biohybrids and their latest progresses for tumor therapy are summarized. Representative engineered photoresponsive biohybrids are highlighted including biomolecules-associated, cell membrane-based, eukaryotic cell-based, bacteria-based, and algae-based photoresponsive biohybrids. Representative tumor therapeutic modalities of the engineered photoresponsive biohybrids are presented, including photothermal therapy, photodynamic therapy, synergistic therapy, and tumor therapy combined with tissue regeneration. Moreover, the challenges and future perspectives of these photoresponsive biohybrids for clinical practice are discussed.
Collapse
Affiliation(s)
- Xiaocheng Wang
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Yazhi Sun
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Daniel Wangpraseurt
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
- Scripps Institution of OceanographyUniversity of California San DiegoSan DiegoCaliforniaUSA
| |
Collapse
|
5
|
Granja A, Lima-Sousa R, Alves CG, de Melo-Diogo D, Nunes C, Sousa CT, Correia IJ, Reis S. Multifunctional targeted solid lipid nanoparticles for combined photothermal therapy and chemotherapy of breast cancer. BIOMATERIALS ADVANCES 2023; 151:213443. [PMID: 37146526 DOI: 10.1016/j.bioadv.2023.213443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 04/23/2023] [Indexed: 05/07/2023]
Abstract
Photothermal therapy has emerged as a new promising strategy for the management of cancer, either alone or combined with other therapeutics, such as chemotherapy. The use of nanoparticles for multimodal therapy can improve treatment performance and reduce drug doses and associated side effects. Here we propose the development of a novel multifunctional nanosystem based on solid lipid nanoparticles co-loaded with gold nanorods and mitoxantrone and functionalized with folic acid for dual photothermal therapy and chemotherapy of breast cancer. Nanoparticles were produced using an economically affordable method and presented suitable physicochemical properties for tumor passive accumulation. Upon Near-Infrared irradiation (808 nm, 1.7 W cm-2, 5 min), nanoparticles could effectively mediate a temperature increase of >20 °C. Moreover, exposure to light resulted in an enhanced release of Mitoxantrone. Furthermore, nanoparticles were non-hemolytic and well tolerated by healthy cells even at high concentrations. The active targeting strategy was found to be successful, as shown by the greater accumulation of the functionalized nanoparticles in MCF-7 cells. Finally, the combined effects of chemotherapy, light-induced drug release and photothermal therapy significantly enhanced breast cancer cell death. Overall, these results demonstrate that the developed lipid nanosystem is an efficient vehicle for breast cancer multimodal therapy.
Collapse
Affiliation(s)
- Andreia Granja
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Rita Lima-Sousa
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Cátia G Alves
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Duarte de Melo-Diogo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Cláudia Nunes
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Célia T Sousa
- IFIMUP and Departamento de Física e Astronomia da Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal; Departamento de Física Aplicada, Facultad de Ciencias, Universidad Autónoma de Madrid (UAM), Campus de Cantoblanco, C/ Francisco Tomás y Valiente, 7, M 12 604 - 28049 Madrid, Spain
| | - Ilídio J Correia
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, 3030-790 Coimbra, Portugal.
| | - Salette Reis
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
6
|
Kola P, Nagesh PKB, Roy PK, Deepak K, Reis RL, Kundu SC, Mandal M. Innovative nanotheranostics: Smart nanoparticles based approach to overcome breast cancer stem cells mediated chemo- and radioresistances. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023:e1876. [PMID: 36600447 DOI: 10.1002/wnan.1876] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
The alarming increase in the number of breast cancer patients worldwide and the increasing death rate indicate that the traditional and current medicines are insufficient to fight against it. The onset of chemo- and radioresistances and cancer stem cell-based recurrence make this problem harder, and this hour needs a novel treatment approach. Competent nanoparticle-based accurate drug delivery and cancer nanotheranostics like photothermal therapy, photodynamic therapy, chemodynamic therapy, and sonodynamic therapy can be the key to solving this problem due to their unique characteristics. These innovative formulations can be a better cargo with fewer side effects than the standard chemotherapy and can eliminate the stability problems associated with cancer immunotherapy. The nanotheranostic systems can kill the tumor cells and the resistant breast cancer stem cells by novel mechanisms like local hyperthermia and reactive oxygen species and prevent tumor recurrence. These theranostic systems can also combine with chemotherapy or immunotherapy approaches. These combining approaches can be the future of anticancer therapy, especially to overcome the breast cancer stem cells mediated chemo- and radioresistances. This review paper discusses several novel theranostic systems and smart nanoparticles, their mechanism of action, and their modifications with time. It explains their relevance and market scope in the current era. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Prithwish Kola
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Rui Luis Reis
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
7
|
Zhang L. The Role of Mesenchymal Stem Cells in Modulating the Breast Cancer Microenvironment. Cell Transplant 2023; 32:9636897231220073. [PMID: 38135917 DOI: 10.1177/09636897231220073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023] Open
Abstract
The role of mesenchymal stem cells (MSCs) in the breast tumor microenvironment (TME) is significant and multifaceted. MSCs are recruited to breast tumor sites through molecular signals released by tumor sites. Once in the TME, MSCs undergo polarization and interact with various cell populations, including immune cells, cancer-associated fibroblasts (CAFs), cancer stem cells (CSCs), and breast cancer cells. In most cases, MSCs play roles in breast cancer therapeutic resistance, but there is also evidence that indicates their abilities to sensitize cancer cells to chemotherapy and radiotherapy. MSCs possess inherent regenerative and homing properties, making them attractive candidates for cell-based therapies. Therefore, MSCs can be engineered to express therapeutic molecules or deliver anti-cancer agents directly to tumor sites. Unraveling the intricate relationship between MSCs and the breast TME has the potential to uncover novel therapeutic targets and advance our understanding of breast cancer biology.
Collapse
Affiliation(s)
- Luxiao Zhang
- Department of Surgical Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
8
|
Swellable hollow periodic mesoporous organosilica capsules with ultrahigh loading capacity for hydrophobic drugs. J Colloid Interface Sci 2023; 630:266-273. [DOI: 10.1016/j.jcis.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/24/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
|
9
|
Tu Z, Karnoub AE. Mesenchymal stem/stromal cells in breast cancer development and management. Semin Cancer Biol 2022; 86:81-92. [PMID: 36087857 DOI: 10.1016/j.semcancer.2022.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) encompass a heterogeneous population of fibroblastic progenitor cells that reside in multiple tissues around the body. They are endowed with capacities to differentiate into multiple connective tissue lineages, including chondrocytes, adipocytes, and osteoblasts, and are thought to function as trophic cells recruited to sites of injury and inflammation where they contribute to tissue regeneration. In keeping with these roles, MSCs also to home to sites of breast tumorigenesis, akin to their migration to wounds, and participate in tumor stroma formation. Mounting evidence over the past two decades has described the critical regulatory roles for tumor-associated MSCs in various aspects of breast tumor pathogenesis, be it tumor initiation, growth, angiogenesis, tumor microenvironment formation, immune evasion, cancer cell migration, invasion, survival, therapeutic resistance, dissemination, and metastatic colonization. In this review, we present a brief summary of the role of MSCs in breast tumor development and progression, highlight some of the molecular frameworks underlying their pro-malignant contributions, and present evidence of their promising utility in breast cancer therapy.
Collapse
Affiliation(s)
- Zhenbo Tu
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Boston Veterans Affairs Research Institute, West Roxbury, MA 02132, USA.
| |
Collapse
|
10
|
Alle M, Sharma G, Lee SH, Kim JC. Next-generation engineered nanogold for multimodal cancer therapy and imaging: a clinical perspectives. J Nanobiotechnology 2022; 20:222. [PMID: 35778747 PMCID: PMC9250257 DOI: 10.1186/s12951-022-01402-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the significant threats to human life. Although various latest technologies are currently available to treat cancer, it still accounts for millions of death each year worldwide. Thus, creating a need for more developed and novel technologies to combat this deadly condition. Nanoparticles-based cancer therapeutics have offered a promising approach to treat cancer effectively while minimizing adverse events. Among various nanoparticles, nanogold (AuNPs) are biocompatible and have proved their efficiency in treating cancer because they can reach tumors via enhanced permeability and retention effect. The size and shape of the AuNPs are responsible for their diverse therapeutic behavior. Thus, to modulate their therapeutic values, the AuNPs can be synthesized in various shapes, such as spheres, cages, flowers, shells, prisms, rods, clusters, etc. Also, attaching AuNPs with single or multiple targeting agents can facilitate the active targeting of AuNPs to the tumor tissue. The AuNPs have been much explored for photothermal therapy (PTT) to treat cancer. In addition to PTT, AuNPs-based nanoplatforms have been investigated for combinational multimodal therapies in the last few years, including photodynamic therapy, chemotherapy, radiotherapy, immunotherapy, etc., to ablate cancer cells. Thus, the present review focuses on the recent advancements in the functionalization of AuNPs-based nanoconstructs for cancer imaging and therapy using combinatorial multimodal approaches to treat various cancers.
Collapse
Affiliation(s)
- Madhusudhan Alle
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Garima Sharma
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Seung-Hwan Lee
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Department of Forest Biomaterials Engineering, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Jin-Chul Kim
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
11
|
Khosravi N, Pishavar E, Baradaran B, Oroojalian F, Mokhtarzadeh A. Stem cell membrane, stem cell-derived exosomes and hybrid stem cell camouflaged nanoparticles: A promising biomimetic nanoplatforms for cancer theranostics. J Control Release 2022; 348:706-722. [PMID: 35732250 DOI: 10.1016/j.jconrel.2022.06.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
Nanomedicine research has advanced dramatically in recent decades. Nonetheless, traditional nanomedicine faces significant obstacles such as the low concentration of the drug at target sites and accelerated removal of the drug from blood circulation. Various techniques of nanotechnology, including cell membrane coating, have been developed to address these challenges and to improve targeted distribution and redcue cell membrane-mediated immunogenicity. Recently, stem cell (SC) membranes, owing to their immunosuppressive and regenerative properties, have grabbed attention as attractive therapeutic carriers for targeting specific tissues or organs. Bioengineering strategies that combine synthetic nanoparticles (NPs) with SC membranes, because of their homing potential and tumor tropism, have recently received a lot of publicity. Several laboratory experiments and clinical trials have indicated that the benefits of SC-based technologies are mostly related to the effects of SC-derived exosomes (SC-Exos). Exosomes are known as nano-sized extracellular vehicles (EVs) that deliver particular bioactive molecules for cell-to-cell communication. In this regard, SC-derived exosome membranes have recently been employed to improve the therapeutic capability of engineered drug delivery vehicles. Most recently, for further enhancing NPs' functionality, a new coating approach has been offered that combines membranes from two separate cells. These hybrid membrane delivery vehicles have paved the way for the development of biocompatible, high-efficiency, biomimetic NPs with varying hybrid capabilities that can overcome the drawbacks of present NP-based treatment techniques. This review explores stem cell membranes, SC-Exos, and hybrid SC-camouflaged NPs preparation methods and their importance in cancer therapy.
Collapse
Affiliation(s)
- Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Pishavar
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
12
|
Pu XQ, Ju XJ, Liu WY, Liu YQ, Li XJ, Li Y, Xie R, Wang W, Liu Z, Chu LY. Stimulus-Responsive Nanoparticle-Integrated Dissolving Microneedles for Synergetic Chemo-Photothermal Therapy of Superficial Skin Tumors. Ind Eng Chem Res 2022. [DOI: 10.1021/acs.iecr.2c00831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Xing-Qun Pu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Xiao-Jie Ju
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Wen-Ying Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Yu-Qiong Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Xin-Jiao Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Yao Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Rui Xie
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Wei Wang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Zhuang Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Liang-Yin Chu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| |
Collapse
|
13
|
Zhang H, Feng Y, Xie X, Song T, Yang G, Su Q, Li T, Li S, Wu C, You F, Liu Y, Yang H. Engineered Mesenchymal Stem Cells as a Biotherapy Platform for Targeted Photodynamic Immunotherapy of Breast Cancer. Adv Healthc Mater 2022; 11:e2101375. [PMID: 34981675 DOI: 10.1002/adhm.202101375] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/12/2021] [Indexed: 01/10/2023]
Abstract
Interleukin-12 (IL12) is a pleiotropic cytokine with promising prospects for cancer immunotherapy. Though IL12 gene-based therapy can overcome the fatal hurdle of severe systemic toxicity, targeted delivery and tumor-located expression of IL12 gene remain the challenging issues yet to be solved. Photo-immunotherapy emerging as a novel and precise therapeutic strategy, which elaborately combines immune-activating agents with light-triggered photosensitizers for potentiated anticancer efficacy. Herein, an engineered stem cell-based biotherapy platform (MB/IL12-MSCs) incorporating immune gene plasmid IL12 (pIL12) and photosensitizer methylene blue (MB) is developed to realize tumor-homing delivery of therapeutic agents and photo-immunotherapy efficacy enhancement. The biotherapy platform retained tumor-tropic migration and penetration functions, which improved the intratumoral distribution of therapeutic agents, thereby promoting photodynamic effects and reinforcing immune responses. Importantly, MB/IL12-MSCs restricted the expression and distribution of IL12 at tumor site, which minimized potential toxicity while eliciting sufficient anticancer immunity. In noteworthy, activation of immunity induced by MB/IL12-MSCs established long-term systemic immunologic memory to prevent tumor relapse. The MB/IL12-MSCs outperform their monotherapy counterparts in breast tumor models, and the growth of tumor significantly arrested as well as re-challenging abscopal tumor growth slowdown. Collectively, this work reveals that MSCs-based strategy may advance more efficient, durable, and safer cancer photo-immunotherapy.
Collapse
Affiliation(s)
- Hanxi Zhang
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Yi Feng
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Xiaoxue Xie
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Ting Song
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Geng Yang
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Qingqing Su
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Tingting Li
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Shun Li
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Chunhui Wu
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine No. 39 Shi‐er‐qiao Road Chengdu Sichuan 610072 P. R. China
| | - Yiyao Liu
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine No. 39 Shi‐er‐qiao Road Chengdu Sichuan 610072 P. R. China
| | - Hong Yang
- Department of Biophysics School of Life Science and Technology University of Electronic Science and Technology of China Chengdu Sichuan 610054 P. R. China
| |
Collapse
|
14
|
Meng Z, Zhang Y, Zhou X, Ji J, Liu Z. Nanovaccines with cell-derived components for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114107. [PMID: 34995678 DOI: 10.1016/j.addr.2021.114107] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/16/2021] [Accepted: 12/29/2021] [Indexed: 12/13/2022]
Abstract
Cancer nanovaccines as one of immunotherapeutic approaches are able to attack tumors by stimulating tumor-specific immunological responses. However, there still exist multiple challenges to be tackled for cancer nanovaccines to evoke potent antitumor immunity. Particularly, the administration of exogenous materials may cause the off-target immunotherapy responses. In recent years, biomimetic nanovaccines by using cell lysates, cell-derived nanovesicles, or extracted cell membranes as the functional components have received extensive attention. Such nanovaccines based on cell-derived components would show many unique advantages including inherent biocompatibility and the ability to trigger immune responses against a range of tumor-associated antigens. In this review article, we will introduce the recent research progresses of those cell-derived biomimetic nanovaccines for cancer immunotherapy, and discuss the perspectives and challenges associated with the future clinical translation of these emerging vaccine platforms.
Collapse
|
15
|
Ning P, Chen Y, Bai Q, Xu C, Deng C, Cheng Q, Cheng Y. Multimodal Imaging-Guided Spatiotemporal Tracking of Photosensitive Stem Cells for Breast Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7551-7564. [PMID: 35107006 DOI: 10.1021/acsami.1c13072] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Stem cell therapy has shown great potential in treating a wide range of diseases including cancer. The real-time tracking of stem cells with high spatiotemporal resolution and stable imaging signals remains the bottleneck to evaluate and monitor therapeutic outcomes once transplanted into patients. Here, we developed a photosensitive mesenchymal stem cell (MSC) loaded with mesoporous silica-coated gold nanostars (MGNSs) integrated with indocyanine green for spatiotemporal tracking and imaging-guided photothermal therapy (PTT) in treating breast cancers. The MGNS served as a stable imaging probe with multifunctional properties for photoacoustic imaging (PAI), fluorescence imaging, and PT imaging. Owing to the excellent PT stability of MGNSs, long-term three-dimensional (3D) PAI was achieved to monitor stem cells in real time at the tumor site, while the tumor structure was imaged using 3D B-mode ultrasound imaging. PAI revealed that the photosensitive stem cells reached the widest distribution area at the tumor site post 24 h of intratumoral injection, which was further confirmed via two-dimensional (2D) PT and fluorescence imaging. With this optimal cell distribution window, in vivo studies showed that the photosensitive stem cells via both intratumoral and intravenous injections successfully inhibited breast cancer cell growth and decreased the tumor recurrence rate post PTT. Our results support that this photo-integrated platform with stable optical properties is promising to achieve real-time tracking and measure the cell distribution quantitatively with high spatiotemporal resolution for stem cell therapy.
Collapse
Affiliation(s)
- Peng Ning
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Yingna Chen
- Institute of Acoustics, School of Physics Science and Engineering, Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, 1239 Siping Road, Shanghai 200092, China
- The Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Qianwen Bai
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Chang Xu
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Cuijun Deng
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, 1239 Siping Road, Shanghai 200092, China
- The Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Yu Cheng
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| |
Collapse
|
16
|
Zheng J, Cheng X, Zhang H, Bai X, Ai R, Shao L, Wang J. Gold Nanorods: The Most Versatile Plasmonic Nanoparticles. Chem Rev 2021; 121:13342-13453. [PMID: 34569789 DOI: 10.1021/acs.chemrev.1c00422] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gold nanorods (NRs), pseudo-one-dimensional rod-shaped nanoparticles (NPs), have become one of the burgeoning materials in the recent years due to their anisotropic shape and adjustable plasmonic properties. With the continuous improvement in synthetic methods, a variety of materials have been attached around Au NRs to achieve unexpected or improved plasmonic properties and explore state-of-the-art technologies. In this review, we comprehensively summarize the latest progress on Au NRs, the most versatile anisotropic plasmonic NPs. We present a representative overview of the advances in the synthetic strategies and outline an extensive catalogue of Au-NR-based heterostructures with tailored architectures and special functionalities. The bottom-up assembly of Au NRs into preprogrammed metastructures is then discussed, as well as the design principles. We also provide a systematic elucidation of the different plasmonic properties associated with the Au-NR-based structures, followed by a discussion of the promising applications of Au NRs in various fields. We finally discuss the future research directions and challenges of Au NRs.
Collapse
Affiliation(s)
- Jiapeng Zheng
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Xizhe Cheng
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Han Zhang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Xiaopeng Bai
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Ruoqi Ai
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Lei Shao
- Beijing Computational Science Research Center, Beijing 100193, China
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| |
Collapse
|
17
|
Liu H, Deng S, Han L, Ren Y, Gu J, He L, Liu T, Yuan ZX. Mesenchymal stem cells, exosomes and exosome-mimics as smart drug carriers for targeted cancer therapy. Colloids Surf B Biointerfaces 2021; 209:112163. [PMID: 34736220 DOI: 10.1016/j.colsurfb.2021.112163] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells with the capacity to differentiate into several cell types under appropriate conditions. They also possess remarkable antitumor features that make them a novel choice to treat cancers. Accumulating evidence suggest that the MSCs-derived extracellular vesicles, known as exosomes, play an essential role in the therapeutic effects of MSCs mainly by carrying biologically active factors. However, limitations such as low yield of exosomes and difficulty in isolation and purification hinder their clinical applications. To overcome these issues, research on development of exosome-mimics has attracted great attention. This systematic review represents, to the best of our knowledge, the first thorough evaluations of the innate antineoplastic features of MSCs-derived exosomes or exosome-mimics, the methods of drug loading, application as drug delivery system and their impacts on targeted cancer therapy. Importantly, we dissect the commonalities and differences as well as address the shortcomings of work accumulated over the last two decades and discuss how this information can serve as a guide map for optimal experimental design implementation ultimately aiding the effective transition into clinical trials.
Collapse
Affiliation(s)
- Hongmei Liu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Shichen Deng
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Yan Ren
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China.
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, Australia.
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
18
|
Takayama Y, Kusamori K, Nishikawa M. Mesenchymal stem/stromal cells as next-generation drug delivery vehicles for cancer therapeutics. Expert Opin Drug Deliv 2021; 18:1627-1642. [PMID: 34311638 DOI: 10.1080/17425247.2021.1960309] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Drug delivery to solid tumors remains a significant therapeutic challenge. Mesenchymal stem/stromal cells (MSCs) home to tumor tissues and can be employed as tumor targeted drug/gene delivery vehicles. Reportedly, therapeutic gene- or anti-cancer drug-loaded MSCs have shown remarkable anti-tumor effects in preclinical studies, and some clinical trials for assessing therapeutic MSCs in patients with cancer have been registered. AREAS COVERED In the present review, we first discuss the source and interdonor heterogeneity of MSCs, their tumor-homing mechanism, and the route of MSC administration in MSC-based cancer therapy. We then summarize the therapeutic applications of MSCs as a drug delivery vehicle for therapeutic genes or anti-cancer drugs and the drug delivery mechanism from drug-loaded MSCs to cancer cells. EXPERT OPINION Although numerous preclinical studies have revealed significant anti-tumor effects, several clinical trials assessing MSC-based cancer gene therapy have failed to demonstrate corroborative results, documenting limited therapeutic effects. Notably, a successful clinical outcome with MSC-based cancer therapy would require the interdonor heterogeneity of administered MSCs to be resolved, along with improved tumor-homing efficiency and optimized drug delivery efficiency from MSCs to cancer cells.
Collapse
Affiliation(s)
- Yukiya Takayama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| | - Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| |
Collapse
|
19
|
Granja A, Pinheiro M, Sousa CT, Reis S. Gold nanostructures as mediators of hyperthermia therapies in breast cancer. Biochem Pharmacol 2021; 190:114639. [PMID: 34077740 DOI: 10.1016/j.bcp.2021.114639] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer is the leading cause of cancer-related deaths among women. Due to the limitations of the current therapeutics, new treatment options are needed. Hyperthermia is a promising approach to improve breast cancer therapy, particularly when combined with chemo and radiotherapy. This area has gained more attention following association with nanotechnology, with the emergence of modalities, such as photothermal therapy (PTT). PTT is a simple, minimally invasive technique that requires a near infrared (NIR) light source and a PTT agent. Gold nanostructures are excellent PTT agents as they offer biocompatibility, versatility, high photothermal conversion efficiency, imaging contrast and an easily-modified surface. In this review, we describe the molecular basis and the current clinical aspects of hyperthermia-based therapies. The emergent area of nanoparticle-induced hyperthermia will be explored, in particular gold nanostructure-mediated PTT, focusing on recent preclinical studies for breast cancer management.
Collapse
Affiliation(s)
- Andreia Granja
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Marina Pinheiro
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Célia T Sousa
- IFIMUP and Dep. Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua Campo Alegre 687, 4169 - 007 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| |
Collapse
|
20
|
Sahu A, Jeon J, Lee MS, Yang HS, Tae G. Nanozyme Impregnated Mesenchymal Stem Cells for Hepatic Ischemia-Reperfusion Injury Alleviation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:25649-25662. [PMID: 33974389 DOI: 10.1021/acsami.1c03027] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mesenchymal stem cell (MSC) based therapy holds great potential for treating numerous diseases owing to their capability to heal injured tissue/organs through paracrine factors secretion and immunomodulation. Despite the high hopes, the low viability of transplanted cells in the injured tissues due to the elevated oxidative stress levels remains the largest obstacle in MSC-based cell therapy. To achieve desired therapeutic efficiency, the survival of the transplanted MSCs in the high oxidative stress environment needs to be ensured. Herein, we proposed the use of a ROS-scavenging nanozyme to protect transplanted MSCs from oxidative stress-mediated apoptosis and thereby improve the therapeutic effect. Prussian blue (PB) nanoparticles as a biocompatible ROS-scavenging nanozyme were incorporated into the MSCs without affecting the stemness and differentiation potential of MSCs. The nanozyme impregnation significantly improved the survival of MSCs in a high oxidative stress condition as well as augmented their paracrine effect and anti-inflammatory properties, resulting in a profound therapeutic effect in vivo in the liver ischemia-reperfusion (I/R) injury animal model. Our results indicated that the nanozyme incorporation into MSCs is a simple but efficient way to improve the therapeutic potential of MSC-based cell therapy.
Collapse
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jin Jeon
- Department of Nanobiomedical Science and BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Min Suk Lee
- Department of Nanobiomedical Science and BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hee Seok Yang
- Department of Nanobiomedical Science and BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Center for Bio-Medical Engineering Core-Facility, Dankook University, Cheonan, 31116, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| |
Collapse
|
21
|
Wang R, Yan H, Yu A, Ye L, Zhai G. Cancer targeted biomimetic drug delivery system. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
22
|
Jeon J, Lee S, Kim H, Kang H, Youn H, Jo S, Youn B, Kim HY. Revisiting Platinum-Based Anticancer Drugs to Overcome Gliomas. Int J Mol Sci 2021; 22:ijms22105111. [PMID: 34065991 PMCID: PMC8151298 DOI: 10.3390/ijms22105111] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Although there are many patients with brain tumors worldwide, there are numerous difficulties in overcoming brain tumors. Among brain tumors, glioblastoma, with a 5-year survival rate of 5.1%, is the most malignant. In addition to surgical operations, chemotherapy and radiotherapy are generally performed, but the patients have very limited options. Temozolomide is the most commonly prescribed drug for patients with glioblastoma. However, it is difficult to completely remove the tumor with this drug alone. Therefore, it is necessary to discuss the potential of anticancer drugs, other than temozolomide, against glioblastomas. Since the discovery of cisplatin, platinum-based drugs have become one of the leading chemotherapeutic drugs. Although many studies have reported the efficacy of platinum-based anticancer drugs against various carcinomas, studies on their effectiveness against brain tumors are insufficient. In this review, we elucidated the anticancer effects and advantages of platinum-based drugs used in brain tumors. In addition, the cases and limitations of the clinical application of platinum-based drugs are summarized. As a solution to overcome these obstacles, we emphasized the potential of a novel approach to increase the effectiveness of platinum-based drugs.
Collapse
Affiliation(s)
- Jaewan Jeon
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea; (J.J.); (S.J.)
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea;
| | - Sunmi Jo
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea; (J.J.); (S.J.)
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
- Correspondence: (B.Y.); (H.Y.K.); Tel.: +82-51-510-2264 (B.Y.); +82-51-797-3923 (H.Y.K.)
| | - Hae Yu Kim
- Department of Neurosurgery, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea
- Correspondence: (B.Y.); (H.Y.K.); Tel.: +82-51-510-2264 (B.Y.); +82-51-797-3923 (H.Y.K.)
| |
Collapse
|
23
|
Zhang T, Huang T, Su Y, Gao J. Mesenchymal Stem Cells‐Based Targeting Delivery System: Therapeutic Promises and Immunomodulation against Tumor. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Tianyuan Zhang
- Zhejiang Province Key Laboratory of Anti‐Cancer Drug Research College of Pharmaceutical Sciences Zhejiang University 866 Yuhangtang Rd Hangzhou 310058 China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University 866 Yuhangtang Rd Hangzhou 310058 China
| | - Ting Huang
- Zhejiang Province Key Laboratory of Anti‐Cancer Drug Research College of Pharmaceutical Sciences Zhejiang University 866 Yuhangtang Rd Hangzhou 310058 China
| | - Yuanqin Su
- Zhejiang Province Key Laboratory of Anti‐Cancer Drug Research College of Pharmaceutical Sciences Zhejiang University 866 Yuhangtang Rd Hangzhou 310058 China
| | - Jianqing Gao
- Zhejiang Province Key Laboratory of Anti‐Cancer Drug Research College of Pharmaceutical Sciences Zhejiang University 866 Yuhangtang Rd Hangzhou 310058 China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University 866 Yuhangtang Rd Hangzhou 310058 China
- Cancer Center of Zhejiang University 866 Yuhangtang Rd Hangzhou 310058 China
| |
Collapse
|
24
|
Wang Z, Dong J, Zhao Q, Ying Y, Zhang L, Zou J, Zhao S, Wang J, Zhao Y, Jiang S. Gold nanoparticle‑mediated delivery of paclitaxel and nucleic acids for cancer therapy (Review). Mol Med Rep 2020; 22:4475-4484. [PMID: 33173972 PMCID: PMC7646735 DOI: 10.3892/mmr.2020.11580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022] Open
Abstract
Paclitaxel is a potent antineoplastic agent, but poor solubility and resistance have limited its use. Gold nanoparticles (AuNPs) are widely studied as drug carriers because they can be engineered to prevent drug insolubility, carry nucleic acid payloads for gene therapy, target specific tumor cell lines, modulate drug release and amplify photothermal therapy. Consequently, the conjugation of paclitaxel with AuNPs to improve antiproliferative and pro‑apoptotic potency may enable improved clinical outcomes. There are currently a number of different AuNPs under development, including simple drug or nucleic acid carriers and targeted AuNPs that are designed to deliver therapeutic payloads to specific cells. The current study reviewed previous research on AuNPs and the development of AuNP‑based paclitaxel delivery.
Collapse
Affiliation(s)
- Zhiguang Wang
- Frontier Institute of Science and Technology, State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Jianyu Dong
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiaojiajie Zhao
- Institute of Hematological Research, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University Medical College, Nanchang, Jiangxi 330006, P.R. China
| | - Lijie Zhang
- Institute of Hematological Research, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Junrong Zou
- Institute of Urology, The First Affiliated Hospital of Gan'nan Medical University, Ganzhou, Jiangxi 341001, P.R. China
| | - Shuqi Zhao
- Institute of Hematological Research, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Jiuju Wang
- Institute of Hematological Research, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Yuan Zhao
- Institute of Hematological Research, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Shanshan Jiang
- Institute of Hematological Research, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710000, P.R. China
| |
Collapse
|
25
|
Sheng M, Zhang L, West JL, Fu S. Multicolor Electrochromic Dye-Doped Liquid Crystal Yolk-Shell Microcapsules. ACS APPLIED MATERIALS & INTERFACES 2020; 12:29728-29736. [PMID: 32508082 DOI: 10.1021/acsami.0c09354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A new system of yolk-shell microcapsules containing two types of dye-doped liquid crystals was prepared via seed emulsion polymerization in which the synthetic process was mimicking plant respiration. The resulting system demonstrated reversible low voltage-driven switching between multispectral colored and transparent states. Moreover, wearable multicolor electrochromic fibers based on calcium alginate were produced via wet spinning to expand the application of yolk-shell dye-doped liquid crystal microcapsules. In addition to its long-term optical stability, the proposed cells and fibers also have satisfactory driving voltage values of color change (4.8 and 9.0 V), which are far lower than the human body safety voltage (12 V). We believe that the prepared microcapsules and fibers are potentially widely applicable in smart windows, electronic paper, and military camouflage clothing.
Collapse
Affiliation(s)
- Mingfei Sheng
- Key Laboratory of Science & Technology of Eco-Textile, Jiangnan University, Ministry of Education, Wuxi, Jiangsu 214122, P.R. China
| | - Liping Zhang
- Key Laboratory of Science & Technology of Eco-Textile, Jiangnan University, Ministry of Education, Wuxi, Jiangsu 214122, P.R. China
| | - John Lawton West
- Liquid Crystal Institute, Kent State University, Kent, Ohio 44242, United States
| | - Shaohai Fu
- Key Laboratory of Science & Technology of Eco-Textile, Jiangnan University, Ministry of Education, Wuxi, Jiangsu 214122, P.R. China
| |
Collapse
|
26
|
Munteanu R, Onaciu A, Moldovan C, Zimta AA, Gulei D, Paradiso AV, Lazar V, Berindan-Neagoe I. Adipocyte-Based Cell Therapy in Oncology: The Role of Cancer-Associated Adipocytes and Their Reinterpretation as Delivery Platforms. Pharmaceutics 2020; 12:E402. [PMID: 32354024 PMCID: PMC7284545 DOI: 10.3390/pharmaceutics12050402] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer-associated adipocytes have functional roles in tumor development through secreted adipocyte-derived factors and exosomes and also through metabolic symbiosis, where the malignant cells take up the lactate, fatty acids and glutamine produced by the neighboring adipocytes. Recent research has demonstrated the value of adipocytes as cell-based delivery platforms for drugs (or prodrugs), nucleic acids or loaded nanoparticles for cancer therapy. This strategy takes advantage of the biocompatibility of the delivery system, its ability to locate the tumor site and also the predisposition of cancer cells to come in functional contact with the adipocytes from the tumor microenvironment for metabolic sustenance. Also, their exosomal content can be used in the context of cancer stem cell reprogramming or as a delivery vehicle for different cargos, like non-coding nucleic acids. Moreover, the process of adipocytes isolation, processing and charging is quite straightforward, with minimal economical expenses. The present review comprehensively presents the role of adipocytes in cancer (in the context of obese and non-obese individuals), the main methods for isolation and characterization and also the current therapeutic applications of these cells as delivery platforms in the oncology sector.
Collapse
Affiliation(s)
- Raluca Munteanu
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Anca Onaciu
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Cristian Moldovan
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Diana Gulei
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Angelo V. Paradiso
- Oncologia Sperimentale, Istituto Tumori G Paolo II, IRCCS, 70125 Bari, Italy
| | - Vladimir Lazar
- Worldwide Innovative Network for Personalized Cancer Therapy, 94800 Villejuif, France
| | - Ioana Berindan-Neagoe
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| |
Collapse
|
27
|
Cao X, Tan T, Zhu D, Yu H, Liu Y, Zhou H, Jin Y, Xia Q. Paclitaxel-Loaded Macrophage Membrane Camouflaged Albumin Nanoparticles for Targeted Cancer Therapy. Int J Nanomedicine 2020; 15:1915-1928. [PMID: 32256068 PMCID: PMC7090179 DOI: 10.2147/ijn.s244849] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/10/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Melanoma is the most common symptom of aggressive skin cancer, and it has become a serious health concern worldwide in recent years. The metastasis rate of malignant melanoma remains high, and it is highly difficult to cure with the currently available treatment options. Effective yet safe therapeutic options are still lacking. Alternative treatment options are in great demand to improve the therapeutic outcome against advanced melanoma. This study aimed to develop albumin nanoparticles (ANPs) coated with macrophage plasma membranes (RANPs) loaded with paclitaxel (PTX) to achieve targeted therapy against malignant melanoma. METHODS Membrane derivations were achieved by using a combination of hypotonic lysis, mechanical membrane fragmentation, and differential centrifugation to empty the harvested cells of their intracellular contents. The collected membrane was then physically extruded through a 400 nm porous polycarbonate membrane to form macrophage cell membrane vesicles. Albumin nanoparticles were prepared through a well-studied nanoprecipitation process. At last, the two components were then coextruded through a 200 nm porous polycarbonate membrane. RESULTS Using paclitaxel as the model drug, PTX-loaded RANPs displayed significantly enhanced cytotoxicity and apoptosis rates compared to albumin nanoparticles without membrane coating in the murine melanoma cell line B16F10. RANPs also exhibited significantly higher internalization efficiency in B16F10 cells than albumin nanoparticles without a membrane coating. Next, a B16F10 tumor xenograft mouse model was established to explore the biodistribution profiles of RANPs, which showed prolonged blood circulation and selective accumulation at the tumor site. PTX-loaded RANPs also demonstrated greatly improved antitumor efficacy in B16F10 tumor-bearing mouse xenografts. CONCLUSION Albumin-based nanoscale delivery systems coated with macrophage plasma membranes offer a highly promising approach to achieve tumor-targeted therapy following systemic administration.
Collapse
Affiliation(s)
- Xi Cao
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Tingfei Tan
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Dongchun Zhu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Haixia Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Yaru Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Haiyun Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Yong Jin
- Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| |
Collapse
|
28
|
Liu Y, Tang Y, Wu J, Sun J, Liao X, Teng Z, Lu G. Facile synthesis of biodegradable flower-like hydroxyapatite for drug and gene delivery. J Colloid Interface Sci 2020; 570:402-410. [PMID: 32224321 DOI: 10.1016/j.jcis.2020.03.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 11/29/2022]
Abstract
Controlled synthesis of hierarchical hydroxyapatite materials is a hot research topic because of the excellent biocompatibility and bioactivity of the materials. In this study, flower-like hydroxyapatite spheres (FHAPS) were facile synthesized in one pot using Al(OH)3 as a structure-directing agent. The prepared FHAPS comprised nanosheets possessing a uniform diameter of approximately 4 µm. Notably, the FHAPS can be degraded in solutions with a pH of 5.5 for 144 h or incubated with cells for 48 h. In addition, the FHAPS have rough surfaces, which exhibit high loading contents for the anticancer drug doxorubicin (DOX, 9.1%) and siRNA (2.0%). Thus, the FHAPS can effectively deliver DOX into drug-resistant breast cancer cells to exert an excellent killing effect compared with free DOX and transfect siRNA into tumor cells to interfere with the expression of the target protein. Taken together, this work successfully prepared FHAPS via a convenient synthesis route that shows high delivery efficiency for anticancer drugs and siRNA.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Jiangsu 210002, PR China
| | - Yuxia Tang
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Jiangsu 210002, PR China.
| | - Jiang Wu
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Jiangsu 210002, PR China
| | - Jing Sun
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Jiangsu 210002, PR China
| | - Xiang Liao
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Jiangsu 210002, PR China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210046, Jiangsu, PR China.
| | - Guangming Lu
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Jiangsu 210002, PR China.
| |
Collapse
|
29
|
Zhong R, Wang R, Hou X, Song L, Zhang Y. Polydopamine-doped virus-like structured nanoparticles for photoacoustic imaging guided synergistic chemo-/photothermal therapy. RSC Adv 2020; 10:18016-18024. [PMID: 35517193 PMCID: PMC9059141 DOI: 10.1039/d0ra02915g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/28/2020] [Indexed: 11/30/2022] Open
Abstract
The therapeutic diagnosis effect of cancer commonly depends on the cellular uptake efficiency of nanomaterials. However, the morphology of nanomaterials significantly affects cellular uptake capability. Herein, we designed a polydopamine-doped virus-like structured nanoparticle (GNR@HPMO@PVMSN) composed of a gold nanorod (GNR) core, hollow periodic mesoporous organosilica (HPMO) shell and polydopamine-doped virus-like mesoporous silica nanoparticle (PVMSN) outer shell. Compared with conventional gold nanorod@hollow periodic mesoporous organosilica core–shell nanoparticles (GNR@HPMO), GNR@HPMO@PVMSN with its virus-like structure was proved to enhance the efficiency of cellular uptake. GNR@HPMO@PVMSN with the virtues of high photothermal conversion efficiency and good photoacoustic imaging (PAI) ability was expected to be a promising nanotheranostic agent for imaging guided cancer treatment. The experiments in vitro and in vivo proved that GNR@HPMO@PVMSN had good biocompatibility as well as photothermal conversion ability. In addition, DOX loading and pH-/NIR-response DOX release abilities of GNR@HPMO@PVMSN were also verified in vitro. Therefore, the GNR@HPMO@PVMSN offers a promising strategy for PAI directed synergistic chemo-/photothermal therapy, which improves the therapeutic effect of the nanomaterial on tumors. This work explores the effects of rough surfaces on cellular uptake and provides a versatile theranostic platform for biomedical applications. The therapeutic diagnosis effect of cancer commonly depends on the cellular uptake efficiency of nanomaterials.![]()
Collapse
Affiliation(s)
- Rong Zhong
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures
- Fujian Provincial Key Laboratory of Nanomaterials
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
| | - Ruoping Wang
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures
- Fujian Provincial Key Laboratory of Nanomaterials
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
| | - Xuemei Hou
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures
- Fujian Provincial Key Laboratory of Nanomaterials
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
| | - Liang Song
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures
- Fujian Provincial Key Laboratory of Nanomaterials
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
| | - Yun Zhang
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures
- Fujian Provincial Key Laboratory of Nanomaterials
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
| |
Collapse
|
30
|
Ouyang X, Wang X, Kraatz HB, Ahmadi S, Gao J, Lv Y, Sun X, Huang Y. A Trojan horse biomimetic delivery strategy using mesenchymal stem cells for PDT/PTT therapy against lung melanoma metastasis. Biomater Sci 2019; 8:1160-1170. [PMID: 31848537 DOI: 10.1039/c9bm01401b] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cell (MSC)-based biomimetic delivery has been actively explored for drug accumulation and penetration into tumors by taking advantage of the tumor-tropic and penetration properties of MSCs. In this work, we further demonstrated the feasibility of MSC-mediated nano drug delivery, which was characterized by the "Trojan horse"-like transport via an endocytosis-exocytosis-endocytosis process between MSCs and cancer cells. Chlorin e6 (Ce6)-conjugated polydopamine nanoparticles (PDA-Ce6) were developed and loaded into the MSCs. Phototherapeutic agents are safe to the MSCs, and their very low dark toxicity causes no impairment of the inherent properties of MSCs, including tumor-homing ability. The MSCs loaded with PDA-Ce6 (MSC-PDA-Ce6) were able to target and penetrate into tumors and exocytose 60% of the payloads in 72 h. The released PDA-Ce6 NPs could penetrate deep and be re-endocytosed by the cancer cells. MSC-PDA-Ce6 tended to accumulate in the lungs and delivered PDA-Ce6 into the tumors after intravenous injection in the mouse model with lung melanoma metastasis. Phototoxicity can be selectively triggered in the tumors by sequentially treating with near-infrared irradiation to induce photodynamic therapy (PDT) and photothermal therapy (PTT). The MSC-based biomimetic delivery of PDA-Ce6 nanoparticles is a potential method for dual phototherapy against lung melanoma metastasis.
Collapse
Affiliation(s)
- Xumei Ouyang
- Department of Pharmacy, Zhejiang University City College, Hangzhou 310015, China. and Department of Pharmacy, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, China and Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoling Wang
- Department of Pharmacy, Zhejiang University City College, Hangzhou 310015, China. and Department of Pharmacy, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Heinz-Bernhard Kraatz
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada
| | - Soha Ahmadi
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada
| | - Jianqing Gao
- Department of Pharmacy, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuanyuan Lv
- Department of Pharmacy, Zhejiang University City College, Hangzhou 310015, China.
| | - Xiaoyi Sun
- Department of Pharmacy, Zhejiang University City College, Hangzhou 310015, China.
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai, 201203, China.
| |
Collapse
|
31
|
Teng Z, Li W, Tang Y, Elzatahry A, Lu G, Zhao D. Mesoporous Organosilica Hollow Nanoparticles: Synthesis and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1707612. [PMID: 30285290 DOI: 10.1002/adma.201707612] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 05/18/2018] [Indexed: 05/22/2023]
Abstract
Hollow periodic mesoporous organosilicas (PMOs) with molecularly homogeneous organic functional groups in the inorganic pore walls are attracting more and more attention due to the high surface areas, tunable pore sizes, low densities, large cavities in the center, permeable thin shells, and versatile organic-inorganic hybrid frameworks, which make them promising in a variety of applications including adsorption, catalysis, drug delivery, and nanotheranostics. Herein, recent advances in the synthesis of hollow PMO nanoparticles with various organic moieties are summarized, and the mechanism and new insights of synthesis approaches, including hard-core templating methods, liquid-interface assembly methods, and the interfacial reassembly and transformation strategy are discussed in-depth. Meanwhile, the design principles, properties, and synthetic strategies for some smart hollow architectures such as multishelled hollow PMOs, yolk-shell structured PMOs, and nonspherical hollow PMOs are discussed. Moreover, the typical applications of hollow PMO nanomaterials as nanoreactors for chemical transformations and nanoplatforms for biomedicine are summarized. Finally, the challenges and prospects for the future development of hollow PMOs are described.
Collapse
Affiliation(s)
- Zhaogang Teng
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, Jiangsu, P.R. China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, Jiangsu, P.R. China
| | - Wei Li
- Department of Chemistry, Laboratory of Advanced Materials, and iChEM, Fudan University, Shanghai, 200433, P.R. China
| | - Yuxia Tang
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, Jiangsu, P.R. China
| | - Ahmed Elzatahry
- Materials Science and Technology Program, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar
| | - Guangming Lu
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, Jiangsu, P.R. China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, Jiangsu, P.R. China
| | - Dongyuan Zhao
- Department of Chemistry, Laboratory of Advanced Materials, and iChEM, Fudan University, Shanghai, 200433, P.R. China
| |
Collapse
|
32
|
Neelgund GM, Okolie MC, Williams FK, Oki A. Ag 2S nanocrystallites deposited over polyamidoamine grafted carbon nanotubes: An efficient NIR active photothermal agent. MATERIALS CHEMISTRY AND PHYSICS 2019; 234:32-37. [PMID: 32123461 PMCID: PMC7051053 DOI: 10.1016/j.matchemphys.2019.05.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A novel NIR (near infrared) active photothermal agent, CNTs-PAMAM-Ag2S has been synthesized by covalent grafting of polyamidoamine (PAMAM) to carbon nanotubes (CNTs) and subsequent deposition of Ag2S nanocrystallites. The potential of CNTs-PAMAM-Ag2S as a NIR active photothermal agent was preliminarily accessed by its electronic absorption spectrum measured in UV-vis-NIR region. The CNTs-PAMAM-Ag2S exhibited excellent photothermal effect and photothermal conversion efficiency of 26% under illumination with 980 nm laser, the efficiency was found to be higher than popular gold and copper based photothermal agents. Temperature attained by CNTs-PAMAM-Ag2S during measurement of its photothermal effect was 64.7 °C at 1 g/mL, which far exceeds the temperature tolerance level of cancer cells. So that CNTs-PAMAM-Ag2S could destroy the cancer cells in an effective manner. Furthermore, it was found the linear dependence of photothermal effect of CNTs-PAMAM-Ag2S over its concentration. CNTs-PAMAM-Ag2S possessed excellent stability against photo-bleaching and photo-corrosiveness. In consideration of its outstanding photothermal effect and photothermal conversion efficiency, CNTs-PAMAM-Ag2S could be a promising photothermal agent to employ in future photothermal therapy.
Collapse
|
33
|
Kwon S, Yoo KH, Sym SJ, Khang D. Mesenchymal stem cell therapy assisted by nanotechnology: a possible combinational treatment for brain tumor and central nerve regeneration. Int J Nanomedicine 2019; 14:5925-5942. [PMID: 31534331 PMCID: PMC6681156 DOI: 10.2147/ijn.s217923] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) intrinsically possess unique features that not only help in their migration towards the tumor-rich environment but they also secrete versatile types of secretomes to induce nerve regeneration and analgesic effects at inflammatory sites. As a matter of course, engineering MSCs to enhance their intrinsic abilities is growing in interest in the oncology and regenerative field. However, the concern of possible tumorigenesis of genetically modified MSCs prompted the development of non-viral transfected MSCs armed with nanotechnology for more effective cancer and regenerative treatment. Despite the fact that a large number of successful studies have expanded our current knowledge in tumor-specific targeting, targeting damaged brain site remains enigmatic due to the presence of a blood–brain barrier (BBB). A BBB is a barrier that separates blood from brain, but MSCs with intrinsic features of transmigration across the BBB can efficiently deliver desired drugs to target sites. Importantly, MSCs, when mediated by nanoparticles, can further enhance tumor tropism and can regenerate the damaged neurons in the central nervous system through the promotion of axon growth. This review highlights the homing and nerve regenerative abilities of MSCs in order to provide a better understanding of potential cell therapeutic applications of non-genetically engineered MSCs with the aid of nanotechnology.
Collapse
Affiliation(s)
- Song Kwon
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
| | - Kwai Han Yoo
- Department of Internal Medicine, Division of Hematology, School of Medicine, Gachon University Gil Medical Center, Incheon, 21565, South Korea
| | - Sun Jin Sym
- Department of Internal Medicine, Division of Hematology, School of Medicine, Gachon University Gil Medical Center, Incheon, 21565, South Korea
| | - Dongwoo Khang
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea.,Department of Gachon Advanced Institute for Health Science & Technology (Gaihst), Gachon University, Incheon 21999, South Korea.,Department of Physiology, School of Medicine, Gachon University, Incheon 21999, South Korea
| |
Collapse
|
34
|
Scioli MG, Storti G, D'Amico F, Gentile P, Fabbri G, Cervelli V, Orlandi A. The Role of Breast Cancer Stem Cells as a Prognostic Marker and a Target to Improve the Efficacy of Breast Cancer Therapy. Cancers (Basel) 2019; 11:1021. [PMID: 31330794 PMCID: PMC6678191 DOI: 10.3390/cancers11071021] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common form of tumor in women and the leading cause of cancer-related mortality. Even though the major cellular burden in breast cancer is constituted by the so-called bulk tumor cells, another cell subpopulation named cancer stem cells (CSCs) has been identified. The latter have stem features, a self-renewal capacity, and the ability to regenerate the bulk tumor cells. CSCs have been described in several cancer types but breast cancer stem cells (BCSCs) were among the first to be identified and characterized. Therefore, many efforts have been put into the phenotypic characterization of BCSCs and the study of their potential as prognostic indicators and therapeutic targets. Many dysregulated pathways in BCSCs are involved in the epithelial-mesenchymal transition (EMT) and are found up-regulated in circulating tumor cells (CTCs), another important cancer cell subpopulation, that shed into the vasculature and disseminate along the body to give metastases. Conventional therapies fail at eliminating BCSCs because of their quiescent state that gives them therapy resistance. Based on this evidence, preclinical studies and clinical trials have tried to establish novel therapeutic regimens aiming to eradicate BCSCs. Markers useful for BCSC identification could also be possible therapeutic methods against BCSCs. New approaches in drug delivery combined with gene targeting, immunomodulatory, and cell-based therapies could be promising tools for developing effective CSC-targeted drugs against breast cancer.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Federico D'Amico
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Giulia Fabbri
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Augusto Orlandi
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Roma, Italy.
| |
Collapse
|
35
|
Scioli MG, Storti G, D'Amico F, Gentile P, Kim BS, Cervelli V, Orlandi A. Adipose-Derived Stem Cells in Cancer Progression: New Perspectives and Opportunities. Int J Mol Sci 2019; 20:3296. [PMID: 31277510 PMCID: PMC6651808 DOI: 10.3390/ijms20133296] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
Growing importance has been attributed to interactions between tumors, the stromal microenvironment and adult mesenchymal stem cells. Adipose-derived stem cells (ASCs) are routinely employed in regenerative medicine and in autologous fat transfer procedures. To date, clinical trials have failed to demonstrate the potential pro-oncogenic role of ASC enrichment. Nevertheless, some pre-clinical studies from in vitro and in vivo models have suggested that ASCs act as a potential tumor promoter for different cancer cell types, and support tumor progression and invasiveness through the activation of several intracellular signals. Interaction with the tumor microenvironment and extracellular matrix remodeling, the exosomal release of pro-oncogenic factors as well as the induction of epithelial-mesenchymal transitions are the most investigated mechanisms. Moreover, ASCs have also demonstrated an elective tumor homing capacity and this tumor-targeting capacity makes them a suitable carrier for anti-cancer drug delivery. New genetic and applied nanotechnologies may help to design promising anti-cancer cell-based approaches through the release of loaded intracellular nanoparticles. These new anti-cancer therapies can more effectively target tumor cells, reaching higher local concentrations even in pharmacological sanctuaries, and thus minimizing systemic adverse drug effects. The potential interplay between ASCs and tumors and potential ASCs-based therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Federico D'Amico
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Bong-Sung Kim
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| |
Collapse
|
36
|
Su X, Tang Y, Li Y, Wang Z, Tao J, Chen K, Liu Y, Wu J, Wang D, Teng Z. Facile Synthesis of Monodisperse Hollow Mesoporous Organosilica/Silica Nanospheres by an in Situ Dissolution and Reassembly Approach. ACS APPLIED MATERIALS & INTERFACES 2019; 11:12063-12069. [PMID: 30789253 DOI: 10.1021/acsami.8b21906] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Hollow structured mesoporous organosilicas are a research hotspot because of their molecularly organic-inorganic hybrid frameworks, large void spaces, permeable shells, high surface areas, uniform pores, and various applications. However, the previous reported hard-core templating method and liquid-interface assembly approach suffered from complex preparation procedures and poor uniformity for the products. In this work, we demonstrate an in situ dissolution and reassembly method to synthesize monodisperse benzene-bridged hollow mesoporous organosilica/silica nanoparticles (HMOSNs) by sequential addition of tetraethoxysilane (TEOS) and 1,4-bis(triethoxysilyl)benzene in a solution containing a cetyltrimethylammonium bromide (CTAB) surfactant. The formation of HMOSNs is completed in one pot, which is very effective and convenient. The formation mechanism of HMOSNs is ascribed to the fact that TEOS first assembles with CTAB to form mesostructured silica cores, which further dissolve and migrate to the outer layers during the deposition of mesostructured organosilica shells. The prepared benzene-bridged HMOSNs possess uniform diameter (140 nm), large pore volume (2.79 m3/g), high specific surface area (2926 m2/g), and a high doxorubicin-loading content of 16.7%. HMOSNs can deliver doxorubicin (Dox) into human breast cancer cells and reduce their excretion. Thus, the Dox-loaded HMOSNs show a high killing effect against the cancer cells.
Collapse
Affiliation(s)
- Xiaodan Su
- Key Laboratory for Organic Electronics & Information Displays and Institute of Advanced Materials , Nanjing University of Posts and Telecommunications , Nanjing , 210046 Jiangsu , P. R. China
| | | | | | - Zhifei Wang
- School of Chemistry and Chemical Engineering , Southeast University , Nanjing , 211189 Jiangsu , P. R. China
| | - Jun Tao
- Key Laboratory for Organic Electronics & Information Displays and Institute of Advanced Materials , Nanjing University of Posts and Telecommunications , Nanjing , 210046 Jiangsu , P. R. China
| | - Kun Chen
- Key Laboratory for Organic Electronics & Information Displays and Institute of Advanced Materials , Nanjing University of Posts and Telecommunications , Nanjing , 210046 Jiangsu , P. R. China
| | | | | | - Dan Wang
- Department of Gynecology & Obstetrics, Affiliated Changzheng Hospital , The Second Military Medical University , 200003 Shanghai , P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics & Information Displays and Institute of Advanced Materials , Nanjing University of Posts and Telecommunications , Nanjing , 210046 Jiangsu , P. R. China
- Key Laboratory of Food Bio-technology, School of Food and Bioengineering , Xihua University , Chengdu , 610039 Sichuan , P. R. China
| |
Collapse
|
37
|
Huang L, Xu C, Xu P, Qin Y, Chen M, Feng Q, Pan J, Cheng Q, Liang F, Wen X, Wang Y, Shi Y, Cheng Y. Intelligent Photosensitive Mesenchymal Stem Cells and Cell-Derived Microvesicles for Photothermal Therapy of Prostate Cancer. Nanotheranostics 2018; 3:41-53. [PMID: 30662822 PMCID: PMC6328305 DOI: 10.7150/ntno.28450] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/08/2018] [Indexed: 01/01/2023] Open
Abstract
Targeted delivery of nanomedicines into the tumor site and improving the intratumoral distribution remain challenging in cancer treatment. Here, we report an effective transportation system utilizing both of mesenchymal stem cells (MSCs) and their secreted microvesicles containing assembled gold nanostars (GNS) for targeted photothermal therapy of prostate cancer. The stem cells act as a cell carrier to actively load and assemble GNS into the lysosomes. Accumulation of GNS in the lysosomes facilitates the close interaction of nanoparticles, which could result in a 20 nm red-shift of surface plasmon resonance of GNS with a broad absorption in the near infrared region. Moreover, the MSCs can behave like an engineering factory to pack and release the GNS clusters into microvesicles. The secretion of GNS can be stimulated via light irradiation, providing an external trigger-assisted approach to encapsulate nanoparticles into cell derived microvesicles. In vivo studies demonstrate that GNS-loaded MSCs have an extensive intratumoral distribution, as monitored via photoacoustic imaging, and efficient antitumor effect under light exposure in a prostate-cancer subcutaneous model by intratumoral and intravenous injection. Our work presents a light-responsive transportation approach for GNS in combination of MSCs and their extracellular microvesicles and holds the promise as an effective strategy for targeted cancer therapy including prostate cancer.
Collapse
Affiliation(s)
- Liqun Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Chang Xu
- Shanghai East Hospital; The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Peng Xu
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Yu Qin
- Institute of Acoustics, Tongji University, Siping Road 1239, Shanghai 200092, China
| | - Mengwei Chen
- Shanghai East Hospital; The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qishuai Feng
- Shanghai East Hospital; The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jing Pan
- Institute of Acoustics, Tongji University, Siping Road 1239, Shanghai 200092, China
| | - Qian Cheng
- Institute of Acoustics, Tongji University, Siping Road 1239, Shanghai 200092, China
| | - Feng Liang
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xiaofei Wen
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ying Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Shi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Cheng
- Shanghai East Hospital; The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| |
Collapse
|
38
|
Xu C, Feng Q, Yang H, Wang G, Huang L, Bai Q, Zhang C, Wang Y, Chen Y, Cheng Q, Chen M, Han Y, Yu Z, Lesniak MS, Cheng Y. A Light-Triggered Mesenchymal Stem Cell Delivery System for Photoacoustic Imaging and Chemo-Photothermal Therapy of Triple Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800382. [PMID: 30356957 PMCID: PMC6193170 DOI: 10.1002/advs.201800382] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/27/2018] [Indexed: 05/12/2023]
Abstract
Targeted therapy is highly challenging and urgently needed for patients diagnosed with triple negative breast cancer (TNBC). Here, a synergistic treatment platform with plasmonic-magnetic hybrid nanoparticle (lipids, doxorubicin (DOX), gold nanorods, iron oxide nanocluster (LDGI))-loaded mesenchymal stem cells (MSCs) for photoacoustic imaging, targeted photothermal therapy, and chemotherapy for TNBC is developed. LDGI can be efficiently taken up into the stem cells with good biocompatibility to maintain the cellular functions. In addition, CXCR4 on the MSCs is upregulated by iron oxide nanoparticles in the LDGI. Importantly, the drug release and photothermal therapy can be simultaneously achieved upon light irradiation. The released drug can enter the cell nucleus and promote cell apoptosis. Interestingly, light irradiation can control the secretion of cellular microvehicles carrying LDGI for targeted treatment. A remarkable in vitro anticancer effect is observed in MDA-MB-231 with near-infrared laser irradiation. In vivo studies show that the MSCs-LDGI has the enhanced migration and penetration abilities in the tumor area via both intratumoral and intravenous injection approaches compared with LDGI. Subsequently, MSCs-LDGI shows the best antitumor efficacy via chemo-photothermal therapy compared to other treatment groups in the TNBC model of nude mice. Thus, MSCs-LDGI multifunctional system represents greatly synergistic potential for cancer treatment.
Collapse
Affiliation(s)
- Chang Xu
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering & Nano ScienceTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Qishuai Feng
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering & Nano ScienceTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Haocheng Yang
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering & Nano ScienceTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Guangxue Wang
- Research Center for Translational MedicineKey Laboratory of Arrhythmias of the Ministry of Education of ChinaEast HospitalTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Liqun Huang
- Department of UrologyShanghai East HospitalTongji University School of MedicineShanghai200120China
| | - Qianwen Bai
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering & Nano ScienceTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Chuyi Zhang
- Research Center for Translational MedicineKey Laboratory of Arrhythmias of the Ministry of Education of ChinaEast HospitalTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Yilong Wang
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering & Nano ScienceTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Yingna Chen
- Institute of AcousticsTongji UniversitySiping Road 1239Shanghai200092China
| | - Qian Cheng
- Institute of AcousticsTongji UniversitySiping Road 1239Shanghai200092China
| | - Mengwei Chen
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering & Nano ScienceTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Yu Han
- Department of Neurological SurgeryThe Feinberg School of MedicineNorthwestern UniversityChicagoIL60611USA
| | - Zuoren Yu
- Research Center for Translational MedicineKey Laboratory of Arrhythmias of the Ministry of Education of ChinaEast HospitalTongji University School of Medicine150 Jimo RoadShanghai200120China
| | - Maciej S. Lesniak
- Department of Neurological SurgeryThe Feinberg School of MedicineNorthwestern UniversityChicagoIL60611USA
| | - Yu Cheng
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering & Nano ScienceTongji University School of Medicine150 Jimo RoadShanghai200120China
| |
Collapse
|
39
|
Croissant JG, Durand JO. Mesoporous Silica-Based Nanoparticles for Light-Actuated Biomedical Applications via Near-Infrared Two-Photon Absorption. Enzymes 2018; 43:67-99. [PMID: 30244809 DOI: 10.1016/bs.enz.2018.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this review, we highlight the design of nanomaterials for two-photon excitation, in order to treat tumors with a high accuracy. Indeed two-photon excitation allows remote control of the nanoparticles with a spatio-temporal resolution. The nanomaterials are based on mesoporous silica-organosilica nanoparticles including core-shell systems. The therapeutic treatments include drug delivery, photodynamic therapy, gene silencing, and their combinations. At first, the nanosystems designed for two-photon-triggered cytotoxic drug delivery are reviewed. Then the nanomaterials prepared for two-photon photodynamic therapy and reactive oxygen species delivery are discussed. Finally, the nanosystems combining drug delivery or gene silencing with two-photon photodynamic therapy are presented. Due to the rapid progresses concerning two-photon-excited nanomaterials and the interest of near-infrared light to treat deep tumors, we believe this technology could be of high interest for the personalized medicine of the future.
Collapse
Affiliation(s)
- Jonas G Croissant
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM, United States; Center for Micro-Engineered Materials, Advanced Materials Laboratory, University of New Mexico, Albuquerque, NM, United States.
| | - Jean-Olivier Durand
- Institut Charles Gerhardt Montpellier, UMR-5253 CNRS-UM-ENSCM, Montpellier, France
| |
Collapse
|
40
|
Scioli MG, Artuso S, D'Angelo C, Porru M, D’Amico F, Bielli A, Gentile P, Cervelli V, Leonetti C, Orlandi A. Adipose-derived stem cell-mediated paclitaxel delivery inhibits breast cancer growth. PLoS One 2018; 13:e0203426. [PMID: 30192811 PMCID: PMC6128546 DOI: 10.1371/journal.pone.0203426] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/21/2018] [Indexed: 01/20/2023] Open
Abstract
Breast cancer represents the main malignancy in women and autologous fat grafting is a diffuse procedure in the management of post-surgical breast defects causing patients' psychosocial problems, with high costs for the public health. Recently, beneficial effects of fat grafting during post-surgical breast reconstruction have been amplified from the enrichment with human adipose-derived stem cells (ASCs) present in the stromal vascular fraction (SVF) of adult adipose tissue isolated during intraoperatory procedures. The major concern about the ASC enrichment during post-surgery breast reconstruction depends on their potential ability to release growth factors and hormones that can promote proliferation of residual or quiescent cancer cells, with the risk of de novo cancer development or recurrence. The recent description that adult stem cells primed in vitro may be vehicle for anti-cancer drug delivery offers a new vision concerning the role of ASCs in breast reconstruction after cancer surgery. Paclitaxel (PTX) is a chemotherapeutic agent acting as a microtubule-stabilizing drug inhibiting cancer cell mitotic activity. We optimized PTX loading and release in cultured ASCs and then analyzed the effects of PTX-loaded ASCs and their conditioned medium on CG5 breast cancer survival, proliferation and apoptosis in vitro, and inCG5 xenograft in vivo. We documented that ASCs can uptake and release PTX in vitro, with slight cytotoxic effects. Interestingly, PTX-loaded ASCs in co-culture, as well as conditioned medium alone, inhibited CG5 cell proliferation and survival in vitro and xenograft tumor growth in vivo. The antitumor effect of PTX-loaded ASCs may offer a new perspective concerning the use of ASCs during breast reconstruction becoming an additional local preventive chemotherapeutic agent against tumor recurrence. However, further experiments in vitro and in vivo are needed to collect more evidence confirming the efficacy and safety in cancer patients.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Simona Artuso
- SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Carmen D'Angelo
- Unit of Oncogenomics and Epigenetics, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Manuela Porru
- SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Federico D’Amico
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Alessandra Bielli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Carlo Leonetti
- SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
- Anatomic Pathology, Department of Biomedical Sciences, Catholic University Our lady of Good Counsel, Tirana, Albania
| |
Collapse
|
41
|
Cheng X, Li D, Lin A, Xu J, Wu L, Gu H, Huang Z, Liu J, Zhang Y, Yin X. Fabrication of multifunctional triple-responsive platform based on CuS-capped periodic mesoporous organosilica nanoparticles for chemo-photothermal therapy. Int J Nanomedicine 2018; 13:3661-3677. [PMID: 29983561 PMCID: PMC6028353 DOI: 10.2147/ijn.s167407] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION For an ideal drug delivery system, the outstanding drug-loading capacity and specific control of the release of therapeutics at the desired lesions are crucial. In this work, we developed a triple-responsive nanoplatform based on copper sulfide (CuS)-capped yolk-shell-structured periodic mesoporous organosilica nanoparticles (YSPMOs) for synergetic chemo-photothermal therapy. METHODS Herein, the YSPMOs were employed as a drug carrier, which exhibited a high doxorubicin (DOX) loading capacity of 386 mg/g. In this controlled-release drug delivery system, CuS serves as a gatekeeper to modify YSPMOs with reduction-cleavable disulfide bond (YSPMOs@CuS). CuS could not only avoid premature leakage in the delivery process, but also endowed the excellent photothermal therapy (PTT) ability. RESULTS Upon entering into cancer cells, the CuS gatekeeper was opened with the breaking of disulfide bonds and the DOX release from YSPMOs(DOX)@CuS in response to the intracellular acidic environment and external laser irradiation. Such a precise control over drug release, combined with the photothermal effect of CuS nanoparticles, is possessed by synergistic chemo-photothermal therapy for cancer treatment. Both in vitro and in vivo experimental data indicated that the synergistic effect of YSPMOs(DOX)@CuS showed efficient antitumor effect. In addition, low systemic toxicity was observed in the pathologic examinations of liver, spleen, lungs, and kidneys. CONCLUSION This versatile nanoplatform combination of PTT, chemotherapeutics, and gating components shows general potential for designing multifunctional drug delivery systems.
Collapse
Affiliation(s)
- Xiangyang Cheng
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| | - Dejian Li
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201301, China
| | - Aiqi Lin
- Department of Retired, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China
| | - Jun Xu
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| | - Liang Wu
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| | - Huijie Gu
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| | - Jiangyi Liu
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| | - Yiming Zhang
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 201199, China,
| |
Collapse
|
42
|
Liao W, Zhang L, Zhong Y, Shen Y, Li C, An N. Fabrication of ultrasmall WS 2 quantum dots-coated periodic mesoporous organosilica nanoparticles for intracellular drug delivery and synergistic chemo-photothermal therapy. Onco Targets Ther 2018; 11:1949-1960. [PMID: 29670370 PMCID: PMC5896670 DOI: 10.2147/ott.s160748] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Introduction The consolidation of different therapies into a single nanoplatform has shown great promise for synergistic tumor treatment. In this study, a multifunctional platform by WS2 quantum dots (WQDs)-coated doxorubicin (DOX)-loaded periodic mesoporous organosilicas (PMOs-DOX@WQDs) nanoparticles were fabricated for the first time, and which exhibits good potential for synergistic chemo-photothermal therapy. Materials and methods The structure, light-mediated drug release behavior, photothermal effect, and synergistic therapeutic efficiency of PMOs-DOX@WQDs to HCT-116 colon cancer cells were investigated. The thioether-bridged PMOs exhibit a high DOX loading capacity of 66.7 μg mg−1. The gating of the PMOs not only improve the drug loading capacity but also introduce the dual-stimuli-responsive performance. Furthermore, the as-synthesized PMOs-DOX@WQDs nanoparticles can efficiently generate heat to the hyperthermia temperature with near infrared laser irradiation. Results It was confirmed that PMOs-DOX@WQDs exhibit remarkable photothermal effect and light-triggered faster release of DOX. More importantly, it was reasonable to attribute the efficient anti-tumor efficiency of PMOs-DOX@WQDs. Conclusion The in vitro experimental results confirm that the fabricated nanocarrier exhibits a significant synergistic effect, resulting in a higher efficacy to kill cancer cells. Therefore, the WQD-coated PMOs present promising applications in cancer therapy.
Collapse
Affiliation(s)
- Wenyun Liao
- Department of Emergency, The First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Li Zhang
- Department of Emergency, The First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Yunhua Zhong
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Yuan Shen
- Department of Emergency, The First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Changlin Li
- Department of Emergency, The First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Na An
- Department of Emergency, The First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, People's Republic of China
| |
Collapse
|
43
|
Chang Y, Cheng Y, Feng Y, Jian H, Wang L, Ma X, Li X, Zhang H. Resonance Energy Transfer-Promoted Photothermal and Photodynamic Performance of Gold-Copper Sulfide Yolk-Shell Nanoparticles for Chemophototherapy of Cancer. NANO LETTERS 2018; 18:886-897. [PMID: 29323915 DOI: 10.1021/acs.nanolett.7b04162] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Gold (Au) core@void@copper sulfide (CuS) shell (Au-CuS) yolk-shell nanoparticles (YSNPs) were prepared in the present study for potential chemo-, photothermal, and photodynamic combination therapy, so-called "chemophototherapy". The resonance energy transfer (RET) process was utilized in Au-CuS YSNPs to achieve both enhanced photothermal and photodynamic performance compared with those of CuS hollow nanoparticles (HNPs). A series of Au nanomaterials as cores that had different localized surface plasmon resonance (LSPR) absorption peaks at 520, 700, 808, 860, and 980 nm were embedded in CuS HNPs to screen the most effective Au-CuS YSNPs according to the RET process. Thermoresponsive polymer was fabricated on these YSNPs' surface to allow for controlled drug release. Au808-CuS and Au980-CuS YSNPs were found capable of inducing the largest temperature elevation and producing the most significant hydroxyl radicals under 808 and 980 nm laser irradiation, respectively, which could accordingly cause the most severe 4T1 cell injury through oxidative stress mechanism. Moreover, doxorubicin-loaded (Dox-loaded) P(NIPAM-co-AM)-coated Au980-CuS (p-Au980-CuS@Dox) YSNPs could more efficiently kill cells than unloaded particles upon 980 nm laser irradiation. After intravenous administration to 4T1 tumor-bearing mice, p-Au980-CuS YSNPs could significantly accumulate in the tumor and effectively inhibit the tumor growth after 980 nm laser irradiation, and p-Au980-CuS@Dox YSNPs could further potentiate the inhibition efficiency and exhibit excellent in vivo biocompatibility. Taken together, this study sheds light on the rational design of Au-CuS YSNPs to offer a promising candidate for chemophototherapy.
Collapse
Affiliation(s)
- Yun Chang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Yan Cheng
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, China
| | - Yanlin Feng
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, China
- University of Science and Technology of China , Hefei, Anhui 230026, China
| | - Hui Jian
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, China
| | - Li Wang
- School of Chemistry and Life Science, Changchun University of Technology , Changchun, Jilin 130012, China
| | - Xiaomin Ma
- School of Chemistry and Life Science, Changchun University of Technology , Changchun, Jilin 130012, China
| | - Xi Li
- School of Chemistry and Life Science, Changchun University of Technology , Changchun, Jilin 130012, China
| | - Haiyuan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, China
- University of Chinese Academy of Sciences , Beijing 100049, China
- University of Science and Technology of China , Hefei, Anhui 230026, China
| |
Collapse
|
44
|
Yan G, Li A, Zhang A, Sun Y, Liu J. Polymer-Based Nanocarriers for Co-Delivery and Combination of Diverse Therapies against Cancers. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E85. [PMID: 29401694 PMCID: PMC5853717 DOI: 10.3390/nano8020085] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/27/2018] [Accepted: 02/01/2018] [Indexed: 02/06/2023]
Abstract
Cancer gives rise to an enormous number of deaths worldwide nowadays. Therefore, it is in urgent need to develop new therapies, among which combined therapies including photothermal therapy (PTT) and chemotherapy (CHT) using polymer-based nanocarriers have attracted enormous interest due to the significantly enhanced efficacy and great progress has been made so far. The preparation of such nanocarriers is a comprehensive task involving the cooperation of nanomaterial science and biomedicine science. In this review, we try to introduce and analyze the structure, preparation and synergistic therapeutic effect of various polymer-based nanocarriers composed of anti-tumor drugs, nano-sized photothermal materials and other possible parts. Our effort may bring benefit to future exploration and potential applications of similar nanocarriers.
Collapse
Affiliation(s)
- Guowen Yan
- School of Materials Science and Engineering, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, China.
| | - Aihua Li
- School of Materials Science and Engineering, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, China.
| | - Aitang Zhang
- School of Materials Science and Engineering, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, China.
| | - Yong Sun
- School of Pharmacy, Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, China.
| | - Jingquan Liu
- School of Materials Science and Engineering, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, China.
| |
Collapse
|
45
|
Croissant JG, Fatieiev Y, Almalik A, Khashab NM. Mesoporous Silica and Organosilica Nanoparticles: Physical Chemistry, Biosafety, Delivery Strategies, and Biomedical Applications. Adv Healthc Mater 2018; 7. [PMID: 29193848 DOI: 10.1002/adhm.201700831] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/30/2017] [Indexed: 01/08/2023]
Abstract
Predetermining the physico-chemical properties, biosafety, and stimuli-responsiveness of nanomaterials in biological environments is essential for safe and effective biomedical applications. At the forefront of biomedical research, mesoporous silica nanoparticles and mesoporous organosilica nanoparticles are increasingly investigated to predict their biological outcome by materials design. In this review, it is first chronicled that how the nanomaterial design of pure silica, partially hybridized organosilica, and fully hybridized organosilica (periodic mesoporous organosilicas) governs not only the physico-chemical properties but also the biosafety of the nanoparticles. The impact of the hybridization on the biocompatibility, protein corona, biodistribution, biodegradability, and clearance of the silica-based particles is described. Then, the influence of the surface engineering, the framework hybridization, as well as the morphology of the particles, on the ability to load and controllably deliver drugs under internal biological stimuli (e.g., pH, redox, enzymes) and external noninvasive stimuli (e.g., light, magnetic, ultrasound) are presented. To conclude, trends in the biomedical applications of silica and organosilica nanovectors are delineated, such as unconventional bioimaging techniques, large cargo delivery, combination therapy, gaseous molecule delivery, antimicrobial protection, and Alzheimer's disease therapy.
Collapse
Affiliation(s)
- Jonas G. Croissant
- Chemical and Biological Engineering; University of New Mexico; 210 University Blvd NE Albuquerque NM 87131-0001 USA
- Center for Micro-Engineered Materials; Advanced Materials Laboratory; University of New Mexico; MSC04 2790, 1001 University Blvd SE Suite 103 Albuquerque NM 87106 USA
| | - Yevhen Fatieiev
- Smart Hybrid Materials Laboratory (SHMs); Advanced Membranes and Porous Materials Center; King Abdullah University of Science and Technology; Thuwal Riyadh KSA 11442 Saudi Arabia
| | - Abdulaziz Almalik
- Life sciences and Environment Research Institute; Center of Excellence in Nanomedicine (CENM); King Abdulaziz City for Science and Technology (KACST); Riyadh 11461 Saudi Arabia
| | - Niveen M. Khashab
- Smart Hybrid Materials Laboratory (SHMs); Advanced Membranes and Porous Materials Center; King Abdullah University of Science and Technology; Thuwal Riyadh KSA 11442 Saudi Arabia
| |
Collapse
|
46
|
Chen YL, Liu FQ, Guo Y, Cheng J, Yang L, Lu M, Li P, Xu J, Yu T, Wang ZG, Cao Y, Ran HT. PA/US dual-modality imaging to guide VEGFR-2 targeted photothermal therapy using ZnPc-/PFH-loaded polymeric nanoparticles. Biomater Sci 2018; 6:2130-2143. [PMID: 29916500 DOI: 10.1039/c8bm00213d] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiogenesis is a common pathological characteristic of many solid tumors and vulnerable atherosclerotic plaques.
Collapse
|
47
|
Yue Q, Li J, Zhang Y, Cheng X, Chen X, Pan P, Su J, Elzatahry AA, Alghamdi A, Deng Y, Zhao D. Plasmolysis-Inspired Nanoengineering of Functional Yolk-Shell Microspheres with Magnetic Core and Mesoporous Silica Shell. J Am Chem Soc 2017; 139:15486-15493. [PMID: 29016118 DOI: 10.1021/jacs.7b09055] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Yolk-shell nanomaterials with a rattle-like structure have been considered ideal carriers and nanoreactors. Traditional methods to constructing yolk-shell nanostructures mainly rely on multistep sacrificial template strategy. In this study, a facile and effective plasmolysis-inspired nanoengineering strategy is developed to controllably fabricate yolk-shell magnetic mesoporous silica microspheres via the swelling-shrinkage of resorcinol-formaldehyde (RF) upon soaking in or removal of n-hexane. Using Fe3O4@RF microspheres as seeds, surfactant-silica mesostructured composite is deposited on the swelled seeds through the multicomponent interface coassembly, followed by solvent extraction to remove surfactant and simultaneously induce shrinkage of RF shell. The obtained yolk-shell microspheres (Fe3O4@RF@void@mSiO2) possess a high magnetization of 40.3 emu/g, high surface area (439 m2/g), radially aligned mesopores (5.4 nm) in the outer shell, tunable middle hollow space (472-638 nm in diameter), and a superparamagnetic core. This simple method allows a simultaneous encapsulation of Au nanoparticles into the hollow space during synthesis, and it leads to spherical Fe3O4@RF@void-Au@mSiO2 magnetic nanocatalysts, which show excellent catalysis efficiency for hydrogenation of 4-nitrophenol by NaBH4 with a high conversion rate (98%) and magnetic recycling stability.
Collapse
Affiliation(s)
- Qin Yue
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and iChEM, Fudan University , Shanghai 200433, China
| | - Jialuo Li
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and iChEM, Fudan University , Shanghai 200433, China
| | - Yu Zhang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and iChEM, Fudan University , Shanghai 200433, China
| | - Xiaowei Cheng
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and iChEM, Fudan University , Shanghai 200433, China
| | - Xiao Chen
- Department of Orthopaedics Trauma, Changhai Hospital, Second Military Medical University , Shanghai 200433, China
| | - Panpan Pan
- Department of Orthopaedics Trauma, Changhai Hospital, Second Military Medical University , Shanghai 200433, China
| | - Jiacan Su
- Department of Orthopaedics Trauma, Changhai Hospital, Second Military Medical University , Shanghai 200433, China
| | - Ahmed A Elzatahry
- Materials Science and Technology Program, College of Arts and Sciences, Qatar University , P.O. Box 2713, Doha, Qatar
| | - Abdulaziz Alghamdi
- Department of Chemistry, College of Science, King Saud University , Riyadh 11451, Saudi Arabia
| | - Yonghui Deng
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and iChEM, Fudan University , Shanghai 200433, China.,State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences , Shanghai 200050, China
| | - Dongyuan Zhao
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and iChEM, Fudan University , Shanghai 200433, China
| |
Collapse
|
48
|
Li G, Chen Y, Zhang L, Zhang M, Li S, Li L, Wang T, Wang C. Facile Approach to Synthesize Gold Nanorod@Polyacrylic Acid/Calcium Phosphate Yolk-Shell Nanoparticles for Dual-Mode Imaging and pH/NIR-Responsive Drug Delivery. NANO-MICRO LETTERS 2017; 10:7. [PMID: 30393656 PMCID: PMC6199055 DOI: 10.1007/s40820-017-0155-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/22/2017] [Indexed: 05/23/2023]
Abstract
A facile strategy to fabricate gold nanorod@polyacrylic acid/calcium phosphate (AuNR@PAA/CaP) yolk-shell nanoparticles (NPs) composed with a PAA/CaP shell and an AuNR yolk is reported. The as-obtained AuNR@PAA/CaP yolk-shell NPs possess ultrahigh doxorubicin (DOX) loading capability (1 mg DOX/mg NPs), superior photothermal conversion property (26%) and pH/near-infrared (NIR) dual-responsive drug delivery performance. The released DOX continuously increased due to the damage of the CaP shell at low pH values. When the DOX-loaded AuNR@PAA/CaP yolk-shell NPs were exposed to NIR irradiation, a burst-like drug release occurs owing to the heat produced by the AuNRs. Furthermore, AuNR@PAA/CaP yolk-shell NPs are successfully employed for synergic dual-mode X-ray computed tomography/photoacoustic imaging and chemo-photothermal cancer therapy. Therefore, this work brings new insights for the synthesis of multifunctional nanomaterials and extends theranostic applications.
Collapse
Affiliation(s)
- Guilan Li
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Yidan Chen
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Lingyu Zhang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Manjie Zhang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Shengnan Li
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Lu Li
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Tingting Wang
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, People's Republic of China.
| | - Chungang Wang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, People's Republic of China.
| |
Collapse
|
49
|
Su X, Wang J, Zhang J, Yuwen L, Zhang Q, Dang M, Tao J, Ma X, Wang S, Teng Z. Synthesis of sandwich-like molybdenum sulfide/mesoporous organosilica nanosheets for photo-thermal conversion and stimuli-responsive drug release. J Colloid Interface Sci 2017; 496:261-266. [DOI: 10.1016/j.jcis.2017.01.068] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 10/20/2022]
|
50
|
Chen L, Feng Y, Zhou X, Zhang Q, Nie W, Wang W, Zhang Y, He C. One-Pot Synthesis of MoS 2 Nanoflakes with Desirable Degradability for Photothermal Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:17347-17358. [PMID: 28471183 DOI: 10.1021/acsami.7b02657] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Developing biodegradable photothermal agent holds great significance for potential clinical translation of photothermal therapy. In the current study, one-pot hydrothermal synthesis of MoS2 nanoflakes with desirable degradation capability was presented. The participation of poly(acrylic acid) (PAA) in hydrothermal process could not only facilitate the modification of polyethylene glycol (PEG), but also bestow degradability to the prepared MoS2 nanoflakes. Moreover, the PEGylated hybrid nanoflakes (MoS2-PPEG) also exhibited excellent stability in various medium and outstanding photothermal properties. Interestingly, MoS2-PPEG behaved distinctly different degradation rate in diverse condition. The rapid degradation of MoS2-PPEG was observed in neutral pH solution, whereas much slower degradation occurred in an acidic tumor microenvironment. Furthermore, data indicated that the major degradation product of MoS2-PPEG was water-soluble Mo-based ion. Meanwhile, the good in vitro biocompatibility of MoS2-PPEG was also confirmed in terms of cytotoxicity and hemolysis. With favorable photothermal performance, MoS2-PPEG can efficiently killing cancer cells in vitro and suppress the tumor growth in vivo. More importantly, the gradual decreasing content of MoS2-PPEG in organs and detectable Mo element in urine of mice suggested that the degradability of MoS2-PPEG might facilitate its excretion to some degree. Hence, the degradable MoS2 nanoflakes prepared by one-pot hydrothermal routine may provide insight for further biomedical applications of inorganic photothermal agent.
Collapse
Affiliation(s)
| | | | - Xiaojun Zhou
- Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine , Shanghai, 200011, China
| | | | | | | | | | | |
Collapse
|