1
|
Kong H, Chen X, Lee W, Xie X, Tao Y, Li M. Dual-color fluorescence detection of tumor-derived extracellular vesicles using a specific and serum-stable membrane-fusion approach. Biosens Bioelectron 2025; 278:117302. [PMID: 40101657 DOI: 10.1016/j.bios.2025.117302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/05/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Tumor-derived extracellular vesicles (tEVs), which are essential mediators for cell-to-cell communication during tumorigenesis and tumor development, have demonstrated significant diagnostic potential in cancer liquid biopsy, particularly through biomarkers like membrane proteins and inner microRNAs. However, traditional detection methods such as ELISA and qRT-PCR encounter challenges with low sensitivity and specificity, complex procedures, and high costs. Although emerging biosensors have been developed, these methods are limited to detecting a single type of tEV biomarker, which may result in misdiagnoses due to false-positive or false-negative signals. Herein, we introduce a specific and serum-stable membrane-fusion approach (SSMFA) capable of simultaneously detecting tEV proteins and microRNAs via dual-color fluorescence analysis. In this strategy, the established epithelial cell adhesion molecule (EpCAM) aptamer-modified serum-stable membrane-fusion liposome (AptSMFL) is labeled with fluorescence resonance energy transfer (FRET) dye pairs, which can specifically recognize EpCAM-overexpressed tEVs and induce serum-stable membrane fusion, allowing the quantification of EpCAM protein levels through red fluorescence changes resulting from FRET alterations. Meanwhile, SSMFA facilitates efficient transfection of the CRISPR/Cas13a probe into tEVs to analyze the levels of microRNA-21 (miR-21) in EpCAM-positive tEVs via green fluorescence detection. When tested on serum samples from hepatocellular carcinoma models, the SSMFA exhibited minimal sample volume requirement and rapid assay time (2 h) to effectively achieve accurate quantification of both tEV EpCAM protein and miR-21 levels. Additionally, this dual-biomarker detection method showed a strong correlation with tumor burden and significantly improved cancer diagnostic accuracy (AUC = 0.98), underscoring the potential of SSMFA as a promising tEV-based liquid biopsy assay for cancer diagnosis.
Collapse
Affiliation(s)
- Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaodie Chen
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Weijen Lee
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-Sen University, Guangzhou 510275, China; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China.
| |
Collapse
|
2
|
Garcia VA, Sarkar CA, Ogle BM. Syncytial therapeutics: Receptor-specific and direct-to-cytosol biologic drug delivery mediated by measles fusion complex. J Control Release 2025; 380:967-975. [PMID: 39956393 PMCID: PMC11967904 DOI: 10.1016/j.jconrel.2025.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 01/17/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
This work explores cell-cell fusion mediated by measles virus (MeV) as a potential new cell therapy modality that achieves direct-to-cytosol (DTC) drug delivery. MeV induces receptor-mediated fusion at the cell surface via its hemagglutinin (H) and fusion glycoproteins (F), bypassing endocytic membrane transport, and enabling direct cytosolic mixing between a fusogenic donor and host target cell. Fusion of this type gives rise to large syncytia formed by the inclusion of additional target cells over time. Fusion receptor specificity was first examined in CHO "non-target" and CHO "target" cells exogenously expressing the measles target SLAM (CHO-SLAM) by mono- or co-transfection of each cell type with plasmids encoding MeV-H and MeV-F. Fusion was observed only in CHO-SLAM cells which were co-transfected with both plasmids, which verified receptor-specificity without false-triggering of fusion in co-transfected "non-target" CHO or in MeV-F mono-transfectants of either cell type. Next, CHO donor cells with constitutive mCherry expression were co-transfected with MeV-H and MeV-F, and mCherry-positive syncytia were observed when cells were mixed with CHO-SLAM demonstrating the ability to deliver the mCherry payload via DTC. Increasing the cell dose does not affect the size distribution of resulting syncytia but contributes to a higher total mCherry delivery. Further, control of MeV stoichiometry can modulate the degree of syncytia formation and protein delivery, demonstrating that limiting MeV-H and increasing MeV-F favors fusion and cytosolic delivery. Taken together, these results demonstrate MeV cell-fusion-based, DTC delivery as a robust and tunable system for achieving targeted cytosolic delivery and controlled syncytia formation.
Collapse
Affiliation(s)
- Victor A Garcia
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Casim A Sarkar
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
3
|
Zhang S, Dong H, Jin X, Sun J, Li Y. The multifaceted roles of macrophages in the transition from hepatitis to hepatocellular carcinoma: From mechanisms to therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167676. [PMID: 39828046 DOI: 10.1016/j.bbadis.2025.167676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Macrophages are central to the progression from hepatitis to hepatocellular carcinoma (HCC), with their remarkable plasticity and ability to adapt to the changing liver microenvironment. Chronic inflammation, fibrosis, and ultimately tumorigenesis are driven by macrophage activation, making them key regulators of liver disease progression. This review explores the diverse roles of macrophages in the transition from hepatitis to HCC. In the early stages of hepatitis, macrophages are essential for pathogen clearance and tissue repair. However, chronic activation leads to prolonged inflammation, which exacerbates liver damage and promotes fibrosis. As the disease progresses to liver fibrosis, macrophages interact with hepatic stellate cells, fostering a pro-tumorigenic microenvironment that supports HCC development. In hepatocarcinogenesis, macrophages contribute to tumor initiation, growth, metastasis, immune evasion, cancer stem cell maintenance, and angiogenesis. Their functional plasticity enables them to adapt to the tumor microenvironment, thereby promoting tumor progression and resistance to therapy. Targeting macrophages represents a promising strategy for preventing and treating HCC. Therapeutic approaches, including reprogramming macrophage phenotypes to enhance anti-tumor immunity, blocking macrophage recruitment and activation, and utilizing nanoparticle-based drug delivery systems, may provide new avenues for combating HCC by modulating macrophage functions and tumor microenvironment dynamics.
Collapse
Affiliation(s)
- Shuairan Zhang
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Hang Dong
- Phase I Clinical Trials Center, The People's Hospital of China Medical University, Shenyang, PR China
| | - Xiuli Jin
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Jing Sun
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Yiling Li
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
4
|
Kang RH, Baek SW, Oh CK, Kim YH, Kim D. Recent Advances of Macrostructural Porous Silicon for Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5609-5626. [PMID: 39818715 PMCID: PMC11788993 DOI: 10.1021/acsami.4c18296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Porous silicon (pSi) has gained substantial attention as a versatile material for various biomedical applications due to its unique structural and functional properties. Initially used as a semiconductor material, pSi has transitioned into a bioactive platform, enabling its use in drug delivery systems, biosensing, tissue engineering scaffolds, and implantable devices. This review explores recent advancements in macrostructural pSi, emphasizing its biocompatibility, biodegradability, high surface area, and tunable properties. In drug delivery, pSi's potential for controlled and sustained release of therapeutic agents has been well-studied, making it suitable for chronic disease treatment. Innovative approaches, like microneedle arrays and hybrid drug delivery systems, are highlighted, along with challenges, such as scalability and stability, in biological environments. pSi-based biosensors offer exceptional sensitivity for detecting biomarkers, benefiting early disease diagnosis. In tissue engineering, fibrous and particulate pSi scaffolds mimic the extracellular matrix, promoting cell proliferation and tissue regeneration. pSi is also gaining momentum in orthopedic implants, demonstrating the potential for bone regeneration. Despite its promise, challenges like mechanical strength, scalability, and long-term stability must be addressed. Looking forward, future research should focus on optimizing production methods, enhancing stability, and exploring hybrid materials for pSi, paving the way for its widespread clinical use in personalized medicine, advanced drug delivery, and next-generation biosensors and implants.
Collapse
Affiliation(s)
- Rae Hyung Kang
- Department
of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Seung Woo Baek
- College
of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Chang-Kyu Oh
- Department
of Anatomy, School of Medicine, Pusan National
University, Yangsan 50612, Republic of Korea
- Institute
for Future Earth, Pusan National University, Busan 46241, Republic of Korea
| | - Yun Hak Kim
- Department
of Anatomy, School of Medicine, Pusan National
University, Yangsan 50612, Republic of Korea
| | - Dokyoung Kim
- College
of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department
of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic
of Korea
- Department
of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic
of Korea
- KHU-KIST
Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic
of Korea
- Department
of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic
of Korea
- Center
for Converging Humanities, Kyung Hee University, Seoul 02447, Republic of Korea
- Medical
Research Center for Bioreaction to Reactive Oxygen Species and Biomedical
Science Institute, School of Medicine, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
- UC San Diego Materials Research Science
and Engineering Center, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
5
|
Gongalsky MB, Tsurikova UA, Kudryavtsev AA, Pervushin NV, Sviridov AP, Kumeria T, Egoshina VD, Tyurin-Kuzmin PA, Naydov IA, Gonchar KA, Kopeina GS, Andreev VG, Zhivotovsky B, Osminkina LA. Amphiphilic Photoluminescent Porous Silicon Nanoparticles as Effective Agents for Ultrasound-Amplified Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:374-385. [PMID: 39701827 DOI: 10.1021/acsami.4c15725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
This study investigates the use of photoluminescent amphiphilic porous silicon nanoparticles (αϕ-pSiNPs) as effective ultrasound (US) amplifiers for cancer sonodynamic theranostics. αϕ-pSiNPs were synthesized via a novel top-down approach involving porous silicon (pSi) films electrochemical etching, borate oxidation, and hydrophobic coating with octadecylsilane (C18), resulting in milling into nanoparticles with hydrophilic exteriors and hydrophobic interiors. These properties promote gas trapping and cavitation nucleation, significantly lowering the US cavitation threshold and resulting in selective destruction of cancer cells in the presence of nanoparticles. Efficient internalization of αϕ-pSiNPs in cell cytoplasm was demonstrated by their intrinsic photoluminescence, activated by partial oxidation of mesoporous silicon films in borate solutions, which resulted in quantum confinement of excitons in 2-5 nm Si quantum dots/wires. Combined with US exposure above the cavitation threshold, αϕ-pSiNPs caused a significant decrease in cell viability through mechanical stretching and microflows generated by oscillating microbubbles. Meanwhile, αϕ-pSiNPs exhibit high biocompatibility up to concentrations of 1 mg/mL without US activation. Their photoluminescent properties facilitate bioimaging, while their US contrast capabilities may enhance both imaging and therapy. The dual functionality of αϕ-pSiNPs supports a theranostic approach, enabling simultaneous diagnostics and treatment with a single agent. This study underscores the potential of αϕ-pSiNPs in sonodynamic therapy and bioimaging, offering a promising strategy for effective and safe anticancer therapy.
Collapse
Affiliation(s)
- Maxim B Gongalsky
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
| | - Uliana A Tsurikova
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Andrey A Kudryavtsev
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Nikolay V Pervushin
- Faculty of Medicine, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - Andrey P Sviridov
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Victoria D Egoshina
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Pyotr A Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Ilia A Naydov
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Kirill A Gonchar
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Gelina S Kopeina
- Faculty of Medicine, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - Valery G Andreev
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| | - Boris Zhivotovsky
- Faculty of Medicine, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institutet, Box 210, Stockholm SE-171 77, Sweden
| | - Liubov A Osminkina
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russian Federation
| |
Collapse
|
6
|
Sanati M, Figueroa-Espada CG, Han EL, Mitchell MJ, Yavari SA. Bioengineered Nanomaterials for siRNA Therapy of Chemoresistant Cancers. ACS NANO 2024; 18:34425-34463. [PMID: 39666006 DOI: 10.1021/acsnano.4c11259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Chemoresistance remains a long-standing challenge after cancer treatment. Over the last two decades, RNA interference (RNAi) has emerged as a gene therapy modality to sensitize cancer cells to chemotherapy. However, the use of RNAi, specifically small-interfering RNA (siRNA), is hindered by biological barriers that limit its intracellular delivery. Nanoparticles can overcome these barriers by protecting siRNA in physiological environments and facilitating its delivery to cancer cells. In this review, we discuss the development of nanomaterials for siRNA delivery in cancer therapy, current challenges, and future perspectives for their implementation to overcome cancer chemoresistance.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 97178, Iran
| | - Christian G Figueroa-Espada
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| |
Collapse
|
7
|
Wang Y, Qu R, Du W, Li W, Wang A, Chen Z, Gao H, Wu D, Geng F, Scherman D, Wang X, Shi S, Zou L, Li H. In Situ Bioorthogonal Repair of the Vascular Endothelium Glycocalyx to Treat Acute Lung Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405092. [PMID: 39324256 DOI: 10.1002/smll.202405092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/11/2024] [Indexed: 09/27/2024]
Abstract
In acute lung injury, destruction of the lung endothelial glycocalyx leads to vessel permeabilization and contributes to pulmonary edema and inflammation. Heparan sulfate, which accounts for >70% of glycosaminoglycans in the endothelial glycocalyx, plays a crucial physiological anti-inflammatory role. To treat acute lung injury, it is explored whether a two-step in vivo bioorthogonal chemistry strategy can covalently link intravenously administered heparan sulfate to the lung vascular endothelium and the damaged glycocalyx. First, fusogenic liposomes (EBP-Tz-FLs) carrying the reactive group tetrazine (Tz), and an E-selectin-binding peptide (EBP) to target the lung inflammatory endothelium are administered intravenously. This step aimed to anchor the tetrazine group to the membrane of inflammatory endothelial cells. Second, heparan sulfate (HS-TCO) conjugated to the trans-cyclooctene (TCO) group, which spontaneously reacts with Tz, is injected intravenously, leading to covalent heparan sulfate addition to the vascular endothelium. In a mouse model of acute lung injury, this approach substantially reduced vascular permeability and attenuated lung tissue infiltration. The EBP-Tz-FLs and HS-TCO showed favorable biocompatibility and safety both in vitro and in vivo. The proposed strategy shows good promise in acute lung injury therapy and covalently anchoring functional molecules onto the membrane of target cells.
Collapse
Affiliation(s)
- Yao Wang
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Rui Qu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Wenxuan Du
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Wei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Anqi Wang
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Zhoujiang Chen
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research, Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Di Wu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Daniel Scherman
- Université Paris Cité, CNRS, Inserm, UTCBS, Paris, 75006, France
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Hanmei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| |
Collapse
|
8
|
Zhao Y, Qin J, Yu D, Liu Y, Song D, Tian K, Chen H, Ye Q, Wang X, Xu T, Xuan H, Sun N, Ma W, Zhong J, Sun P, Song Y, Hu J, Zhao Y, Hou X, Meng X, Jiang C, Cai J. Polymer-locking fusogenic liposomes for glioblastoma-targeted siRNA delivery and CRISPR-Cas gene editing. NATURE NANOTECHNOLOGY 2024; 19:1869-1879. [PMID: 39209994 DOI: 10.1038/s41565-024-01769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
In patients with glioblastoma (GBM), upregulated midkine (MDK) limits the survival benefits conferred by temozolomide (TMZ). RNA interference (RNAi) and CRISPR-Cas9 gene editing technology are attractive approaches for regulating MDK expression. However, delivering these biologics to GBM tissue is challenging. Here we demonstrate a polymer-locking fusogenic liposome (Plofsome) that can be transported across the blood-brain barrier (BBB) and deliver short interfering RNA or CRISPR-Cas9 ribonucleoprotein complexes into the cytoplasm of GBM cells. Plofsome is designed by integrating a 'lock' into the fusogenic liposome using a traceless reactive oxygen species (ROS)-cleavable linker so that fusion occurs only after crossing the BBB and entering the GBM tissue with high ROS levels. Our results showed that MDK suppression by Plofsomes significantly reduced TMZ resistance and inhibited GBM growth in orthotopic brain tumour models. Importantly, Plofsomes are effective only at tumour sites and not in normal tissues, which improves the safety of combined RNAi and CRISPR-Cas9 therapeutics.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA.
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin, China.
| | - Jie Qin
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Daohan Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuxiang Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dan Song
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kaifu Tian
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qile Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianye Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanwen Xuan
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nan Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Junzhe Zhong
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Penggang Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Song
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingze Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunlei Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintong Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
- The Sixth Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
9
|
Xu C, Ma C, Zhang W, Wei Y, Yang K, Yuan B. Membrane Fusion Mediated by Cationic Helical Peptide L-MMBen through Phosphatidylglycerol Recruitment. J Phys Chem Lett 2024; 15:11027-11034. [PMID: 39466831 DOI: 10.1021/acs.jpclett.4c02558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Membrane fusion is the basis for many biological processes, which holds promise in biomedical applications including the creation of engineered hybrid cells and cell membrane functionalization. Extensive research efforts, including investigations into DNA zippers and carbon nanotubes, have been dedicated to the development of membrane fusion strategies inspired by natural SNARE proteins; nevertheless, achieving a delicate balance between membrane selectivity and high fusion efficiency through precise molecular engineering remains unclear. In our recent study, we successfully designed L-MMBen, a cationic helical antimicrobial peptide that exhibits remarkable antimicrobial efficacy while demonstrating moderate cytotoxicity. In this work, we demonstrate the effective and selective induction of fusion between phosphatidylglycerol (PG)-containing membranes by L-MMBen. By combining biophysical assays at the single-vesicle level with computer simulations at the molecular level, we discovered that L-MMBen can stably adsorb onto the surface of PG-containing membranes, leading to the formation of stalk structures between vesicles and ultimately resulting in membrane fusion. Furthermore, the occurrence of fusion is attributed to the unique ability of L-MMBen to recruit PG lipids and bridge adjacent vesicles. In contrast, its nonhelical counterpart DL-MMBen was found to lack this capability despite possessing an identical positive charge. These findings present an alternative molecule for achieving selective membrane fusion and provide insights for designing helical peptides with diverse applications.
Collapse
Affiliation(s)
- Cheng Xu
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Chiyun Ma
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Wanting Zhang
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, Jiangsu, China
| | - Yushuang Wei
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| |
Collapse
|
10
|
Huang J, Fu Y, Wang A, Shi K, Peng Y, Yi Y, Yu R, Gao J, Feng J, Jiang G, Song Q, Jiang J, Chen H, Gao X. Brain Delivery of Protein Therapeutics by Cell Matrix-Inspired Biomimetic Nanocarrier. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405323. [PMID: 38718295 DOI: 10.1002/adma.202405323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Indexed: 05/24/2024]
Abstract
Protein therapeutics are anticipated to offer significant treatment options for central nervous system (CNS) diseases. However, the majority of proteins are unable to traverse the blood-brain barrier (BBB) and reach their CNS target sites. Inspired by the natural environment of active proteins, the cell matrix components hyaluronic acid (HA) and protamine (PRTM) are used to self-assemble with proteins to form a protein-loaded biomimetic core and then incorporated into ApoE3-reconstituted high-density lipoprotein (rHDL) to form a protein-loaded biomimetic nanocarrier (Protein-HA-PRTM-rHDL). This cell matrix-inspired biomimetic nanocarrier facilitates the penetration of protein therapeutics across the BBB and enables their access to intracellular target sites. Specifically, CAT-HA-PRTM-rHDL facilitates rapid intracellular delivery and release of catalase (CAT) via macropinocytosis-activated membrane fusion, resulting in improved spatial learning and memory in traumatic brain injury (TBI) model mice (significantly reduces the latency of TBI mice and doubles the number of crossing platforms), and enhances motor function and prolongs survival in amyotrophic lateral sclerosis (ALS) model mice (extended the median survival of ALS mice by more than 10 days). Collectively, this cell matrix-inspired nanoplatform enables the efficient CNS delivery of protein therapeutics and provides a novel approach for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Jialin Huang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yuli Fu
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Antian Wang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kexing Shi
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yidong Peng
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yao Yi
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Renhe Yu
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinchao Gao
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Junfeng Feng
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiyao Jiang
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shuguang Lab for Future Health, Academy of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
11
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
12
|
Ren X, Xue R, Luo Y, Wang S, Ge X, Yao X, Li L, Min J, Li M, Luo Z, Wang F. Programmable melanoma-targeted radio-immunotherapy via fusogenic liposomes functionalized with multivariate-gated aptamer assemblies. Nat Commun 2024; 15:5035. [PMID: 38866788 PMCID: PMC11169524 DOI: 10.1038/s41467-024-49482-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
Radio-immunotherapy exploits the immunostimulatory features of ionizing radiation (IR) to enhance antitumor effects and offers emerging opportunities for treating invasive tumor indications such as melanoma. However, insufficient dose deposition and immunosuppressive microenvironment (TME) of solid tumors limit its efficacy. Here we report a programmable sequential therapeutic strategy based on multifunctional fusogenic liposomes (Lip@AUR-ACP-aptPD-L1) to overcome the intrinsic radio-immunotherapeutic resistance of solid tumors. Specifically, fusogenic liposomes are loaded with gold-containing Auranofin (AUR) and inserted with multivariate-gated aptamer assemblies (ACP) and PD-L1 aptamers in the lipid membrane, potentiating melanoma-targeted AUR delivery while transferring ACP onto cell surface through selective membrane fusion. AUR amplifies IR-induced immunogenic death of melanoma cells to release antigens and damage-associated molecular patterns such as adenosine triphosphate (ATP) for triggering adaptive antitumor immunity. AUR-sensitized radiotherapy also upregulates matrix metalloproteinase-2 (MMP-2) expression that combined with released ATP to activate ACP through an "and" logic operation-like process (AND-gate), thus triggering the in-situ release of engineered cytosine-phosphate-guanine aptamer-based immunoadjuvants (eCpG) for stimulating dendritic cell-mediated T cell priming. Furthermore, AUR inhibits tumor-intrinsic vascular endothelial growth factor signaling to suppress infiltration of immunosuppressive cells for fostering an anti-tumorigenic TME. This study offers an approach for solid tumor treatment in the clinics.
Collapse
Affiliation(s)
- Xijiao Ren
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, PR China
| | - Rui Xue
- School of Life Science, Chongqing University, Chongqing, 400044, PR China
| | - Yan Luo
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, 400030, PR China
| | - Shuang Wang
- School of Life Science, Chongqing University, Chongqing, 400044, PR China
| | - Xinyue Ge
- School of Life Science, Chongqing University, Chongqing, 400044, PR China
| | - Xuemei Yao
- School of Life Science, Chongqing University, Chongqing, 400044, PR China
| | - Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, PR China
| | - Junxia Min
- The Second Affiliated Hospital, The First Affiliated Hospital School of Public Health Institute of Translational Medicine State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, PR China.
| | - Zhong Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, PR China.
- School of Life Science, Chongqing University, Chongqing, 400044, PR China.
| | - Fudi Wang
- The Second Affiliated Hospital, The First Affiliated Hospital School of Public Health Institute of Translational Medicine State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
- The First Affiliated Hospital Basic Medical Sciences, School of Public Health Hengyang Medical School University of South China, Hengyang, 421001, PR China.
| |
Collapse
|
13
|
Kong H, Zheng C, Yi K, Mintz RL, Lao YH, Tao Y, Li M. An antifouling membrane-fusogenic liposome for effective intracellular delivery in vivo. Nat Commun 2024; 15:4267. [PMID: 38769317 PMCID: PMC11106281 DOI: 10.1038/s41467-024-46533-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 03/01/2024] [Indexed: 05/22/2024] Open
Abstract
The membrane-fusion-based internalization without lysosomal entrapment is advantageous for intracellular delivery over endocytosis. However, protein corona formed on the membrane-fusogenic liposome surface converts its membrane-fusion performance to lysosome-dependent endocytosis, causing poorer delivery efficiency in biological conditions. Herein, we develop an antifouling membrane-fusogenic liposome for effective intracellular delivery in vivo. Leveraging specific lipid composition at an optimized ratio, such antifouling membrane-fusogenic liposome facilitates fusion capacity even in protein-rich conditions, attributed to the copious zwitterionic phosphorylcholine groups for protein-adsorption resistance. Consequently, the antifouling membrane-fusogenic liposome demonstrates robust membrane-fusion-mediated delivery in the medium with up to 38% fetal bovine serum, outclassing two traditional membrane-fusogenic liposomes effective at 4% and 6% concentrations. When injected into mice, antifouling membrane-fusogenic liposomes can keep their membrane-fusion-transportation behaviors, thereby achieving efficient luciferase transfection and enhancing gene-editing-mediated viral inhibition. This study provides a promising tool for effective intracellular delivery under complex physiological environments, enlightening future nanomedicine design.
Collapse
Affiliation(s)
- Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- School of Chemistry, South China Normal University, Guangzhou, 510006, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Rachel L Mintz
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
14
|
Broc B, Varini K, Sonnette R, Pecqueux B, Benoist F, Masse M, Mechioukhi Y, Ferracci G, Temsamani J, Khrestchatisky M, Jacquot G, Lécorché P. LDLR-Mediated Targeting and Productive Uptake of siRNA-Peptide Ligand Conjugates In Vitro and In Vivo. Pharmaceutics 2024; 16:548. [PMID: 38675209 PMCID: PMC11054735 DOI: 10.3390/pharmaceutics16040548] [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: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Small RNA molecules such as microRNA and small interfering RNA (siRNA) have become promising therapeutic agents because of their specificity and their potential to modulate gene expression. Any gene of interest can be potentially up- or down-regulated, making RNA-based technology the healthcare breakthrough of our era. However, the functional and specific delivery of siRNAs into tissues of interest and into the cytosol of target cells remains highly challenging, mainly due to the lack of efficient and selective delivery systems. Among the variety of carriers for siRNA delivery, peptides have become essential candidates because of their high selectivity, stability, and conjugation versatility. Here, we describe the development of molecules encompassing siRNAs against SOD1, conjugated to peptides that target the low-density lipoprotein receptor (LDLR), and their biological evaluation both in vitro and in vivo.
Collapse
Affiliation(s)
- Baptiste Broc
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Karine Varini
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Rose Sonnette
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Belinda Pecqueux
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Florian Benoist
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Maxime Masse
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Yasmine Mechioukhi
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Géraldine Ferracci
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Jamal Temsamani
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | | | - Guillaume Jacquot
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Pascaline Lécorché
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| |
Collapse
|
15
|
Yang C, Lin ZI, Zhang X, Xu Z, Xu G, Wang YM, Tsai TH, Cheng PW, Law WC, Yong KT, Chen CK. Recent Advances in Engineering Carriers for siRNA Delivery. Macromol Biosci 2024; 24:e2300362. [PMID: 38150293 DOI: 10.1002/mabi.202300362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/29/2023] [Indexed: 12/28/2023]
Abstract
RNA interference (RNAi) technology has been a promising treatment strategy for combating intractable diseases. However, the applications of RNAi in clinical are hampered by extracellular and intracellular barriers. To overcome these barriers, various siRNA delivery systems have been developed in the past two decades. The first approved RNAi therapeutic, Patisiran (ONPATTRO) using lipids as the carrier, for the treatment of amyloidosis is one of the most important milestones. This has greatly encouraged researchers to work on creating new functional siRNA carriers. In this review, the recent advances in siRNA carriers consisting of lipids, polymers, and polymer-modified inorganic particles for cancer therapy are summarized. Representative examples are presented to show the structural design of the carriers in order to overcome the delivery hurdles associated with RNAi therapies. Finally, the existing challenges and future perspective for developing RNAi as a clinical modality will be discussed and proposed. It is believed that the addressed contributions in this review will promote the development of siRNA delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zheng-Ian Lin
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Xinmeng Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yu-Min Wang
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Tzu-Hsien Tsai
- Division of Cardiology and Department of Internal Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, 60002, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
- Department of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, P. R. China
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Chih-Kuang Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| |
Collapse
|
16
|
Dumontel B, Rosso G, Cauda V. Natural and artificial phospholipid bilayer coatings on solid-state nanoparticles, current and future perspectives. Nanomedicine (Lond) 2024; 19:653-655. [PMID: 38406890 DOI: 10.2217/nnm-2023-0358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Affiliation(s)
- Bianca Dumontel
- Department of Applied Science & Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
| | - Giada Rosso
- Department of Applied Science & Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
| | - Valentina Cauda
- Department of Applied Science & Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
| |
Collapse
|
17
|
Li SL, Hou HY, Chu X, Zhu YY, Zhang YJ, Duan MD, Liu J, Liu Y. Nanomaterials-Involved Tumor-Associated Macrophages' Reprogramming for Antitumor Therapy. ACS NANO 2024; 18:7769-7795. [PMID: 38420949 DOI: 10.1021/acsnano.3c12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Tumor-associated macrophages (TAMs) play pivotal roles in tumor development. As primary contents of tumor environment (TME), TAMs secrete inflammation-related substances to regulate tumoral occurrence and development. There are two kinds of TAMs: the tumoricidal M1-like TAMs and protumoral M2-like TAMs. Reprogramming TAMs from immunosuppressive M2 to immunocompetent M1 phenotype is considered a feasible way to improve immunotherapeutic efficiency. Notably, nanomaterials show great potential for biomedical fields due to their controllable structures and properties. There are many types of nanomaterials that exhibit great regulatory activities for TAMs' reprogramming. In this review, the recent progress of nanomaterials-involved TAMs' reprogramming is comprehensively discussed. The various nanomaterials for TAMs' reprogramming and the reprogramming strategies are summarized and introduced. Additionally, the challenges and perspectives of TAMs' reprogramming for efficient therapy are discussed, aiming to provide inspiration for TAMs' regulator design and promote the development of TAMs-mediated immunotherapy.
Collapse
Affiliation(s)
- Shu-Lan Li
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Hua-Ying Hou
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Xu Chu
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Ying Zhu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Juan Zhang
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Meng-Die Duan
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Junyi Liu
- Albany Medical College, New York 12208, United States
| | - Yi Liu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
| |
Collapse
|
18
|
Xia Y, Wu K, Liu C, Zhao X, Wang J, Cao J, Chen Z, Fang M, Yu J, Zhu C, Zhang X, Wang Z. Filamentous-Actin-Mimicking Nanoplatform for Enhanced Cytosolic Protein Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305600. [PMID: 38152963 PMCID: PMC10933650 DOI: 10.1002/advs.202305600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/29/2023] [Indexed: 12/29/2023]
Abstract
Despite the potential of protein therapeutics, the cytosolic delivery of proteins with high efficiency and bioactivity remains a significant challenge owing to exocytosis and lysosomal degradation after endocytosis. Therefore, it is important to develop a safe and efficient strategy to bypass endocytosis. Inspired by the extraordinary capability of filamentous-actin (F-actin) to promote cell membrane fusion, a cyanine dye assembly-containing nanoplatform mimicking the structure of natural F-actin is developed. The nanoplatform exhibits fast membrane fusion to cell membrane mimics and thus enters live cells through membrane fusion and bypasses endocytosis. Moreover, it is found to efficiently deliver protein cargos into live cells and quickly release them into the cytosol, leading to high protein cargo transfection efficiency and bioactivity. The nanoplatform also results in the superior inhibition of tumor cells when loaded with anti-tumor proteins. These results demonstrate that this fusogenic nanoplatform can be valuable for cytosolic protein delivery and tumor treatment.
Collapse
Affiliation(s)
- Yuqiong Xia
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Keyun Wu
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Chang Liu
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Xuejuan Zhao
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Jun Wang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Jianxia Cao
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Zhaoxu Chen
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Minchao Fang
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin University92 Weijin Road, Nankai DistrictTianjin300072P. R. China
| | - Jie Yu
- School of Biology and EngineeringGuizhou Medical UniversityGuizhouGuiyang550025P. R. China
| | - Cheng Zhu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin University92 Weijin Road, Nankai DistrictTianjin300072P. R. China
| | - Xianghan Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Guangzhou Institute of TechnologyXidian UniversityGuangzhouGuangdong510555P. R. China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| |
Collapse
|
19
|
Yan J, Zhang H, Li G, Su J, Wei Y, Xu C. Lipid nanovehicles overcome barriers to systemic RNA delivery: Lipid components, fabrication methods, and rational design. Acta Pharm Sin B 2024; 14:579-601. [PMID: 38322344 PMCID: PMC10840434 DOI: 10.1016/j.apsb.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/24/2023] [Accepted: 10/08/2023] [Indexed: 02/08/2024] Open
Abstract
Lipid nanovehicles are currently the most advanced vehicles used for RNA delivery, as demonstrated by the approval of patisiran for amyloidosis therapy in 2018. To illuminate the unique superiority of lipid nanovehicles in RNA delivery, in this review, we first introduce various RNA therapeutics, describe systemic delivery barriers, and explain the lipid components and methods used for lipid nanovehicle preparation. Then, we emphasize crucial advances in lipid nanovehicle design for overcoming barriers to systemic RNA delivery. Finally, the current status and challenges of lipid nanovehicle-based RNA therapeutics in clinical applications are also discussed. Our objective is to provide a comprehensive overview showing how to utilize lipid nanovehicles to overcome multiple barriers to systemic RNA delivery, inspiring the development of more high-performance RNA lipid nanovesicles in the future.
Collapse
Affiliation(s)
- Jing Yan
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Institute of Medicine, Shanghai University, Shanghai 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guangfeng Li
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Shanghai 200433, China
| |
Collapse
|
20
|
Dai W, Chen Y, Xue Y, Wan M, Mao C, Zhang K. Progress in the Treatment of Peritoneal Metastatic Cancer and the Application of Therapeutic Nanoagents. ACS APPLIED BIO MATERIALS 2023; 6:4518-4548. [PMID: 37916787 DOI: 10.1021/acsabm.3c00662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Peritoneal metastatic cancer is a cancer caused by the direct growth of cancer cells from the primary site through the bloodstream, lymph, or peritoneum, which is a difficult part of current clinical treatment. In the abdominal cavity of patients with metastatic peritoneal cancer, there are usually nodules of various sizes and malignant ascites. Among them, nodules of different sizes can obstruct intestinal movement and form intestinal obstruction, while malignant ascites can cause abdominal distension and discomfort, and even cause patients to have difficulty in breathing. The pathology and physiology of peritoneal metastatic cancer are complex and not fully understood. The main hypothesis is "seed" and "soil"; i.e., cells from the primary tumor are shed and implanted in the peritoneal cavity (peritoneal metastasis). In the last two decades, the main treatment modalities used clinically are cytoreductive surgery (CRS), systemic chemotherapy, intraperitoneal chemotherapy, and combined treatment, all of which help to improve patient survival and quality of life (QOL). However, the small-molecule chemotherapeutic drugs used clinically still have problems such as rapid drug metabolism and systemic toxicity. With the rapid development of nanotechnology in recent years, therapeutic nanoagents for the treatment of peritoneal metastatic cancer have been gradually developed, which has improved the therapeutic effect and reduced the systemic toxicity of small-molecule chemotherapeutic drugs to a certain extent. In addition, nanomaterials have been developed not only as therapeutic agents but also as imaging agents to guide peritoneal tumor CRS. In this review, we describe the etiology and pathological features of peritoneal metastatic cancer, discuss in detail the clinical treatments that have been used for peritoneal metastatic cancer, and analyze the advantages and disadvantages of the different clinical treatments and the QOL of the treated patients, followed by a discussion focusing on the progress, obstacles, and challenges in the use of therapeutic nanoagents in peritoneal metastatic cancer. Finally, therapeutic nanoagents and therapeutic tools that may be used in the future for the treatment of peritoneal metastatic cancer are prospected.
Collapse
Affiliation(s)
- Wenjun Dai
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yidan Chen
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yunxin Xue
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ke Zhang
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
21
|
Chen G, Wang L, He P, Su T, Lai Q, Kuo HC, Wu W, Chen SL, Tu CC. Biodistributions and Imaging of Poly(ethylene glycol)-Conjugated Silicon Quantum Dot Nanoparticles in Osteosarcoma Models via Intravenous and Intratumoral Injections. ACS APPLIED BIO MATERIALS 2023; 6:4856-4866. [PMID: 37843986 DOI: 10.1021/acsabm.3c00595] [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: 10/18/2023]
Abstract
Osteosarcoma is a malignant tumor with relatively high mortality rates in children and adolescents. While nanoparticles have been widely used in assisting the diagnosis and treatment of cancers, the biodistributions of nanoparticles in osteosarcoma models have not been well studied. Herein, we synthesize biocompatible and highly photoluminescent silicon quantum dot nanoparticles (SiQDNPs) and investigate their biodistributions in osteosarcoma mouse models after intravenous and intratumoral injections by fluorescence imaging. The bovine serum albumin (BSA)-coated and poly(ethylene glycol) (PEG)-conjugated SiQDNPs, when dispersed in phosphate-buffered saline (PBS), can emit red photoluminescence with the photoluminescence quantum yield more than 30% and have very low in vitro and in vivo toxicity. The biodistributions after intravenous injections reveal that the SiQDNPs are mainly metabolized through the livers in mice, while only slight accumulation in the osteosarcoma tumor is observed. Furthermore, the PEG conjugation can effectively extend the circulation time. Finally, a mixture of SiQDNPs and indocyanine green (ICG), which complement each other in the spectral range and diffusion length, is directly injected into the tumor for imaging. After the injection, the SiQDNPs with relatively large particle sizes stay around the injection site, while the ICG molecules diffuse over a broad range, especially in the muscular tissue. By taking advantage of this property, the difference between the osteosarcoma tumor and normal muscular tissue is demonstrated.
Collapse
Affiliation(s)
- Guo Chen
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Wang
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Pengbo He
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Taiyu Su
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qingxuan Lai
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao-Chung Kuo
- Hon Hai Research Institute, Foxconn Technology Group, Shenzhen 518109, China
| | - Wen Wu
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, Shanghai 200030, China
| | - Sung-Liang Chen
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
- Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, Shanghai 200030, China
| | - Chang-Ching Tu
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
- Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, Shanghai 200030, China
- Hon Hai Research Institute, Foxconn Technology Group, Shenzhen 518109, China
| |
Collapse
|
22
|
Lee B, Park OK, Pan L, Kim K, Kang T, Kim H, Lee N, Choi SH, Hyeon T. Co-Delivery of Metabolic Modulators Leads to Simultaneous Lactate Metabolism Inhibition and Intracellular Acidification for Synergistic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305512. [PMID: 37487702 DOI: 10.1002/adma.202305512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Simultaneous lactate metabolism inhibition and intracellular acidification (LIIA) is a promising approach for inducing tumor regression by depleting ATP. However, given the limited efficacy of individual metabolic modulators, a combination of various modulators is required for highly efficient LIIA. Herein, a co-delivery system that combines lactate transporter inhibitor, glucose oxidase, and O2 -evolving nanoparticles is proposed. As a vehicle, a facile room-temperature synthetic method for large-pore mesoporous silica nanoparticles (L-MSNs) is developed. O2 -evolving nanoparticles are then conjugated onto L-MSNs, followed by immobilizing the lactate transporter inhibitor and glucose oxidase inside the pores of L-MSNs. To load the lactate transporter inhibitor, which is too small to be directly loaded into the large pores, it is encapsulated in albumin by controlling the albumin conformation before being loaded into L-MSNs. Notably, inhibiting lactate efflux shifts the glucose consumption mechanism from lactate metabolism to glucose oxidase reaction, which eliminates glucose and produces acid. This leads to synergistic LIIA and subsequent ATP depletion in cancer cells. Consequently, L-MSN-based co-delivery of modulators for LIIA shows high anticancer efficacy in several mouse tumor models without toxicity in normal tissues. This study provides new insights into co-delivery of small-molecule drugs, proteins, and nanoparticles for synergistic metabolic modulation in tumors.
Collapse
Affiliation(s)
- Bowon Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ok Kyu Park
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Limin Pan
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kang Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taegyu Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyunjoong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul, 02707, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
23
|
Picchetti P, Volpi S, Sancho-Albero M, Rossetti M, Dore MD, Trinh T, Biedermann F, Neri M, Bertucci A, Porchetta A, Corradini R, Sleiman H, De Cola L. Supramolecular Nucleic Acid-Based Organosilica Nanoparticles Responsive to Physical and Biological Inputs. J Am Chem Soc 2023; 145:22903-22912. [PMID: 37844092 PMCID: PMC10603779 DOI: 10.1021/jacs.3c04345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Indexed: 10/18/2023]
Abstract
Organosilica nanoparticles that contain responsive organic building blocks as constitutive components of the silica network offer promising opportunities for the development of innovative drug formulations, biomolecule delivery, and diagnostic tools. However, the synthetic challenges required to introduce dynamic and multifunctional building blocks have hindered the realization of biomimicking nanoparticles. In this study, capitalizing on our previous research on responsive nucleic acid-based organosilica nanoparticles, we combine the supramolecular programmability of nucleic acid (NA) interactions with sol-gel chemistry. This approach allows us to create dynamic supramolecular bridging units of nucleic acids in a silica-based scaffold. Two peptide nucleic acid-based monoalkoxysilane derivatives, which self-assemble into a supramolecular bis-alkoxysilane through direct base pairing, were chosen as the noncovalent units inserted into the silica network. In addition, a bridging functional NA aptamer leads to the specific recognition of ATP molecules. In a one-step bottom-up approach, the resulting supramolecular building blocks can be used to prepare responsive organosilica nanoparticles. The supramolecular Watson-Crick-Franklin interactions of the organosilica nanoparticles result in a programmable response to external physical (i.e., temperature) and biological (i.e., DNA and ATP) inputs and thus pave the way for the rational design of multifunctional silica materials with application from drug delivery to theranostics.
Collapse
Affiliation(s)
- Pierre Picchetti
- Karlsruhe
Institute of Technology (KIT), Institute
of Nanotechnology (INT), Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen 76344, Germany
| | - Stefano Volpi
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - María Sancho-Albero
- Department
of Molecular Biochemistry and Pharmacology, Instituto di Ricerche Farmacologiche Mario Negri, IRCCS, 20156 Milano, Italy
| | - Marianna Rossetti
- Department
of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, Rome 00133, Italy
| | - Michael D. Dore
- Department
of Chemistry, McGill University, 801 Sherbrooke St. W., Montreal, Québec City H3A 0B8, Canada
| | - Tuan Trinh
- Department
of Chemistry, McGill University, 801 Sherbrooke St. W., Montreal, Québec City H3A 0B8, Canada
| | - Frank Biedermann
- Karlsruhe
Institute of Technology (KIT), Institute
of Nanotechnology (INT), Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen 76344, Germany
| | - Martina Neri
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Alessandro Bertucci
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Alessandro Porchetta
- Department
of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, Rome 00133, Italy
| | - Roberto Corradini
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Hanadi Sleiman
- Department
of Chemistry, McGill University, 801 Sherbrooke St. W., Montreal, Québec City H3A 0B8, Canada
| | - Luisa De Cola
- Karlsruhe
Institute of Technology (KIT), Institute
of Nanotechnology (INT), Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen 76344, Germany
- Department
of Molecular Biochemistry and Pharmacology, Instituto di Ricerche Farmacologiche Mario Negri, IRCCS, 20156 Milano, Italy
- Dipartimento
DISFARM, University of Milano, via Camillo Golgi 19, 20133 Milano, Italy
| |
Collapse
|
24
|
Picchetti P, Volpi S, Rossetti M, Dore MD, Trinh T, Biedermann F, Neri M, Bertucci A, Porchetta A, Corradini R, Sleiman H, De Cola L. Responsive Nucleic Acid-Based Organosilica Nanoparticles. J Am Chem Soc 2023; 145:22896-22902. [PMID: 37734737 PMCID: PMC10603775 DOI: 10.1021/jacs.3c00393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Indexed: 09/23/2023]
Abstract
The development of smart nanoparticles (NPs) that encode responsive features in the structural framework promises to extend the applications of NP-based drugs, vaccines, and diagnostic tools. New nanocarriers would ideally consist of a minimal number of biocompatible components and exhibit multiresponsive behavior to specific biomolecules, but progress is limited by the difficulty of synthesizing suitable building blocks. Through a nature-inspired approach that combines the programmability of nucleic acid interactions and sol-gel chemistry, we report the incorporation of synthetic nucleic acids and analogs, as constitutive components, into organosilica NPs. We prepared different nanomaterials containing single-stranded nucleic acids that are covalently embedded in the silica network. Through the incorporation of functional nucleic acids into the organosilica framework, the particles respond to various biological, physical, and chemical inputs, resulting in detectable physicochemical changes. The one-step bottom-up approach used to prepare organosilica NPs provides multifunctional systems that combine the tunability of oligonucleotides with the stiffness, low cost, and biocompatibility of silica for different applications ranging from drug delivery to sensing.
Collapse
Affiliation(s)
- Pierre Picchetti
- Karlsruhe
Institute of Technology (KIT), Institute of Nanotechnology (INT), Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen 76344, Germany
| | - Stefano Volpi
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Marianna Rossetti
- Department
of Sciences and Chemical Technologies, University
of Rome, Tor Vergata,
Via della Ricerca Scientifica, Rome 00133, Italy
| | - Michael D. Dore
- Department
of Chemistry, McGill University, 801 Sherbrooke Street W., Montreal, Québec City H3A 0B8, Canada
| | - Tuan Trinh
- Department
of Chemistry, McGill University, 801 Sherbrooke Street W., Montreal, Québec City H3A 0B8, Canada
| | - Frank Biedermann
- Karlsruhe
Institute of Technology (KIT), Institute of Nanotechnology (INT), Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen 76344, Germany
| | - Martina Neri
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Alessandro Bertucci
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Alessandro Porchetta
- Department
of Sciences and Chemical Technologies, University
of Rome, Tor Vergata,
Via della Ricerca Scientifica, Rome 00133, Italy
| | - Roberto Corradini
- Department
of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Hanadi Sleiman
- Department
of Chemistry, McGill University, 801 Sherbrooke Street W., Montreal, Québec City H3A 0B8, Canada
| | - Luisa De Cola
- Karlsruhe
Institute of Technology (KIT), Institute of Nanotechnology (INT), Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen 76344, Germany
- Dipartimento
DISFARM, University of Milano, via Camillo Golgi 19, 20133 Milano, Italy
- Department
of Molecular Biochemistry and Pharmacology, Instituto di Ricerche Farmacologiche Mario Negri, IRCCS, 20156 Milano, Italy
| |
Collapse
|
25
|
Byun J, Wu Y, Park J, Kim JS, Li Q, Choi J, Shin N, Lan M, Cai Y, Lee J, Oh YK. RNA Nanomedicine: Delivery Strategies and Applications. AAPS J 2023; 25:95. [PMID: 37784005 DOI: 10.1208/s12248-023-00860-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/04/2023] [Indexed: 10/04/2023] Open
Abstract
Delivery of RNA using nanomaterials has emerged as a new modality to expand therapeutic applications in biomedical research. However, the delivery of RNA presents unique challenges due to its susceptibility to degradation and the requirement for efficient intracellular delivery. The integration of nanotechnologies with RNA delivery has addressed many of these challenges. In this review, we discuss different strategies employed in the design and development of nanomaterials for RNA delivery. We also highlight recent advances in the pharmaceutical applications of RNA delivered via nanomaterials. Various nanomaterials, such as lipids, polymers, peptides, nucleic acids, and inorganic nanomaterials, have been utilized for delivering functional RNAs, including messenger RNA (mRNA), small interfering RNA, single guide RNA, and microRNA. Furthermore, the utilization of nanomaterials has expanded the applications of functional RNA as active pharmaceutical ingredients. For instance, the delivery of antigen-encoding mRNA using nanomaterials enables the transient expression of vaccine antigens, leading to immunogenicity and prevention against infectious diseases. Additionally, nanomaterial-mediated RNA delivery has been investigated for engineering cells to express exogenous functional proteins. Nanomaterials have also been employed for co-delivering single guide RNA and mRNA to facilitate gene editing of genetic diseases. Apart from the progress made in RNA medicine, we discuss the current challenges and future directions in this field.
Collapse
Affiliation(s)
- Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Qiaoyun Li
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Namjo Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Meng Lan
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
26
|
Liu B, Wang B, Wang Z, Meng Y, Li Y, Li L, Wang J, Zhai M, Liu R, Wei F. Near-Infrared Light-Controlled MicroRNA-21-Loaded Upconversion Nanoparticles to Promote Bone Formation in the Midpalatal Suture. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43503-43514. [PMID: 37694956 DOI: 10.1021/acsami.3c08616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Rapid maxillary expansion (RME) is a common therapy for maxillary transverse deficiency. However, relapses after RME usually occur because of insufficient bone formation. MicroRNA-21 (miR-21) was reported as an important post-transcriptional modulator for osteogenesis. Herein, a photocontrolled miR-21 (PC-miR-21)-loaded nanosystem using upconversion nanoparticles (UCNPs) modified with poly(ether imide) (PEI), i.e., UCNPs@PEI@PC-miR-21, was constructed to promote bone formation in the midpalatal suture. UCNPs@PEI was constructed as the light transducer and delivery carrier. The UCNPs@PEI@PC-miR-21 nanocomplexes have good aqueous dispersibility and biocompatibility. The in vitro cell experiment suggested that UCNPs@PEI could protect PC-miR-21 from biodegradation and release PC-miR-21 into the cytoplasm under near-infrared light (NIR) irradiation. Furthermore, UCNPs@PEI@PC-miR-21 upregulated the expression of the osteogenic key markers, ALP, RUNX2, and COL1A1, at the levels of both genes and proteins. Besides, the results of the in vivo RME mice models further corroborated that photocontrollable UCNPs@PEI@PC-miR-21 accelerated bone formation with upregulating osteogenic markers of ALP, RUNX2, and osteoprotegerin and inducing fewer osteoclasts formation. In conclusion, UCNPs@PEI@PC-miR-21 nanoparticles with a NIR light could facilitate the remote and precise delivery of exogenous miR-21 to the midpalatal suture to promote bone formation during RME. This work represents a cutting-edge approach of gene therapy to promote osteogenesis in the midpalatal suture during RME and provides a frontier scientific basis for later clinical treatment.
Collapse
Affiliation(s)
- Bohui Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
- Department of Stomatology, Qingdao West Coast New Area Central Hospital, Qingdao 266555, China
| | - Bing Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Ziyao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Yiling Meng
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Yixuan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Lan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Jixiao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Mingrui Zhai
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Rui Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| |
Collapse
|
27
|
Fletcher RB, Stokes LD, Kelly IB, Henderson KM, Vallecillo-Viejo IC, Colazo JM, Wong BV, Yu F, d'Arcy R, Struthers MN, Evans BC, Ayers J, Castanon M, Weirich MJ, Reilly SK, Patel SS, Ivanova YI, Silvera Batista CA, Weiss SM, Gersbach CA, Brunger JM, Duvall CL. Nonviral In Vivo Delivery of CRISPR-Cas9 Using Protein-Agnostic, High-Loading Porous Silicon and Polymer Nanoparticles. ACS NANO 2023; 17:16412-16431. [PMID: 37582231 PMCID: PMC11129837 DOI: 10.1021/acsnano.2c12261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The complexity of CRISPR machinery is a challenge to its application for nonviral in vivo therapeutic gene editing. Here, we demonstrate that proteins, regardless of size or charge, efficiently load into porous silicon nanoparticles (PSiNPs). Optimizing the loading strategy yields formulations that are ultrahigh loading─>40% cargo by volume─and highly active. Further tuning of a polymeric coating on the loaded PSiNPs yields nanocomposites that achieve colloidal stability under cryopreservation, endosome escape, and gene editing efficiencies twice that of the commercial standard Lipofectamine CRISPRMAX. In a mouse model of arthritis, PSiNPs edit cells in both the cartilage and synovium of knee joints, and achieve 60% reduction in expression of the therapeutically relevant MMP13 gene. Administered intramuscularly, they are active over a broad dose range, with the highest tested dose yielding nearly 100% muscle fiber editing at the injection site. The nanocomposite PSiNPs are also amenable to systemic delivery. Administered intravenously in a model that mimics muscular dystrophy, they edit sites of inflamed muscle. Collectively, the results demonstrate that the PSiNP nanocomposites are a versatile system that can achieve high loading of diverse cargoes and can be applied for gene editing in both local and systemic delivery applications.
Collapse
Affiliation(s)
- R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Larry D Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Isom B Kelly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Katelyn M Henderson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Isabel C Vallecillo-Viejo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Benjamin V Wong
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Richard d'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Morgan N Struthers
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Brian C Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Jacob Ayers
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Matthew Castanon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Michael J Weirich
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Sarah K Reilly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Shrusti S Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Yoanna I Ivanova
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Carlos A Silvera Batista
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Sharon M Weiss
- Department of Electrical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235-1631, United States
| |
Collapse
|
28
|
Zheng C, Zhong Q, Yi K, Kong H, Cao F, Zhuo C, Xu Y, Shi R, Ju E, Song W, Tao Y, Chen X, Li M. Anti-phagocytosis-blocking repolarization-resistant membrane-fusogenic liposome (ARMFUL) for adoptive cell immunotherapy. SCIENCE ADVANCES 2023; 9:eadh2413. [PMID: 37556535 PMCID: PMC10411906 DOI: 10.1126/sciadv.adh2413] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023]
Abstract
Equipping multiple functionalities on adoptive effector cells is essential to overcome the complex immunological barriers in solid tumors for superior antitumor efficacy. However, current cell engineering technologies cannot endow these functionalities to cells within a single step because of the different spatial distributions of targets in one cell. Here, we present a core-shell anti-phagocytosis-blocking repolarization-resistant membrane-fusogenic liposome (ARMFUL) to achieve one-step multiplexing cell engineering for multifunctional cell construction. Through fusing with the M1 macrophage membrane, ARMFUL inserts an anti-CD47 (aCD47)-modified lipid shell onto the surface and simultaneously delivers colony-stimulating factor 1 receptor inhibitor BLZ945-loaded core into the cytoplasm. The surface-presenting aCD47 boosts macrophage's phagocytosis against the tumor by blocking CD47. The cytoplasm-located BLZ945 prompts its polarization resistance to M2 phenotype in the immunosuppressive microenvironment via inactivating the intracellular M2 polarization signaling pathway. This ARMFUL provides a versatile cell engineering platform to customize multimodal cellular functions for enhanced adoptive cell therapy.
Collapse
Affiliation(s)
- Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Chenya Zhuo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
29
|
Hotta M, Hayase K, Kitanaka A, Li T, Takeoka S. Development of the observation of membrane fusion with label-free liposomes by calcium imaging. Biochem Biophys Rep 2023; 34:101483. [PMID: 37250982 PMCID: PMC10209117 DOI: 10.1016/j.bbrep.2023.101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/31/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Liposomes are artificial vesicles composed of lipid bilayers that have enabled drugs to be encapsulated and delivered to tumor tissue. Membrane-fusogenic liposomes fuse with the plasma membranes of cells to deliver encapsulated drugs directly to the cytosol, which makes it a promising method for rapid and highly efficient drug delivery. In a previous study, liposomal lipid bilayers were labeled with fluorescent probes, and colocalization of labeled lipids with plasma membrane was observed under a microscope. However, there was concern that fluorescent labeling would affect lipid dynamics and cause liposomes to acquire membrane fusogenic ability. In addition, encapsulation of hydrophilic fluorescent substances in the inner aqueous phase sometimes requires an additional step of removing unencapsulated substances after preparation, and there is a risk of leakage. Herein, we propose a new method to observe cell interaction with liposomes without labeling. Our laboratory has developed two types of liposomes with different cellular internalization pathways, i.e., endocytosis and membrane fusion. We found that cytosolic calcium influx would be triggered following the internalization of cationic liposomes, and different cell entry routes led to different calcium responses. Thus, the correlation between cell entry routes and calcium responses could be utilized to study liposome-cell interactions without fluorescent labeling lipids. Briefly, liposomes were added to phorbol 12-myristate 13-acetate (PMA)-primed THP-1 cells, and calcium influx was measured by time-lapse imaging using a fluorescent indicator (Fura 2-AM). Liposomes with high membrane fusogenic ability elicited a strong transient calcium response immediately after adding liposomes, whereas those taken up mainly by endocytosis elicited multiple weak calcium responses. In order to verify the cell entry routes, we also tracked the intracellular distribution of fluorescent-labeled liposomes in PMA-primed THP-1 cells using a confocal laser scanning microscope. It was shown that for fusogenic liposomes, colocalization with plasma membrane occurred at the same time as calcium elevation, whereas for liposomes with a high endocytosis potential, fluorescent dots were observed in the cytoplasm, suggesting the cell internalization by endocytosis. These results suggested that the calcium response patterns correspond to cell entry routes, and membrane fusion can be observed by calcium imaging.
Collapse
Affiliation(s)
- Morihiro Hotta
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Kengo Hayase
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Aya Kitanaka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Tianshu Li
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| |
Collapse
|
30
|
Qiu C, Xia F, Zhang J, Shi Q, Meng Y, Wang C, Pang H, Gu L, Xu C, Guo Q, Wang J. Advanced Strategies for Overcoming Endosomal/Lysosomal Barrier in Nanodrug Delivery. RESEARCH (WASHINGTON, D.C.) 2023; 6:0148. [PMID: 37250954 PMCID: PMC10208951 DOI: 10.34133/research.0148] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/27/2023] [Indexed: 05/31/2023]
Abstract
Nanocarriers have therapeutic potential to facilitate drug delivery, including biological agents, small-molecule drugs, and nucleic acids. However, their efficiency is limited by several factors; among which, endosomal/lysosomal degradation after endocytosis is the most important. This review summarizes advanced strategies for overcoming endosomal/lysosomal barriers to efficient nanodrug delivery based on the perspective of cellular uptake and intracellular transport mechanisms. These strategies include promoting endosomal/lysosomal escape, using non-endocytic methods of delivery to directly cross the cell membrane to evade endosomes/lysosomes and making a detour pathway to evade endosomes/lysosomes. On the basis of the findings of this review, we proposed several promising strategies for overcoming endosomal/lysosomal barriers through the smarter and more efficient design of nanodrug delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chong Qiu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiaoli Shi
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuqing Meng
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chen Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Huanhuan Pang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liwei Gu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chengchao Xu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
- Department of Nephrology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital,
Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| |
Collapse
|
31
|
Zheng C, Zhong Q, Song W, Yi K, Kong H, Wang H, Tao Y, Li M, Chen X. Membrane-Fusion-Mediated Multiplex Engineering of Tumor Cell Surface Glycans for Enhanced NK Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206989. [PMID: 36566024 DOI: 10.1002/adma.202206989] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Natural killer (NK) cell therapies show potential for tumor treatment but are immunologically resisted by the overexpressed immunosuppressing tumor cell surface glycans. To reverse this glycan-mediated immunosuppression, the surface NK-inhibitory glycan expressions need to be downregulated and NK-activating glycan levels should be elevated synchronously with optimal efficiency. Here, a core-shell membrane-fusogenic liposome (MFL) is designed to simultaneously achieve the physical modification of NK-activating glycans and biological inhibition of immunosuppressing glycans on the tumor cell surface via a membrane-fusion manner. Loaded into a tumor-microenvironment-triggered-degradable thermosensitive hydrogel, MFLs could be conveniently injected and controllably released into local tumor. Through fusion with tumor cell membrane, the released MFLs could simultaneously deliver sialyltransferase-inhibitor-loaded core into cytoplasm, and anchor NK-activating-glycan-modified shell onto tumor surface. This spatially-differential distribution of core and shell in one cell ensures the effective inhibition of intracellular sialyltransferase to downregulate immunosuppressing sialic acid, and direct presentation of NK-activating Lewis X trisaccharide (LeX) on tumor surface simultaneously. Consequentially, the sialic acid-caused immunosuppression of tumor surface is reprogrammed to be LeX-induced NK activation, resulting in sensitive susceptibility to NK-cell-mediated recognition and lysis for improved tumor elimination. This MFL provides a novel platform for multiplex cell engineering and personalized regulation of intercellular interactions for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
32
|
Cheng R, Santos HA. Smart Nanoparticle-Based Platforms for Regulating Tumor Microenvironment and Cancer Immunotherapy. Adv Healthc Mater 2023; 12:e2202063. [PMID: 36479842 PMCID: PMC11468886 DOI: 10.1002/adhm.202202063] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/18/2022] [Indexed: 12/12/2022]
Abstract
Tumor development and metastasis are closely related to the tumor microenvironment (TME). Recently, several studies indicate that modulating TME can enhance cancer immunotherapy. Among various approaches to modulating TME, nanoparticles (NPs) with unique inherent advantages and smart modified characteristics are promising candidates in delivering drugs to cancer cells, amplifying the therapeutic effects, and leading to a cascade of immune responses. In this review, several smart NP-based platforms are briefly introduced, such as responsive NPs, targeting NPs, and the composition of TME, including dendritic cells, macrophages, fibroblasts, endothelial cells, myeloid-derived suppressor cells, and regulatory T cells. Moreover, the recent applications of smart NP-based platforms in regulating TME and cancer immunotherapy are briefly introduced. Last, the advantages and disadvantages of these smart NP-based platforms in potential clinical translation are discussed.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Department of Biomedical EngineeringUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- W. J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Hélder A. Santos
- Department of Biomedical EngineeringUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- W. J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| |
Collapse
|
33
|
Zhao Y, Gao S, Song D, Ye Z, Xu R, Luo Y, Xu Q. Lipidoid Artificial Compartments for Bidirectional Regulation of Enzyme Activity through Nanomechanical Action. J Am Chem Soc 2023; 145:551-559. [PMID: 36537880 DOI: 10.1021/jacs.2c11004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Photoresponsive inhibitor and noninhibitor systems have been developed to achieve on-demand enzyme activity control. However, inhibitors are only effective for a specific and narrow range of enzymes. Noninhibitor systems usually require mutation and modification of the enzymes, leading to irreversible loss of enzymatic activities. Inspired by biological membranes, we herein report a lipidoid-based artificial compartment composed of azobenzene (Azo) lipidoids and helper lipids, which can bidirectionally regulate the activity of the encapsulated enzymes by light. In this system, the reversible photoisomerization of Azo lipidoids triggered by UV/vis light creates a continuous rotation-inversion movement, thereby enhancing the permeability of the compartment membrane and allowing substrates to pass through. Moreover, the membrane can revert to its impermeable state when light is removed. Thus, enzyme activity can be switched on and off when encapsulating enzymes in the compartments. Importantly, since neither mutation nor modification is required, negligible loss of activity is observed for the encapsulated enzymes after repeated activation and inhibition. Furthermore, this approach provides a generic strategy for controlling multiple enzymes by forgoing the use of inhibitors and may broaden the applications of enzymes in biological mechanism research and precision medicine.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Shuliang Gao
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Donghui Song
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Ruijie Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Ying Luo
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
34
|
Li J, Fan J, Gao Y, Huang S, Huang D, Li J, Wang X, Santos HA, Shen P, Xia B. Porous Silicon Nanocarriers Boost the Immunomodulation of Mitochondria-Targeted Bovine Serum Albumins on Macrophage Polarization. ACS NANO 2023; 17:1036-1053. [PMID: 36598186 PMCID: PMC9878978 DOI: 10.1021/acsnano.2c07439] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/29/2022] [Indexed: 05/31/2023]
Abstract
The development of nanosystems with intrinsic immunomodulatory effects on macrophage polarization is important for the macrophage-targeted immunotherapy. Here, mitochondria-targeted bovine serum albumins (BSAs) via the conjugation of fluorescent, lipophilic, and cationic rhodamine 110 molecules can efficiently enhance the gene expression of the proinflammatory phenotype of macrophages and correspondingly inhibit the gene expression of their anti-inflammatory phenotype. On this basis, porous silicon nanocarriers can further boost the immunomodulation of these mitochondria-targeted BSAs in vitro or in vivo, accompanied by the secretion of proinflammatory mediators including tumor necrosis factor α, nitric oxide, and reactive oxygen species (ROS). Meanwhile, BSA coatings can also improve the biocompatibility of porous silicon nanoparticulate cores on macrophages. Finally, the mechanism investigations demonstrate that porous silicon nanocarriers can efficiently deliver mitochondria-targeted BSA into macrophages to generate mitochondrial ROS via the interference with mitochondrial respiratory chains, which can further trigger the downstream signaling transduction pathways for the proinflammatory transition. Considering the good biosafety and versatile loading capability, this developed porous silicon@BSA nanosystem with a strong proinflmmatory regulatory effect has important potential on the combinatorial chemoimmunotherapy against cancer or viral/bacterial-related infectious diseases.
Collapse
Affiliation(s)
- Jialiang Li
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Jiqiang Fan
- State
Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive
Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital
of Nanjing University Medical School, Nanjing
University, Nanjing210023, China
| | - Yan Gao
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Shuodan Huang
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Di Huang
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Jiachen Li
- Department
of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AVGroningen, The Netherlands
- W.
J. Kolff Institute for Biomedical Engineering and Materials Science,
University Medical Center Groningen, University
of Groningen, Antonius
Deusinglaan 1, 9713 AVGroningen, The Netherlands
| | - Xiaoyu Wang
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Hélder A. Santos
- Department
of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AVGroningen, The Netherlands
- W.
J. Kolff Institute for Biomedical Engineering and Materials Science,
University Medical Center Groningen, University
of Groningen, Antonius
Deusinglaan 1, 9713 AVGroningen, The Netherlands
| | - Pingping Shen
- Department
of Geriatric Medicine, The Second Affiliated
Hospital and Yuying Children’s Hospital of Wenzhou Medical
University, Wenzhou325027, China
- State
Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive
Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital
of Nanjing University Medical School, Nanjing
University, Nanjing210023, China
| | - Bing Xia
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| |
Collapse
|
35
|
Recent advances in targeted gene silencing and cancer therapy by nanoparticle-based delivery systems. Biomed Pharmacother 2023; 157:114065. [PMID: 36481408 DOI: 10.1016/j.biopha.2022.114065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Nanomedicine has emerged as a promising platform for disease treatment and much progress has been achieved in the clinical translation for cancer treatment. Several types of nanomedicines have been approved for therapeutic application. However, many nanoparticles still suffer from challenges in the translation from bench to bedside. Currently, nanoparticle-based delivery systems have been developed to explore their functions in targeted gene silencing and cancer therapy. This review describes the research progress of different nano-carriers in targeted gene editing, and the recent progress in co-delivery of anticancer drugs and small ribonucleic acid. We also summarize the strategies for improving the specificity of carrier systems. Finally, we discuss the functions of targeted nano-carriers in overcoming chemotherapeutic drug resistance in cancer therapy. As research continues to advance, a better understanding of the safety including long-term toxicity, immunogenicity, and body metabolism may impel nanoparticle translation.
Collapse
|
36
|
Vijayakumar S, Nasr SH, Davis JE, Wang E, Zuidema JM, Lu YS, Lo YH, Sicklick JK, Sailor MJ, Ray P. Anti-KIT DNA aptamer-conjugated porous silicon nanoparticles for the targeted detection of gastrointestinal stromal tumors. NANOSCALE 2022; 14:17700-17713. [PMID: 36416809 PMCID: PMC9744628 DOI: 10.1039/d2nr03905b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Evaluation of Gastrointestinal Stromal Tumors (GIST) during initial clinical staging, surgical intervention, and postoperative management can be challenging. Current imaging modalities (e.g., PET and CT scans) lack sensitivity and specificity. Therefore, advanced clinical imaging modalities that can provide clinically relevant images with high resolution would improve diagnosis. KIT is a tyrosine kinase receptor overexpressed on GIST. Here, the application of a specific DNA aptamer targeting KIT, decorated onto a fluorescently labeled porous silicon nanoparticle (pSiNP), is used for the in vitro & in vivo imaging of GIST. This nanoparticle platform provides high-fidelity GIST imaging with minimal cellular toxicity. An in vitro analysis shows greater than 15-fold specific KIT protein targeting compared to the free KIT aptamer, while in vivo analyses of GIST-burdened mice that had been injected intravenously (IV) with aptamer-conjugated pSiNPs show extensive nanoparticle-to-tumor signal co-localization (>90% co-localization) compared to control particles. This provides an effective platform for which aptamer-conjugated pSiNP constructs can be used for the imaging of KIT-expressing cancers or for the targeted delivery of therapeutics.
Collapse
Affiliation(s)
- Sanahan Vijayakumar
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
| | - Seyedmehdi H Nasr
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California, 92093, USA
| | - Jacob E Davis
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, San Diego, California, 92093, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, California, 92093, USA
| | - Edward Wang
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
| | - Jonathan M Zuidema
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California, 92093, USA
- Department of Neurosciences, University of California, San Diego, San Diego, California, 92093, USA
| | - Yi-Sheng Lu
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
| | - Yu-Hwa Lo
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
- Department of Electrical & Computer Engineering, University of California, San Diego, La Jolla, California, 92093, USA
| | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, San Diego, California, 92093, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, California, 92093, USA
- Department of Pharmacology, University of California, San Diego, San Diego, California, 92093, USA
| | - Michael J Sailor
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California, 92093, USA
| | - Partha Ray
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, San Diego, California, 92093, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, California, 92093, USA
| |
Collapse
|
37
|
Xu T, Liu Z, Huang L, Jing J, Liu X. Modulating the tumor immune microenvironment with nanoparticles: A sword for improving the efficiency of ovarian cancer immunotherapy. Front Immunol 2022; 13:1057850. [PMID: 36532066 PMCID: PMC9751906 DOI: 10.3389/fimmu.2022.1057850] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
With encouraging antitumor effects, immunotherapy represented by immune checkpoint blockade has developed into a mainstream cancer therapeutic modality. However, only a minority of ovarian cancer (OC) patients could benefit from immunotherapy. The main reason is that most OC harbor a suppressive tumor immune microenvironment (TIME). Emerging studies suggest that M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), myeloid-derived suppressor cells (MDSCs), and cancer-associated fibroblasts (CAFs) are enriched in OC. Thus, reversing the suppressive TIME is considered an ideal candidate for improving the efficiency of immunotherapy. Nanoparticles encapsulating immunoregulatory agents can regulate immunocytes and improve the TIME to boost the antitumor immune response. In addition, some nanoparticle-mediated photodynamic and photothermal therapy can directly kill tumor cells and induce tumor immunogenic cell death to activate antigen-presenting cells and promote T cell infiltration. These advantages make nanoparticles promising candidates for modulating the TIME and improving OC immunotherapy. In this review, we analyzed the composition and function of the TIME in OC and summarized the current clinical progress of OC immunotherapy. Then, we expounded on the promising advances in nanomaterial-mediated immunotherapy for modulating the TIME in OC. Finally, we discussed the obstacles and challenges in the clinical translation of this novel combination treatment regimen. We believe this resourceful strategy will open the door to effective immunotherapy of OC and benefit numerous patients.
Collapse
Affiliation(s)
| | | | | | - Jing Jing
- *Correspondence: Xiaowei Liu, ; Jing Jing,
| | | |
Collapse
|
38
|
Grondek JF, Huffman K, Lee EJ, Cavichini M, Warter A, P Kalaw FG, Heinke A, Fan R, Cheng L, Sailor MJ, Freeman WR. Effective treatment of retinal neovascular leakage with fusogenic porous silicon nanoparticles delivering VEGF-siRNA. Nanomedicine (Lond) 2022; 17:2089-2108. [PMID: 36748946 PMCID: PMC10031552 DOI: 10.2217/nnm-2022-0255] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/13/2023] [Indexed: 02/08/2023] Open
Abstract
Aim: To evaluate an intravitreally injected nanoparticle platform designed to deliver VEGF-A siRNA to inhibit retinal neovascular leakage as a new treatment for proliferative diabetic retinopathy and diabetic macular edema. Materials & methods: Fusogenic lipid-coated porous silicon nanoparticles loaded with VEGF-A siRNA, and pendant neovascular integrin-homing iRGD, were evaluated for efficacy by intravitreal injection in a rabbit model of retinal neovascularization. Results: For 12 weeks post-treatment, a reduction in vascular leakage was observed for treated diseased eyes versus control eyes (p = 0.0137), with a corresponding reduction in vitreous VEGF-A. Conclusion: Fusogenic lipid-coated porous silicon nanoparticles siRNA delivery provides persistent knockdown of VEGF-A and reduced leakage in a rabbit model of retinal neovascularization as a potential new intraocular therapeutic.
Collapse
Affiliation(s)
- Joel F Grondek
- Department of Chemistry & Biochemistry, University of California, San Diego, CA 92093, USA
| | - Kristyn Huffman
- Department of Ophthalmology, Jacobs Retinal Center at Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| | - Ella Jiyoon Lee
- Department of Chemistry & Biochemistry, University of California, San Diego, CA 92093, USA
| | - Melina Cavichini
- Department of Ophthalmology, Jacobs Retinal Center at Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| | - Alexandra Warter
- Department of Ophthalmology, Jacobs Retinal Center at Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| | - Fritz Gerald P Kalaw
- Department of Ophthalmology, Jacobs Retinal Center at Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| | - Anna Heinke
- Department of Ophthalmology, Jacobs Retinal Center at Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| | - Ruhan Fan
- Materials Science & Engineering, University of California, San Diego, CA 92093, USA
| | - Lingyun Cheng
- Department of Ophthalmology, Jacobs Retinal Center at Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| | - Michael J Sailor
- Department of Chemistry & Biochemistry, University of California, San Diego, CA 92093, USA
- Materials Science & Engineering, University of California, San Diego, CA 92093, USA
| | - William R Freeman
- Department of Ophthalmology, Jacobs Retinal Center at Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| |
Collapse
|
39
|
Nie W, Chen J, Wang B, Gao X. Nonviral vector system for cancer immunogene therapy. MEDCOMM – BIOMATERIALS AND APPLICATIONS 2022. [DOI: 10.1002/mba2.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu PR China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu PR China
| | - Bilan Wang
- Department of Pharmacy West China Second University Hospital of Sichuan University Chengdu PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu PR China
| |
Collapse
|
40
|
Meng J, Zhang P, Chen Q, Wang Z, Gu Y, Ma J, Li W, Yang C, Qiao Y, Hou Y, Jing L, Wang Y, Gu Z, Zhu L, Xu H, Lu X, Gao M. Two-Pronged Intracellular Co-Delivery of Antigen and Adjuvant for Synergistic Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202168. [PMID: 35362203 DOI: 10.1002/adma.202202168] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/22/2022] [Indexed: 05/27/2023]
Abstract
Nanovaccines have emerged as promising alternatives or complements to conventional cancer treatments. Despite the progresses, specific co-delivery of antigen and adjuvant to their corresponding intracellular destinations for maximizing the activation of antitumor immune responses remains a challenge. Herein, a lipid-coated iron oxide nanoparticle is delivered as nanovaccine (IONP-C/O@LP) that can co-deliver peptide antigen and adjuvant (CpG DNA) into cytosol and lysosomes of dendritic cells (DCs) through both membrane fusion and endosome-mediated endocytosis. Such two-pronged cellular uptake pattern enables IONP-C/O@LP to synergistically activate immature DCs. Iron oxide nanoparticle also exhibits adjuvant effects by generating intracellular reactive oxygen species, which further promotes DC maturation. IONP-C/O@LP accumulated in the DCs of draining lymph nodes effectively increases the antigen-specific T cells in both tumor and spleen, inhibits tumor growth, and improves animal survival. Moreover, it is demonstrated that this nanovaccine is a general platform of delivering clinically relevant peptide antigens derived from human papilloma virus 16 to trigger antigen-specific immune responses in vivo.
Collapse
Affiliation(s)
- Junli Meng
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peisen Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qizhe Chen
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zihua Wang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuan Gu
- Center for Molecular Imaging and Nuclear Medicine School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215123, China
| | - Jie Ma
- Center for Molecular Imaging and Nuclear Medicine School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215123, China
| | - Wang Li
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chen Yang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanyuan Qiao
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yi Hou
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Lihong Jing
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yong Wang
- Center for Molecular Imaging and Nuclear Medicine School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215123, China
| | - Zi Gu
- School of Chemical Engineering and Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW, 2052, Australia
| | - Lichong Zhu
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Haozhen Xu
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, 85287-1804, USA
| | - Xueguang Lu
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Mingyuan Gao
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- Center for Molecular Imaging and Nuclear Medicine School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215123, China
| |
Collapse
|
41
|
Lin F, Li X, Wang X, Sun H, Wang Z, Wang X. Stanniocalcin 1 promotes metastasis, lipid metabolism and cisplatin chemoresistance via the FOXC2/ITGB6 signaling axis in ovarian cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:129. [PMID: 35392966 PMCID: PMC8988421 DOI: 10.1186/s13046-022-02315-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 03/08/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Stanniocalcin 1 (STC1) plays an integral role in ovarian cancer (OC). However, the functional role of STC1 in metastasis, lipid metabolism and cisplatin (DDP) chemoresistance in OC is not fully understood. METHODS Single-cell sequencing and IHC analysis were performed to reveal STC1 expression profiles in patient tissues. Metastasis, lipid metabolism and DDP chemoresistance were subsequently assessed. Cell-based in vitro and in vivo assays were subsequently conducted to gain insight into the underlying mechanism of STC1 in OC. RESULTS Single-cell sequencing assays and IHC analysis verified that STC1 expression was significantly enhanced in OC tissues compared with para-carcinoma tissues, and it was further up-regulated in peritoneal metastasis tissues compared with OC tissues. In vitro and in vivo experiments demonstrated that STC1 promoted metastasis, lipid metabolism and DDP chemoresistance in OC. Simultaneously, STC1 promoted lipid metabolism by up-regulating lipid-related genes such as UCP1, TOM20 and perilipin1. Mechanistically, STC1 directly bound to integrin β6 (ITGB6) to activate the PI3K signaling pathway. Moreover, STC1 was directly regulated by Forkhead box C2 (FOXC2) in OC. Notably, targeting STC1 and the FOXC2/ITGB6 signaling axis was related to DDP chemoresistance in vitro. CONCLUSIONS Overall, these findings revealed that STC1 promoted metastasis, lipid metabolism and DDP chemoresistance via the FOXC2/ITGB6 signaling axis in OC. Thus, STC1 may be used as a prognostic indicator in patients with metastatic OC. Meanwhile, STC1 could be a therapeutic target in OC patients, especially those who have developed chemoresistance to DDP.
Collapse
Affiliation(s)
- Feikai Lin
- Department of Gynecology and Obstetrics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Xiaoduan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, People's Republic of China
| | - Xinjing Wang
- Department of Gynecology and Obstetrics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Huizhen Sun
- Department of Gynecology and Obstetrics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Ziliang Wang
- Department of Gynecology and Obstetrics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China.
| | - Xipeng Wang
- Department of Gynecology and Obstetrics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
42
|
Li H, Yang YG, Sun T. Nanoparticle-Based Drug Delivery Systems for Induction of Tolerance and Treatment of Autoimmune Diseases. Front Bioeng Biotechnol 2022; 10:889291. [PMID: 35464732 PMCID: PMC9019755 DOI: 10.3389/fbioe.2022.889291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune disease is a chronic inflammatory disease caused by disorders of immune regulation. Antigen-specific immunotherapy has the potential to inhibit the autoreactivity of inflammatory T cells and induce antigen-specific immune suppression without impairing normal immune function, offering an ideal strategy for autoimmune disease treatment. Tolerogenic dendritic cells (Tol DCs) with immunoregulatory functions play important roles in inducing immune tolerance. However, the effective generation of tolerogenic DCs in vivo remains a great challenge. The application of nanoparticle-based drug delivery systems in autoimmune disease treatment can increase the efficiency of inducing antigen-specific tolerance in vivo. In this review, we discuss multiple nanoparticles, with a focus on their potential in treatment of autoimmune diseases. We also discuss how the physical properties of nanoparticles influence their therapeutic efficacy.
Collapse
Affiliation(s)
- He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- Department of Rehabilitation Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| |
Collapse
|
43
|
Yuan H, Huang X, Li Q, Luo C, Lin C, Zhang S, Zhang Y, Yan Z, Du N, Liu Z, Jiang H, Chen B. SiRNA-circFARSA-loaded porous silicon nanomaterials for pancreatic cancer treatment via inhibition of CircFARSA expression. Pharmacotherapy 2022; 147:112672. [DOI: 10.1016/j.biopha.2022.112672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/02/2022]
|
44
|
Xu W, Zhao Z, Falconer J, Whittaker AK, Popat A, Smith MT, Kumeria T, Han FY. Sustained release ketamine-loaded porous silicon-PLGA microparticles prepared by an optimized supercritical CO 2 process. Drug Deliv Transl Res 2022; 12:676-694. [PMID: 33907987 DOI: 10.1007/s13346-021-00991-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 12/15/2022]
Abstract
Ketamine in sub-anaesthetic doses has analgesic properties and an opioid-sparing effect. Intrathecal (i.t.) delivery of analgesics bypasses systemic metabolism and delivers the analgesic agent adjacent to the target receptors in the spinal cord and so small doses are required to achieve effective pain relief. In order to relieve intractable cancer-related pain, sustained-release ketamine formulations are required in combination with a strong opioid because frequent i.t. injection is not practical. In this study, ketamine or ketamine-loaded porous silicon (pSi) were encapsulated into poly(lactic-co-glycolic acid) (PLGA) microparticles by a novel supercritical carbon dioxide (scCO2) method, thereby avoiding the use of organic solvent. Multiple parameters including theoretical drug loading (DL), presence of pSi, size of scCO2 vessel, PLGA type, and use of co-solvent were investigated with a view to obtaining high DL and a sustained-release for an extended period. The most important finding was that the use of a large scCO2 vessel (60 mL) resulted in a much higher encapsulation efficiency (EE) compared with a small vessel (12 mL). In addition, pre-loading ketamine into pSi slightly improved the level of drug incorporation (i.e. EE and DL). Although the in vitro release was mainly affected by the drug payload, the use of the large scCO2 vessel reduced the burst release and extended the release period for PLGA microparticles with 10% or 20% ketamine loading. Together, our findings provide valuable information for optimization of drug delivery systems prepared with the aid of scCO2.
Collapse
Affiliation(s)
- Weizhi Xu
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Zonglan Zhao
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - James Falconer
- School of Pharmacy, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Andrew K Whittaker
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- ARC Centre of Excellence in Convergent Bio Nano Science and Technology, The University of Queensland, Brisbane, QLD, Australia
| | - Amirali Popat
- School of Pharmacy, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, Australia
- Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Tushar Kumeria
- School of Pharmacy, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, Australia.
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, Australia.
| | - Felicity Y Han
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
45
|
Ottonelli I, Caraffi R, Tosi G, Vandelli MA, Duskey JT, Ruozi B. Tunneling Nanotubes: A New Target for Nanomedicine? Int J Mol Sci 2022; 23:ijms23042237. [PMID: 35216348 PMCID: PMC8878036 DOI: 10.3390/ijms23042237] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/01/2023] Open
Abstract
Tunneling nanotubes (TNTs), discovered in 2004, are thin, long protrusions between cells utilized for intercellular transfer and communication. These newly discovered structures have been demonstrated to play a crucial role in homeostasis, but also in the spreading of diseases, infections, and metastases. Gaining much interest in the medical research field, TNTs have been shown to transport nanomedicines (NMeds) between cells. NMeds have been studied thanks to their advantageous features in terms of reduced toxicity of drugs, enhanced solubility, protection of the payload, prolonged release, and more interestingly, cell-targeted delivery. Nevertheless, their transfer between cells via TNTs makes their true fate unknown. If better understood, TNTs could help control NMed delivery. In fact, TNTs can represent the possibility both to improve the biodistribution of NMeds throughout a diseased tissue by increasing their formation, or to minimize their formation to block the transfer of dangerous material. To date, few studies have investigated the interaction between NMeds and TNTs. In this work, we will explain what TNTs are and how they form and then review what has been published regarding their potential use in nanomedicine research. We will highlight possible future approaches to better exploit TNT intercellular communication in the field of nanomedicine.
Collapse
Affiliation(s)
- Ilaria Ottonelli
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.C.); (G.T.); (M.A.V.); (B.R.)
| | - Riccardo Caraffi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.C.); (G.T.); (M.A.V.); (B.R.)
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.C.); (G.T.); (M.A.V.); (B.R.)
| | - Maria Angela Vandelli
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.C.); (G.T.); (M.A.V.); (B.R.)
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.C.); (G.T.); (M.A.V.); (B.R.)
- Correspondence:
| | - Barbara Ruozi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.C.); (G.T.); (M.A.V.); (B.R.)
| |
Collapse
|
46
|
Yan Y, Liu XY, Lu A, Wang XY, Jiang LX, Wang JC. Non-viral vectors for RNA delivery. J Control Release 2022; 342:241-279. [PMID: 35016918 PMCID: PMC8743282 DOI: 10.1016/j.jconrel.2022.01.008] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/13/2022]
Abstract
RNA-based therapy is a promising and potential strategy for disease treatment by introducing exogenous nucleic acids such as messenger RNA (mRNA), small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides (ASO) to modulate gene expression in specific cells. It is exciting that mRNA encoding the spike protein of COVID-19 (coronavirus disease 2019) delivered by lipid nanoparticles (LNPs) exhibits the efficient protection of lungs infection against the virus. In this review, we introduce the biological barriers to RNA delivery in vivo and discuss recent advances in non-viral delivery systems, such as lipid-based nanoparticles, polymeric nanoparticles, N-acetylgalactosamine (GalNAc)-siRNA conjugate, and biomimetic nanovectors, which can protect RNAs against degradation by ribonucleases, accumulate in specific tissue, facilitate cell internalization, and allow for the controlled release of the encapsulated therapeutics.
Collapse
Affiliation(s)
- Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiao-Yu Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiang-Yu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Lin-Xia Jiang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China..
| |
Collapse
|
47
|
Zhao Y, Hou X, Chai J, Zhang Z, Xue X, Huang F, Liu J, Shi L, Liu Y. Stapled Liposomes Enhance Cross-Priming of Radio-Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107161. [PMID: 34767279 DOI: 10.1002/adma.202107161] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/09/2021] [Indexed: 06/13/2023]
Abstract
The release of tumor-associated antigens (TAAs) and their cross-presentation in dendritic cells (DCs) are crucial for radio-immunotherapy. However, the irradiation resistance of tumor cells usually results in limited TAA generation and release. Importantly, TAAs internalized by DCs are easily degraded in lysosomes, resulting in unsatisfactory extent of TAA cross-presentation. Herein, an antigen-capturing stapled liposome (ACSL) with a robust structure and bioactive surface is developed. The ACSLs capture and transport TAAs from lysosomes to the cytoplasm in DCs, thereby enhancing TAA cross-presentation. l-arginine encapsulated in ACSLs induces robust T cell-dependent antitumor response and immune memory in 4T1 tumor-bearing mice after local irradiation, resulting in significant tumor suppression and an abscopal effect. Replacing l-arginine with radiosensitizers, photosensitizers, and photothermal agents may make ACSL a universal platform for the rapid development of various combinations of anticancer therapies.
Collapse
Affiliation(s)
- Yu Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Xiaoxue Hou
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Jingshan Chai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Zhanzhan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Fan Huang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| |
Collapse
|
48
|
Kong H, Yi K, Zheng C, Lao YH, Zhou H, Chan HF, Wang H, Tao Y, Li M. Membrane-fusogenic biomimetic particles: a new bioengineering tool learned from nature. J Mater Chem B 2022; 10:6841-6858. [PMID: 35781483 DOI: 10.1039/d2tb00632d] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Membrane fusion, a fundamental biological process of the fusion of the membrane composition between cells within natural organisms, is vital for cell-cell communication and cargo transport between the living cells....
Collapse
Affiliation(s)
- Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Huicong Zhou
- College of Science, Changchun Institute of Technology, Changchun 130012, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
49
|
Wu L, Zhou W, Lin L, Chen A, Feng J, Qu X, Zhang H, Yue J. Delivery of therapeutic oligonucleotides in nanoscale. Bioact Mater 2022; 7:292-323. [PMID: 34466734 PMCID: PMC8379367 DOI: 10.1016/j.bioactmat.2021.05.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/28/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Therapeutic oligonucleotides (TOs) represent one of the most promising drug candidates in the targeted cancer treatment due to their high specificity and capability of modulating cellular pathways that are not readily druggable. However, efficiently delivering of TOs to cancer cellular targets is still the biggest challenge in promoting their clinical translations. Emerging as a significant drug delivery vector, nanoparticles (NPs) can not only protect TOs from nuclease degradation and enhance their tumor accumulation, but also can improve the cell uptake efficiency of TOs as well as the following endosomal escape to increase the therapeutic index. Furthermore, targeted and on-demand drug release of TOs can also be approached to minimize the risk of toxicity towards normal tissues using stimuli-responsive NPs. In the past decades, remarkable progresses have been made on the TOs delivery based on various NPs with specific purposes. In this review, we will first give a brief introduction on the basis of TOs as well as the action mechanisms of several typical TOs, and then describe the obstacles that prevent the clinical translation of TOs, followed by a comprehensive overview of the recent progresses on TOs delivery based on several various types of nanocarriers containing lipid-based nanoparticles, polymeric nanoparticles, gold nanoparticles, porous nanoparticles, DNA/RNA nanoassembly, extracellular vesicles, and imaging-guided drug delivery nanoparticles.
Collapse
Affiliation(s)
- Lei Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Lihua Lin
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Anhong Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Jing Feng
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Xiangmeng Qu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
| | - Jun Yue
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| |
Collapse
|
50
|
Xu J, Chen T, Sun T, Yu C, Yan D, Zhu L. Erythrocyte membrane camouflaged siRNA/chemodrug nanoassemblies for cancer combination therapy. Biomater Sci 2022; 10:6601-6613. [DOI: 10.1039/d2bm01478e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Erythrocyte membrane camouflaged nanoassemblies of siRNA/chemodrugs were constructed, in which cationic amphiphilic chemodrugs interact with negatively charged siRNA and self-assemble into siRNA/chemodrug nanoparticles for combination cancer therapy.
Collapse
Affiliation(s)
- Jie Xu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200217, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tianbao Chen
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200217, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tingting Sun
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunyang Yu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Deyue Yan
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200217, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lijuan Zhu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200217, China
| |
Collapse
|