1
|
Szefczyk M, Szulc N, Bystranowska D, Szczepańska A, Lizandra Pérez J, Dudek A, Pawlak A, Ożyhar A, Berlicki Ł. Construction and cytotoxicity evaluation of peptide nanocarriers based on coiled-coil structures with a cyclic β-amino acid at the knob-into-hole interaction site. J Mater Chem B 2025. [PMID: 40364573 DOI: 10.1039/d5tb00752f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Peptides are highly attractive as nanocarriers for drug delivery and other biomedical applications due to their unique combination of biocompatibility, efficacy, safety, and versatility-qualities that are difficult to achieve with other nanocarrier types. Particularly promising in this context are peptide foldamers containing non-canonical residues, which can yield nanostructures with diverse physicochemical properties. Additionally, the introduction of non-proteinogenic amino acids into the sequence enhances conformational stability and resistance to proteolysis, critical features for bioapplications. In this article, we report the development of novel foldameric bundles based on a coiled-coil structure incorporating trans-(1S,2S)-2-aminocyclopentanecarboxylic acid (trans-ACPC) at the key interacting site. We also provide both theoretical and experimental analyses of how this cyclic β-residue affects the thermodynamic and proteolytic stability, oligomerization state, and encapsulation properties of the resulting foldamers compared to standard coiled-coils. Additionally, we assessed the cytotoxicity of these foldamers using the MTT assay on 3T3 cells. The results demonstrate that neither the foldamers nor trans-ACPC exhibit toxic effects on the 3T3 cell line, highlighting their potential as safe and effective nanocarriers.
Collapse
Affiliation(s)
- Monika Szefczyk
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| | - Natalia Szulc
- Department of Physics and Biophysics, Faculty of Biotechnology and Food Sciences, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland
| | - Dominika Bystranowska
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Anna Szczepańska
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| | - Juan Lizandra Pérez
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| | - Anita Dudek
- Department of Physics and Biophysics, Faculty of Biotechnology and Food Sciences, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland
| | - Aleksandra Pawlak
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Norwida 31, 50-375, Wrocław, Poland
| | - Andrzej Ożyhar
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Łukasz Berlicki
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| |
Collapse
|
2
|
Hu B, Ji C, Zhou Z, Xu X, Wang L, Cao T, Cheng J, Sun R. Bioorthogonal reaction-mediated photosensitizer-peptide conjugate anchoring on cell membranes for enhanced photodynamic therapy. Biomater Sci 2025; 13:1233-1242. [PMID: 39846132 DOI: 10.1039/d4bm01602e] [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: 01/24/2025]
Abstract
Photodynamic therapy (PDT), utilizing a photosensitizer (PS) to induce tumor cell death, is an effective modality for cancer treatment. PS-peptide conjugates have recently demonstrated remarkable antitumor potential in preclinical trials. However, the limited cell membrane binding affinity and rapid systemic clearance have hindered their transition to clinical applications. To address these challenges, we investigated whether in vivo covalent chemistry could enhance tumor accumulation and potentiate antitumor efficacy. Specifically, we synthesized a PS-peptide conjugate termed P-DBCO-Ce6, with chlorin e6 (Ce6) and dibenzocyclooctyne (DBCO) conjugated to a negatively charged short peptide. By employing metabolic glycoengineering and bioorthogonal reactions, P-DBCO-Ce6 achieves covalent bonding to the cell membrane, enabling prolonged retention of the PS on the cell surface and the in situ generation of reactive oxygen species (ROS) on cell membranes to kill tumor cells. In vivo studies demonstrated a 3.3-fold increase in tumor accumulation of the PS through bioorthogonal reactions compared to the control group, confirming that click chemistry can effectively enhance PS tumor accumulation. This approach allows for the effective elimination of tumors with a single treatment. The improved efficiency of this strategy provides new insights into the design of PDT systems for potential clinical applications.
Collapse
Affiliation(s)
- Buwei Hu
- Department of Materials Science, Fudan University, Shanghai 200433, China
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
| | - Chenlin Ji
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310023, China
| | - Zhuohang Zhou
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
| | - Xuehan Xu
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310023, China
| | - Luyi Wang
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310023, China
| | - Tingting Cao
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310023, China
| | - Jianjun Cheng
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang 310030, China
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310023, China.
| | - Rui Sun
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310023, China.
| |
Collapse
|
3
|
Zhang C, Fan J, Wu L. Application of nanomaterials in precision treatment of lung cancer. iScience 2025; 28:111704. [PMID: 39886464 PMCID: PMC11780121 DOI: 10.1016/j.isci.2024.111704] [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] [Indexed: 02/01/2025] Open
Abstract
Lung cancer remains one of the most prevalent and lethal malignancies worldwide, characterized by high mortality rates due to its aggressive nature, metastatic potential, and drug resistance. Despite advancements in conventional therapies, their efficacy is often limited by systemic toxicity, poor tumor specificity, and the emergence of resistance mechanisms. Nanomedicine has emerged as a promising approach to address these challenges, leveraging the unique physicochemical properties of nanomaterials to enhance drug delivery, reduce off-target effects, and enable combination therapies. This review provides a comprehensive overview of the applications of nanomaterials in lung cancer treatment, focusing on advancements in chemotherapy, phototherapy, and immunotherapy. Key strategies include the development of stimuli-responsive nanoparticles, active targeting mechanisms, and multifunctional platforms for co-delivery of therapeutic agents. Notable successes, such as liposomal formulations and polymeric nanoparticles, highlight the potential to overcome biological barriers and improve therapeutic outcomes. However, significant challenges remain, including limited tumor penetration, immunogenicity, scalability in manufacturing, and regulatory complexities. Addressing these limitations through innovative design, advanced manufacturing technologies, and interdisciplinary collaboration will be critical to translating nanomedicine from bench to bedside. Overall, nanomedicine represents a transformative frontier in lung cancer therapy, offering the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Chengcheng Zhang
- Department of Thoracic Surgery, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Jiang Fan
- Department of Thoracic Surgery, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Liang Wu
- Department of Thoracic Surgery, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| |
Collapse
|
4
|
Xu W, Qin X, Liu Y, Chen J, Wang Y. Advances in Enzyme-responsive Supramolecular In situ Self-assembled Peptide for Drug Delivery. Curr Drug Deliv 2025; 22:374-386. [PMID: 37496133 DOI: 10.2174/1567201820666230726151607] [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: 03/29/2023] [Revised: 05/17/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023]
Abstract
Because of low immunogenicity, ease of modification, and inherent biosafety, peptides have been well recognized as vehicles to deliver therapeutic agents to targeted regions with improved pharmacokinetic characteristics. Enzyme-responsive self-assembled peptides (ERSAPs) show superiority over their naive forms due to their enhanced targeting efficacy and long-retention property. In this review, we have summarized recent advances in the therapeutic application of ERSAPs, mainly focusing on their self-therapeutic properties and potential as vehicles to deliver different drugs.
Collapse
Affiliation(s)
- Wentao Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiaowen Qin
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yang Liu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jun Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuguang Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
5
|
Wang H, Hao D, Wu Q, Sun T, Xie Z. A morphologically transformable hypoxia-induced radical anion for tumor-specific photothermal therapy. Acta Pharm Sin B 2024; 14:5407-5417. [PMID: 39807323 PMCID: PMC11725169 DOI: 10.1016/j.apsb.2024.09.017] [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: 05/22/2023] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 01/16/2025] Open
Abstract
Tumor microenvironment activatable therapeutic agents and their effective tumor accumulation are significant for selective tumor treatment. Herein, we provide an unadulterated nanomaterial combining the above advantages. We synthesize a perylene diimide (PDI) molecule substituted by glutamic acid (Glu), which can self-assemble into small spherical nanoparticles (PDI-SG) in aqueous solution. PDI-SG can not only be transformed into nanofibers at low pH conditions but also be reduced to PDI radical anion (PDI·‒), which exhibits strong near-infrared absorption and excellent photothermal performance. More importantly, PDI-SG can also be reduced to PDI·‒ in hypoxic tumors to ablate the tumors and minimize the damage to normal tissues. The morphological transformation from small nanoparticles to nanofibers makes for better tumor accumulation and retention. This work sheds light on the design of tumor microenvironment activatable therapeutics with precise structures for high-performance tumor therapy.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Dengyuan Hao
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Qihang Wu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Tingting Sun
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
6
|
Ma J, Yang H, Tian X, Meng F, Zhai X, Li A, Li C, Wang M, Wang G, Lu C, Bai J. Matrix metalloproteinase 2-responsive dual-drug-loaded self-assembling peptides suppress tumor growth and enhance breast cancer therapy. Bioeng Transl Med 2024; 9:e10702. [PMID: 39545088 PMCID: PMC11558207 DOI: 10.1002/btm2.10702] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/07/2024] [Accepted: 07/04/2024] [Indexed: 11/17/2024] Open
Abstract
Conventional chemotherapeutic agents are limited by their lack of targeting and penetration and their short retention time, and chemotherapy might induce an immune suppressive environment. Peptide self-assembly can result in a specific morphology, and the resulting morphological changes are stimuli responsive to the external environment, which is important for drug permeation and retention of encapsulated chemotherapeutic agents. In this study, a polypeptide (Pep1) containing the peptide sequences PLGLAG and RGD that is responsive to matrix metalloproteinase 2 (MMP-2) was successfully developed. Pep1 underwent a morphological transformation from a spherical structure to aggregates with a high aspect ratio in response to MMP-2 induction. This drug delivery system (DI/Pep1) can transport doxorubicin (DOX) and indomethacin (IND) simultaneously to target tumor cells for subsequent drug release while extending drug retention within tumor cells, which increases immunogenic cell death and facilitates the immunotherapeutic effect of CD4+ T cells. Ultimately, DI/Pep1 attenuated tumor-associated inflammation, enhanced the body's immune response, and inhibited breast cancer growth by combining the actions of DOX and IND. Our research offers an approach to hopefully enhance the effectiveness of cancer treatment.
Collapse
Affiliation(s)
- Jihong Ma
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Haiyan Yang
- Emergency DepartmentYantaishan HospitalYantaiChina
| | - Xue Tian
- School of Basic Medical SciencesShandong Second Medical UniversityWeifangChina
| | - Fanhu Meng
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Xiaoqing Zhai
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Aimei Li
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Chuntao Li
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Min Wang
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Guohui Wang
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Chunbo Lu
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Jingkun Bai
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| |
Collapse
|
7
|
Wang D, Chen X, Chen S, Wang H, Yang J, He J, Liu X, Zhao Y, Zhang J. Enzyme-activated binary assembly for targeted, controlled delivery of anti-liver cancer compounds. Carbohydr Res 2024; 544:109229. [PMID: 39154417 DOI: 10.1016/j.carres.2024.109229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
Liver cancer is the third leading cause of cancer deaths globally. The use of Hydroxycamptothecin (HCPT) as a first-line chemotherapeutic agent for liver, lung, and gastric cancers is often hampered by its low activity, limited targeting, and poor water solubility. This results in a low accumulation of HCPT in tumor cells, as well as the inability to maintain continuous treatment. Consequently, there is an urgent need to develop an accessory method that can enhance the therapeutic efficacy of HCPT while exhibiting good biocompatibility and targeted delivery ability. To address this critical issue, an enzyme-triggered supramolecular nanocarrier, refer as SCD/LCC SNCs, has been successfully developed, leveraging the aggregation of the negatively charged sulfate-modified β-CDs and positively charged lauroylcholine chloride (LCC). This nanocarrier demonstrates acetylcholinesterase (LCC) triggered decomposition behavior, making it a promising drug carrier for HCPT. The cellular assays conducted have demonstrated that HCPT loaded into these SCD/LCC SNCs exhibit reduced cytotoxicity towards normal cells while maintaining robust tumor inhibitory activity and inducing apoptosis. Therefore, this study offers a promising strategy for the effective use of HCPT in the treatment of liver cancer.
Collapse
Affiliation(s)
- Dandan Wang
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China
| | - Xiangyu Chen
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China
| | - Shuai Chen
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China
| | - Hongxia Wang
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China
| | - Jianmei Yang
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China
| | - Junnan He
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China
| | - Xiaoqing Liu
- Shenzhen Kewode Technology Co., Ltd, Shenzhen, 518028, People's Republic of China
| | - Yan Zhao
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China.
| | - Jin Zhang
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, People's Republic of China.
| |
Collapse
|
8
|
Cao Z, Liu J, Yang X. Deformable nanocarriers for enhanced drug delivery and cancer therapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230037. [PMID: 39439489 PMCID: PMC11491306 DOI: 10.1002/exp.20230037] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/28/2024] [Indexed: 10/25/2024]
Abstract
Recently, the field of nanomedicine has witnessed substantial advancements in the development of nanocarriers for targeted drug delivery, emerges as promising platforms to enhance therapeutic efficacy and minimize adverse effects associated with conventional chemotherapy. Notably, deformable nanocarriers have garnered considerable attention due to their unique capabilities of size changeable, tumor-specific aggregation, stimuli-triggered disintegration, and morphological transformations. These deformable nanocarriers present significant opportunities for revolutionizing drug delivery strategies, by responding to specific stimuli or environmental cues, enabling achieved various functions at the tumor site, including size-shrinkage nanocarriers enhance drug penetration, aggregative nanocarriers enhance retention effect, disintegrating nanocarriers enable controlled drug release, and shape-changing nanocarriers improve cellular uptake, allowing for personalized treatment approaches and combination therapies. This review provides an overview of recent developments and applications of deformable nanocarriers for enhancing tumor therapy, underscores the diverse design strategies employed to create deformable nanocarriers and elucidates their remarkable potential in targeted tumor therapy.
Collapse
Affiliation(s)
- Ziyang Cao
- Department of General SurgeryGuangzhou First People's Hospitalthe Second Affiliated HospitalSouth China University of TechnologyGuangzhouPeople's Republic of China
- Center for Medical Research on Innovation and TranslationInstitute of Clinical MedicineSchool of MedicineGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouPeople's Republic of China
| | - Jing Liu
- School of ChemistryChemical Engineering and Biotechnology Nanyang Technological UniversitySingaporeSingapore
| | - Xianzhu Yang
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdongPeople's Republic of China
| |
Collapse
|
9
|
Wu C, Jiang P, Su W, Yan Y. Alkaline Phosphatase-Instructed Peptide Assemblies for Imaging and Therapeutic Applications. Biomacromolecules 2024; 25:5609-5629. [PMID: 39185628 DOI: 10.1021/acs.biomac.4c00795] [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: 08/27/2024]
Abstract
Self-assembly, a powerful strategy for constructing highly stable and well-ordered supramolecular structures, widely exists in nature and in living systems. Peptides are frequently used as building blocks in the self-assembly process due to their advantageous characteristics, such as ease of synthesis, tunable mechanical stability, good biosafety, and biodegradability. Among the initiators for peptide self-assembly, enzymes are excellent candidates for guiding this process under mild reaction conditions. As a crucial and commonly used biomarker, alkaline phosphatase (ALP) cleaves phosphate groups, triggering a hydrophilicity-to-hydrophobicity transformation that induces peptide self-assembly. In recent years, ALP-instructed peptide self-assembly has made breakthroughs in biological imaging and therapy, inspiring the development of self-assembly biomaterials for diagnosis and therapeutics. In this review, we highlight the most recent advancements in ALP-instructed peptide assemblies and provide perspectives on their potential impact. Finally, we briefly discuss the ongoing challenges for future research in this field.
Collapse
Affiliation(s)
- Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Pingge Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Wen Su
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| |
Collapse
|
10
|
Peng F, Hu M, Su Z, Hu L, Guo L, Yang K. Intratumoral Microbiota as a Target for Advanced Cancer Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405331. [PMID: 39054925 DOI: 10.1002/adma.202405331] [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: 04/15/2024] [Revised: 06/21/2024] [Indexed: 07/27/2024]
Abstract
In recent years, advancements in microbial sequencing technology have sparked an increasing interest in the bacteria residing within solid tumors and its distribution and functions in various tumors. Intratumoral bacteria critically modulate tumor oncogenesis and development through DNA damage induction, chronic inflammation, epigenetic alterations, and metabolic and immune regulation, while also influencing cancer treatment efficacy by affecting drug metabolism. In response to these discoveries, a variety of anti-cancer therapies targeting these microorganisms have emerged. These approaches encompass oncolytic therapy utilizing tumor-associated bacteria, the design of biomaterials based on intratumoral bacteria, the use of intratumoral bacterial components for drug delivery systems, and comprehensive strategies aimed at the eradication of tumor-promoting bacteria. Herein, this review article summarizes the distribution patterns of bacteria in different solid tumors, examines their impact on tumors, and evaluates current therapeutic strategies centered on tumor-associated bacteria. Furthermore, the challenges and prospects for developing drugs that target these bacterial communities are also explored, promising new directions for cancer treatment.
Collapse
Affiliation(s)
- Fei Peng
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Mengyuan Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhiyue Su
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Lingchuan Guo
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Kai Yang
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Alkene-carbon Fibres-based Technology & Application for Detection of Major Infectious Diseases, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
11
|
Srivastava G, Mukherjee E, Mittal R, Ganjewala D. Geraniol and citral: recent developments in their anticancer credentials opening new vistas in complementary cancer therapy. Z NATURFORSCH C 2024; 79:163-177. [PMID: 38635829 DOI: 10.1515/znc-2023-0150] [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: 11/11/2023] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
About 10 million people are diagnosed with cancer each year. Globally, it is the second leading cause of death after heart disease, and by 2035, the death toll could reach 14.6 million. Several drugs and treatments are available to treat cancer, but survival rates remain low. Many studies in recent years have shown that plant-derived monoterpenes, particularly geraniol and citral, are effective against various cancers, including breast, liver, melanoma, endometrial, colon, prostate, and skin cancers. This trend has opened new possibilities for the development of new therapeutics or adjuvants in the field of cancer therapy. These monoterpenes can improve the efficacy of chemotherapy by modulating many signaling molecules and pathways within tumors. Analysis of reports on the anticancer effects published in the past 5 years provided an overview of the most important results of these and related properties. Also, the molecular mechanisms by which they exert their anticancer effects in cell and animal studies have been explained. Therefore, this review aims to highlight the scope of geraniol and citral as complementary or alternative treatment options in cancer therapy.
Collapse
Affiliation(s)
- Gauri Srivastava
- Amity Institute of Biotechnology, 77282 Amity University , Sector-125, Noida 201303, Uttar Pradesh, India
| | - Esha Mukherjee
- Amity Institute of Biotechnology, 77282 Amity University , Sector-125, Noida 201303, Uttar Pradesh, India
| | - Ruchika Mittal
- Amity Institute of Biotechnology, 77282 Amity University , Sector-125, Noida 201303, Uttar Pradesh, India
| | - Deepak Ganjewala
- Amity Institute of Biotechnology, 77282 Amity University , Sector-125, Noida 201303, Uttar Pradesh, India
| |
Collapse
|
12
|
Yang P, Li Y, Qian K, Zhou L, Cheng Y, Wu J, Xu M, Wang T, Yang X, Mu Y, Liu X, Zhang Q. Precise Modulation of Pericyte Dysfunction by a Multifunctional Nanoprodrug to Ameliorate Alzheimer's Disease. ACS NANO 2024; 18:14348-14366. [PMID: 38768086 DOI: 10.1021/acsnano.4c00480] [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: 05/22/2024]
Abstract
Pericyte dysfunction severely undermines cerebrovascular integrity and exacerbates neurodegeneration in Alzheimer's disease (AD). However, pericyte-targeted therapy is a yet-untapped frontier for AD. Inspired by the elevation of vascular cell adhesion molecule-1 (VCAM-1) and reactive oxygen species (ROS) levels in pericyte lesions, we fabricated a multifunctional nanoprodrug by conjugating the hybrid peptide VLC, a fusion of the VCAM-1 high-affinity peptide VHS and the neuroprotective apolipoprotein mimetic peptide COG1410, to curcumin (Cur) through phenylboronic ester bond (VLC@Cur-NPs) to alleviate complex pericyte-related pathological changes. Importantly, VLC@Cur-NPs effectively homed to pericyte lesions via VLC and released their contents upon ROS stimulation to maximize their regulatory effects. Consequently, VLC@Cur-NPs markedly increased pericyte regeneration to form a positive feedback loop and thus improved neurovascular function and ultimately alleviated memory defects in APP/PS1 transgenic mice. We present a promising therapeutic strategy for AD that can precisely modulate pericytes and has the potential to treat other cerebrovascular diseases.
Collapse
Affiliation(s)
- Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Xiyu Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yongkang Mu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Xuan Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| |
Collapse
|
13
|
Gong Z, Zhao H, Bai J. pH-responsive drug-loaded peptides enhance drug accumulation and promote apoptosis in tumor cells. Colloids Surf B Biointerfaces 2024; 239:113954. [PMID: 38744076 DOI: 10.1016/j.colsurfb.2024.113954] [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: 03/25/2024] [Revised: 04/26/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
The efficacy of chemotherapeutic drugs in tumor treatment is limited by their toxicity and side effects due to their inability to selectively accumulate in tumor tissue. In addition, chemotherapeutic agents are easily pumped out of tumor cells, resulting in their inadequate accumulation. To overcome these challenges, a drug delivery system utilizing the amphiphilic peptide Pep1 was designed. Pep1 can self-assemble into spherical nanoparticles (PL/Pep1) and encapsulate paclitaxel (PTX) and lapatinib (LAP). PL/Pep1 transformed into nanofibers in an acidic environment, resulting in longer drug retention and higher drug concentrations within tumor cells. Ultimately, PL/Pep1 inhibited tumor angiogenesis and enhanced tumor cell apoptosis. The use of shape-changing peptides as drug carriers to enhance cancer cell apoptosis is promising.
Collapse
Affiliation(s)
- Zhongying Gong
- College of Economics and Management, Qingdao University of Science and Technology, Qingdao, China
| | - Hongxia Zhao
- College of Economics and Management, Qingdao University of Science and Technology, Qingdao, China.
| | - Jingkun Bai
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
14
|
Shaw I, Boafo GF, Ali YS, Liu Y, Mlambo R, Tan S, Chen C. Advancements and prospects of lipid-based nanoparticles: dual frontiers in cancer treatment and vaccine development. J Microencapsul 2024; 41:226-254. [PMID: 38560994 DOI: 10.1080/02652048.2024.2326091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024]
Abstract
Cancer is a complex heterogeneous disease that poses a significant public health challenge. In recent years, lipid-based nanoparticles (LBNPs) have expanded drug delivery and vaccine development options owing to their adaptable, non-toxic, tuneable physicochemical properties, versatile surface functionalisation, and biocompatibility. LBNPs are tiny artificial structures composed of lipid-like materials that can be engineered to encapsulate and deliver therapeutic agents with pinpoint accuracy. They have been widely explored in oncology; however, our understanding of their pharmacological mechanisms, effects of their composition, charge, and size on cellular uptake, tumour penetration, and how they can be utilised to develop cancer vaccines is still limited. Hence, we reviewed LBNPs' unique characteristics, biochemical features, and tumour-targeting mechanisms. Furthermore, we examined their ability to enhance cancer therapies and their potential contribution in developing anticancer vaccines. We critically analysed their advantages and challenges impeding swift advancements in oncology and highlighted promising avenues for future research.
Collapse
Affiliation(s)
- Ibrahim Shaw
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - George Frimpong Boafo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Yimer Seid Ali
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
- Department of Pharmacy, College of Medicine and Health Science, Wollo University, Dessie, Ethiopia
| | - Yang Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Ronald Mlambo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Chuanpin Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
15
|
Yan H, Xu P, Cong H, Yu B, Shen Y. Research progress in construction of organic carrier drug delivery platform using tumor microenvironment. MATERIALS TODAY CHEMISTRY 2024; 37:101997. [DOI: 10.1016/j.mtchem.2024.101997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Liu Y, Liu Y, Sun X, Wang Y, Du C, Bai J. Morphologically transformable peptide nanocarriers coloaded with doxorubicin and curcumin inhibit the growth and metastasis of hepatocellular carcinoma. Mater Today Bio 2024; 24:100903. [PMID: 38130427 PMCID: PMC10733681 DOI: 10.1016/j.mtbio.2023.100903] [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: 09/23/2023] [Revised: 11/03/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
In tumor treatment, the highly disordered vascular system and lack of accumulation of chemotherapeutic drugs in tumors severely limit the therapeutic role of nanocarriers. Smaller drug-containing nanoparticles (NPs) can better penetrate the tumor but are easily removed, which severely limits the tumor-killing properties of the drug. The chemotherapeutic medication doxorubicin (DOX) is highly toxic to the heart, but this toxicity can be effectively mitigated and the combined anticancer effect can be enhanced by clinically incorporating curcumin (CUR) as part of the dual therapy. We designed a small-molecule peptide, Pep1, containing a targeting peptide (CREKA) and a pH-responsive moiety. These NPs can target the blood vessels in tumor microthrombi and undergo a morphological shift in the tumor microenvironment. This process enhances the penetration and accumulation of drugs, ultimately improving the effectiveness of cancer treatment. In vitro and in vivo experiments demonstrated that this morphological transformation allowed rapid and effective drug release into tumors, the effective inhibition of tumor angiogenesis, and the promotion of tumor cell apoptosis, thus effectively killing tumor cells. Our findings provide a novel and simple approach to nhibit the growth and metastasis of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yun Liu
- School of Stomatology, Weifang Medical University, Weifang, 261053, China
| | - Yunxia Liu
- School of Stomatology, Weifang Medical University, Weifang, 261053, China
- Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, 261035, China
| | - Xinyu Sun
- School of Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Yue Wang
- School of Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Changqing Du
- Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, 261035, China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, China
| |
Collapse
|
17
|
Gong Z, Peng S, Cao J, Tan H, Zhao H, Bai J. Advances in the variations and biomedical applications of stimuli-responsive nanodrug delivery systems. NANOTECHNOLOGY 2024; 35:132001. [PMID: 38198449 DOI: 10.1088/1361-6528/ad170b] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
Chemotherapy is an important cancer treatment modality, but the clinical utility of chemotherapeutics is limited by their toxic side effects, inadequate distribution and insufficient intracellular concentrations. Nanodrug delivery systems (NDDSs) have shown significant advantages in cancer diagnosis and treatment. Variable NDDSs that respond to endogenous and exogenous triggers have attracted much research interest. Here, we summarized nanomaterials commonly used for tumor therapy, such as peptides, liposomes, and carbon nanotubes, as well as the responses of NDDSs to pH, enzymes, magnetic fields, light, and multiple stimuli. Specifically, well-designed NDDSs can change in size or morphology or rupture when induced by one or more stimuli. The varying responses of NDDSs to stimulation contribute to the molecular design and development of novel NDDSs, providing new ideas for improving drug penetration and accumulation, inhibiting tumor resistance and metastasis, and enhancing immunotherapy.
Collapse
Affiliation(s)
- Zhongying Gong
- College of Economics and Management, Qingdao University of Science and Technology, Qingdao 266061, People's Republic of China
| | - Shan Peng
- School of Stomatology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Juanjuan Cao
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, Jinan 250012, People's Republic of China
| | - Hongxia Zhao
- College of Economics and Management, Qingdao University of Science and Technology, Qingdao 266061, People's Republic of China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| |
Collapse
|
18
|
Meng F, Zhai X, Ma J, Li A, Wang X, Bai J. Enzyme-Induced Shape-Shifting Peptide Nanocarrier Coloaded with Paclitaxel and Dipyridamole Inhibits Platelet Function and Tumor Metastasis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:166-177. [PMID: 38143309 DOI: 10.1021/acsami.3c13855] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
Tumor-associated platelets can bind to tumor cells and protect circulating tumor cells from NK-mediated immune surveillance. Tumor-associated platelets secrete cytokines to induce the epithelial-mesenchymal transition (EMT) in tumor cells, which promotes tumor metastasis. Combining chemotherapeutic agents with antiplatelet drugs can reduce the occurrence of metastasis, but the systemic application of chemotherapeutic agents and antiplatelet drugs is prone to causing serious side effects. Therefore, delivering drugs to the tumor microthrombus site for long-lasting inhibition is a problem that needs to be addressed. Here, we show that small molecule peptide nanoparticles containing the Cys-Arg-Glu-Lys-Ala (CREKA) peptide can deliver the platelet inhibitor dipyridamole (DIP) and the chemotherapeutic drug paclitaxel (PTX) to tumor tissues, thereby inhibiting tumor-associated platelet function while killing tumor cells. The drug-loaded nanoparticles PD/Pep1 inhibited platelet-tumor cell interactions, were effectively taken up by tumor cells, and underwent morphological transformation induced by alkaline phosphatase (ALP) to prolong the retention time of the drugs. After intravenous injection, PD/Pep1 can target tumors and inhibit tumor metastasis. Thus, this small molecule peptide nanoformulation provides a simple strategy for efficient drug delivery and shows promise as a novel cancer therapy platform.
Collapse
Affiliation(s)
- Fanhu Meng
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Xiaoqing Zhai
- School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Jihong Ma
- School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Aimei Li
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Xizhen Wang
- Medical Imaging Center, Affiliated Hospital of Weifang Medical University, Weifang 261053, China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
19
|
Wang Y, Xie L, Li X, Wang L, Yang Z. Chemo-immunotherapy by dual-enzyme responsive peptide self-assembling abolish melanoma. Bioact Mater 2024; 31:549-562. [PMID: 37746663 PMCID: PMC10511343 DOI: 10.1016/j.bioactmat.2023.09.006] [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: 06/25/2023] [Revised: 09/10/2023] [Accepted: 09/10/2023] [Indexed: 09/26/2023] Open
Abstract
Herein, we designed Comp. 1 to simultaneously respond to two enzymes: alkaline phosphatase and matrix metalloproteinase 2, which is commonly found in highly malignant cancer cell lines containing B16-F10 murine melanoma cells and CT26 murine colon carcinoma cells. We used the regional differences in the expression levels of dual-markers to accurately release immune molecule IND into tumor microenvironment for the activation of anti-tumor related immune effects, while in-situ self-assembly occurs. The dual-enzyme response process can further regulate the peptide precursors' self-assembly in the form of short rod-shaped nanofibers, enabling the delivery of the loaded chemotherapeutic drug HCPT into the cancer cells and further allowing the peptide assemblies to escape from lysosomes and return to cytoplasm in the form of tiny nanoparticles to induce apoptosis of cancer cells. This process does not occur in the single-positive breast cancer cell line MCF-7 or the normal hepatocytes cell line LO2, indicating the selectivity of the cancer cells exhibited using our strategy. In vivo studies revealed that Comp. 1 can effectively cooperate with chemotherapy to enhance the immunotherapy effect and induce immune responses associated with elevated pro-inflammatory cytokines in vivo to inhibit malignant tumors growth. Our dual-enzyme responsive chemo-immunotherapy strategy feasible in anti-tumor treatment, provides a new avenue for regulating peptide self-assembly to adapt to diverse tumor properties and may eventually be used for the development of novel multifunctional anti-tumor nanomedicines.
Collapse
Affiliation(s)
- Yuhan Wang
- Tianjin Key Laboratory of Inflammation Biology, Department of Pharmacology, School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, PR China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Limin Xie
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Xinxin Li
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Ling Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300071, PR China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
20
|
Zhang L, Zhao L, Lin X, Zhao S, Pan W, Wang D, Sun Z, Li J, Liang Z, Zhang R, Jiang H. Comparison of Tumor Non-specific and PD-L1 Specific Imaging by Near-Infrared Fluorescence/Cerenkov Luminescence Dual-Modality In-situ Imaging. Mol Imaging 2024; 23:15353508241261473. [PMID: 38952401 PMCID: PMC11208884 DOI: 10.1177/15353508241261473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024] Open
Abstract
Background Labeled antibodies are excellent imaging agents in oncology to non-invasively visualize cancer-related antigens expression levels. However, tumor tracer uptake (TTU) of specific antibodies in-vivo may be inferior to non-specific IgG in some cases. Objectives To explore factors affecting labeled antibody visualization by PD-L1 specific and non-specific imaging of nude mouse tumors. Methods TTU was observed in RKO model on Cerenkov luminescence (CL) and near-infrared fluorescence (NIRF) imaging of radionuclide 131I or NIRF dyes labeled Atezolizumab and IgG. A mixture of NIRF dyes labeled Atezolizumab and 131I-labeled IgG was injected, and TTU was observed in the RKO and HCT8 model by NIRF/CL dual-modality in-situ imaging. TTU were observed by 131I-labeled Atezolizumab and IgG in-vitro distribution. Results Labeled IgG concentrated more in tumors than Atezolizumab. NIRF/CL imaging in 24 to 168 h showed that TTU gradually decreased over time, which decreased more slowly on CL imaging compared to NIRF imaging. The distribution data in-vitro showed that TTU of 131I-labeled IgG was higher than that of 131I-labeled Atezolizumab at any time point. Conclusion Non-specific IgG may not be suitable as a control for Atezolizumab in comparing tumor PD-L1 expression in nude mice via labeled antibody optical imaging under certain circumstances.
Collapse
Affiliation(s)
- Linhan Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lianmeng Zhao
- Ultrasound Department, Heilongjiang Provincial Hospital, Harbin, China
| | - Xue Lin
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Sheng Zhao
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Pan
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dandan Wang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhongqi Sun
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinping Li
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zonghui Liang
- Department of Radiology, Jing’an District Centre Hospital (Jing’an Branch of Huashan Hospital), Shanghai, China
| | | | - Huijie Jiang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Zhai X, Tang S, Meng F, Ma J, Li A, Zou X, Zhou B, Peng F, Bai J. A dual drug-loaded peptide system with morphological transformation prolongs drug retention and inhibits breast cancer growth. BIOMATERIALS ADVANCES 2023; 154:213650. [PMID: 37857084 DOI: 10.1016/j.bioadv.2023.213650] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
The treatment of breast cancer relies heavily on chemotherapy, but chemotherapy is limited by the disadvantages of poor targeting, susceptibility to extracellular matrix (ECM) interference and a short duration of action in tumor cells. To address these limitations, we developed an amphipathic peptide containing an RGD motif, Pep1, that encapsulated paclitaxel (PTX) and losartan potassium (LP) to form the drug-loaded peptide PL/Pep1. PL/Pep1 self-assembled into spherical nanoparticles (NPs) under normal physiological conditions and transformed into aggregates containing short nanofibers at acidic pH. The RGD peptide facilitated tumor targeting and the aggregates prolonged drug retention in the tumor, which allowed more drug to reach and accumulate in the tumor tissue to promote apoptosis and remodel the tumor microenvironment. The results of in vitro and in vivo experiments confirmed the superiority of PL/Pep1 in terms of targeting, prolonged retention and facilitated penetration for antitumor therapy. In conclusion, amphipathic peptides as coloaded drug carriers are a new platform and strategy for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Xiaoqing Zhai
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang 261053, China
| | - Shusen Tang
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang 261053, China
| | - Fanhu Meng
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Jihong Ma
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang 261053, China
| | - Aimei Li
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Xiangyu Zou
- School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| | - Fujun Peng
- School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China.
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
22
|
Kim HS, Kang JH, Jang J, Lee EJ, Kim JH, Byun J, Shin US. Dual stimuli-responsive mesoporous silica nanoparticles for efficient loading and smart delivery of doxorubicin to cancer with RGD-integrin targeting. Eur J Pharm Sci 2023; 188:106525. [PMID: 37437854 DOI: 10.1016/j.ejps.2023.106525] [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: 03/15/2023] [Revised: 06/15/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023]
Abstract
The recent progress in nanoparticle applications, such as tumor-targeting, has enabled specific delivery of chemotherapeutics to malignant tissues with enhanced local efficacy while limiting side effects. However, existing delivery systems leave much room for improvement in terms of achieving enhanced colloidal stability in fluid medium, efficient targeting of intended sites, and effective release of therapeutic drugs into diseased cells. Here, an efficient stimuli-responsive nanocarrier for mammalian cells, termed RGD-NAMs, was developed, which enabled temperature- and pH-sensitive release of drug loads. The RGD-NAMs comprise two parts: a stimuli-responsive copolymer shell (NIBIm-AA-RGD) and drug-container core (MSNs). The RGD-NAMs have a stable drug-loading capacity with a marked difference in the release rate depending on the temperature and pH conditions. The RGD-NAMs also exhibit high colloidal stability in SBF (Stimulated body fluid) solutions and minimal toxicity in skeletal myoblasts (C2C12) and bovine arterial endothelial cells (BAEC). The doxorubicin-loaded RGD-NAMs induced a cytotoxic effect in a dose-dependent manner, which was furthered by an increase in temperature from 37 to 40 °C. Moreover, significant control of the release rate and the amount were achieved through pH change. This novel, smart drug-delivery system with high responsiveness to temperature and pH changes has wide application prospects in biomedical fields, including the theragnosis of tumors and vascular diseases.
Collapse
Affiliation(s)
- Han-Sem Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, South Korea
| | - Ji-Hye Kang
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, South Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 31116, Republic of Korea
| | - JunHwee Jang
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 31116, Republic of Korea
| | - Eun-Jung Lee
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 31116, Republic of Korea
| | - Jin Hee Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, South Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 31116, Republic of Korea; Department of Molecular Biology, Division of Biological Sciences, Institute of Nanosensor and Biotechnology, Dankook University, Cheonan-si, Chungnam, 31116, Republic of Korea
| | - Jonghoe Byun
- Department of Molecular Biology, Division of Biological Sciences, Institute of Nanosensor and Biotechnology, Dankook University, Cheonan-si, Chungnam, 31116, Republic of Korea.
| | - Ueon Sang Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, South Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 31116, Republic of Korea.
| |
Collapse
|
23
|
Huang YY, Lee ZH, Chang KC, Wu ZY, Lee CC, Tsou MH, Lin HM. Mesoporous silica nanoparticles with dual-targeting agricultural sources for enhanced cancer treatment via tritherapy. RSC Adv 2023; 13:19079-19090. [PMID: 37362343 PMCID: PMC10288219 DOI: 10.1039/d3ra02068a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023] Open
Abstract
In this study, we introduced dual-targeting folic acid (FA) and hyaluronic acid (HA) modified on the surface of rice husk mesoporous silica nanoparticles (rMSNs). The rMSNs were employed as a drug delivery system loaded with camptothecin (CPT) as a model drug, Eu3+ ions as a photosensitizer for photodynamic therapy (PDT), bismuth (Bi) for photothermal therapy (PTT), and Gd3+ ions for magnetic resonance imaging (MRI) to develop novel nanoparticles, rMSN-EuGd-Bi@CPT-HA-FA, with dual-targeted function and triple therapy for cancer treatment. The results of the cell cytotoxicity experiment showed that the A549 cancer cells had a survival rate of approximately 35% when treated with 200 μg mL-1 of rMSN-EuGd-Bi@CPT-HA-FA under 808 nm irradiation for 15 min. The dual-targeted function and synergistic treatment of CPT, PTT, and PDT were also responsible for the 20% survival rate of the A549 cancer cells treated with 200 μg mL-1 of rMSN-EuGd-Bi@CPT-HA-FA under 808 nm irradiation for 30 min. The results showed that rMSN-EuGd-Bi@CPT-HA-FA can effectively combine chemotherapy (through CPT), PDT, and PTT for cancer treatment.
Collapse
Affiliation(s)
- Yu-Ya Huang
- National Taiwan Ocean University, Department of Bioscience and Biotechnology No. 2, Beining Rd., Zhongzheng Dist. Keelung City 202 Taiwan Republic of China +886-2-2462-2192 +886-2-2462-2192
| | - Zui-Harng Lee
- National Taiwan Ocean University, Department of Bioscience and Biotechnology No. 2, Beining Rd., Zhongzheng Dist. Keelung City 202 Taiwan Republic of China +886-2-2462-2192 +886-2-2462-2192
| | - Kai-Chi Chang
- National Taiwan Ocean University, Bachelor Degree Program in Marine Biotechenology Taiwan
| | - Zhi-Yuan Wu
- National Taiwan Ocean University, Department of Bioscience and Biotechnology No. 2, Beining Rd., Zhongzheng Dist. Keelung City 202 Taiwan Republic of China +886-2-2462-2192 +886-2-2462-2192
| | - Cheng-Chang Lee
- National Taiwan Ocean University, Department of Bioscience and Biotechnology No. 2, Beining Rd., Zhongzheng Dist. Keelung City 202 Taiwan Republic of China +886-2-2462-2192 +886-2-2462-2192
| | - Min-Hsuan Tsou
- National Taiwan Ocean University, Department of Bioscience and Biotechnology No. 2, Beining Rd., Zhongzheng Dist. Keelung City 202 Taiwan Republic of China +886-2-2462-2192 +886-2-2462-2192
| | - Hsiu-Mei Lin
- National Taiwan Ocean University, Department of Bioscience and Biotechnology No. 2, Beining Rd., Zhongzheng Dist. Keelung City 202 Taiwan Republic of China +886-2-2462-2192 +886-2-2462-2192
- National Taiwan Ocean University, Center of Excellence for the Oceans Taiwan
- National Taiwan Ocean University, Center of Excellence for Ocean Engineering Taiwan
| |
Collapse
|
24
|
A multi-bioresponsive self-assembled nano drug delivery system based on hyaluronic acid and geraniol against liver cancer. Carbohydr Polym 2023; 310:120695. [PMID: 36925236 DOI: 10.1016/j.carbpol.2023.120695] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023]
Abstract
Herein, a multi-bioresponsive self-assembled nano-drug delivery system (HSSG) was constructed by conjugating the anticancer drug Geraniol (GER) to hyaluronic acid (HA) via a disulfide bond. The HSSG NPs displayed a uniform spherical shape with an average diameter of ∼110 nm, maintained high stability, and realized controlled drug release in the tumor microenvironment (pH/glutathione/hyaluronidase). Results of fluorescence microscopy and flow cytometry verified that HSSG NPs were selectively uptaken by human hepatocellular carcinoma cell lines HepG2 and Huh7 via CD44 receptor-mediated internalization. Studies on H22 tumor-bearing mice demonstrate that HSSG NPs could effectively accumulate at the tumor site for a long period. In vitro and in vivo studies show that HSSG NPs significantly promoted the death of cancer cells while reducing the toxicity as compared to GER. Therefore, the HSSG NPs have great potential in the treatment of tumors.
Collapse
|
25
|
Li X, Huang Z, Liao Z, Liu A, Huo S. Transformable nanodrugs for overcoming the biological barriers in the tumor environment during drug delivery. NANOSCALE 2023; 15:8532-8547. [PMID: 37114478 DOI: 10.1039/d2nr06621a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Drug delivery systems have been studied massively with explosive growth in the last few decades. However, challenges such as biological barriers are still obstructing the delivery efficiency of nanomedicines. Reports have shown that the physicochemical properties, such as the morphologies of nanodrugs, could highly affect their biodistribution and bioavailability. Therefore, transformable nanodrugs that take advantage of different sizes and shapes allow for overcoming multiple biological barriers, providing promising prospects for drug delivery. This review aims to present an overview of the most recent developments of transformable nanodrugs in this emerging field. First, the design principles and transformation mechanisms which serve as guidelines for smart nanodrugs are summarized. Afterward, their applications in overcoming biological barriers, including the bloodstream, intratumoral pressure, cellular membrane, endosomal wrapping, and nuclear membrane, are highlighted. Finally, discussions on the current developments and future perspectives of transformable nanodrugs are given.
Collapse
Affiliation(s)
- Xuejian Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhenkun Huang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhihuan Liao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Aijie Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
26
|
Xiao L, Sun Y, Liao L, Su X. Response of mesenchymal stem cells to surface topography of scaffolds and the underlying mechanisms. J Mater Chem B 2023; 11:2550-2567. [PMID: 36852826 DOI: 10.1039/d2tb01875f] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) serve as essential components of regenerative medicine. Their destiny is influenced by the interaction of the cells with the external environment. In addition to the biochemical cues in a microenvironment, physical cues of the topography of the surrounding materials such as the extracellular matrix emerge as a crucial regulator of stem cell destiny and function. With recent advances in technologies of materials production and surface modification, surfaces with micro/nanotopographical characteristics can be fabricated to mimic the micro/nanoscale mechanical stimuli of the extracellular matrix environment and regulate the biological behavior of cells. Understanding the interaction of cells with the topography of a surface is conducive to the control of stem cell fate for application in regenerative medicine. However, the mechanisms by which topography affects the biological behavior of stem cells have not been fully elucidated. This review will present the effects of surface topography at the nano/micrometer scale on stem cell adhesion, morphology, proliferation, migration, and differentiation. It also focuses on discussing current theories about the sensing and recognition of surface topology cues, the transduction of the extracellular cues into plasma, and the final activation of related signaling pathways and downstream gene expression in MSCs. These insights will provide a theoretical basis for the future design of biomaterial scaffolds for application in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Li Xiao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatrics & Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| | - Yanping Sun
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatrics & Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| | - Li Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatrics & Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| | - Xiaoxia Su
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatrics & Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
27
|
Recent progress in nanocarrier-based drug delivery systems for antitumour metastasis. Eur J Med Chem 2023; 252:115259. [PMID: 36934485 DOI: 10.1016/j.ejmech.2023.115259] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Tumour metastasis is one of the major factors leading to poor prognosis as well as lower survival among cancer patients. A number of studies investigating the inhibition of tumour metastasis have been conducted. It is difficult to achieve satisfactory results with surgery alone for distant metastatic tumours, and chemotherapy can boost the healing rate and prognosis of patients. However, the poor therapeutic efficacy of chemotherapy drugs due to their low solubility, lack of tumour targeting, instability in vivo, high toxicity and multidrug resistance hinder their application. Immunotherapy is beneficial to the treatment of metastatic cancers, but it also has disadvantages such as adverse reactions and acquired resistance. Fortunately, delivery of chemotherapeutic drugs with nanocarriers can reduce systemic reactions caused by chemotherapeutic agents and inhibit metastasis. This review discusses the underlying mechanisms of metastasis, therapeutic approaches for antitumour metastasis, the advantages of nanodrug delivery systems and their application in reducing metastasis.
Collapse
|
28
|
Ha W, Yang JL, Shi YP. Antibacterial metal-phenolic nanosheets as smart carriers for the controlled release of epirubicin hydrochloride. NANOSCALE 2022; 14:9806-9817. [PMID: 35770915 DOI: 10.1039/d2nr02066a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bacterial infections can cause serious complications in cancer treatment and have been proven to weaken therapeutic benefits. Recently, antibacterial nanomaterials that serve as carriers for anticancer drug delivery have been attracting extensive interest due to their combined antimicrobial and anticancer activities. In this study, antibacterial metal-phenolic nanosheets (Cu-TA) were successfully prepared via the self-assembly of the metal-phenolic coordination complexes formed between copper ions and tannic acid, and the structure, morphology, and formation mechanism of Cu-TA nanosheets were explored. The antibacterial activity of Cu-TA nanosheets against both Gram-positive and Gram-negative bacteria was detected using the minimum inhibitory concentration (MIC), zone of inhibition and plate counting methods. The MIC values of both bacterial strains were about 0.4 mg mL-1, and the killing rates of Cu-TA samples were close to 100% at the concentration of 2 and 0.2 mg mL-1 after 12-hour incubation. Epirubicin hydrochloride (EPI) molecules were successfully loaded on the porous Cu-TA nanosheets mainly through the formation of the Cu-EPI chelate complex and strong electrostatic interactions. The Cu-EPI complex and Cu-TA nanosheets could be disassembled under acidic conditions or in the presence of high levels of glutathione (GSH) after uptake by cancer cells, which triggered the unique pH and GSH-responsive controlled release behaviors of EPI and copper ions. The MTT assay results revealed that the presence of bacteria in Hep G2 cells can greatly impair the cell death rate induced by free EPI, but the resultant EPI-loaded Cu-TA nanosheets can significantly enhance cell death both in the presence and absence of bacteria.
Collapse
Affiliation(s)
- Wei Ha
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, People's Republic of China.
| | - Jun-Li Yang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, People's Republic of China.
| | - Yan-Ping Shi
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, People's Republic of China.
| |
Collapse
|
29
|
Wang Z, Wang Y, Sun X, Zhou J, Chen X, Xi J, Fan L, Han J, Guo R. Supramolecular Core-Shell Nanoassemblies with Tumor Microenvironment-Triggered Size and Structure Switch for Improved Photothermal Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200588. [PMID: 35277929 DOI: 10.1002/smll.202200588] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/26/2022] [Indexed: 06/14/2023]
Abstract
Photothermal therapy (PTT) is demonstrated to be an effective methodology for cancer treatment. However, the relatively low photothermal conversion efficiency, limited tumor accumulation, and penetration still remain to be challenging issues that hinder the clinical application of PTT. Herein, the core-shell hierarchical nanostructures induced by host-guest interaction between water-soluble pillar[5]arene (WP5) and polyethylene glycol-modified aniline tetramer (TAPEG) are constructed. The pH-responsive performance endows the core-shell nanostructures with size switchable property, with an average diameter of 200 nm in the neutral pH and 60 nm in the acidic microenvironment, which facilitates not only tumor accumulation but also tumor penetration. Moreover, the structure switch of WP5⊃TAPEG under acidic microenvironment and the dual mechanism regulated extending of п conjugate, inclusion in the hydrophobic cavity of WP5 and the dense distribution in the core-shell structured assemblies, dramatically enhance the absorption in the near-infrared-II region and, further, the photothermal conversion efficiency (60.2%). The as-designed intelligent nanoplatform is demonstrated for improved antitumor efficacy via PTT.
Collapse
Affiliation(s)
- Ziyao Wang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Yanqiu Wang
- School of Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, P. R. China
| | - Xiaohuan Sun
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Jinfeng Zhou
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Xiaolin Chen
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Juqun Xi
- School of Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, P. R. China
| | - Lei Fan
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Jie Han
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Rong Guo
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| |
Collapse
|