1
|
Wang T, Sun Y, Zeng D, Wang M, Zhang Y, Liu G, Chen X, Liu L. Integrated cell membrane encapsulated PQDs-TK quantum dot nanoclusters with ROS-responsive triggering for efficient and visualized DNA delivery. J Colloid Interface Sci 2025; 683:393-410. [PMID: 39740557 DOI: 10.1016/j.jcis.2024.12.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
With the unique photo-physical properties and strong bio-compatibility. Quantum dots (QDs) have sparked interest in biomedical fields such as imaging, biosensing and therapeutics. However, the low stability and insufficient tumor specificity have largely constrained their potential biomedical applications. Here, we reported a cell membrane encapsulated PQDs-TK quantum dots nanoclusters (CM-PQDs-TK) gene delivery system with ROS-responsive triggering for efficient and visualized DNA delivery. CM-PQDs-TK possessed excellent DNA loading capacity and protective ability. The particle size and morphology change suggested that CM-PQDs-TK displayed superior ROS responsiveness. As expected, CM-PQDs-TK had an obvious nanocluster structure, and the particle size was stabilized in the range of 200-300 nm. Compared with PQDs and PQDs-TK, HlM-PQDs-TK/DNA encapsulated by Hela cell membrane has higher uptake efficiency and transfection ability, reaching 67.14 % and 63.41 % in 293 T cells, and the DNA transfection efficiency could still reach 43.98 % even in cancer cells (Hela cells). Moreover, flow cytometry and fluorescence microscopy showedthat nanoclusters could effectively enter tumor cells, and the internal DNA could be effectively released. In this process, PQDs-TK responded to high ROS in tumor cells and greatly improved DNA delivery efficiency. After loading DNA, cell membrane encapsulation technology was used to enhance its biocompatibility and targeting further. This work was anticipated to provide an in-depth understanding of the important role of PEI quantum dots in the field of ROS-responsive intelligent gene carriers and laid a foundation for the design and preparation of novel quantum dots gene carriers.
Collapse
Affiliation(s)
- Tiange Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yanlin Sun
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Dong Zeng
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Mengying Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yajing Zhang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Gang Liu
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xin Chen
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Liang Liu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
2
|
Medeiros TS, Bezerra de Lima LE, Alves-Pereira EL, Alves-Silva MF, Dourado D, Fernandes-Pedrosa MDF, Figueiredo RCBQD, da Silva-Junior AA. Cationic and anionic PLGA-cholesterol hybrid nanoparticles as promising platforms to enhance the trypanocidal efficacy of benznidazole and drug delivery in Trypanosoma cruzi-infected cells. Biomed Pharmacother 2025; 183:117782. [PMID: 39755025 DOI: 10.1016/j.biopha.2024.117782] [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: 10/25/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/06/2025] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan Trypanosoma cruzi, remains a significant global health challenge. Currently, benznidazole (BNZ) is the primary treatment in many countries. However, this drug is limited by low bioavailability, significant host toxicity, and reduced efficacy in chronic disease phase. Additionally, cases of parasite resistance to treatment and low efficacy in in chronic disease phase have been reported. In this context, nanotechnology formulations for intracellular drug delivery have emerged as a promising alternative to improve the pharmacological properties of BNZ. In this study, we developed and evaluated cationic and anionic PLGA-cholesterol hybrid nanoparticles (HNPs) as innovative drug delivery systems for BNZ. These HNPs, functionalized with polyethyleneimine, were synthesized using a composition-dependent self-assembly method, yielding stable nanosystems with tuneable physicochemical properties. Furthermore, four release kinetic models were applied and Peppas-Sahlin demonstrated the best fit. In vitro assays confirmed the biocompatibility of HNPs with cardiomyoblasts at tested concentrations and revealed significantly enhanced trypanocidal activity against intracellular amastigotes compared to free BNZ. Transmission electron microscopy and fluorescence microscopy analyses highlighted effective nanoparticle internalization, with superior performance trypanocidal observed in anionic HNPs, which can be attributed to the residence of cationic in endo/lysosomal vesicles. Taken together, our results demonstrate the successful development of HNPs, underscoring their potential as a promising platform for the intracellular delivery of therapeutic agents.
Collapse
Affiliation(s)
- Thayse Silva Medeiros
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil; Laboratory of Cellular Biology of Pathogens, Department of Microbiology, Aggeu Magalhães Institute/FIOCRUZ-PE, Recife, PE, Brazil
| | - Lucas Eduardo Bezerra de Lima
- Laboratory of Cellular Biology of Pathogens, Department of Microbiology, Aggeu Magalhães Institute/FIOCRUZ-PE, Recife, PE, Brazil
| | - Eron Lincoln Alves-Pereira
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil
| | - Mariana Farias Alves-Silva
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil
| | - Douglas Dourado
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute/FIOCRUZ-PE, Recife, PE, Brazil
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil
| | | | - Arnóbio Antônio da Silva-Junior
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil.
| |
Collapse
|
3
|
Yang M, Zhou J, Lu L, Deng D, Huang J, Tang Z, Shi X, Lo P, Lovell JF, Zheng Y, Jin H. Tumor cell membrane-based vaccines: A potential boost for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230171. [PMID: 39713208 PMCID: PMC11655317 DOI: 10.1002/exp.20230171] [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: 12/15/2023] [Accepted: 03/08/2024] [Indexed: 12/24/2024]
Abstract
Because therapeutic cancer vaccines can, in theory, eliminate tumor cells specifically with relatively low toxicity, they have long been considered for application in repressing cancer progression. Traditional cancer vaccines containing a single or a few discrete tumor epitopes have failed in the clinic, possibly due to challenges in epitope selection, target downregulation, cancer cell heterogeneity, tumor microenvironment immunosuppression, or a lack of vaccine immunogenicity. Whole cancer cell or cancer membrane vaccines, which provide a rich source of antigens, are emerging as viable alternatives. Autologous and allogenic cellular cancer vaccines have been evaluated as clinical treatments. Tumor cell membranes (TCMs) are an intriguing antigen source, as they provide membrane-accessible targets and, at the same time, serve as integrated carriers of vaccine adjuvants and other therapeutic agents. This review provides a summary of the properties and technologies for TCM cancer vaccines. Characteristics, categories, mechanisms, and preparation methods are discussed, as are the demonstrable additional benefits derived from combining TCM vaccines with chemotherapy, sonodynamic therapy, phototherapy, and oncolytic viruses. Further research in chemistry, biomedicine, cancer immunology, and bioinformatics to address current drawbacks could facilitate the clinical adoption of TCM vaccines.
Collapse
Affiliation(s)
- Muyang Yang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jie Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Liseng Lu
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Deqiang Deng
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Zijian Tang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Xiujuan Shi
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Pui‐Chi Lo
- Department of Biomedical SciencesCity University of Hong KongKowloonHong KongChina
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNew YorkUSA
| | - Yongfa Zheng
- Department of OncologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
4
|
Peng Y, Bai J, Li W, Su Z, Cheng X. Advancements in p53-Based Anti-Tumor Gene Therapy Research. Molecules 2024; 29:5315. [PMID: 39598704 PMCID: PMC11596491 DOI: 10.3390/molecules29225315] [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: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
The p53 gene is one of the genes most closely associated with human tumors and has become a popular target for tumor drug design. Currently, p53-based gene therapy techniques have been developed, but these therapies face challenges such as immaturity, high safety hazards, limited efficacy, and low patient acceptance. However, researchers are no less enthusiastic about the treatment because of its theoretical potential to treat cancer. In this paper, the advances in p53-based gene therapy and related nucleic acid delivery technologies were reviewed and prospected in order to support further development in this field.
Collapse
Affiliation(s)
- Yuanwan Peng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Jinping Bai
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Wang Li
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Zhengding Su
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Xiyao Cheng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| |
Collapse
|
5
|
Afonso I, Neves AR, Eusébio D, Albuquerque T, Vivès E, Boisguérin P, Santos AO, Sousa Â, Costa D. Design of Experiments to Tailor the Potential of BSA-Coated Peptide Nanocomplexes for Temozolomide/p53 Gene Co-Delivery. Pharmaceutics 2024; 16:1389. [PMID: 39598513 PMCID: PMC11597296 DOI: 10.3390/pharmaceutics16111389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Gene therapy can be viewed as a promising/valuable therapeutic approach directed to cancer treatment, including glioblastoma. Concretely, the combination of gene therapy with chemotherapy could increase its therapeutic index due to a synergistic effect. In this context, bovine serum albumin (BSA)-coated temozolomide (TMZ)-peptide (WRAP5)/p53 gene-based plasmid DNA complexes were developed to promote payload co-delivery. Methods: Design of experiments (DoE) was employed to unravel the BSA-coated TMZ-WRAP5/p53 nanocomplexes with the highest potential by considering the nitrogen to phosphate groups ratio (N/P), and the BSA concentration as inputs and the size, polydispersity index, surface charge and p53-based plasmid complexation capacity (CC) as DoE outputs. Results: The obtained quadratic models were statistically significant (p-value < 0.05) with an adequate coefficient of determination, and the correspondent optimal points were successfully validated. The optimal complex formulation had N/P of 1.03, a BSA concentration of 0.08%, a size of approximately 182 nm, a zeta potential of +9.8 mV, and a pDNA CC of 96.5%. The optimal nanocomplexes are approximately spherical. A cytotoxicity assay showed that these BSA-coated TMZ-WRAP5/p53 complexes did not elicit toxicity in normal brain cells, and a hemolysis study demonstrated the hemocompatibility of the complexes. The complexes were stable in cell culture medium and fetal bovine serum and assured pDNA protection and release. Moreover, the optimal BSA-coated complexes were able of gene transcription and promoted a significant inhibition of glioblastoma cell viability. Conclusions: The reported findings instigate the development of future research to evaluate their potential utility to TMZ/p53 co-delivery. The DoE tool proved to be a powerful approach to explore and tailor the composition of BSA-coated TMZ-WRAP5/p53 complexes, which are expected to contribute to the progress toward a more efficient therapy against cancer and, more specifically, against glioblastoma.
Collapse
Affiliation(s)
- Inês Afonso
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; (I.A.); (A.R.N.); (D.E.); (T.A.); (A.O.S.); (Â.S.)
| | - Ana R. Neves
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; (I.A.); (A.R.N.); (D.E.); (T.A.); (A.O.S.); (Â.S.)
| | - Dalinda Eusébio
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; (I.A.); (A.R.N.); (D.E.); (T.A.); (A.O.S.); (Â.S.)
| | - Tânia Albuquerque
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; (I.A.); (A.R.N.); (D.E.); (T.A.); (A.O.S.); (Â.S.)
| | - Eric Vivès
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Prisca Boisguérin
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Adriana O. Santos
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; (I.A.); (A.R.N.); (D.E.); (T.A.); (A.O.S.); (Â.S.)
| | - Ângela Sousa
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; (I.A.); (A.R.N.); (D.E.); (T.A.); (A.O.S.); (Â.S.)
| | - Diana Costa
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; (I.A.); (A.R.N.); (D.E.); (T.A.); (A.O.S.); (Â.S.)
| |
Collapse
|
6
|
Kachanov A, Kostyusheva A, Brezgin S, Karandashov I, Ponomareva N, Tikhonov A, Lukashev A, Pokrovsky V, Zamyatnin AA, Parodi A, Chulanov V, Kostyushev D. The menace of severe adverse events and deaths associated with viral gene therapy and its potential solution. Med Res Rev 2024; 44:2112-2193. [PMID: 38549260 DOI: 10.1002/med.22036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, in vivo gene replacement therapy has significantly advanced, resulting in market approval of numerous therapeutics predominantly relying on adeno-associated viral vectors (AAV). While viral vectors have undeniably addressed several critical healthcare challenges, their clinical application has unveiled a range of limitations and safety concerns. This review highlights the emerging challenges in the field of gene therapy. At first, we discuss both the role of biological barriers in viral gene therapy with a focus on AAVs, and review current landscape of in vivo human gene therapy. We delineate advantages and disadvantages of AAVs as gene delivery vehicles, mostly from the safety perspective (hepatotoxicity, cardiotoxicity, neurotoxicity, inflammatory responses etc.), and outline the mechanisms of adverse events in response to AAV. Contribution of every aspect of AAV vectors (genomic structure, capsid proteins) and host responses to injected AAV is considered and substantiated by basic, translational and clinical studies. The updated evaluation of recent AAV clinical trials and current medical experience clearly shows the risks of AAVs that sometimes overshadow the hopes for curing a hereditary disease. At last, a set of established and new molecular and nanotechnology tools and approaches are provided as potential solutions for mitigating or eliminating side effects. The increasing number of severe adverse reactions and, sadly deaths, demands decisive actions to resolve the issue of immune responses and extremely high doses of viral vectors used for gene therapy. In response to these challenges, various strategies are under development, including approaches aimed at augmenting characteristics of viral vectors and others focused on creating secure and efficacious non-viral vectors. This comprehensive review offers an overarching perspective on the present state of gene therapy utilizing both viral and non-viral vectors.
Collapse
Affiliation(s)
- Artyom Kachanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Ivan Karandashov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Andrey Tikhonov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Vadim Pokrovsky
- Laboratory of Biochemical Fundamentals of Pharmacology and Cancer Models, Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People's Friendship University, Russia (RUDN University), Moscow, Russia
| | - Andrey A Zamyatnin
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Research, Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Vladimir Chulanov
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Infectious Diseases, Sechenov University, Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
7
|
Liang L, Jia M, Zhao M, Deng Y, Tang J, He X, Liu Y, Yan K, Yu X, Yang H, Li C, Li Y, Li T. Progress of Nanomaterials Based on Manganese Dioxide in the Field of Tumor Diagnosis and Therapy. Int J Nanomedicine 2024; 19:8883-8900. [PMID: 39224196 PMCID: PMC11368147 DOI: 10.2147/ijn.s477026] [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: 05/07/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
As a pivotal transition metal oxide, manganese dioxide (MnO2) has garnered significant attention owing to its abundant reserves, diverse crystal structures and exceptional performance. Nanosizing MnO2 results in smaller particle sizes, larger specific surface areas, optimized material characteristics, and expanded application possibilities. With the burgeoning research efforts in this field, MnO2 has emerged as a promising nanomaterial for tumor diagnosis and therapy. The distinctive properties of MnO2 in regulating the tumor microenvironment (TME) have attracted considerable interest, leading to a rapid growth in research on MnO2-based nanomaterials for tumor diagnosis and treatment. Additionally, MnO2 nanomaterials are also gradually showing up in the regulation of chronic inflammatory diseases. In this review, we mainly summarized the recent advancements in various MnO2 nanomaterials for tumor diagnosis and therapy. Furthermore, we discuss the current challenges and future directions in the development of MnO2 nanomaterials, while also envisaging their potential for clinical translation.
Collapse
Grants
- This work was supported by the Sichuan Science and Technology Program (grant numbers 2023NSFSC0620, 2022YFS0614, 2022YFS0622, 2022YFS0627), the Luzhou Municipal People’s Government-Southwest Medical University Joint Scientific Research Project (grant number 2023LZXNYDHZ003), the Open fund for Key Laboratory of Medical Electrophysiology of Ministry of Education (grant numbers KeyME-2023-07), the Youth Science Foundation Project of Southwest Medical University (grant numbers 2023QN075, 2022QN025), the Southwest Medical University Science and Technology Project (No.2021ZKMS034), the Hejiang County People’s Hospital-Southwest Medical University Joint Scientific Research Project (grant numbers 2023HJXNYD03, 2022HJXNYD03, 2022HJXNYD14), Chinese student innovation and entrepreneurship project (202310632027)
Collapse
Affiliation(s)
- Lijuan Liang
- Department of Pharmacy, Hejiang County People’s Hospital, Luzhou, Sichuan, People’s Republic of China
| | - Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Nanchong Institute for Food and Drug Control, Nanchong, Sichuan, People’s Republic of China
| | - Min Zhao
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Kexin Yan
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Hong Yang
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yao Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Science and Technology department, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
8
|
Huang Z, Wang Y, Su C, Li W, Wu M, Li W, Wu J, Xia Q, He H. Mn-Anti-CTLA4-CREKA-Sericin Nanotheragnostics for Enhanced Magnetic Resonance Imaging and Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306912. [PMID: 38009480 DOI: 10.1002/smll.202306912] [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: 08/12/2023] [Revised: 10/16/2023] [Indexed: 11/29/2023]
Abstract
The integration of magnetic resonance imaging (MRI), cGAS-STING, and anti-CTLA-4 (aCTLA-4) based immunotherapy offers new opportunities for tumor precision therapy. However, the precise delivery of aCTLA-4 and manganese (Mn), an activator of cGAS, to tumors remains a major challenge for enhanced MRI and active immunotherapy. Herein, a theragnostic nanosphere Mn-CREKA-aCTLA-4-SS (MCCS) is prepared by covalently assembling Mn2+, silk sericin (SS), pentapeptide CREKA, and aCTLA-4. MCCS are stable with an average size of 160 nm and is almost negatively charged or neutral at pH 5.5/7.4. T1-weighted images showed MCCS actively targeted tumors to improve the relaxation rate r1 and contrast time of MRI. This studies demonstrated MCCS raises reactive oxygen species levels, activates the cGAS-STING pathway, stimulates effectors CD8+ and CD80+ T cells, reduces regulatory T cell numbers, and increases IFN-γ and granzyme secretion, thereby inducing tumor cells autophagy and apoptosis in vitro and in vivo. Also, MCCS are biocompatible and biosafe. These studies show the great potential of Mn-/SS-based integrative material MCCS for precision and personalized tumor nanotheragnostics.
Collapse
Affiliation(s)
- Zixuan Huang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing, 400715, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yejing Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing, 400715, China
| | - Can Su
- School of medical imaging, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Wanting Li
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Min Wu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Wuling Li
- College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Jun Wu
- School of medical imaging, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing, 400715, China
| | - Huawei He
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing, 400715, China
| |
Collapse
|
9
|
Mohammadi N, Fayazi Hosseini N, Nemati H, Moradi-Sardareh H, Nabi-Afjadi M, Kardar GA. Revisiting of Properties and Modified Polyethylenimine-Based Cancer Gene Delivery Systems. Biochem Genet 2024; 62:18-39. [PMID: 37394575 DOI: 10.1007/s10528-023-10416-7] [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: 04/23/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023]
Abstract
A new era of medical technology in cancer treatment is a directly specific modification of gene expression in tumor cells by nucleic acid delivery. Currently, the main challenge to achieving this goal is to find a non-toxic, safe, and effective strategy for gene transfer to cancer cells. Synthetic composites based on cationic polymers have historically been favored in bioengineering due to their ability to mimic bimolecular structures. Among them, polyethylenimines (PEIs) with superior properties such as a wide range of molecular weight and a flexible structure may propel the development of functional combinations in the biomedical and biomaterial fields. Here, in this review, we will focus on the recent progressions in the formulation optimization of PEI-based polyplex in gene delivery to treat cancer. Also, the effect of PEI's intrinsic characteristics such as structure, molecular weight, and positive charges which influence the gene delivery efficiency will be discussed.
Collapse
Affiliation(s)
- Nejad Mohammadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunology Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nashmin Fayazi Hosseini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Nemati
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Gholam Ali Kardar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Immunology Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Wang M, Huang H, Sun Y, Wang M, Yang Z, Shi Y, Liu L. PEI functionalized cell membrane for tumor targeted and glutathione responsive gene delivery. Int J Biol Macromol 2024; 255:128354. [PMID: 37995795 DOI: 10.1016/j.ijbiomac.2023.128354] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023]
Abstract
Polyethylenimine (PEI) is a broadly exploited cationic polymer due to its remarkable gene-loading capacity. However, the high cytotoxicity caused by its high surface charge density has been reported in many cell lines, limiting its application significantly. In this study, two different molecular weights of PEI (PEI10k and PEI25k) were crosslinked with red blood cell membranes (RBCm) via disulfide bonds to form PEI derivatives (RMPs) with lower charge density. Furthermore, the targeting molecule folic acid (FA) molecules were further grafted onto the polymers to obtain FA-modified PEI-RBCm copolymers (FA-RMP25k) with tumor cell targeting and glutathione response. In vitro experiments showed that the FA-RMP25k/DNA complex had satisfactory uptake efficiency in both HeLa and 293T cells, and did not cause significant cytotoxicity. Furthermore, the uptake and transfection efficiency of the FA-RMP25k/DNA complex was significantly higher than that of the PEI25k/DNA complex, indicating that FA grafting can increase transfection efficiency by 15 %. These results suggest that FA-RMP25k may be a promising non-viral gene vector with potential applications in gene therapy.
Collapse
Affiliation(s)
- Mengying Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Haoxiang Huang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yanlin Sun
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mingjie Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhaojun Yang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yong Shi
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Liang Liu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
11
|
Lin Y, Guan X, Su J, Chen S, Fu X, Xu X, Deng X, Chang J, Qin A, Shen A, Zhang L. Cell Membrane-Camouflaged Nanoparticles Mediated Nucleic Acids Delivery. Int J Nanomedicine 2023; 18:8001-8021. [PMID: 38164266 PMCID: PMC10758188 DOI: 10.2147/ijn.s433737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024] Open
Abstract
Nucleic acids have emerged as promising therapeutic agents for many diseases because of their potential in modulating gene expression. However, the delivery of nucleic acids remains a significant challenge in gene therapy. Although viral vectors have shown high transfection efficiency, concerns regarding teratogenicity or carcinogenicity have been raised. Non-viral vehicles, including cationic polymers, liposomes, and inorganic materials possess advantages in terms of safety, ease of preparation, and low cost. Nevertheless, they also face limitations related to immunogenicity, quick clearance in vivo, and lack of targeting specificity. On the other hand, bioinspired strategies have shown increasing potential in the field of drug delivery, yet there is a lack of comprehensive reviews summarizing the rapid development of bioinspired nanoparticles based on the cell membrane camouflage to construct the nucleic acids vehicles. Herein, we enumerated the current difficulties in nucleic acid delivery with various non-viral vehicles and provided an overview of bioinspired strategies for nucleic acid delivery.
Collapse
Affiliation(s)
- Yinshan Lin
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xiaoling Guan
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Jianfen Su
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Sheng Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xihua Fu
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Xiaowei Xu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xiaohua Deng
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Jishuo Chang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Aiping Qin
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Ao Shen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Lingmin Zhang
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| |
Collapse
|
12
|
Zhang Y, Luo J, Gui X, Zheng Y, Schaar E, Liu G, Shi J. Bioengineered nanotechnology for nucleic acid delivery. J Control Release 2023; 364:124-141. [PMID: 37879440 PMCID: PMC10838211 DOI: 10.1016/j.jconrel.2023.10.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/15/2023] [Accepted: 10/21/2023] [Indexed: 10/27/2023]
Abstract
Nucleic acid-based therapy has emerged as a promising therapeutic approach for treating various diseases, such as genetic disorders, cancers, and viral infections. Diverse nucleic acid delivery systems have been reported, and some, including lipid nanoparticles, have exhibited clinical success. In parallel, bioengineered nucleic acid delivery nanocarriers have also gained significant attention due to their flexible functional design and excellent biocompatibility. In this review, we summarize recent advances in bioengineered nucleic acid delivery nanocarriers, focusing on exosomes, cell membrane-derived nanovesicles, protein nanocages, and virus-like particles. We highlight their unique features, advantages for nucleic acid delivery, and biomedical applications. Furthermore, we discuss the challenges that bioengineered nanocarriers face towards clinical translation and the possible avenues for their further development. This review ultimately underlines the potential of bioengineered nanotechnology for the advancement of nucleic acid therapy.
Collapse
Affiliation(s)
- Yang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China; Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Luo
- Department of Urology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiran Gui
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yating Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Eric Schaar
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|