1
|
Wang JL, Ji WW, Huang AL, Liu Z, Chen DF. CEBPA Restrains the Malignant Progression of Breast Cancer by Prompting the Transcription of SOCS2. Mol Biotechnol 2025; 67:2127-2137. [PMID: 38775935 DOI: 10.1007/s12033-024-01189-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/23/2024] [Indexed: 04/10/2025]
Abstract
The suppressor of cytokine signaling 2 (SOCS2) has been identified to act as a tumor suppressor in breast cancer (BC) progression. However, the action of SOCS2 in macrophage polarization in BC cells has not been reported yet. The qRT-PCR and western blotting were adopted for detecting the levels of mRNAs and proteins. The macrophage M2 polarization was analyzed by flow cytometry. Analyses of cell oncogenic phenotypes and tumor growth were conducted using 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, scratch, Transwell, tube formation assays in vitro, and tumor xenograft assay in vivo, respectively. The interaction between CEBPA (CCAAT Enhancer Binding Protein Alpha) and SOCS2 was confirmed using bioinformatics analysis and dual-luciferase reporter assay. SOCS2 was lowly expressed in BC tissues and cells. Functionally, overexpression of SOCS2 inhibited macrophage M2 polarization, and impaired BC cell proliferation, angiogenesis, and metastasis. Mechanistically, CEBPA bound to the promoter region of SOCS2, and promoted its transcription. A low CEBPA expression was observed in BC tissues and cells. Forced expression of CEBPA also suppressed macrophage M2 polarization, BC cell proliferation, angiogenesis, and metastasis. Moreover, the anticancer effects mediated by CEBPA were abolished by SOCS2 knockdown. In addition, CEBPA overexpression impeded BC growth in nude mice by regulating SOCS2. CEBPA suppressed macrophage M2 polarization, BC cell proliferation, angiogenesis, and metastasis by promoting SOCS2 transcription in a targeted manner.
Collapse
Affiliation(s)
- Jin-Li Wang
- Department of Galactophore, Jingzhou Central Hospital, The Second Clinical Medical College, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, Hubei, China
| | - Wei-Wei Ji
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Ao-Li Huang
- Department of Galactophore, Jingzhou Central Hospital, The Second Clinical Medical College, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, Hubei, China
| | - Zhen Liu
- Department of Galactophore, Jingzhou Central Hospital, The Second Clinical Medical College, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, Hubei, China
| | - Deng-Feng Chen
- Department of Galactophore, Jingzhou Central Hospital, The Second Clinical Medical College, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, Hubei, China.
| |
Collapse
|
2
|
Esmaeilzadeh A, Hadiloo K, Yaghoubi S, Makoui MH, Mostanadi P. State of the art in CAR-based therapy: In vivo CAR production as a revolution in cell-based cancer treatment. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01056-7. [PMID: 40261561 DOI: 10.1007/s13402-025-01056-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Chimeric antigen receptor (CAR) therapy has successfully treated relapsed/refractory hematological cancers. This strategy can effectively target tumor cells. However, despite positive outcomes in clinical applications, challenges remain to overcome. These hurdles pertain to the production of the drugs, solid tumor resistance, and side effects related to the treatment. Some cases have been missed during the drug preparation due to manufacturing issues, prolonged production times, and high costs. These challenges mainly arise from the in vitro manufacturing process, so reevaluating this process could minimize the number of missed patients. The immune cells are traditionally collected and sent to the laboratory; after several steps, the cells are modified to express the CAR gene before being injected back into the patient's body. During the in vivo method, the CAR gene is introduced to the immune cells inside the body. This allows for treatment to begin sooner, avoiding potential failures in drug preparation and the associated high costs. In this review, we will elaborate on the production and treatment process using in vivo CAR, examine the benefits and challenges of this approach, and ultimately present the available solutions for incorporating this treatment into clinical practice.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Pficell R&D Canadian Institution & Corporation, Profound Future Focused Innovative Cell and Gene Therapy, Pficell Canadian Institution and Corporation, Ontario, Canada.
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Kaveh Hadiloo
- Pficell R&D Canadian Institution & Corporation, Profound Future Focused Innovative Cell and Gene Therapy, Pficell Canadian Institution and Corporation, Ontario, Canada
- Department of Surgery, Velayat Clinical Research Development Unit, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
| | - Sara Yaghoubi
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Parsa Mostanadi
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
3
|
Ma R, Yang Z, Miao X, Hu J, Zhang T, Ma LT, Lin JY, Zhao LN. Dual-Mode Radiosensitization of Esophageal Squamous Cell Carcinoma via SOCS6-Loaded Virus-Inspired Manganese-Bismuth Bimetallic Oxide Nanoparticles. Adv Healthc Mater 2025:e2404737. [PMID: 40159874 DOI: 10.1002/adhm.202404737] [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: 11/26/2024] [Revised: 03/21/2025] [Indexed: 04/02/2025]
Abstract
Radioresistance poses a significant obstacle to controlling the recurrence of esophageal squamous cell carcinoma (ESCC) during radiotherapy. It is urgent to develop innovative radiosensitization strategies to improve the prognosis of patients with ESCC. Here, a novel dual-mode radiosensitizer: a virus-inspired hollow mesoporous manganese-bismuth bimetallic oxide nanoparticles (vHMMn-Bi) encapsulating the radiosensitizing plasmids (suppressor of cytokine signaling 6, SOCS6) is developed, designed to significantly amplify ESCC radiotherapy under hypoxic conditions. After intravenous injection, the SOCS6@vHMMn-Bi nanoparticles can be efficiently delivered to the tumor site and rapidly invade tumor cells by virus-like surface-assisted adhesion. Under X-ray irradiation, the nanoparticles exhibits a unique dual-mode sensitization effect, encompassing exogenous and endogenous mechanisms, thereby significantly augmenting the ESCC radiotherapeutic effectiveness. First, the Bi2O3 within the shell can enhance the radiosensitivity owing to its robust X-ray attenuation characteristics. Second, the SOCS6 released from the interior can inhibit both HIF-1α and JAK2/STAT3 signaling pathways, triggering ROS upregulation and intensifying radiation-mediated DNA damage inside ESCC cells. Furthermore, the shell employs MnO2 to catalyze the decomposition of endogenous H2O2 to increase oxygen generation, alleviating hypoxia within the tumor microenvironment. These nanoparticles demonstrates considerable potential as dual-mode radiosensitizers with no systemic toxicity and low immunogenicity for amplifying radiotherapeutic efficacy in ESCC.
Collapse
Affiliation(s)
- Rui Ma
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Zhi Yang
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Xia Miao
- Department of Radiation Medicine, The Faculty of Preventive Medicine, The Fourth Military Medical University, Xi'an, 710000, P. R. China
| | - Jing Hu
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Te Zhang
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Li-Tian Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710000, P. R. China
| | - Jin-Yan Lin
- Department of Occupational and Environmental Health, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710000, P. R. China
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, P. R. China
| | - Li-Na Zhao
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| |
Collapse
|
4
|
Feng S, Li Y, Tan Z, Shen S. Current landscape of metal-organic framework-mediated nucleic acid delivery and therapeutics. Int J Pharm 2025; 672:125295. [PMID: 39914507 DOI: 10.1016/j.ijpharm.2025.125295] [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: 11/12/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Nucleic acid drugs utilize DNA or RNA molecules to modulate abnormal gene expression or protein translation in cells, enabling precise treatment for specific conditions. In recent years, nucleic acid drugs have demonstrated tremendous potential in vaccine development and treating genetic disorders. Currently, the primary carriers for clinically approved nucleic acid therapies include lipid nanoparticles and viral vectors. Beyond that, metal-organic frameworks (MOFs) are highly ordered, porous nanomaterials formed through the self-assembly of metal ions and organic ligands via coordination bonds. Their porosity structure offers great loading efficiency, stability, tunability, and biocompatibility, making them an attractive option for nucleic acid delivery. Given the research on MOFs as nucleic acid carriers has garnered significant attention in recent years, this review provides an overview of the therapeutic strategies and advancements in MOF-mediated nucleic acid delivery. The unique properties of various MOF carriers are introduced, and different approaches for nucleic acid loading are parallelly compared. Moreover, a systematic classification based on the type of nucleic acid cargo loaded in MOFs and corresponding applications is thoroughly described. This summary outlines the unique mechanisms through MOFs enhance nucleic acid delivery and emphasizes their substantial impact on therapeutic efficacy. In addition, the utilization of MOF-mediated nucleic acid treatment in combination with other therapies against malignant tumors is discussed in particular. Finally, an outlook on the challenges and potential opportunities of this technology in future translational production and clinical implementation is presented and explored.
Collapse
Affiliation(s)
- Shiwei Feng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Zheng Tan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Shiyang Shen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Pharmaceutical Preparation Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
5
|
Hosseini-Kharat M, Bremmell KE, Grubor-Bauk B, Prestidge CA. Enhancing non-viral DNA delivery systems: Recent advances in improving efficiency and target specificity. J Control Release 2025; 378:170-194. [PMID: 39647508 DOI: 10.1016/j.jconrel.2024.12.002] [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: 06/07/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
DNA-based therapies are often limited by challenges such as stability, long-term integration, low transfection efficiency, and insufficient targeted DNA delivery. This review focuses on recent progress in the design of non-viral delivery systems for enhancing targeted DNA delivery and modulation of therapeutic efficiency. Cellular uptake and intracellular trafficking mechanisms play a crucial role in optimizing gene delivery efficiency. There are two main strategies employed to improve the efficiency of gene delivery vectors: (i) explore different administration routes (e.g., mucosal, intravenous, intramuscular, subcutaneous, intradermal, intratumoural, and intraocular) that best facilitates optimal uptake into the targeted cells and organs and (ii) modify the delivery vectors with cell-specific ligands (e.g., natural ligands, antibodies, peptides, carbohydrates, or aptamers) that enable targeted uptake to specific cells with higher specificity and improved biodistribution. We describe how recent progress in employing these DNA delivery strategies is advancing the field and increasing the clinical translation and ultimate clinical application of DNA therapies.
Collapse
Affiliation(s)
- Mahboubeh Hosseini-Kharat
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Kristen E Bremmell
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Branka Grubor-Bauk
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Clive A Prestidge
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
6
|
Liu S, Li H, Xi S, Zhang Y, Sun T. Advancing CNS Therapeutics: Enhancing Neurological Disorders with Nanoparticle-Based Gene and Enzyme Replacement Therapies. Int J Nanomedicine 2025; 20:1443-1490. [PMID: 39925682 PMCID: PMC11806685 DOI: 10.2147/ijn.s457393] [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: 12/31/2023] [Accepted: 12/12/2024] [Indexed: 02/11/2025] Open
Abstract
Given the complexity of the central nervous system (CNS) and the diversity of neurological conditions, the increasing prevalence of neurological disorders poses a significant challenge to modern medicine. These disorders, ranging from neurodegenerative diseases to psychiatric conditions, not only impact individuals but also place a substantial burden on healthcare systems and society. A major obstacle in treating these conditions is the blood-brain barrier (BBB), which restricts the passage of therapeutic agents to the brain. Nanotechnology, particularly the use of nanoparticles (NPs), offers a promising solution to this challenge. NPs possess unique properties such as small size, large surface area, and modifiable surface characteristics, enabling them to cross the BBB and deliver drugs directly to the affected brain regions. This review focuses on the application of NPs in gene therapy and enzyme replacement therapy (ERT) for neurological disorders. Gene therapy involves altering or manipulating gene expression and can be enhanced by NPs designed to carry various genetic materials. Similarly, NPs can improve the efficacy of ERT for lysosomal storage disorders (LSDs) by facilitating enzyme delivery to the brain, overcoming issues like immunogenicity and instability. Taken together, this review explores the potential of NPs in revolutionizing treatment options for neurological disorders, highlighting their advantages and the future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Haisong Li
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Shiwen Xi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- International Center of Future Science, Jilin University, Changchun, People’s Republic of China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
7
|
Bhattacharjee K, Ghosh A. Identification of key regulators in pancreatic ductal adenocarcinoma using network theoretical approach. PLoS One 2025; 20:e0313738. [PMID: 39869563 PMCID: PMC11771905 DOI: 10.1371/journal.pone.0313738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/30/2024] [Indexed: 01/29/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a devastating disease with poor clinical outcomes, which is mainly because of delayed disease detection, resistance to chemotherapy, and lack of specific targeted therapies. The disease's development involves complex interactions among immunological, genetic, and environmental factors, yet its molecular mechanism remains elusive. A major challenge in understanding PDAC etiology lies in unraveling the genetic profiling that governs the PDAC network. To address this, we examined the gene expression profile of PDAC and compared it with that of healthy controls, identifying differentially expressed genes (DEGs). These DEGs formed the basis for constructing the PDAC protein interaction network, and their network topological properties were calculated. It was found that the PDAC network self-organizes into a scale-free fractal state with weakly hierarchical organization. Newman and Girvan's algorithm (leading eigenvector (LEV) method) of community detection enumerated four communities leading to at least one motif defined by G (3,3). Our analysis revealed 33 key regulators were predominantly enriched in neuroactive ligand-receptor interaction, Cell adhesion molecules, Leukocyte transendothelial migration pathways; positive regulation of cell proliferation, positive regulation of protein kinase B signaling biological functions; G-protein beta-subunit binding, receptor binding molecular functions etc. Transcription Factor and mi-RNA of the key regulators were obtained. Recognizing the therapeutic potential and biomarker significance of PDAC Key regulators, we also identified approved drugs for specific genes. However, it is imperative to subject Key regulators to experimental validation to establish their efficacy in the context of PDAC.
Collapse
Affiliation(s)
| | - Aryya Ghosh
- Department of Chemistry, Ashoka University, Sonipat, Haryana, India
| |
Collapse
|
8
|
Rezaei S, Moncada-Restrepo M, Leng S, Chambers JW, Leng F. Synthesizing unmodified, supercoiled circular DNA molecules in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634800. [PMID: 39896529 PMCID: PMC11785245 DOI: 10.1101/2025.01.24.634800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Supercoiled (Sc) circular DNA, such as plasmids, has shown therapeutic potential since the 1990s, but is limited by bacterial modifications, unnecessary DNA sequences, and contaminations that may trigger harmful responses. To overcome these challenges, we have developed two novel scalable biochemical methods to synthesize unmodified Sc circular DNA. Linear DNA with two loxP sites in the same orientation is generated via PCR or rolling circle amplification. Cre recombinase then converts this linear DNA into relaxed circular DNA. After T5 exonuclease removes unwanted linear DNA, topoisomerases are employed to generate Sc circular DNA. We have synthesized EGFP-FL, a 2,002 bp mini-circular DNA carrying essential EGFP expression elements. EGFP-FL transfected human HeLa and mouse C2C12 cells with much higher efficiency than E. coli-derived plasmids. These new biochemical methods can produce unmodified Sc circular DNA, in length from 196 base pairs to several kilobases and in quantities from micrograms to milligrams, providing a promising platform for diverse applications.
Collapse
Affiliation(s)
- Sepideh Rezaei
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, United States
| | - Monica Moncada-Restrepo
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, United States
| | - Sophia Leng
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, United States
- Top Biosciences, LLC, 7405 SW 157 Terrace, Palmetto Bay, FL 33157
| | - Jeremy W. Chambers
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, United States
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199
| | - Fenfei Leng
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, United States
- Top Biosciences, LLC, 7405 SW 157 Terrace, Palmetto Bay, FL 33157
| |
Collapse
|
9
|
Majerciak V, Zheng ZM. Induction of translation-suppressive G3BP1 + stress granules and interferon-signaling cGAS condensates by transfected plasmid DNA. HLIFE 2025; 3:21-37. [PMID: 40078969 PMCID: PMC11902918 DOI: 10.1016/j.hlife.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Plasmid DNA transfection is one of the fundamental tools of biomedical research. Here, we found that plasmid DNA transfection mediated by liposomes activates multiple innate immune responses in several widely used cell lines. Their activations were visible by detection of stress granules (SG) and cGAS-DNA condensates (cGC) in the transfected cells in a plasmid DNA dose-dependent manner. The elevated levels of phosphorylated eukaryotic translation initiation factor 2 subunit alpha (eIF2α), interferon regulatory factor 3 (IRF3), and signal transducer and activator of transcription 1 (STAT1) were induced in plasmid DNA-transfected cells. The formation of SG but not cGC required active transcription and formation of dsRNA in transfected cells. Plasmid DNA-induced SG or cGC were mutually exclusive because of triggering two distinct pathways. Knockdown (KD) of PKR before plasmid DNA transfection led to abolish SG without affecting cGC formation. Conversely, cGAS KD could prevent cGC without affecting SG formation. In addition, plasmid DNA-induced SG and cGC formation could be prevented, respectively, by co-expression of KSHV proteins ORF57 (PKR inhibitor) and ORF52 (cGAS inhibitor). Inhibition of SG formation mediated by PKR KD, but not cGC KD, also led to increased expression of transgenes, indicating that PKR activation represents a major roadblock to gene expression. Together, these data indicate that plasmid DNA triggers innate immune responses in the transfected cells and causes a significant cellular perturbation that should be considered during experiment design and data interpretation.
Collapse
Affiliation(s)
- Vladimir Majerciak
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Maryland, USA
| | | |
Collapse
|
10
|
Tarab-Ravski D, Stotsky-Oterin L, Elisha A, Kundoor GR, Ramishetti S, Hazan-Halevy I, Haas H, Peer D. The future of genetic medicines delivered via targeted lipid nanoparticles to leukocytes. J Control Release 2024; 376:286-302. [PMID: 39401676 DOI: 10.1016/j.jconrel.2024.10.014] [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: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Genetic medicines hold vast therapeutic potential, offering the ability to silence or induce gene expression, knock out genes, and even edit DNA fragments. Applying these therapeutic modalities to leukocytes offers a promising path for treating various conditions yet overcoming the obstacles of specific and efficient delivery to leukocytes remains a major bottleneck in their clinical translation. Lipid nanoparticles (LNPs) have emerged as the leading delivery system for nucleic acids due to their remarkable versatility and ability to improve their in vivo stability, pharmacokinetics, and therapeutic benefits. Equipping LNPs with targeting moieties can promote their specific cellular uptake and internalization to leukocytes, making targeted LNPs (tLNPs) an inseparable part of developing leukocyte-targeted gene therapy. However, despite the significant advancements in research, genetic medicines for leukocytes using targeted delivery approaches have not been translated into the clinic yet. Herein, we discuss the important aspects of designing tLNPs and highlight the considerations for choosing an appropriate bioconjugation strategy and targeting moiety. Furthermore, we provide our insights on limiting challenges and identify key areas for further research to advance these exciting therapies for patient care.
Collapse
Affiliation(s)
- Dana Tarab-Ravski
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Govinda Reddy Kundoor
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | | | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Heinrich Haas
- NeoVac Ltd. 127 Olympic Ave., OX14 4SA, Milton Park, Oxfordshire, UK; Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
11
|
HUANG YONGJIAN, WANG JINZHOU, XU JIUHUA, RUAN NING. Remodeling tumor microenvironment using pH-sensitive biomimetic co-delivery of TRAIL/R848 liposomes against colorectal cancer. Oncol Res 2024; 32:1765-1776. [PMID: 39449815 PMCID: PMC11497182 DOI: 10.32604/or.2024.045564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/11/2024] [Indexed: 10/26/2024] Open
Abstract
Background Despite significant advancements in the development of anticancer therapies over the past few decades, the clinical management of colorectal cancer remains a challenging task. This study aims to investigate the inhibitory effects of cancer-targeting liposomes against colorectal cancer. Materials and Methods Liposomes consisting of 3β-[N-(N', N'-dimethylamino ethane)carbamoyl]-cholesterol (DC-CHOL), cholesterol (CHOL), and dioleoylphosphatidylethanolamine (DOPE) at a molar ratio of 1:1:0.5 were created and used as carriers to deliver an apoptosis-inducing plasmid encoding the tumor necrosis factor-related apoptosis-inducing ligand (pTRAIL) gene, along with the toll-like receptor (TLR7) agonist Rsiquimod (R848). The rationale behind this design is that pTRAIL can trigger cancer cell apoptosis by activating the DR4/5 receptor, while R848 can stimulate the immune microenvironment. Results Experimental results demonstrated the synergistic effects of R848 and pTRAIL encapsulated by liposomes (RTL) in suppressing the proliferation of colorectal cancer cells. Moreover, further in vivo investigations revealed the strong anti-tumor efficacy of RTL in xenograft and orthotropic in situ models of colorectal cancer. Conclusions These findings collectively highlight the therapeutic potential of R848/pTRAIL-loaded liposomes in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- YONGJIAN HUANG
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - JINZHOU WANG
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - JIUHUA XU
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - NING RUAN
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| |
Collapse
|
12
|
Lee M, Lee M, Song Y, Kim S, Park N. Recent Advances and Prospects of Nucleic Acid Therapeutics for Anti-Cancer Therapy. Molecules 2024; 29:4737. [PMID: 39407665 PMCID: PMC11477775 DOI: 10.3390/molecules29194737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Nucleic acid therapeutics are promising alternatives to conventional anti-cancer therapy, such as chemotherapy and radiation therapy. While conventional therapies have limitations, such as high side effects, low specificity, and drug resistance, nucleic acid therapeutics work at the gene level to eliminate the cause of the disease. Nucleic acid therapeutics treat diseases in various forms and using different mechanisms, including plasmid DNA (pDNA), small interfering RNA (siRNA), anti-microRNA (anti-miR), microRNA mimics (miRNA mimic), messenger RNA (mRNA), aptamer, catalytic nucleic acid (CNA), and CRISPR cas9 guide RNA (gRNA). In addition, nucleic acids have many advantages as nanomaterials, such as high biocompatibility, design flexibility, low immunogenicity, small size, relatively low price, and easy functionalization. Nucleic acid therapeutics can have a high therapeutic effect by being used in combination with various nucleic acid nanostructures, inorganic nanoparticles, lipid nanoparticles (LNPs), etc. to overcome low physiological stability and cell internalization efficiency. The field of nucleic acid therapeutics has advanced remarkably in recent decades, and as more and more nucleic acid therapeutics have been approved, they have already demonstrated their potential to treat diseases, including cancer. This review paper introduces the current status and recent advances in nucleic acid therapy for anti-cancer treatment and discusses the tasks and prospects ahead.
Collapse
Affiliation(s)
- Minhyuk Lee
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Minjae Lee
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| | - Youngseo Song
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| | - Sungjee Kim
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Nokyoung Park
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| |
Collapse
|
13
|
Edo GI, Yousif E, Al-Mashhadani MH. Modified chitosan: Insight on biomedical and industrial applications. Int J Biol Macromol 2024; 275:133526. [PMID: 38960250 DOI: 10.1016/j.ijbiomac.2024.133526] [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: 04/22/2024] [Revised: 06/22/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Chitosan (CS), a by -product of chitin deacetylation can be useful in a broad range of purposes, to mention agriculture, pharmaceuticals, material science, food and nutrition, biotechnology and of recent, in gene therapy. Chitosan is a highly desired biomolecule due to the existence of many sensitive functional groups inside the molecule and also because of its net cationicity. The latter provides flexibility for creating a wide range of derivatives for particular end users across various industries. This overview aims to compile some of the most recent research on the bio-related applications that chitosan and its derivatives can be used for. However, chitosan's reactive functional groups are amendable to chemical reaction. Modifying the material to show enhanced solubility, a greater range of application options and pH-sensitive targeting and others have been a major focus of chitosan research. This review describes the modifications of chitosan that have been made to improve its water solubility, pH sensitivity, and capacity to target chitosan derivatives. Applying the by-products of chitosan as antibacterial, in targeting, extended release and as delivery systems is also covered. The by-products of chitosan will be important and potentially useful in developing new biomedical drugs in time to come.
Collapse
Affiliation(s)
- Great Iruoghene Edo
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq.
| | - Emad Yousif
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | | |
Collapse
|
14
|
Soroudi S, Jaafari MR, Arabi L. Lipid nanoparticle (LNP) mediated mRNA delivery in cardiovascular diseases: Advances in genome editing and CAR T cell therapy. J Control Release 2024; 372:113-140. [PMID: 38876358 DOI: 10.1016/j.jconrel.2024.06.023] [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: 01/09/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of global mortality among non-communicable diseases. Current cardiac regeneration treatments have limitations and may lead to adverse reactions. Hence, innovative technologies are needed to address these shortcomings. Messenger RNA (mRNA) emerges as a promising therapeutic agent due to its versatility in encoding therapeutic proteins and targeting "undruggable" conditions. It offers low toxicity, high transfection efficiency, and controlled protein production without genome insertion or mutagenesis risk. However, mRNA faces challenges such as immunogenicity, instability, and difficulty in cellular entry and endosomal escape, hindering its clinical application. To overcome these hurdles, lipid nanoparticles (LNPs), notably used in COVID-19 vaccines, have a great potential to deliver mRNA therapeutics for CVDs. This review highlights recent progress in mRNA-LNP therapies for CVDs, including Myocardial Infarction (MI), Heart Failure (HF), and hypercholesterolemia. In addition, LNP-mediated mRNA delivery for CAR T-cell therapy and CRISPR/Cas genome editing in CVDs and the related clinical trials are explored. To enhance the efficiency, safety, and clinical translation of mRNA-LNPs, advanced technologies like artificial intelligence (AGILE platform) in RNA structure design, and optimization of LNP formulation could be integrated. We conclude that the strategies to facilitate the extra-hepatic delivery and targeted organ tropism of mRNA-LNPs (SORT, ASSET, SMRT, and barcoded LNPs) hold great prospects to accelerate the development and translation of mRNA-LNPs in CVD treatment.
Collapse
Affiliation(s)
- Setareh Soroudi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Garza Treviño EN, Quiroz Reyes AG, Delgado Gonzalez P, Rojas Murillo JA, Islas JF, Alonso SS, Gonzalez Villarreal CA. Applications of Modified Mesenchymal Stem Cells as Targeted Systems against Tumor Cells. Int J Mol Sci 2024; 25:7791. [PMID: 39063032 PMCID: PMC11276748 DOI: 10.3390/ijms25147791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Combined gene and cell therapy are promising strategies for cancer treatment. Given the complexity of cancer, several approaches are actively studied to fight this disease. Using mesenchymal stem cells (MSCs) has demonstrated dual antitumor and protumor effects as they exert massive immune/regulatory effects on the tissue microenvironment. MSCs have been widely investigated to exploit their antitumor target delivery system. They can be genetically modified to overexpress genes and selectively or more efficiently eliminate tumor cells. Current approaches tend to produce more effective and safer therapies using MSCs or derivatives; however, the effect achieved by engineered MSCs in solid tumors is still limited and depends on several factors such as the cell source, transgene, and tumor target. This review describes the progress of gene and cell therapy focused on MSCs as a cornerstone against solid tumors, addressing the different MSC-engineering methods that have been approached over decades of research. Furthermore, we summarize the main objectives of engineered MSCs against the most common cancers and discuss the challenges, limitations, risks, and advantages of targeted treatments combined with conventional ones.
Collapse
Affiliation(s)
- Elsa N. Garza Treviño
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Adriana G. Quiroz Reyes
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Paulina Delgado Gonzalez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Juan Antonio Rojas Murillo
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Jose Francisco Islas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Santiago Saavedra Alonso
- Departamento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico
| | - Carlos A. Gonzalez Villarreal
- Departamento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico
| |
Collapse
|
16
|
Yu HP, Liu FC, Chung YK, Alalaiwe A, Sung CT, Fang JY. Nucleic acid-based nanotherapeutics for treating sepsis and associated organ injuries. Theranostics 2024; 14:4411-4437. [PMID: 39113804 PMCID: PMC11303080 DOI: 10.7150/thno.98487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
In recent years, gene therapy has been made possible with the success of nucleic acid drugs against sepsis and its related organ dysfunction. Therapeutics based on nucleic acids such as small interfering RNAs (siRNAs), microRNAs (miRNAs), messenger RNAs (mRNAs), and plasmid DNAs (pDNAs) guarantee to treat previously undruggable diseases. The advantage of nucleic acid-based therapy against sepsis lies in the development of nanocarriers, achieving targeted and controlled gene delivery for improved efficacy with minimal adverse effects. Entrapment into nanocarriers also ameliorates the poor cellular uptake of naked nucleic acids. In this study, we discuss the current state of the art in nanoparticles for nucleic acid delivery to treat hyperinflammation and apoptosis associated with sepsis. The optimized design of the nanoparticles through physicochemical property modification and ligand conjugation can target specific organs-such as lung, heart, kidney, and liver-to mitigate multiple sepsis-associated organ injuries. This review highlights the nanomaterials designed for fabricating the anti-sepsis nanosystems, their physicochemical characterization, the mechanisms of nucleic acid-based therapy in working against sepsis, and the potential for promoting the therapeutic efficiency of the nucleic acids. The current investigations associated with nanoparticulate nucleic acid application in sepsis management are summarized in this paper. Noteworthily, the potential application of nanotherapeutic nucleic acids allows for a novel strategy to treat sepsis. Further clinical studies are required to confirm the findings in cell- and animal-based experiments. The capability of large-scale production and reproducibility of nanoparticle products are also critical for commercialization. It is expected that numerous anti-sepsis possibilities will be investigated for nucleic acid-based nanotherapeutics in the future.
Collapse
Affiliation(s)
- Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Yu-Kuo Chung
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Ahmed Alalaiwe
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Calvin T. Sung
- Department of Dermatology, University of California, Irvine, United States
| | - Jia-You Fang
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan
| |
Collapse
|
17
|
Mantooth SM, Abdou Y, Saez-Ibañez AR, Upadhaya S, Zaharoff DA. Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies. Front Immunol 2024; 15:1385484. [PMID: 38803496 PMCID: PMC11128577 DOI: 10.3389/fimmu.2024.1385484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Breast cancer poses one of the largest threats to women's health. Treatment continues to improve for all the subtypes of breast cancer, but some subtypes, such as triple negative breast cancer, still present a significant treatment challenge. Additionally, metastasis and local recurrence are two prevalent problems in breast cancer treatment. A newer type of therapy, immunotherapy, may offer alternatives to traditional treatments for difficult-to-treat subtypes. Immunotherapy engages the host's immune system to eradicate disease, with the potential to induce long-lasting, durable responses. However, systemic immunotherapy is only approved in a limited number of indications, and it benefits only a minority of patients. Furthermore, immune related toxicities following systemic administration of potent immunomodulators limit dosing and, consequently, efficacy. To address these safety considerations and improve treatment efficacy, interest in local delivery at the site of the tumor has increased. Numerous intratumorally delivered immunotherapeutics have been and are being explored clinically and preclinically, including monoclonal antibodies, cellular therapies, viruses, nucleic acids, cytokines, innate immune agonists, and bacteria. This review summarizes the current and past intratumoral immunotherapy clinical landscape in breast cancer as well as current progress that has been made in preclinical studies, with a focus on delivery parameters and considerations.
Collapse
Affiliation(s)
- Siena M. Mantooth
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
| | - Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - David A. Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
18
|
Peccoud S, Berezin CT, Hernandez SI, Peccoud J. PlasCAT: Plasmid Cloud Assembly Tool. Bioinformatics 2024; 40:btae299. [PMID: 38696761 PMCID: PMC11101281 DOI: 10.1093/bioinformatics/btae299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/04/2024] Open
Abstract
SUMMARY PlasCAT (Plasmid Cloud Assembly Tool) is an easy-to-use cloud-based bioinformatics tool that enables de novo plasmid sequence assembly from raw sequencing data. Nontechnical users can now assemble sequences from long reads and short reads without ever touching a line of code. PlasCAT uses high-performance computing servers to reduce run times on assemblies and deliver results faster. AVAILABILITY AND IMPLEMENTATION PlasCAT is freely available on the web at https://sequencing.genofab.com. The assembly pipeline source code and server code are available for download at https://bitbucket.org/genofabinc/workspace/projects/PLASCAT. Click the Cancel button to access the source code without authenticating. Web servers implemented in React.js and Python, with all major browsers supported.
Collapse
Affiliation(s)
| | - Casey-Tyler Berezin
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States
| | - Sarah I Hernandez
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States
| | - Jean Peccoud
- GenoFAB, Inc., Fort Collins, CO 80528, United States
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
19
|
Qin Y, Rouatbi N, Wang JTW, Baker R, Spicer J, Walters AA, Al-Jamal KT. Plasmid DNA ionisable lipid nanoparticles as non-inert carriers and potent immune activators for cancer immunotherapy. J Control Release 2024; 369:251-265. [PMID: 38493950 PMCID: PMC11464404 DOI: 10.1016/j.jconrel.2024.03.018] [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: 01/16/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Immunotherapy is currently a standard of care in the treatment of many malignancies. However, predictable side effects caused by systemic administration of highly immunostimulatory molecules have been a serious concern within this field. Intratumoural expression or silencing of immunogenic and immunoinhibitory molecules using nucleic acid-based approaches such as plasmid DNA (pDNA) and small interfering RNA (siRNA), respectively, could represent a next generation of cancer immunotherapy. Here, we employed lipid nanoparticles (LNPs) to deliver either non-specific pDNA and siRNA, or constructs targeting two prominent immunotherapeutic targets OX40L and indoleamine 2,3-dioxygenase-1 (IDO), to tumours in vivo. In the B16F10 mouse model, intratumoural delivery of LNP-formulated non-specific pDNA and siRNA led to strong local immune activation and tumour growth inhibition even at low doses due to the pDNA immunogenic nature. Replacement of these non-specific constructs by pOX40L and siIDO resulted in more prominent immune activation as evidenced by increased immune cell infiltration in tumours and tumour-draining lymph nodes. Consistently, pOX40L alone or in combination with siIDO could prolong overall survival, resulting in complete tumour regression and the formation of immunological memory in tumour rechallenge models. Our results suggest that intratumoural administration of LNP-formulated pDNA and siRNA offers a promising approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Yue Qin
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Nadia Rouatbi
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Rafal Baker
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - James Spicer
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust (GSTT), London SE1 9RT, UK; School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 9RT, UK
| | - Adam A Walters
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
20
|
Ko T, Fumoto S, Kurosaki T, Nakashima M, Miyamoto H, Sasaki H, Nishida K. Interaction of γ-Polyglutamic Acid/Polyethyleneimine/Plasmid DNA Ternary Complexes with Serum Components Plays a Crucial Role in Transfection in Mice. Pharmaceutics 2024; 16:522. [PMID: 38675183 PMCID: PMC11053868 DOI: 10.3390/pharmaceutics16040522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/20/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Typical examples of non-viral vectors are binary complexes of plasmid DNA with cationic polymers such as polyethyleneimine (PEI). However, problems such as cytotoxicity and hemagglutination, owing to their positively charged surfaces, hinder their in vivo use. Coating binary complexes with anionic polymers, such as γ-polyglutamic acid (γ-PGA), can prevent cytotoxicity and hemagglutination. However, the role of interactions between these complexes and serum components in in vivo gene transfer remains unclear. In this study, we analyzed the contribution of serum components to in vivo gene transfer using PEI/plasmid DNA binary complexes and γ-PGA/PEI/plasmid DNA ternary complexes. In binary complexes, heat-labile components in the serum greatly contribute to the hepatic and splenic gene expression of the luciferase gene. In contrast, serum albumin and salts affected the hepatic and splenic gene expression in the ternary complexes. Changes in physicochemical characteristics, such as increased particle size and decreased absolute values of ζ-potential, might be involved in the enhanced gene expression. These findings would contribute to a better understanding of in vivo non-viral gene transfer using polymers, such as PEI and γ-PGA.
Collapse
Affiliation(s)
- Tomotaka Ko
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Tomoaki Kurosaki
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Moe Nakashima
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Hirotaka Miyamoto
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Hitoshi Sasaki
- Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| |
Collapse
|
21
|
Guan X, Pei Y, Song J. DNA-Based Nonviral Gene Therapy─Challenging but Promising. Mol Pharm 2024; 21:427-453. [PMID: 38198640 DOI: 10.1021/acs.molpharmaceut.3c00907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Over the past decades, significant progress has been made in utilizing nucleic acids, including DNA and RNA molecules, for therapeutic purposes. For DNA molecules, although various DNA delivery systems have been established, viral vector systems are the go-to choice for large-scale commercial applications. However, viral systems have certain disadvantages such as immune response, limited payload capacity, insertional mutagenesis and pre-existing immunity. In contrast, nonviral systems are less immunogenic, not size limited, safer, and easier for manufacturing compared with viral systems. What's more, nonviral DNA vectors have demonstrated their capacity to mediate specific protein expression in vivo for diverse therapeutic objectives containing a wide range of diseases such as cancer, rare diseases, neurodegenerative diseases, and infectious diseases, yielding promising therapeutic outcomes. However, exogenous plasmid DNA is prone to degrade and has poor immunogenicity in vivo. Thus, various strategies have been developed: (i) designing novel plasmids with special structures, (ii) optimizing plasmid sequences for higher expression, and (iii) developing more efficient nonviral DNA delivery systems. Based on these strategies, many interesting clinical results have been reported. This Review discusses the development of DNA-based nonviral gene therapy, including novel plasmids, nonviral delivery systems, clinical advances, and prospects. These developments hold great potential for enhancing the efficacy and safety of nonviral gene therapy and expanding its applications in the treatment of various diseases.
Collapse
Affiliation(s)
- Xiaocai Guan
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yufeng Pei
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jie Song
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
22
|
V R, Chacko AM, Abdulla N, Annamalai M, Kandi V. Medication Adherence in Cancer Patients: A Comprehensive Review. Cureus 2024; 16:e52721. [PMID: 38384629 PMCID: PMC10880514 DOI: 10.7759/cureus.52721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Cancer is a complex disease that can affect different parts of the body. The rates of cancer have shown an increasing trend in the past decade. A majority of cancers are detected late, therefore becoming untreatable and resulting in significant mortality. Additionally, the lack of awareness about cancers, their risk factors, diagnostic modalities, and preventive measures contributes to increased burden among people. Despite significant developments in the therapeutic and comprehensive management of cancers, the cause for concern is the lack of medication adherence. This is majorly attributed to the adverse effects of the medication, the cost of the drugs, and other reasons. This review comprehensively discusses various aspects of cancer medication adherence that include therapeutic modalities for treating cancers, factors influencing medication adherence, barriers, and facilitators to medication adherence.
Collapse
Affiliation(s)
- Reshma V
- Pharmacology, Shri Sathya Sai Medical College and Research Institute (SSSMCRI), Chennai, IND
| | - Arun M Chacko
- Biochemistry, Sri Balaji Vidyapeeth, Puducherry, IND
- Biochemistry, Azeezia Institute of Medical Sciences and Research, Kollam, IND
| | - Naseeha Abdulla
- Pharmacology and Therapeutics, Kunhitharuvai Memorial Charitable Trust (KMCT) Medical College, Calicut, IND
| | - Maduram Annamalai
- Pharmacology, Shri Sathya Sai Medical College and Research Institute (SSSMCRI), Chennai, IND
| | - Venkataramana Kandi
- Clinical Microbiology, Prathima Institute of Medical Sciences, Karimnagar, IND
| |
Collapse
|
23
|
Pang J, Zhuang B, Zhang LM. A co-carrier for plasmid DNA and curcumin delivery to treat pancreatic cancer via dendritic poly(l-lysine) modified amylose. Int J Biol Macromol 2023; 253:127467. [PMID: 37863141 DOI: 10.1016/j.ijbiomac.2023.127467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 10/22/2023]
Abstract
Pancreatic cancer is one of the most lethal malignancies in the world and remains one of the leading causes of cancer related death. For its treatment, a lot of investigations have dealt not only with individual chemotherapy by using polymeric carriers to deliver anticancer drugs, but also with individual gene therapy by using polymeric carriers to deliver nucleic acids such as small interfering RNA (siRNA) and plasmid DNA. However, relatively few studies have been focused on the co-delivery of gene and anticancer drug by multifunctional polymeric carriers for its synergistic therapy. In this work, a DPLL-functionalized amylose (ADP) was prepared by the click reaction between azidized amylose and propargyl focal point poly(l-lysine) dendrons, and then used to co-deliver plasmid pIRES2-EGFP-TNFα and curcumin for pancreatic cancer treatment. Due to the internal hydrophobic cavity of amylose component, ADP could load efficiently curcumin with anticancer activity and showed a sustained release behavior. Moreover, the curcumin-loaded ADP could form colloidally stable nanocomplexes with plasmid DNA in aqueous system due to the existence of cationic poly(l-lysine) dendrons and exhibited high gene transfection efficiency. The in vitro and in vivo tests confirmed the effectiveness of using ADP to co-deliver plasmid pIRES2-EGFP-TNFα and curcumin for synergistic therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Jiadong Pang
- DSAPM Lab and PCFM Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; Medical Intelligence and Innovation Academy, South University of Science and Technology Hospital, Shenzhen 518000, China
| | - Baoxiong Zhuang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li-Ming Zhang
- DSAPM Lab and PCFM Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
24
|
Pagliari S, Dema B, Sanchez-Martinez A, Montalvo Zurbia-Flores G, Rollier CS. DNA Vaccines: History, Molecular Mechanisms and Future Perspectives. J Mol Biol 2023; 435:168297. [PMID: 37797831 DOI: 10.1016/j.jmb.2023.168297] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/07/2023]
Abstract
The history of DNA vaccine began as early as the 1960s with the discovery that naked DNA can transfect mammalian cells in vivo. In 1992, the evidence that such transfection could lead to the generation of antigen-specific antibody responses was obtained and supported the development of this technology as a novel vaccine platform. The technology then attracted immense interest and high hopes in vaccinology, as evidence of high immunogenicity and protection against virulent challenges accumulated from several animal models for several diseases. In particular, the capacity to induce T-cell responses was unprecedented in non-live vaccines. However, the technology suffered its major knock when the success in animals failed to translate to humans, where DNA vaccine candidates were shown to be safe but remained poorly immunogenic, or not associated with clinical benefit. Thanks to a thorough exploration of the molecular mechanisms of action of these vaccines, an impressive range of approaches have been and are currently being explored to overcome this major challenge. Despite limited success so far in humans as compared with later genetic vaccine technologies such as viral vectors and mRNA, DNA vaccines are not yet optimised for human use and may still realise their potential.
Collapse
Affiliation(s)
- Sthefany Pagliari
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Barbara Dema
- Pandemic Science Institute, Institute of Developmental and Regenerative Medicine (IDRM), University of Oxford, Oxford, UK
| | | | | | - Christine S Rollier
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.
| |
Collapse
|
25
|
Skolnik JM, Morrow MP. Vaccines for HPV-associated diseases. Mol Aspects Med 2023; 94:101224. [PMID: 37931422 DOI: 10.1016/j.mam.2023.101224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/08/2023]
Abstract
Human papillomavirus (HPV) infection represents a significant global health concern owing to its role in the etiology of conditions ranging from benign low-grade lesions to cancers of the cervix, head and neck, anus, vagina, vulva, and penis. Prophylactic vaccination programs, primarily targeting adolescent girls, have achieved dramatic reductions in rates of HPV infection and cervical cancer in recent years. However, there is a clear demand for a strategy to manage the needs of the many people who are already living with persistent HPV infection and/or HPV-associated conditions. Unlike prophylactic vaccines, which act to prevent HPV infection, therapeutic vaccination presents an opportunity to induce cellular immunity against established HPV infections and lesions and prevent progression to cancer. Several HPV vaccines are undergoing clinical development, using a range of platforms. Peptide- or protein-based vaccines, vector-based vaccines, whole-cell vaccines, and nucleic acid vaccines each offer relative merits and limitations for the delivery of HPV antigens and the subsequent generation of targeted immune responses. There has been particular interest in DNA-based vaccines, which elicit both cellular and humoral immune responses to provide long-lasting immunity. DNA vaccines offer several practical advantages over other vaccine platforms, including the potential for rapid and scalable manufacturing, targeting of many different antigens, and potential for repeat boosting. Furthermore, unlike vectored approaches, DNA vaccines are thermostable over extended time periods, which may enable shipping and storage. Several delivery strategies are available to address the main challenge of DNA vaccines, namely their relatively low transfection efficiency. We review the latest clinical data supporting the development of DNA vaccines and reflect on this exciting prospect in the management of HPV-related disease.
Collapse
|
26
|
Kieu Doan TN, Croyle MA. Physical characteristics and stability profile of recombinant plasmid DNA within a film matrix. Eur J Pharm Biopharm 2023; 190:270-283. [PMID: 37567395 DOI: 10.1016/j.ejpb.2023.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Plasmids are essential source material for production of biological drugs, vaccines and vectors for gene therapy. They are commonly formulated as frozen solutions. Considering the cost associated with maintenance of cold chain conditions during storage and transport, there is a significant need for alternative methods for stabilization of plasmids at ambient temperature. The objective of these studies was to identify a film-based formulation that preserved transfection efficiency of plasmids at 25 °C. A model plasmid, pAAVlacZ, was used for these studies. Transfection efficiency and agarose gel electrophoresis were utilized to assess bioactivity and changes in physical conformation of plasmid during storage. An amino acid, capable of sustaining a positive charge while supporting an alkaline environment within the film matrix, preserved transfection efficiency for 9 months at 25 °C. Addition of sugar and a plasticizer to the formulation preserved the plasmid in an amorphous state and improved handling properties of the film. The manner in which excipients were incorporated into bulk formulations and environmental humidity in which films were stored significantly impacted transfection efficiency of plasmid in the rehydrated solution. Taken together, these results suggest that plasmids can be stored for extended periods of time without refrigeration within a film matrix.
Collapse
Affiliation(s)
- Trang Nguyen Kieu Doan
- The University of Texas at Austin College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX 78712, United States
| | - Maria A Croyle
- The University of Texas at Austin College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX 78712, United States; John R. LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
27
|
Ren S, Zhang Z, Li M, Wang D, Guo R, Fang X, Chen F. Cancer testis antigen subfamilies: Attractive targets for therapeutic vaccine (Review). Int J Oncol 2023; 62:71. [PMID: 37144487 PMCID: PMC10198712 DOI: 10.3892/ijo.2023.5519] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Cancer‑testis antigen (CTA) is a well‑accepted optimal target library for cancer diagnosis and treatment. Most CTAs are located on the X chromosome and aggregate into large gene families, such as the melanoma antigen, synovial sarcoma X and G antigen families. Members of the CTA subfamily are usually co‑expressed in tumor tissues and share similar structural characteristics and biological functions. As cancer vaccines are recommended to induce specific antitumor responses, CTAs, particularly CTA subfamilies, are widely used in the design of cancer vaccines. To date, DNA, mRNA and peptide vaccines have been commonly used to generate tumor‑specific CTAs in vivo and induce anticancer effects. Despite promising results in preclinical studies, the antitumor efficacy of CTA‑based vaccines is limited in clinical trials, which may be partially attributed to weak immunogenicity, low efficacy of antigen delivery and presentation processes, as well as a suppressive immune microenvironment. Recently, the development of nanomaterials has enhanced the cancer vaccination cascade, improved the antitumor performance and reduced off‑target effects. The present study provided an in‑depth review of the structural characteristics and biofunctions of the CTA subfamilies, summarised the design and utilisation of CTA‑based vaccine platforms and provided recommendations for developing nanomaterial‑derived CTA‑targeted vaccines.
Collapse
Affiliation(s)
- Shengnan Ren
- Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhanyi Zhang
- Bethune Third Clinical Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Mengyuan Li
- Traditional Chinese Medicine College, Jilin Agricultural University, Changchun, Jilin 130118, P.R. China
| | - Daren Wang
- Bethune Third Clinical Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruijie Guo
- Bethune Third Clinical Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuedong Fang
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Fangfang Chen
- Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
28
|
Dormeshkin D, Katsin M, Stegantseva M, Golenchenko S, Shapira M, Dubovik S, Lutskovich D, Kavaleuski A, Meleshko A. Design and Immunogenicity of SARS-CoV-2 DNA Vaccine Encoding RBD-PVXCP Fusion Protein. Vaccines (Basel) 2023; 11:1014. [PMID: 37376403 DOI: 10.3390/vaccines11061014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 06/29/2023] Open
Abstract
The potential of immune-evasive mutation accumulation in the SARS-CoV-2 virus has led to its rapid spread, causing over 600 million confirmed cases and more than 6.5 million confirmed deaths. The huge demand for the rapid development and deployment of low-cost and effective vaccines against emerging variants has renewed interest in DNA vaccine technology. Here, we report the rapid generation and immunological evaluation of novel DNA vaccine candidates against the Wuhan-Hu-1 and Omicron variants based on the RBD protein fused with the Potato virus X coat protein (PVXCP). The delivery of DNA vaccines using electroporation in a two-dose regimen induced high-antibody titers and profound cellular responses in mice. The antibody titers induced against the Omicron variant of the vaccine were sufficient for effective protection against both Omicron and Wuhan-Hu-1 virus infections. The PVXCP protein in the vaccine construct shifted the immune response to the favorable Th1-like type and provided the oligomerization of RBD-PVXCP protein. Naked DNA delivery by needle-free injection allowed us to achieve antibody titers comparable with mRNA-LNP delivery in rabbits. These data identify the RBD-PVXCP DNA vaccine platform as a promising solution for robust and effective SARS-CoV-2 protection, supporting further translational study.
Collapse
Affiliation(s)
- Dmitri Dormeshkin
- Institute of Bioorganic Chemistry of the National Academy of Sciences of Belarus, 220084 Minsk, Belarus
| | - Mikalai Katsin
- Immunofusion, LLC, 210004 Vitebsk, Belarus
- Imunovakcina, UAB, LT-08102 Vilnius, Lithuania
| | | | | | - Michail Shapira
- Institute of Bioorganic Chemistry of the National Academy of Sciences of Belarus, 220084 Minsk, Belarus
| | - Simon Dubovik
- Institute of Bioorganic Chemistry of the National Academy of Sciences of Belarus, 220084 Minsk, Belarus
| | | | - Anton Kavaleuski
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Alexander Meleshko
- Immunofusion, LLC, 210004 Vitebsk, Belarus
- Imunovakcina, UAB, LT-08102 Vilnius, Lithuania
| |
Collapse
|
29
|
Quraish RU, Hirahata T, Quraish AU, ul Quraish S. An Overview: Genetic Tumor Markers for Early Detection and Current Gene Therapy Strategies. Cancer Inform 2023; 22:11769351221150772. [PMID: 36762284 PMCID: PMC9903029 DOI: 10.1177/11769351221150772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/24/2022] [Indexed: 02/04/2023] Open
Abstract
Genomic instability is considered a fundamental factor involved in any neoplastic disease. Consequently, the genetically unstable cells contribute to intratumoral genetic heterogeneity and phenotypic diversity of cancer. These genetic alterations can be detected by several diagnostic techniques of molecular biology and the detection of alteration in genomic integrity may serve as reliable genetic molecular markers for the early detection of cancer or cancer-related abnormal changes in the body cells. These genetic molecular markers can detect cancer earlier than any other method of cancer diagnosis, once a tumor is diagnosed, then replacement or therapeutic manipulation of these cancer-related abnormal genetic changes can be possible, which leads toward effective and target-specific cancer treatment and in many cases, personalized treatment of cancer could be performed without the adverse effects of chemotherapy and radiotherapy. In this review, we describe how these genetic molecular markers can be detected and the possible ways for the application of this gene diagnosis for gene therapy that can attack cancerous cells, directly or indirectly, which lead to overall improved management and quality of life for a cancer patient.
Collapse
Affiliation(s)
| | - Tetsuyuki Hirahata
- Tetsuyuki Hirahata, Hirahata Gene Therapy Laboratory, HIC Clinic #1105, Itocia Office Tower 11F, 2-7-1, Yurakucho, Chiyoda-ku, Tokyo 100-0006, Japan.
| | | | | |
Collapse
|