1
|
Anurogo D, Chen CY, Lin CC, Pawitan JA, Qiu DW, Qiu JT. Codon optimized influenza H1 HA sequence but not CTLA-4 targeting of HA antigen to enhance the efficacy of DNA vaccines in an animal model. J Immunotoxicol 2024; 21:2400624. [PMID: 39319829 DOI: 10.1080/1547691x.2024.2400624] [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: 06/26/2024] [Revised: 06/05/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Infections caused by the influenza virus lead to both epidemic and pandemic outbreaks in humans and animals. Owing to their rapid production, safety, and stability, DNA vaccines represent a promising avenue for eliciting immunity and thwarting viral infections. While DNA vaccines have demonstrated substantial efficacy in murine models, their effectiveness in larger animals remains subdued. This limitation may be addressed by augmenting the immunogenicity of DNA-based vaccines. In the investigation here, protein expression was enhanced via codon optimization and then mouse cytotoxic T-lymphocyte antigen 4 (CTLA-4) was harnessed as a modulatory adjunct to bind directly to antigen-presenting cells. Further, the study evaluated the immunogenicity of two variants of the hemagglutinin (HA) antigen, i.e. the full-length and the C-terminal deletion versions. The study findings revealed that the codon-optimized HA gene (pcHA) led to increased protein synthesis, as evidenced by elevated mRNA levels. Codon optimization also significantly bolstered both cellular and humoral immune responses. In cytokine assays, all plasmid constructs, particularly pCTLA4-cHA, induced robust interferon (IFN)-γ production, while interleukin (IL)-4 levels remained uniformly non-significant. Mice immunized with pcHA displayed an augmented presence of IFNγ+ T-cells, underscoring the enhanced potency of the codon-optimized HA vaccine. Contrarily, CTLA-4-fused DNA vaccines did not significantly amplify the immune response.
Collapse
MESH Headings
- Animals
- Vaccines, DNA/immunology
- Vaccines, DNA/genetics
- Mice
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Codon/genetics
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Humans
- Female
- Mice, Inbred BALB C
- Disease Models, Animal
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza A Virus, H1N1 Subtype/immunology
Collapse
Affiliation(s)
- Dito Anurogo
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Makassar, Makassar City, Indonesia
| | - Chia-Yuan Chen
- Department of Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Chu-Chi Lin
- Department of Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Universitas Indonesia, Jakarta, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Universitas Indonesia, Jakarta, Indonesia
- Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Daniel W Qiu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - J Timothy Qiu
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
2
|
Liu Y, Wu Y, Li Z, Wan D, Pan J. Targeted Drug Delivery Strategies for the Treatment of Hepatocellular Carcinoma. Molecules 2024; 29:4405. [PMID: 39339402 PMCID: PMC11434448 DOI: 10.3390/molecules29184405] [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: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent malignant tumors, exhibiting a high incidence rate that presents a substantial threat to human health. The use of sorafenib and lenvatinib, commonly employed as single-agent targeted inhibitors, complicates the treatment process due to the absence of definitive targeting. Nevertheless, the advent of nanotechnology has injected new optimism into the domain of liver cancer therapy. Nanocarriers equipped with active targeting or passive targeting mechanisms have demonstrated the capability to deliver drugs to tumor cells with high efficiency. This approach not only facilitates precise delivery to the affected site but also enables targeted drug release, thereby enhancing therapeutic efficacy. As medical technology progresses, there is an increasing call for innovative treatment modalities, including novel chemotherapeutic agents, gene therapy, phototherapy, immunotherapy, and combinatorial treatments for HCC. These emerging therapies are anticipated to yield improved clinical outcomes for patients, while minimizing systemic toxicity and adverse effects. Consequently, the application of nanotechnology is poised to significantly improve HCC treatment. This review focused on targeted strategies for HCC and the application of nanotechnology in this area.
Collapse
Affiliation(s)
- Yonghui Liu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Yanan Wu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Zijian Li
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Dong Wan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Jie Pan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| |
Collapse
|
3
|
Yang G, Zhou D, Dai Y, Li Y, Wu J, Liu Q, Deng X. Construction of PEI-EGFR-PD-L1-siRNA dual functional nano-vaccine and therapeutic efficacy evaluation for lung cancer. Thorac Cancer 2022; 13:2941-2950. [PMID: 36117149 PMCID: PMC9626337 DOI: 10.1111/1759-7714.14618] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND PD-1/PD-L1 tumor immunotherapy shows effective anticancer in treatment of solid tumors, so PEI lipid nanoparticles (PEI-LNP)/siRNA complex (EPV-PEI-LNP-SiRNA) with the therapeutic function of PD-L1-siRNA and EGFR short peptide/PD-L1 double immune-enhancing function were constructed for the prevention and treatment of EGFR-positive lung cancer in this study. METHOD In this study, PEI lipid nanoparticles (PEI-LNP)/siRNA complex (EPV-PEI-LNP-siRNA) with the therapeutic function of PD-L1-siRNA and EGFR short peptide/PD-L1 double immune-enhancing function were constructed for the prevention and treatment of EGFR-positive lung cancer and functional evaluation was conducted. RESULTS On the basis of the construction of the composite nano-drug delivery system, the binding capacity, cytotoxicity, apoptosis and uptake capacity of siRNA and EPV-PEI-LNP were tested in vitro, and the downregulation effect of PD-L1 on A549 cancer cells and the cytokine levels of cocultured T cells were tested. Lipid nanoparticles delivered siRNA and EGFR short peptide vaccine to non-small cell lung cancer (NSCLC), increasing tumor invasion and activation of CD8 + T cells. Combination therapy is superior to single target therapy. CONCLUSION Our constructed lipid nanoparticles of tumor targeted therapy gene siRNA combination had the ability to target cells in vitro and downregulate the expression of PD-L1, realizing the tumor-specific expression of immune-stimulating cytokines, which is a highly efficient and safe targeted therapy nano-vaccine.
Collapse
Affiliation(s)
- Guixue Yang
- Department of Thoracic Surgery, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Dong Zhou
- Department of Thoracic Surgery, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yin Dai
- Department of Information, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yanqi Li
- Department of Thoracic Surgery, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Jiang Wu
- Department of Thoracic Surgery, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Quanxing Liu
- Department of Thoracic Surgery, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Xufeng Deng
- Department of Thoracic Surgery, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
4
|
Lin X, Lin L, Wu J, Jiang W, Wu J, Yang J, Chen C. A targeted siRNA-loaded PDL1-exosome and functional evaluation against lung cancer. Thorac Cancer 2022; 13:1691-1702. [PMID: 35545838 PMCID: PMC9161323 DOI: 10.1111/1759-7714.14445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND As an endocytic nanosicle involved in intercellular communication, an exosome can efficiently deliver drugs from one cell to another and deliver therapeutic short interfering RNA (siRNA) to target cells. This is conducive to gene therapy for cancers. In this study, an exosome was used as the siRNA-loaded substrate to prepare a targeted siRNA-loaded PD-L1 exosome and evaluate its function against lung cancer. METHODS The optimal preparation process and binding ratio of the targeted nanovesicle/siRNA complex was determined by detecting the particle size, potential, and other physical parameters in combination with cell binding and uptake capacity of exosome complexes. The biological cell behavior of targeted exosome nanosicles was evaluated through cytotoxicity, apoptosis, and the cell uptake capacity. RESULTS A targeted exosome nanovesicle capable of loading siRNA and characterized with low toxicity, high loading rate, and the ability to be used for targeted tumor cell gene therapy was constructed. CONCLUSION The PD-L1 targeting exosome can be used as an efficient siRNA delivery carrier, which is an efficient and safe nanocarrier for tumor targeted gene therapy.
Collapse
Affiliation(s)
- Xianbin Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Liangan Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jingyang Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wentan Jiang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jiayun Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jianshen Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fuzhou, China
| |
Collapse
|
5
|
Chen YP, Lin CC, Xie YX, Chen CY, Qiu JT. Enhancing immunogenicity of HPV16 E 7 DNA vaccine by conjugating codon-optimized GM-CSF to HPV16 E 7 DNA. Taiwan J Obstet Gynecol 2021; 60:700-705. [PMID: 34247810 DOI: 10.1016/j.tjog.2021.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE To generate immunity against human papillomavirus (HPV), the use of a recombinant DNA vaccine to carry an appropriate target gene is a promising and cost-effective approach. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a potent immunomodulatory cytokine that enhances the efficacy of vaccines by promoting the development and prolongation of humoral and cellular immunity. In this study, we linked codon-optimized GM-CSF (cGM-CSF) to the HPV16 E7 sequence as fused protein and evaluated the immunogenic potential of this DNA vaccine. MATERIALS AND METHODS We have demonstrated that cGM-CSF enhanced immunity against tumor challenges by generating and promoting the proliferation of HPV16 E7-specific CD8+ T cells, which secrete IFN-γ in the murine model. In this study, we aimed to evaluate the immunogenic potential of DNA vaccine that constructed by linking codon-optimized GM-CSF to HPV16 E7 sequence in the animal model. We study the half-life of RNA decay and cellular location of HPV16 E7 by Q-PCR and Western blot. We also assess immune response in the animal model by flow cytometry and ELISA. RESULTS The cGM-CSF-E7 sequence increased and extended the expression of E7 mRNA, in comparison with the E7 sequence alone. Mice vaccinated with the cGM-CSF-E7 DNA vaccine exhibited a slower rate of tumor growth than those vaccinated with the unconjugated E7 DNA vaccine. We also found that the CD4 and CD8+ T cells from these mice showed strong secretion of IFN-γ. CONCLUSION Through in vivo antibody depletion experiments, we demonstrated that both CD4+ and CD8+ T cells play an important role in the suppression of tumor growth.
Collapse
Affiliation(s)
- Yi-Pin Chen
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan, ROC; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chu-Chi Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Yu-Xin Xie
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chia-Yuan Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - J Timothy Qiu
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC; College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
6
|
Chang CC, Dinh TK, Lee YA, Wang FN, Sung YC, Yu PL, Chiu SC, Shih YC, Wu CY, Huang YD, Wang J, Lu TT, Wan D, Chen Y. Nanoparticle Delivery of MnO 2 and Antiangiogenic Therapy to Overcome Hypoxia-Driven Tumor Escape and Suppress Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44407-44419. [PMID: 32865389 DOI: 10.1021/acsami.0c08473] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Antiangiogenic therapy is widely administered in many cancers, and the antiangiogenic drug sorafenib offers moderate benefits in advanced hepatocellular carcinoma (HCC). However, antiangiogenic therapy can also lead to hypoxia-driven angiogenesis and immunosuppression in the tumor microenvironment (TME) and metastasis. Here, we report the synthesis and evaluation of NanoMnSor, a tumor-targeted, nanoparticle drug carrier that efficiently codelivers oxygen-generating MnO2 and sorafenib into HCC. We found that MnO2 not only alleviates hypoxia by catalyzing the decomposition of H2O2 to oxygen but also enhances pH/redox-responsive T1-weighted magnetic resonance imaging and drug-release properties upon decomposition into Mn2+ ions in the TME. Moreover, macrophages exposed to MnO2 displayed increased mRNA associated with the immunostimulatory M1 phenotype. We further show that NanoMnSor treatment leads to sorafenib-induced decrease in tumor vascularization and significantly suppresses primary tumor growth and distal metastasis, resulting in improved overall survival in a mouse orthotopic HCC model. Furthermore, NanoMnSor reprograms the immunosuppressive TME by reducing the hypoxia-induced tumor infiltration of tumor-associated macrophages, promoting macrophage polarization toward the immunostimulatory M1 phenotype, and increasing the number of CD8+ cytotoxic T cells in tumors, thereby augmenting the efficacy of anti-PD-1 antibody and whole-cell cancer vaccine immunotherapies. Our study demonstrates the potential of oxygen-generating nanoparticles to deliver antiangiogenic agents, efficiently modulate the hypoxic TME, and overcome hypoxia-driven drug resistance, thereby providing therapeutic benefit in cancer.
Collapse
Affiliation(s)
- Chih-Chun Chang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Trinh Kieu Dinh
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-An Lee
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Fu-Nien Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Pei-Lun Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Shao-Chieh Chiu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yu-Chuan Shih
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Cheng-Yun Wu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-Da Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Dehui Wan
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
7
|
Huang KW, Hsu FF, Qiu JT, Chern GJ, Lee YA, Chang CC, Huang YT, Sung YC, Chiang CC, Huang RL, Lin CC, Dinh TK, Huang HC, Shih YC, Alson D, Lin CY, Lin YC, Chang PC, Lin SY, Chen Y. Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer. SCIENCE ADVANCES 2020; 6:eaax5032. [PMID: 31998834 PMCID: PMC6962042 DOI: 10.1126/sciadv.aax5032] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 11/15/2019] [Indexed: 05/07/2023]
Abstract
While immunotherapy holds great promise for combating cancer, the limited efficacy due to an immunosuppressive tumor microenvironment and systemic toxicity hinder the broader application of cancer immunotherapy. Here, we report a combinatorial immunotherapy approach that uses a highly efficient and tumor-selective gene carrier to improve anticancer efficacy and circumvent the systemic toxicity. In this study, we engineered tumor-targeted lipid-dendrimer-calcium-phosphate (TT-LDCP) nanoparticles (NPs) with thymine-functionalized dendrimers that exhibit not only enhanced gene delivery capacity but also immune adjuvant properties by activating the stimulator of interferon genes (STING)-cGAS pathway. TT-LDCP NPs delivered siRNA against immune checkpoint ligand PD-L1 and immunostimulatory IL-2-encoding plasmid DNA to hepatocellular carcinoma (HCC), increased tumoral infiltration and activation of CD8+ T cells, augmented the efficacy of cancer vaccine immunotherapy, and suppressed HCC progression. Our work presents nanotechnology-enabled dual delivery of siRNA and plasmid DNA that selectively targets and reprograms the immunosuppressive tumor microenvironment to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Kuan-Wei Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Fu-Fei Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jiantai Timothy Qiu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Guann-Jen Chern
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yi-An Lee
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chih-Chun Chang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-Ting Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Chin Chiang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Rui-Lin Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chu-Chi Lin
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Trinh Kieu Dinh
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hsi-Chien Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-Chuan Shih
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Donia Alson
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yen Lin
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Hepatology, Department of Gastroenterology and Hepatology, Linkou Medical Center; Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yung-Chang Lin
- Division of Medical Oncology/Hematology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Po-Chiao Chang
- Division of Hepatology, Department of Gastroenterology and Hepatology, Linkou Medical Center; Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Yi Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
8
|
Sung YC, Jin PR, Chu LA, Hsu FF, Wang MR, Chang CC, Chiou SJ, Qiu JT, Gao DY, Lin CC, Chen YS, Hsu YC, Wang J, Wang FN, Yu PL, Chiang AS, Wu AYT, Ko JJS, Lai CPK, Lu TT, Chen Y. Delivery of nitric oxide with a nanocarrier promotes tumour vessel normalization and potentiates anti-cancer therapies. NATURE NANOTECHNOLOGY 2019; 14:1160-1169. [PMID: 31740794 DOI: 10.1038/s41565-019-0570-3] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/03/2019] [Indexed: 05/28/2023]
Abstract
Abnormal tumour vasculature has a significant impact on tumour progression and response to therapy. Nitric oxide (NO) regulates angiogenesis and maintains vascular homeostasis and, thus, can be delivered to normalize tumour vasculature. However, a NO-delivery system with a prolonged half-life and a sustained release mechanism is currently lacking. Here we report the development of NanoNO, a nanoscale carrier that enables sustained NO release to efficiently deliver NO into hepatocellular carcinoma. Low-dose NanoNO normalizes tumour vessels and improves the delivery and effectiveness of chemotherapeutics and tumour necrosis factor-related, apoptosis-inducing, ligand-based therapy in both primary tumours and metastases. Furthermore, low-dose NanoNO reprogrammes the immunosuppressive tumour microenvironment toward an immunostimulatory phenotype, thereby improving the efficacy of cancer vaccine immunotherapy. Our findings demonstrate the ability of nanoscale NO delivery to efficiently reprogramme tumour vasculature and immune microenvironments to overcome resistance to cancer therapy, resulting in a therapeutic benefit.
Collapse
Affiliation(s)
- Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Ru Jin
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-An Chu
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Fu-Fei Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Mei-Ren Wang
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Chih-Chun Chang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Show-Jen Chiou
- Department of Applied Chemistry, National Chiayi University, Chiayi, Taiwan
| | - Jiantai Timothy Qiu
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Dong-Yu Gao
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Chu-Chi Lin
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Sing Chen
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Fu-Nien Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Lun Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ann-Shyn Chiang
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Anthony Yan-Tang Wu
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei, Taiwan
| | - John Jun-Sheng Ko
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, Taiwan.
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
9
|
Effect of Multiple Vaccinations with Tumor Cell-Based Vaccine with Codon-Modified GM-CSF on Tumor Growth in a Mouse Model. Cancers (Basel) 2019; 11:cancers11030368. [PMID: 30875953 PMCID: PMC6468346 DOI: 10.3390/cancers11030368] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/07/2019] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
Ectopic expression of codon-modified granulocyte-macrophage colony-stimulating factor (cGM-CSF) in TC-1 cells (TC-1/cGM-CSF), a model cell line for human papillomavirus (HPV)-infected cervical cancer cells, increased the expression level of GM-CSF and improved the efficacy of tumor cell-based vaccines in a cervical cancer mouse model. The number of vaccine doses required to induce a long-term immune response in a cervical cancer mouse model is poorly understood. Here, we investigated one, three, and five doses of the irradiated TC-1/cGM-CSF vaccine to determine which dose was effective in inducing a greater immune response and the suppression of tumors. Our findings showed that three doses of irradiated TC-1/cGM-CSF vaccine elicited slower tumor growth rates and enhanced survival rates compared with one dose or five doses of irradiated TC-1/cGM-CSF vaccine. Consistently, mice vaccinated with three doses of irradiated TC-1/cGM-CSF vaccine exhibited stronger interferon gamma (IFN-γ) production in HPV E7-specific CD8⁺ T cells and CD4⁺ T cells. A higher percentage of natural killer cells and interferon-producing killer dendritic cells (IKDCs) appeared in the splenocytes of the mice vaccinated with three doses of irradiated TC-1/cGM-CSF vaccine compared with those of the mice vaccinated with one dose or five doses of irradiated TC-1/cGM-CSF vaccine. Our findings demonstrate that single or multiple vaccinations, such as five doses, with irradiated TC-1/cGM-CSF vaccine suppressed the immune response, whereas three doses of irradiated TC-1/cGM-CSF vaccine elicited a greater immune response and subsequent tumor suppression.
Collapse
|
10
|
Chen YP, Liu YW, Lee D, Qiu JT, Lee TY, Liu SJ. Biodegradable andrographolide-eluting nanofibrous membranes for the treatment of cervical cancer. Int J Nanomedicine 2019; 14:421-429. [PMID: 30666104 PMCID: PMC6331077 DOI: 10.2147/ijn.s186714] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND In this study, we developed biodegradable andrographolide (AG)-eluting nanofibrous mats and evaluated their efficacy in treating cervical cancer. MATERIALS AND METHODS Membranes of two different poly[(d,l)-lactide-co-glycolide] (PLGA)-to-AG ratios (6:1 and 3:1) were prepared via electrospinning technology. The liberation behavior of AG was evaluated. A cervical cancer model with C57BL/6J mice was created and employed for an in vivo efficacy assessment of the drug-eluting nanofibers. Twelve mice with cervical cancer were stochastically divided into three different groups (four animals per group): group A received no treatment as the control, group B was treated with pure PLGA mats, and group C was treated with AG-loaded nanofibrous membranes. The changes in tumor sizes were recorded. RESULTS All membranes eluted high concentrations of AG at the target area for three weeks, while the systemic drug concentration in the blood remained low. Histological analysis showed no obvious tissue inflammation. Compared with the mice in groups A and B, the tumor size of the mice in group C decreased with time until day 25, when the daily drug concentration reduced to 3 µg/mL. CONCLUSION Biodegradable nanofibers with a sustainable release of AG exhibit adequate efficacy and durability for the treatment of mice with cervical cancer.
Collapse
Affiliation(s)
- Yi-Pin Chen
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yen-Wei Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan,
| | - Demei Lee
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan,
| | - Jiantai Timothy Qiu
- Department of Obstetrics and Gynecology, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Traditional Chinese Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan,
- Department of Orthopedic Surgery, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan,
| |
Collapse
|
11
|
Wu J, Zhao C, Liu Q, Huang W, Wang Y. Development and application of a bioluminescent imaging mouse model for Chikungunya virus based on pseudovirus system. Vaccine 2017; 35:6387-6394. [PMID: 29031692 DOI: 10.1016/j.vaccine.2017.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/09/2017] [Accepted: 10/03/2017] [Indexed: 01/28/2023]
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne virus that is transmitted to humans primarily via the bite of an infected mosquito. Infection of humans by CHIKV can cause chikungunya fever which is an acute febrile illness associated with severe, often debilitating polyarthralgias. Since a re-emergence of CHIKV in 2004, the virus has spread into novel locations in nearly 40 countries including non-endemic regions and has led to millions of cases of disease throughout countries. Handling of CHIKV is restricted to the high-containment Biosafety Level 3 (BSL-3) facilities, which greatly impede the research progress of this virus. In this study, an envelope-pseudotyped virus expressing the firefly luciferase reporter protein (pHIV-CHIKV-Fluc) was generated. An in vitro sensitive neutralizing assay and an in vivo bioluminescent-imaging-based mouse infection model had been developed based on the CHIKV pseudovirus. Utilizing the platform, protection effect of DNA vaccine was evaluated. Therefore, this study provides a safe, sensitive and visualizing model for evaluating vaccines and antiviral therapies against CHIKV in low containment BSL-2 laboratories.
Collapse
Affiliation(s)
- Jiajing Wu
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Qiang Liu
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China.
| |
Collapse
|
12
|
Ruan J, Duan Y, Li F, Wang Z. Enhanced synergistic anti-Lewis lung carcinoma effect of a DNA vaccine harboring a MUC1-VEGFR2 fusion gene used with GM-CSF as an adjuvant. Clin Exp Pharmacol Physiol 2016; 44:71-78. [PMID: 27562635 DOI: 10.1111/1440-1681.12654] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/05/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Junzhong Ruan
- Department of Thoracic Surgery; Beijing Chest Hospital; Capital Medical University; Beijing Tuberculosis and Thoracic Tumour Research Institute; Beijing China
| | - Yong Duan
- Department of Thoracic Surgery; Beijing Chest Hospital; Capital Medical University; Beijing Tuberculosis and Thoracic Tumour Research Institute; Beijing China
| | - Fugen Li
- Department of Thoracic Surgery; Beijing Chest Hospital; Capital Medical University; Beijing Tuberculosis and Thoracic Tumour Research Institute; Beijing China
| | - Zitong Wang
- Department of Thoracic Surgery; Beijing Chest Hospital; Capital Medical University; Beijing Tuberculosis and Thoracic Tumour Research Institute; Beijing China
| |
Collapse
|
13
|
Yu TW, Chueh HY, Tsai CC, Lin CT, Qiu JT. Novel GM-CSF-based vaccines: One small step in GM-CSF gene optimization, one giant leap for human vaccines. Hum Vaccin Immunother 2016; 12:3020-3028. [PMID: 27560197 DOI: 10.1080/21645515.2016.1221551] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Granulocyte macrophage-colony stimulating factor (GM-CSF) is a potent immunomodulatory cytokine that is known to facilitate vaccine efficacy by promoting the development and prolongation of both humoral and cellular immunity. In the past years we have generated a novel codon-optimized GM-CSF gene as an adjuvant. The codon-optimized GM-CSF gene significantly increased protein expression levels in all cells tested and helped in generating a strong immune responses against HIV-1 Gag and HPV-associated cancer. Here, we review the literature dealing with the adjuvant activity of GM-CSF both in animal models and clinical trials. We anticipate that the codon-optimized GM-CSF gene offers a practical molecular strategy for potentiating immune responses to tumor cell-based vaccinations as well as other immunotherapeutic strategies.
Collapse
Affiliation(s)
- Ting-Wei Yu
- a School of Medicine , Chang Gung University , Taoyuan , Taiwan , ROC.,b Department of Obstetrics and Gynecology , Chang Gung Memorial Hospital , Taoyuan , Taiwan , ROC
| | - Ho-Yen Chueh
- a School of Medicine , Chang Gung University , Taoyuan , Taiwan , ROC.,b Department of Obstetrics and Gynecology , Chang Gung Memorial Hospital , Taoyuan , Taiwan , ROC
| | - Ching-Chou Tsai
- a School of Medicine , Chang Gung University , Taoyuan , Taiwan , ROC.,c Department of Obstetrics and Gynecology , Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine , Kaohsiung , Taiwan , ROC
| | - Cheng-Tao Lin
- a School of Medicine , Chang Gung University , Taoyuan , Taiwan , ROC.,b Department of Obstetrics and Gynecology , Chang Gung Memorial Hospital , Taoyuan , Taiwan , ROC
| | - Jiantai Timothy Qiu
- a School of Medicine , Chang Gung University , Taoyuan , Taiwan , ROC.,b Department of Obstetrics and Gynecology , Chang Gung Memorial Hospital , Taoyuan , Taiwan , ROC.,d Department of Biomedical Sciences , School of Medicine, Chang Gung University , Taoyuan , Taiwan , ROC
| |
Collapse
|