1
|
Chen CH, Weng TH, Huang HH, Huang LY, Huang KY, Chen PR, Yeh KY, Huang CT, Chien YT, Chuang PY, Lin YL, Tsai NM, Liu SJ, Su YC, Weng SL, Liao KW. A flexible liposomal polymer complex as a platform of specific and regulable immune regulation for individual cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:29. [PMID: 36691089 PMCID: PMC9869520 DOI: 10.1186/s13046-023-02601-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND The applicability and therapeutic efficacy of specific personalized immunotherapy for cancer patients is limited by the genetic diversity of the host or the tumor. Side-effects such as immune-related adverse events (IRAEs) derived from the administration of immunotherapy have also been observed. Therefore, regulatory immunotherapy is required for cancer patients and should be developed. METHODS The cationic lipo-PEG-PEI complex (LPPC) can stably and irreplaceably adsorb various proteins on its surface without covalent linkage, and the bound proteins maintain their original functions. In this study, LPPC was developed as an immunoregulatory platform for personalized immunotherapy for tumors to address the barriers related to the heterogenetic characteristics of MHC molecules or tumor associated antigens (TAAs) in the patient population. Here, the immune-suppressive and highly metastatic melanoma, B16F10 cells were used to examine the effects of this platform. Adsorption of anti-CD3 antibodies, HLA-A2/peptide, or dendritic cells' membrane proteins (MP) could flexibly provide pan-T-cell responses, specific Th1 responses, or specific Th1 and Th2 responses, depending on the host needs. Furthermore, with regulatory antibodies, the immuno-LPPC complex properly mediated immune responses by adsorbing positive or negative antibodies, such as anti-CD28 or anti-CTLA4 antibodies. RESULTS The results clearly showed that treatment with LPPC/MP/CD28 complexes activated specific Th1 and Th2 responses, including cytokine release, CTL and prevented T-cell apoptosis. Moreover, LPPC/MP/CD28 complexes could eliminate metastatic B16F10 melanoma cells in the lung more efficiently than LPPC/MP. Interestingly, the melanoma resistance of mice treated with LPPC/MP/CD28 complexes would be reversed to susceptible after administration with LPPC/MP/CTLA4 complexes. NGS data revealed that LPPC/MP/CD28 complexes could enhance the gene expression of cytokine and chemokine pathways to strengthen immune activation than LPPC/MP, and that LPPC/MP/CTLA4 could abolish the LPPC/MP complex-mediated gene expression back to un-treatment. CONCLUSIONS Overall, we proved a convenient and flexible immunotherapy platform for developing personalized cancer therapy.
Collapse
Affiliation(s)
- Chia-Hung Chen
- grid.413593.90000 0004 0573 007XDepartment of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan
| | - Tzu-Han Weng
- grid.413593.90000 0004 0573 007XDepartment of Dermatology, MacKay Memorial Hospital, Taipei City, 10449 Taiwan
| | - Hsiao-Hsuan Huang
- grid.260539.b0000 0001 2059 7017Industrial Development Graduate Program of College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Ling-Ya Huang
- grid.260539.b0000 0001 2059 7017Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Kai-Yao Huang
- grid.413593.90000 0004 0573 007XDepartment of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan ,grid.452449.a0000 0004 1762 5613Department of Medicine, MacKay Medical College, 25245 New Taipei City, Taiwan
| | - Pin-Rong Chen
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Kuang-Yu Yeh
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Chi-Ting Huang
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Yu-Tzu Chien
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Po-Ya Chuang
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Yu-Ling Lin
- grid.28665.3f0000 0001 2287 1366Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 11529 Taiwan
| | - Nu-Man Tsai
- grid.411641.70000 0004 0532 2041Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, 40201 Taiwan ,grid.411645.30000 0004 0638 9256Department of Pathology and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201 Taiwan
| | - Shih-Jen Liu
- grid.59784.370000000406229172National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 350401 Miaoli, Taiwan
| | - Yu-Cheng Su
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Shun-Long Weng
- grid.452449.a0000 0004 1762 5613Department of Medicine, MacKay Medical College, 25245 New Taipei City, Taiwan ,grid.413593.90000 0004 0573 007XDepartment of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan ,grid.507991.30000 0004 0639 3191MacKay Junior College of Medicine, Nursing and Management, Taipei City, 11260 Taiwan
| | - Kuang-Wen Liao
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan ,grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan ,grid.412019.f0000 0000 9476 5696Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708 Taiwan ,grid.412019.f0000 0000 9476 5696College of Dental Medicine, Kaohsiung Medical University School of Dentistry, Kaohsiung City, 80708 Taiwan ,grid.64523.360000 0004 0532 3255Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City, 70101 Taiwan ,grid.260539.b0000 0001 2059 7017Center for Intelligent Drug Systems and Smart Bio-Devices, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| |
Collapse
|
2
|
Chen CH, Weng TH, Chuang CH, Huang KY, Huang SC, Chen PR, Huang HH, Huang LY, Shen PC, Chuang PY, Huang HY, Wu YS, Chang HC, Weng SL, Liao KW. Transdermal nanolipoplex simultaneously inhibits subcutaneous melanoma growth and suppresses systemically metastatic melanoma by activating host immunity. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 47:102628. [PMID: 36400317 DOI: 10.1016/j.nano.2022.102628] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/22/2022] [Accepted: 11/06/2022] [Indexed: 11/17/2022]
Abstract
Benefit for clinical melanoma treatments, the transdermal neoadjuvant therapy could reduce surgery region and increase immunotherapy efficacy. Using lipoplex (Lipo-PEG-PEI-complex, LPPC) encapsulated doxorubicin (DOX) and carrying CpG oligodeoxynucleotide; the transdermally administered nano-liposomal drug complex (LPPC-DOX-CpG) would have high cytotoxicity and immunostimulatory activity to suppress systemic metastasis of melanoma. LPPC-DOX-CpG dramatically suppressed subcutaneous melanoma growth by inducing tumor cell apoptosis and recruiting immune cells into the tumor area. Animal studies further showed that the colonization and growth of spontaneously metastatic melanoma cells in the liver and lung were suppressed by transdermal LPPC-DOX-CpG. Furthermore, NGS analysis revealed IFN-γ and NF-κB pathways were triggered to recruit and activate the antigen-presenting-cells and effecter cells, which could activate the anti-tumor responses as the major mechanism responsible for the therapeutic effect of LPPC-DOX-CpG. Finally, we have successfully proved transdermal LPPC-DOX-CpG as a promising penetrative carrier to activate systemic anti-tumor immunity against subcutaneous and metastatic tumor.
Collapse
Affiliation(s)
- Chia-Hung Chen
- Department of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City 30071, Taiwan, ROC
| | - Tzu-Han Weng
- Dependent of Medical Education, MacKay Memorial Hospital, Taipei 10449, Taiwan, ROC
| | - Cheng-Hsun Chuang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Kai-Yao Huang
- Department of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City 30071, Taiwan, ROC; Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan, ROC
| | - Sih-Cheng Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Pin-Rong Chen
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Hsiao-Hsuan Huang
- Industrial Development Graduate Program of College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Ling-Ya Huang
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Pei-Chun Shen
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Po-Ya Chuang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Hsiao-Yen Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Yi-Syuan Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Hao-Chiun Chang
- Ph.D. Degree Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC
| | - Shun-Long Weng
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan, ROC; Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City 30071, Taiwan, ROC; MacKay Junior College of Medicine, Nursing and Management, Taipei City 11260, Taiwan, ROC.
| | - Kuang-Wen Liao
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC; Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC; Drug Development and Value Creation Research Center, College of Dental Medicine, Kaohsiung Medical University School of Dentistry, Graduate Institute of Medicine, College of Medicine, Center for Cancer Research, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan, ROC; Center for Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC; Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City 70101, Taiwan, ROC; Ph.D. Degree Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu City 30068, Taiwan, ROC.
| |
Collapse
|
3
|
Wu Y, Jiang L, Dong Z, Chen S, Yu XY, Tang S. Intracellular Delivery of Proteins into Living Cells by Low-Molecular-Weight Polyethyleneimine. Int J Nanomedicine 2021; 16:4197-4208. [PMID: 34188469 PMCID: PMC8232877 DOI: 10.2147/ijn.s315444] [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: 04/12/2021] [Accepted: 06/01/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Intracellular protein delivery is emerging as a potential strategy to revolutionize therapeutics in the field of biomedicine, aiming at treating a wide range of diseases including cancer, inflammatory diseases and other oxidative stress-related disorders with high specificity. However, the current challenges and limitations are addressed to either synthetically or biologically through multipotency of engineering, such as protein modification, insufficient delivery of large-size proteins, deficiency or mutation of proteins, and high cytotoxicity. Methods We prepared the nanocomposites by mixing protein with PEI1200 at a certain molar ratio and demonstrated that it can deliver proteins into living cells in high efficiency and safety through the following experiments, such as dynamic light scattering, fluorescent detection, agarose gel electrophoresis, ß-Galactosidase activity detection, immunofluorescence staining, digital fluorescent detection, cell viability assay and flow cytometry. Results The self-assembly of PEI1200/protein nanocomposites with appropriate molar ratio (4:1 and 8:1) could provide efficiently delivery of active proteins to a variety of cell types in the presence of serum. The nanocomposites could continuously release protein up to 96 h in their desired intracellular locations. In addition, these nanocomposites were able to preserve protein activity while maintain low cytotoxicity (when final concentration <1 μg/mL). Conclusion Collectively, PEI1200-based delivery system provided an alternative strategy to direct protein delivery in high efficiency and safety, offering increased potential applications in clinical biomedicine.
Collapse
Affiliation(s)
- Yueheng Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Institute of Medical Instruments, Biomedical Engineering Institute, Jinan University, Guangzhou, 510632, People's Republic of China.,Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510080, People's Republic of China
| | - Lin Jiang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510080, People's Republic of China
| | - Zixuan Dong
- National Engineering Research Center for Healthcare Devices, Guangdong Institute of Medical Instruments, Biomedical Engineering Institute, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Shaoxian Chen
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510080, People's Republic of China
| | - Xi-Yong Yu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510080, People's Republic of China
| | - Shunqing Tang
- National Engineering Research Center for Healthcare Devices, Guangdong Institute of Medical Instruments, Biomedical Engineering Institute, Jinan University, Guangzhou, 510632, People's Republic of China
| |
Collapse
|
4
|
Positively Charged Nanoparticle Delivery of n-Butylidenephthalide Enhances Antitumor Effect in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8817875. [PMID: 33791383 PMCID: PMC7997748 DOI: 10.1155/2021/8817875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/28/2021] [Accepted: 03/06/2021] [Indexed: 01/04/2023]
Abstract
Hepatocellular carcinoma (HCC) is the second and sixth leading cause of cancer death in men and woman in 185 countries statistics, respectively. n-Butylidenephthalide (BP) has shown anti-HCC activity, but it also has an unstable structure that decreases its potential antitumor activity. The aim of this study was to investigate the cell uptake, activity protection, and antitumor mechanism of BP encapsulated in the novel liposome LPPC in HCC cells. BP/LPPC exhibited higher cell uptake and cytotoxicity than BP alone, and combined with clinical drug etoposide (VP-16), BP/LPPC showed a synergistic effect against HCC cells. Additionally, BP/LPPC increased cell cycle regulators (p53, p-p53, and p21) and decreased cell cycle-related proteins (Rb, p-Rb, CDK4, and cyclin D1), leading to cell cycle arrest at the G0/G1 phase in HCC cells. BP/LPPC induced cell apoptosis through activation of both the extrinsic (Fas-L and Caspase-8) and intrinsic (Bax and Caspase-9) apoptosis pathways and activated the caspase cascade to trigger HCC cell death. In conclusion, the LPPC complex improved the antitumor activity of BP in terms of cytotoxicity, cell cycle regulation and cell apoptosis, and BP/LPPC synergistically inhibited cell growth during combination treatment with VP-16 in HCC cells. Therefore, BP/LPPC is potentially a good candidate for clinical drug development or for use as an adjuvant for clinical drugs as a combination therapy for hepatocellular carcinoma.
Collapse
|
5
|
Huang XF, Chang KF, Lin YL, Liao KW, Hsiao CY, Sheu GT, Tsai NM. Enhancement of cytotoxicity and induction of apoptosis by cationic nano-liposome formulation of n-butylidenephthalide in breast cancer cells. Int J Med Sci 2021; 18:2930-2942. [PMID: 34220320 PMCID: PMC8241786 DOI: 10.7150/ijms.51439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 05/26/2021] [Indexed: 12/09/2022] Open
Abstract
Breast cancer is the second most common malignancy in women. Current clinical therapy for breast cancer has many disadvantages, including metastasis, recurrence, and poor quality of life. Furthermore, it is necessary to find a new therapeutic drug for breast cancer patients to meet clinical demand. n-Butylidenephthalide (BP) is a natural and hydrophobic compound that can inhibit several tumors. However, BP is unstable in aqueous or protein-rich environments, which reduces the activity of BP. Therefore, we used an LPPC (Lipo-PEG-PEI complex) that can encapsulate both hydrophobic and hydrophilic compounds to improve the limitation of BP. The purpose of this study is to investigate the anti-tumor mechanisms of BP and BP/LPPC and further test the efficacy of BP encapsulated by LPPC on SK-BR-3 cells. BP inhibited breast cancer cell growth, and LPPC encapsulation (BP/LPPC complex) enhanced the cytotoxicity on breast cancer by stabilizing the BP activity and offering endocytic pathways. Additionally, BP and LPPC-encapsulated BP induced cell cycle arrest at the G0/G1 phase and might trigger both extrinsic as well as intrinsic cell apoptosis pathway, resulting in cell death. Moreover, the BP/LPPC complex had a synergistic effect with doxorubicin of enhancing the inhibitory effect on breast cancer cells. Consequently, LPPC-encapsulated BP could improve the anti-cancer effects on breast cancer in vitro. In conclusion, BP exhibited an anti-cancer effect on breast cancer cells, and LPPC encapsulation efficiently improved the cytotoxicity of BP via an acceleration of entrapment efficiency to induce cell cycle block and apoptosis. Furthermore, BP/LPPC exhibited a synergistic effect in combination with doxorubicin.
Collapse
Affiliation(s)
- Xiao-Fan Huang
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - Kai-Fu Chang
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - Yu-Ling Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 11529, Taiwan, ROC
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 30010, Taiwan, ROC.,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30010, Taiwan, ROC
| | - Chih-Yen Hsiao
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, 60002, Taiwan, ROC.,Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan, ROC
| | - Gwo-Tarng Sheu
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| |
Collapse
|
6
|
Huang XF, Chen PT, Lin YL, Lee MS, Chang KF, Liao KW, Sheu GT, Hsieh MC, Tsai NM. Enhanced anticancer activity and endocytic mechanisms by polymeric nanocarriers of n-butylidenephthalide in leukemia cells. Clin Transl Oncol 2020; 23:1142-1151. [PMID: 32989675 DOI: 10.1007/s12094-020-02500-w] [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: 07/06/2020] [Accepted: 09/15/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE The purpose of this study was to investigate the antitumor mechanisms of n-butylidenephthalide (BP) and to further examine the delivery efficacy of polycationic liposome containing PEI and polyethylene glycol complex (LPPC)-encapsulated BP in leukemia cells. METHODS MTS, flow cytometric and TUNEL assays were performed to assess cell viability and apoptosis. BP and BP/LPPC complex delivery efficiency was analyzed by full-wavelength fluorescent scanner and fluorescence microscope. The expressions of cell cycle- and apoptosis-related proteins were conducted by Western blotting. RESULTS The results showed that BP inhibited leukemia cell growth by inducing cell cycle arrest and cell apoptosis. LPPC-encapsulated BP rapidly induced endocytic pathway activation, resulting in the internalization of BP into leukemia cells, causing cell apoptosis within 1 h. CONCLUSIONS LPPC encapsulation enhanced the cytotoxic activity of BP and did not influence the effects of BP induction that suggested LPPC-encapsulated BP might be developed as anti-leukemia drugs in future.
Collapse
Affiliation(s)
- X-F Huang
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - P-T Chen
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| | - Y-L Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 11529, Taiwan, ROC
| | - M-S Lee
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| | - K-F Chang
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - K-W Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 30010, Taiwan, ROC.,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30010, Taiwan, ROC
| | - G-T Sheu
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - M-C Hsieh
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| | - N-M Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC. .,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC.
| |
Collapse
|
7
|
Chang KF, Chang JT, Huang XF, Lin YL, Liao KW, Huang CW, Tsai NM. Antitumor Effects of N-Butylidenephthalide Encapsulated in Lipopolyplexs in Colorectal Cancer Cells. Molecules 2020; 25:molecules25102394. [PMID: 32455622 PMCID: PMC7288114 DOI: 10.3390/molecules25102394] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 01/15/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common type of cancer and the second most common cause of cancer-related death in the world. N-Butylidenephthalide (BP), a natural compound, inhibits several cancers, such as hepatoma, brain tumor and colon cancer. However, due to the unstable structure, the activity of BP is quickly lost after dissolution in an aqueous solution. A polycationic liposomal polyethylenimine and polyethylene glycol complex (LPPC), a new drug carrier, encapsulates both hydrophobic and hydrophilic compounds, maintains the activity of the compound, and increases uptake of cancer cells. The purpose of this study is to investigate the antitumor effects and protection of BP encapsulated in LPPC in CRC cells. The LPPC encapsulation protected BP activity, increased the cytotoxicity of BP and enhanced cell uptake through clathrin-mediated endocytosis. Moreover, the BP/LPPC-regulated the expression of the p21 protein and cell cycle-related proteins (CDK4, Cyclin B1 and Cyclin D1), resulting in an increase in the population of cells in the G0/G1 and subG1 phases. BP/LPPC induced cell apoptosis by activating the extrinsic (Fas, Fas-L and Caspase-8) and intrinsic (Bax and Caspase-9) apoptosis pathways. Additionally, BP/LPPC combined with 5-FU synergistically inhibited the growth of HT-29 cells. In conclusion, LPPC enhanced the antitumor activity and cellular uptake of BP, and the BP/LPPC complex induced cell cycle arrest and apoptosis, thereby causing death. These findings suggest the putative use of BP/LPPC as an adjuvant cytotoxic agent for colorectal cancer.
Collapse
Affiliation(s)
- Kai-Fu Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (K.-F.C.); (J.T.C.); (X.-F.H.)
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (K.-F.C.); (J.T.C.); (X.-F.H.)
| | - Xiao-Fan Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (K.-F.C.); (J.T.C.); (X.-F.H.)
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Yu-Ling Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan;
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30068, Taiwan;
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu 30068, Taiwan
| | - Chien-Wei Huang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Correspondence: (C.-W.H.); (N.-M.T.)
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: (C.-W.H.); (N.-M.T.)
| |
Collapse
|
8
|
Ho SY, Chen PR, Chen CH, Tsai NM, Lin YH, Lin CS, Chuang CH, Huang XF, Chan YL, Liu YK, Chung CH, Weng SL, Liao KW. Lipoplex-based targeted gene therapy for the suppression of tumours with VEGFR expression by producing anti-angiogenic molecules. J Nanobiotechnology 2020; 18:58. [PMID: 32272948 PMCID: PMC7144055 DOI: 10.1186/s12951-020-00610-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/16/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The anti-angiogenic fusion protein RBDV-IgG1 Fc (RBDV), which comprises the receptor-binding domain of vascular endothelial growth factor-A (VEGF-A), has shown antitumour effects by reducing angiogenesis in vivo. This study used the cationic lipoplex lipo-PEG-PEI-complex (LPPC) to simultaneously encapsulate both the RBDV targeting protein and the RBDV plasmid (pRBDV) without covalent bonds to assess VEGFR targeting gene therapy in mice with melanoma in vivo. RESULTS LPPC protected the therapeutic transgene from degradation by DNase, and the LPPC/RBDV complexes could specifically target VEGFR-positive B16-F10 cells both in vitro and in vivo. With or without RBDV protein-targeting direction, the pRBDV-expressing RBDV proteins were expressed and reached a maximal concentration on the 7th day in the sera after transfection in vivo and significantly elicited growth suppression against B16-F10 melanoma but not IgG1 control proteins. In particular, LPPC/pRBDV/RBDV treatment with the targeting molecules dramatically inhibited B16-F10 tumour growth in vivo to provide better therapeutic efficacy than the treatments with gene therapy with IgG1 protein targeting or administration of a protein drug with RBDV. CONCLUSIONS The simultaneous combination of the LPPC complex with pRBDV gene therapy and RBDV protein targeting might be a potential tool to conveniently administer targeted gene therapy for cancer therapy.
Collapse
Affiliation(s)
- Shu-Yi Ho
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC
| | - Pin-Rong Chen
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC
| | - Chia-Hung Chen
- Department of Medical Research, Hsinchu Mackay Memorial Hospital, Hsinchu City, 30071, Taiwan, ROC
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, 40201, Taiwan, ROC.,Department of Pathology and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan, ROC
| | - Yu-Hsin Lin
- Ph.D. Program in Industrial Development of College of Biological Science and Technology, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC
| | - Chen-Si Lin
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei City, 10617, Taiwan, ROC.,Animal Cancer Center, College of Bioresources and Agriculture, National Taiwan University, Taipei City, 10617, Taiwan, ROC
| | - Cheng-Hsun Chuang
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC
| | - Xiao-Fan Huang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, 40201, Taiwan, ROC.,Institute of Medicine of Chung, Shan Medical University, Taichung City, 40201, Taiwan, ROC
| | - Yi-Lin Chan
- Department of Life Science, Chinese Culture University, Taipei City, 11114, Taiwan, ROC
| | - Yen-Ku Liu
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC
| | - Chen-Han Chung
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC.,Hank Clinic Orthopedics Surgery, Miaoli County, 35157, Taiwan, ROC
| | - Shun-Long Weng
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan, ROC. .,Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071, Taiwan, ROC.
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC. .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC. .,Ph.D. Program in Industrial Development of College of Biological Science and Technology, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan, ROC. .,Center for Intelligent Drug Systems and Smart Bio-Devices, National Chiao Tung University, Hsinchu City, 30068, Taiwan, ROC.
| |
Collapse
|
9
|
Lin YL, Huang XF, Chang KF, Liao KW, Tsai NM. Encapsulated n-Butylidenephthalide Efficiently Crosses the Blood-Brain Barrier and Suppresses Growth of Glioblastoma. Int J Nanomedicine 2020; 15:749-760. [PMID: 32099363 PMCID: PMC6999785 DOI: 10.2147/ijn.s235815] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/10/2020] [Indexed: 12/30/2022] Open
Abstract
Background n-Butylidenephthalide (BP) has anti-tumor effects on glioblastoma. However, the limitation of BP for clinical application is its unstable structure. A polycationic liposomal polyethylenimine (PEI) and polyethylene glycol (PEG) complex (LPPC) has been developed to encapsulate BP for drug structure protection. The purpose of this study was to investigate the anti-cancer effects of the BP/LPPC complex on glioblastoma in vitro and in vivo. Methods DBTRG-05MG tumor bearing xenograft mice were treated with BP and BP/LPPC and then their tumor sizes, survival, drug biodistribution were measured. RG2 tumor bearing F344 rats also treated with BP and BP/LPPC and then their tumor sizes by magnetic resonance imaging for evaluation blood–brain barrier (BBB) across and drug therapeutic effects. After treated with BP/LPPC in vitro, cell uptake, cell cycle and apoptotic regulators were analyzed for evaluation the therapeutic mechanism. Results In athymic mice, BP/LPPC could efficiently suppress tumor growth and prolong survival. In F334 rats, BP/LPPC crossed the BBB and led to tumor shrinkage. BP/LPPC promoted cell cycle arrest at the G0/G1 phase and triggered the extrinsic and intrinsic cell apoptosis pathways resulting cell death. BP/LPPC also efficiently suppressed VEGF, VEGFR1, VEGFR2, MMP2 and MMP9 expression. Conclusion BP/LPPC was rapidly and efficiently transported to the tumor area across the BBB and induced cell apoptosis, anti-angiogenetic and anti-metastatic effects in vitro and in vivo.
Collapse
Affiliation(s)
- Yu-Ling Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan, Republic of China
| | - Xiao-Fan Huang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, Republic of China.,Institute of Medicine of Chung Shun Medical University, Taichung 40201, Taiwan, Republic of China
| | - Kai-Fu Chang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, Republic of China.,Institute of Medicine of Chung Shun Medical University, Taichung 40201, Taiwan, Republic of China
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan, Republic of China.,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu 30010, Taiwan, Republic of China.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, Republic of China
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, Republic of China.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, Republic of China
| |
Collapse
|
10
|
Oliveira Pinho J, Matias M, Gaspar MM. Emergent Nanotechnological Strategies for Systemic Chemotherapy against Melanoma. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1455. [PMID: 31614947 PMCID: PMC6836019 DOI: 10.3390/nano9101455] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022]
Abstract
Melanoma is an aggressive form of skin cancer, being one of the deadliest cancers in the world. The current treatment options involve surgery, radiotherapy, targeted therapy, immunotherapy and the use of chemotherapeutic agents. Although the last approach is the most used, the high toxicity and the lack of efficacy in advanced stages of the disease have demanded the search for novel bioactive molecules and/or efficient drug delivery systems. The current review aims to discuss the most recent advances on the elucidation of potential targets for melanoma treatment, such as aquaporin-3 and tyrosinase. In addition, the role of nanotechnology as a valuable strategy to effectively deliver selective drugs is emphasized, either incorporating/encapsulating synthetic molecules or natural-derived compounds in lipid-based nanosystems such as liposomes. Nanoformulated compounds have been explored for their improved anticancer activity against melanoma and promising results have been obtained. Indeed, they displayed improved physicochemical properties and higher accumulation in tumoral tissues, which potentiated the efficacy of the compounds in pre-clinical experiments. Overall, these experiments opened new doors for the discovery and development of more effective drug formulations for melanoma treatment.
Collapse
Affiliation(s)
- Jacinta Oliveira Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Mariana Matias
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
11
|
Lin YL, Chen CH, Liu YK, Huang TH, Tsai NM, Tzou SC, Liao KW. Lipo-PEG-PEI complex as an intracellular transporter for protein therapeutics. Int J Nanomedicine 2019; 14:1119-1130. [PMID: 30863049 PMCID: PMC6391157 DOI: 10.2147/ijn.s188970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Protein or peptide drugs are emerging therapeutics for treating human diseases. However, current protein drugs are typically limited to acting on extracellular/cell membrane components associated with the diseases, while intracellular delivery of recombinant proteins replaces or replenishes faulty/missing proteins and remains inadequate. In this study, we developed a convenient and efficient intracellular protein delivery vehicle. Materials and methods A cationic liposomal polyethylenimine and polyethylene glycol complex (LPPC) was developed to noncovalently capture proteins for protein transfer into cells via endocytosis. β-glucuronidase (βG) was used in vitro and in vivo as a model enzyme to demonstrate the enzymatic activity of the intracellular transport of a protein. Results The endocytosed protein/LPPC complexes escaped from lysosomes, and the bound protein dissociated from LPPC in the cytosol. The enzymatic activity of βG was well preserved after intracellular delivery in vitro and in vivo. Conclusion Using LPPC as an intracellular protein transporter for protein therapeutics, we illustrated that LPPC may be an effective and convenient tool for studying diseases and developing therapeutics.
Collapse
Affiliation(s)
- Yu-Ling Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Chia-Hung Chen
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC, ;
| | - Yen-Ku Liu
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC, ;
| | - Tse-Hung Huang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan, ROC.,School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan, ROC.,Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan, ROC.,School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, ROC
| | - Nu-Man Tsai
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University.,Department of Pathology and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Shey-Cherng Tzou
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC, ; .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC, ;
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC, ; .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC, ; .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC, .,Center for Intelligent Drug Systems and Smart Bio-devices, National Chiao Tung University, Hsinchu, Taiwan, ROC,
| |
Collapse
|
12
|
Lin YL, Tsai NM, Chen CH, Liu YK, Lee CJ, Chan YL, Wang YS, Chang YC, Lin CH, Huang TH, Wang CC, Chi KH, Liao KW. Specific drug delivery efficiently induced human breast tumor regression using a lipoplex by non-covalent association with anti-tumor antibodies. J Nanobiotechnology 2019; 17:25. [PMID: 30728015 PMCID: PMC6364477 DOI: 10.1186/s12951-019-0457-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/14/2019] [Indexed: 11/18/2022] Open
Abstract
Background A cationic liposome-PEG-PEI complex (LPPC) was employed as a carrier for achieving targeted delivery of drug to human epidermal growth factor receptor-2 (HER2/neu)-expressing breast cancer cells. LPPC can be easily loaded with an anti-tumor drug and non-covalently associated with an anti-tumor antibody such as Herceptin that is clinically used to rapidly form immunoparticles within 1 h. Results Drug-loaded LPPC have an average size about 250 nm and a zeta potential of about 40 mV. Herceptin was complexed onto surface of the LPPC to form the drug/LPPC/Herceptin complexes. The size of curcumin/LPPC/Herceptin complexes were 280 nm and the zeta potentials were about 23 mV. Targeting ability of this delivery system was demonstrated through specific binding on surface of cells and IVIS images in vivo, which showed specific binding in HER2-positive SKBR3 cells as compared to HER2-negative Hs578T cells. Only the drug/LPPC/Herceptin complexes displayed dramatically increased the cytotoxic activity in cancer cells. Both in vitro and in vivo results indicated that Herceptin adsorbed on LPPC directed the immunocomplex towards HER2/neu-positive cells but not HER2/neu-negative cells. The complexes with either component (curcumin or doxorubicin) used in the LPPC-delivery system provided a better therapeutic efficacy compared to the drug treatment alone and other treatment groups, including clinical dosages of Herceptin and LipoDox, in a xenografted model. Conclusions LPPC displays important clinical implications by easily introducing a specific targeting characteristic to drugs utilized for breast cancer therapy. Electronic supplementary material The online version of this article (10.1186/s12951-019-0457-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu-Ling Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 11529, Taiwan, ROC
| | - Nu-Man Tsai
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Pathology and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| | - Chia-Hung Chen
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30068, Taiwan, ROC
| | - Yen-Ku Liu
- Department of Biological Science and Technology, National Chiao Tung University, No.75 Po-Ai Street, Hsinchu, 30068, Taiwan, ROC
| | - Chung-Jen Lee
- Department of Nursing, Tzu Chi College of Technology, Hualien, 97005, Taiwan, ROC
| | - Yi-Lin Chan
- Department of Life Science, Chinese Culture University, Taipei, 11114, Taiwan, ROC
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin-Kong Wu Ho-Su Memorial Hospital, No.95, Wenchang Rd., Shilin Dist., Taipei City, 11101, Taiwan, ROC
| | - Yuan-Ching Chang
- Department of Surgery, Mackay Memorial Hospital, Taipei, 10491, Taiwan, ROC
| | - Chi-Hsin Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei, 10491, Taiwan, ROC
| | - Tse-Hung Huang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, 20401, Taiwan, ROC.,School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, 33302, Taiwan, ROC.,School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, 11219, Taiwan, ROC
| | - Chao Ching Wang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, 20401, Taiwan, ROC
| | - Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin-Kong Wu Ho-Su Memorial Hospital, No.95, Wenchang Rd., Shilin Dist., Taipei City, 11101, Taiwan, ROC.
| | - Kuang-Wen Liao
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30068, Taiwan, ROC. .,Department of Biological Science and Technology, National Chiao Tung University, No.75 Po-Ai Street, Hsinchu, 30068, Taiwan, ROC. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan, ROC. .,Center for Intelligent Drug Systems and Smart Bio-devices, National Chiao Tung University, Hsinchu, 30068, Taiwan, ROC.
| |
Collapse
|
13
|
Antitumor Effect of n-Butylidenephthalide Encapsulated on B16/F10 Melanoma Cells In Vitro with a Polycationic Liposome Containing PEI and Polyethylene Glycol Complex. Molecules 2018; 23:molecules23123224. [PMID: 30563276 PMCID: PMC6321413 DOI: 10.3390/molecules23123224] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/01/2018] [Accepted: 12/05/2018] [Indexed: 11/29/2022] Open
Abstract
Advanced melanoma can metastasize to distal organs from the skin and yield an aggressive disease and poor prognosis even after treatment with chemotherapeutic agents. The compound n-Butylidenephthalide (BP) is isolated from Angelica sinensis, which is used to treat anemia and gynecological dysfunction in traditional Chinese medicine. Studies have indicated that BP can inhibit cancers, including brain, lung, prostate, liver, and colon cancers. However, because BP is a natural hydrophobic compound, it is quickly metabolized by the liver within 24 h, and thus has limited potential for development in cancer therapy. This study investigated the anticancer mechanisms of BP through encapsulation with a novel polycationic liposome containing polyethylenimine (PEI) and polyethylene glycol complex (LPPC) in melanoma cells. The results demonstrated that BP/LPPC had higher cytotoxicity than BP alone and induced cell cycle arrest at the G0/G1 phase in B16/F10 melanoma cells. The BP/LPPC-treated cell indicated an increase in subG1 percentage and TUNEL positive apoptotic morphology through induction of extrinsic and intrinsic apoptosis pathways. The combination of BP and LPPC and clinical drug 5-Fluorouracil had a greater synergistic inhibition effect than did a single drug. Moreover, LPPC encapsulation improved the uptake of BP values through enhancement of cell endocytosis and maintained BP cytotoxicity activity within 24 h. In conclusion, BP/LPPC can inhibit growth of melanoma cells and induce cell arrest and apoptosis, indicating that BP/LPPC has great potential for development of melanoma therapy agents.
Collapse
|
14
|
Ho SY, Chang BH, Chung CH, Lin YL, Chuang CH, Hsieh PJ, Huang WC, Tsai NM, Huang SC, Liu YK, Lo YC, Liao KW. Development of a computational promoter with highly efficient expression in tumors. BMC Cancer 2018; 18:480. [PMID: 29703163 PMCID: PMC5924487 DOI: 10.1186/s12885-018-4421-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/22/2018] [Indexed: 11/25/2022] Open
Abstract
Background Gene therapy is a potent method to increase the therapeutic efficacy against cancer. However, a gene that is specifically expressed in the tumor area has not been identified. In addition, nonspecific expression of therapeutic genes in normal tissues may cause side effects that can harm the patients’ health. Certain promoters have been reported to drive therapeutic gene expression specifically in cancer cells; however, low expression levels of the target gene are a problem for providing good therapeutic efficacy. Therefore, a specific and highly expressive promoter is needed for cancer gene therapy. Methods Bioinformatics approaches were utilized to analyze transcription factors (TFs) from high-throughput data. Reverse transcription polymerase chain reaction, western blotting and cell transfection were applied for the measurement of mRNA, protein expression and activity. C57BL/6JNarl mice were injected with pD5-hrGFP to evaluate the expression of TFs. Results We analyzed bioinformatics data and identified three TFs, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), cyclic AMP response element binding protein (CREB), and hypoxia-inducible factor-1α (HIF-1α), that are highly active in tumor cells. Here, we constructed a novel mini-promoter, D5, that is composed of the binding sites of the three TFs. The results show that the D5 promoter specifically drives therapeutic gene expression in tumor tissues and that the strength of the D5 promoter is directly proportional to tumor size. Conclusions Our results show that bioinformatics may be a good tool for the selection of appropriate TFs and for the design of specific mini-promoters to improve cancer gene therapy. Electronic supplementary material The online version of this article (10.1186/s12885-018-4421-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shu-Yi Ho
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China
| | - Bo-Hau Chang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China
| | - Chen-Han Chung
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30050, Taiwan, Republic of China
| | - Yu-Ling Lin
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China.,Center for Bioinformatics Research, National Chiao Tung University, Hsinchu, Taiwan, Republic of China
| | - Cheng-Hsun Chuang
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30050, Taiwan, Republic of China
| | - Pei-Jung Hsieh
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China
| | - Wei-Chih Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan, Republic of China
| | - Nu-Man Tsai
- School of Medical and Laboratory Biotechnology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Sheng-Chieh Huang
- Department of Surgery, National Yang Ming University, Taipei, Taiwan, Republic of China.,Division of Colon and Rectal surgery, Department of surgery, Taipei Veteran General Hospital, Taipei, Taiwan, Republic of China
| | - Yen-Ku Liu
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30050, Taiwan, Republic of China
| | - Yu-Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China. .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, 30050, Taiwan, Republic of China. .,College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China.
| |
Collapse
|
15
|
Lin YL, Chen CH, Wu HY, Tsai NM, Jian TY, Chang YC, Lin CH, Wu CH, Hsu FT, Leung TK, Liao KW. Inhibition of breast cancer with transdermal tamoxifen-encapsulated lipoplex. J Nanobiotechnology 2016; 14:11. [PMID: 26892504 PMCID: PMC4759757 DOI: 10.1186/s12951-016-0163-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/08/2016] [Indexed: 12/21/2022] Open
Abstract
Background Tamoxifen is currently used for the treatment of both early and advanced estrogen receptor (ER) positive breast cancer in pre- and post-menopausal women. However, using tamoxifen routinely to inhibit endogenous or exogenous estrogen effects is occasionally difficult because of its potential side effects. Objectives The aim of this study is to design a local drug delivery system to encapsulate tamoxifen for observing their efficacy of skin penetration, drug accumulation and cancer therapy. Methods A cationic liposome-PEG-PEI complex (LPPC) was used as a carrier for the encapsulation of tamoxifen and forming ‘LPPC/TAM’ for transdermal release. The cytotoxicity of LPPC/TAM was analyzed by MTT. The skin penetration, tumor growth inhibition and organ damages were measured in xenograft mice following transdermal treatment. Results LPPC/TAM had an average size less than 270 nm and a zeta-potential of approximately 40 mV. LPPC/TAM displayed dramatically increased the cytotoxic activity in all breast cancer cells, especially in ER-positive breast cancer cells. In vivo, LPPC drug delivery helped the fluorescent dye penetrating across the skim and accumulating rapidly in tumor area.
Administration of LPPC/TAM by transdermal route inhibited about 86 % of tumor growth in mice bearing BT474 tumors. This local treatment of LPPC/TAM did not injury skin and any organs. Conclusion LPPC-delivery system provided a better skin penetration and drug accumulation and therapeutic efficacy. Therefore, LPPC/TAM drug delivery maybe a useful transdermal tool of drugs utilization for breast cancer therapy.
Collapse
Affiliation(s)
- Yu-Ling Lin
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC. .,Center for Bioinformatics Research, National Chiao Tung University, Hsinchu, Taiwan, ROC.
| | - Chia-Hung Chen
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC.
| | - Hsin-Yi Wu
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC.
| | - Nu-Man Tsai
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan, ROC. .,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC.
| | - Ting-Yan Jian
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC.
| | - Yuan-Ching Chang
- Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan, ROC.
| | - Chi-Hsin Lin
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan, ROC.
| | - Chih-Hsiung Wu
- Department of Surgery, En Chu Kong Hospital, New Taipei City, Taiwan, ROC.
| | - Fei-Ting Hsu
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan, ROC. .,Translational Imaging Research Center, Taipei Medical University, Taipei, Taiwan, ROC.
| | - Ting Kai Leung
- Department of Diagnostic Radiology, Taipei Medical University Hospital, Taipei, Taiwan, ROC. .,Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC. .,Department of Diagnostic Radiology, Taipei Hospital, Ministry of Health and Welfare, Taipei, Taiwan, ROC. .,College of Science and Engineering, Fu Jen Catholic University, Hsinchuang, Taiwan, ROC.
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC. .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC.
| |
Collapse
|
16
|
Lin YL, Chang KF, Huang XF, Hung CL, Chen SC, Chao WR, Liao KW, Tsai NM. Liposomal n-butylidenephthalide protects the drug from oxidation and enhances its antitumor effects in glioblastoma multiforme. Int J Nanomedicine 2015; 10:6009-20. [PMID: 26451107 PMCID: PMC4592058 DOI: 10.2147/ijn.s85790] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background The natural compound n-butylidenephthalide (BP) can pass through the blood–brain barrier to inhibit the growth of glioblastoma multiforme tumors. However, BP has an unstable structure that reduces its antitumor activity and half-life in vivo. Objective The aim of this study is to design a drug delivery system to encapsulate BP to enhance its efficacy by improving its protection and delivery. Methods To protect its structural stability against protein-rich and peroxide solutions, BP was encapsulated into a lipo-PEG-PEI complex (LPPC). Then, the cytotoxicity of BP/LPPC following preincubation in protein-rich, acid/alkaline, and peroxide solutions was analyzed by MTT. Cell uptake of BP/LPPC was also measured by confocal microscopy. The therapeutic effects of BP/LPPC were analyzed in xenograft mice following intratumoral and intravenous injections. Results When BP was encapsulated in LPPC, its cytotoxicity was maintained following preincubation in protein-rich, acid/alkaline, and peroxide solutions. The cytotoxic activity of encapsulated BP was higher than that of free BP (~4.5- to 8.5-fold). This increased cytotoxic activity of BP/LPPC is attributable to its rapid transport across the cell membrane. In an animal study, a subcutaneously xenografted glioblastoma multiforme mouse that was treated with BP by intratumoral and intravenous administration showed inhibited tumor growth. The same dose of BP/LPPC was significantly more effective in terms of tumor inhibition. Conclusion LPPC encapsulation technology is able to protect BP’s structural stability and enhance its antitumor effects, thus providing a better tool for use in cancer therapy.
Collapse
Affiliation(s)
- Yu-Ling Lin
- College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan ; Center for Bioinformatics Research, National Chiao Tung University, Hsinchu, Taiwan
| | - Kai-Fu Chang
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Xiao-Fan Huang
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Che-Lun Hung
- Department of Computer Science and Communication Engineering, Providence University, Taichung, Taiwan
| | - Shyh-Chang Chen
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Chung Shan Medical University, Taichung, Taiwan
| | - Wan-Ru Chao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan ; Department of Pathology, Chung Shan Medical University and Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kuang-Wen Liao
- College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan ; Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Nu-Man Tsai
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan ; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
17
|
Zhu QL, Zhou Y, Guan M, Zhou XF, Yang SD, Liu Y, Chen WL, Zhang CG, Yuan ZQ, Liu C, Zhu AJ, Zhang XN. Low-density lipoprotein-coupled N-succinyl chitosan nanoparticles co-delivering siRNA and doxorubicin for hepatocyte-targeted therapy. Biomaterials 2014; 35:5965-76. [DOI: 10.1016/j.biomaterials.2014.03.088] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 03/31/2014] [Indexed: 12/25/2022]
|
18
|
Chen CH, Lin YL, Liu YK, He PJ, Lin CM, Chiu YH, Wu CJ, Cheng TL, Liu SJ, Liao KW. Liposome-based polymer complex as a novel adjuvant: enhancement of specific antibody production and isotype switch. Int J Nanomedicine 2012; 7:607-21. [PMID: 22346354 PMCID: PMC3277439 DOI: 10.2147/ijn.s28097] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of vaccination is to induce appropriate immunity against pathogens. Antibody-mediated immunity is critical for protection against many virus diseases, although it is becoming more evident that coordinated, multifunctional immune responses lead to the most effective defense. Specific antibody (Ab) isotypes are more efficient at protecting against pathogen invasion in different locations in the body. For example, compared to other Ab isotypes, immunoglobulin (Ig) A provides more protection at mucosal areas. In this study, we developed a cationic lipopolymer (liposome-polyethylene glycol-polyethyleneimine complex [LPPC]) adjuvant that strongly adsorbs antigens or immunomodulators onto its surface to enhance or switch immune responses. The results demonstrate that LPPC enhances uptake ability, surface marker expression, proinflammatory cytokine release, and antigen presentation in mouse phagocytes. In contrast to Freund’s adjuvant, LPPC preferentially activates Th1- immunity against antigens in vivo. With lipopolysaccharides or CpG oligodeoxynucleotides, LPPC dramatically enhances the IgA or IgG2A proportion of total Ig, even in hosts that have developed Th2 immunities and high IgG1 serum titers. Taken together, the results demonstrate that the LPPC adjuvant not only increases the immunogenicity of antigens but also modulates host immunity to produce an appropriate Ab isotype by combining with immunomodulators.
Collapse
Affiliation(s)
- Chia-Hung Chen
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lin YL, Liu YK, Tsai NM, Hsieh JH, Chen CH, Lin CM, Liao KW. A Lipo-PEG-PEI complex for encapsulating curcumin that enhances its antitumor effects on curcumin-sensitive and curcumin-resistance cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 8:318-27. [PMID: 21704596 DOI: 10.1016/j.nano.2011.06.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 04/19/2011] [Accepted: 06/06/2011] [Indexed: 10/18/2022]
Abstract
A cationic liposome-PEG-PEI complex (LPPC) was used as a carrier for the encapsulation of hydrophobic curcumin to give curcumin/LPPC. Curcumin/LPPC had an average size less than 270 nm and a zeta potential of approximately 40 mV. The LPPC encapsulation efficiency for curcumin was about 45%. The authors found it surprising that the cytotoxic activity of the curcumin/LPPC was fivefold higher than curcumin when tested on curcumin-sensitive cells and 20-fold more active against curcumin-resistant cells. Curcumin/LPPC treatment caused a cell cycle arrest at G2/M phase, which rapidly resulted in apoptosis. The increased cytotoxic activity of curcumin/LPPC is likely attributable to its rapid accumulation in the cell. In vivo, administration of curcumin/LPPC inhibited about 60 - 90% of tumor growth in mice bearing CT-26 or B16F10 cells. These results demonstrate LPPC encapsulation technology is able to enhance the effects of antitumor drugs. Use of this technology may provide a new tool for cancer therapy, especially for drug-resistant cancer. From the Clinical Editor: This team of investigators used a cationic liposome-PEG-PEI complex (LPPC) to encapsulate curcumin. The different delivery method resulted in the five-fold increase of cytotoxic activity against curcumin-sensitive cells and twenty-fold against curcumin-resistant cells.
Collapse
Affiliation(s)
- Yu-Ling Lin
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | | | | | | | | | | | | |
Collapse
|