1
|
Yang S, Lan J, Li Z, Li M, Wu Y, Sun L, Zhang T, Ding Y. Bufonis venenum extract loaded novel cholesterol-free liposome for the treatment of hepatocellular carcinoma. Front Pharmacol 2024; 15:1486742. [PMID: 39654615 PMCID: PMC11625546 DOI: 10.3389/fphar.2024.1486742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Background This study aims to improve the solubility and the toxicity of Bufonis venenum, and finally enhance the therapeutic outcomes of hepatocellular carcinoma (HCC). Methods The cholesterol-free liposomes simultaneously encapsulate bufadienolides and indolealkylamines (Non-Cholesterol-Bufonis Venenum Extract-Liposome, Non-Chol-BVE-LP) was prepared by the thin-film evaporation technique. In vitro, the cytotoxicity, cell apoptosis study, cellular uptake and hemolysis studies were evaluated in HepG2 cells. In vivo, the biodistribution and anti-tumor activity studies were conducted in BALB/C mice with HepG2 cells. Results The liposomes showed good size distribution, encapsulation efficiency drug loading capacity and slower drug release. Non-Chol-BVE-LP had higher cytotoxicity on HepG2 cells and induced more apoptosis on HepG2 Cells compared with BVE. In addition, the liposomes could accumulate in tumor by passive targeting, thus facilitating the anti-tumor effects. In vivo, Non-Chol-BVE-LP showed equivalent anti-tumor efficacy to the first-line anti-HCC drug sorafenib. Conclusion The study provided new ideas for the development and clinical application of Bufonis venenum related formulation and offered new drug for the treatment of HCC.
Collapse
Affiliation(s)
- Siqi Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ya Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liyan Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- National Innovation Platform for medical industry-education integration, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Silva MLS. Lectin-modified drug delivery systems - Recent applications in the oncology field. Int J Pharm 2024; 665:124685. [PMID: 39260750 DOI: 10.1016/j.ijpharm.2024.124685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Chemotherapy with cytotoxic drugs remains the core treatment for cancer but, due to the difficulty to find general and usable biochemical differences between cancer cells and normal cells, many of these drugs are associated with lack of specificity, resulting in side effects and collateral cytotoxicity that impair patients' adherence to therapy. Novel cancer treatments in which the cytotoxic effect is maximized while adverse effects are reduced can be implemented by developing targeted therapies that exploit the specific features of cancer cells, such as the typical expression of aberrant glycans. Modification of drug delivery systems with lectins is one of the strategies to implement targeted chemotherapies, as lectins are able to specifically recognize and bind to cancer-associated glycans expressed at the surface of cancer cells, guiding the drug treatment towards these cells and not affecting healthy ones. In this paper, recent advances on the development of lectin-modified drug delivery systems for targeted cancer treatments are thoroughly reviewed, with a focus on their properties and performance in diverse applications, as well as their main advantages and limitations. The synthesis and analytical characterization of the cited lectin-modified drug delivery systems is also briefly described. A comparison with free-drug treatments and with antibody-modified drug delivery systems is presented, emphasizing the advantages of lectin-modified drug delivery systems. Main constraints and potential challenges of lectin-modified drug delivery systems, including key difficulties for clinical translation of these systems, and the required developments in this area, are also signalled.
Collapse
Affiliation(s)
- Maria Luísa S Silva
- Centro de Estudos Globais, Universidade Aberta, Rua da Escola Politécnica 147, 1269-001 Lisboa, Portugal.
| |
Collapse
|
3
|
Riet K, Adegoke A, Mashele S, Sekhoacha M. Effective Use of Euphorbia milii DCM Root Extract Encapsulated by Thermosensitive Immunoliposomes for Targeted Drug Delivery in Prostate Cancer Cells. Curr Issues Mol Biol 2024; 46:12037-12060. [PMID: 39590308 PMCID: PMC11593239 DOI: 10.3390/cimb46110714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
The delivery of anticancer drugs using nanotechnology is a promising approach aimed at improving the therapeutic efficacy and reducing the toxicity of chemotherapeutic agents. Liposomes were prepared using HSPC: DSPE-PEG-2000: DSPE-PEG2000-maleimide in the ratio of 4:1:0.2 and conjugated with a PSA antibody. Euphorbia milii extract (EME), doxorubicin (Dox), and docetaxel (Doc) encapsulated in temperature-sensitive immunoliposomes were investigated for their activities against the prostate cancer LNCap and DU145 cell lines. Organic extracts of EME leaves, roots, and stems were screened against both cell lines, inhibiting more than 50% of cell culture at concentrations of 10 μg/mL. The immunoliposomes incorporating the EME and docetaxel were active against the LNCap cells when exposed to heat at 39-40 °C. The liposomes not exposed to heat were inactive against the LNCap cells. The developed heat-sensitive immunoliposomes used for the delivery of both the EME and chemotherapeutic agents was able to successfully release the entrapped contents upon heat exposure above the phase transition temperature of the liposome membrane. The heat-sensitive immunoliposomes conjugated with a PSA antibody encapsulated the extract successfully and showed better cell antiproliferation efficacy against the prostate cancer cell lines in the presence of heat.
Collapse
Affiliation(s)
- Keamogetswe Riet
- Department of Health Sciences, Central University of Technology, Bloemfontein 9300, South Africa; (K.R.); (S.M.)
| | - Ayodeji Adegoke
- Department of Pharmacology, University of the Free State, Bloemfontein 9300, South Africa;
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan 200005, Nigeria
| | - Samson Mashele
- Department of Health Sciences, Central University of Technology, Bloemfontein 9300, South Africa; (K.R.); (S.M.)
| | - Mamello Sekhoacha
- Department of Pharmacology, University of the Free State, Bloemfontein 9300, South Africa;
| |
Collapse
|
4
|
Tu H, Zhou X, Zhou H, Luo Z, Yan Y, Luo Z, Qi Q. Anti-tumor effect and mechanisms of Timosaponin AIII across diverse cancer progression. Biochem Pharmacol 2024; 228:116080. [PMID: 38402911 DOI: 10.1016/j.bcp.2024.116080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/29/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Timosaponin AIII (TAIII), a steroidal saponin derived from Anemarrhena asphodeloides Bunge, has gained attention for its versatile therapeutic properties. While well-established for its anti-inflammatory, antidepressant, and anticoagulant properties, emerging research highlights its potent anti-tumor capabilities. This review synthesizes recent findings on the intricate mechanisms and diverse functions of TAIII in cancer therapy, elucidating its impact on various tumor cells, encompassing the effects of TAIII on critical aspects of cancer progression, including metastasis, apoptosis, and autophagy. Additionally, the shared features of TAIII-induced anti-tumor activities, the factors contributing to the multifaceted anti-cancer activities of TAIII, and an exploration of the advantages and disadvantages associated with the regulation of multiple anti-tumor pathways by TAIII are discussed. Furthermore, the detailed regulation of signaling pathways is delineated and tailored to specific cancer types, providing a comprehensive overview of the potential development of TAIII as a promising anti-tumor agent.
Collapse
Affiliation(s)
- Hanyun Tu
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Department of Pharmacology, Jinan University, Guangzhou 510632, China
| | - Xiaofeng Zhou
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Department of Pharmacology, Jinan University, Guangzhou 510632, China
| | - Haixia Zhou
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Department of Pharmacology, Jinan University, Guangzhou 510632, China
| | - Zepeng Luo
- Neurosurgery, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou 423000, China
| | - Yu Yan
- Functional Experimental Teaching Center, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Zhongping Luo
- Neurosurgery, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou 423000, China.
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Department of Pharmacology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
5
|
Li S, Liu Y, Sui X, Zhuo Y, Siqi H, Sijia Z, Hui Z, Dihua L, Dapeng Z, Lei Y. Novel Tubeimoside I liposomal drug delivery system in combination with gemcitabine for the treatment of pancreatic cancer. Nanomedicine (Lond) 2024; 19:1977-1993. [PMID: 39225145 PMCID: PMC11485868 DOI: 10.1080/17435889.2024.2382076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Aim: To evaluate the anti-pancreatic cancer effect of novel Tubeimoside I multifunctional liposomes combined with gemcitabine.Methods: Liposomes were prepared through the thin film hydration method, with evaluations conducted on parameters including encapsulation efficiency (EE%), particle size, polydispersity index (PDI), zeta potential (ZP), storage stability, and release over a 7-day period. The cellular uptake rate, therapeutic efficacy in vitro and in vivo and the role of immune microenvironment modulation were evaluated.Results: The novel Tubeimoside I multifunctional liposomal exhibited good stability, significant anti-cancer activity, and immune microenvironment remodeling effects. Furthermore, it showed a safety profile.Conclusion: This study underscores the potential of Novel Tubeimoside I multifunctional liposomal as a promising treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Shuhui Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuansheng Liu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300000, China
| | - Xiaojun Sui
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
- Graduate School ofTianjin Medical University, Tianjin, 300270, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - He Siqi
- Graduate School ofTianjin Medical University, Tianjin, 300270, China
| | - Zhang Sijia
- Graduate School ofTianjin Medical University, Tianjin, 300270, China
| | - Zhang Hui
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Li Dihua
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Zhang Dapeng
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Yang Lei
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| |
Collapse
|
6
|
Ercelen S, Bulkurcuoglu B, Oksuz M, Nalbantsoy A, Sarikahya NB. Development and Characterization of Plant-derived Aristatoside C and Davisianoside B Saponin-loaded Phytosomes with Suppressed Hemolytic Activity. ChemistryOpen 2024; 13:e202300254. [PMID: 38466160 PMCID: PMC11633355 DOI: 10.1002/open.202300254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/20/2024] [Indexed: 03/12/2024] Open
Abstract
Saponins are glycosides widely distributed in the plant kingdom and have many pharmacological activities. However, their tendency to bind to cell membranes can cause cell rupture, limiting their clinical use. In the previous study, aristatoside C and davisianoside B were isolated from Cephalaria species. Cytotoxicity assays showed that they are more active on A-549 cell lines than doxorubicin but caused hemolysis. In the current research, aristatoside C and davisianoside B were loaded to phytosomes called ALPs and DLPs respectively, and characterized for particle size, zeta potential, encapsulation efficiency, release kinetic, hemolytic activity, and cytotoxicity on A-549 cell line. DLPs maintained the cytotoxic activity of the free saponins against A-549 cells with IC50 of 9,64±0,02 μg/ml but dramatically reduced their hemolytic activity. Furthermore, temperature and time-dependent stability studies based on the size and zeta potential of ALPs and DLPs revealed that the phytosomes have sustained release properties over 2 weeks. Overall, DLPs displayed cytotoxicity against A-549 cells with minimal hemolysis and sustained release, highlighting their potential as nanotherapeutics for clinical applications.
Collapse
Affiliation(s)
- Sebnem Ercelen
- Life SciencesBionanotechnology Lab.Scientific And Technological Research Council of Türkiye (TUBITAK)Marmara Research Center (MRC)Gebze41470Kocaeli/Türkiye
- Hamidiye Faculty of MedicineDepartment of BiophysicsUniversity of Health SciencesÜsküdar34668İstanbul/Türkiye
| | - Bunyamin Bulkurcuoglu
- Life SciencesBionanotechnology Lab.Scientific And Technological Research Council of Türkiye (TUBITAK)Marmara Research Center (MRC)Gebze41470Kocaeli/Türkiye
- Institute of BiotechnologyGebze Technical UniversityGebze41400Kocaeli/Türkiye
| | - Mustafa Oksuz
- Life SciencesBionanotechnology Lab.Scientific And Technological Research Council of Türkiye (TUBITAK)Marmara Research Center (MRC)Gebze41470Kocaeli/Türkiye
- Faculty of PharmacyBiochemistry DepartmentMersin UniversityYenişehir33160Mersin/Türkiye
| | - Ayse Nalbantsoy
- Faculty of EngineeringDepartment of BioengineeringEge UniversityBornova35040İzmir/Türkiye
| | - Nazli Boke Sarikahya
- Faculty of ScienceDepartment of ChemistryEge UniversityBornova35040İzmir/Türkiye
| |
Collapse
|
7
|
Zhang YB, Wang JF, Wang MX, Peng J, Kong XD, Tian J. Nano-based drug delivery systems for active ingredients from traditional Chinese medicine: Harnessing the power of nanotechnology. Front Pharmacol 2024; 15:1405252. [PMID: 38910887 PMCID: PMC11190311 DOI: 10.3389/fphar.2024.1405252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction: Traditional Chinese medicine (TCM) is gaining worldwide popularity as a complementary and alternative medicine. The isolation and characterization of active ingredients from TCM has become optional strategies for drug development. In order to overcome the inherent limitations of these natural products such as poor water solubility and low bioavailability, the combination of nanotechnology with TCM has been explored. Taking advantage of the benefits offered by the nanoscale, various drug delivery systems have been designed to enhance the efficacy of TCM in the treatment and prevention of diseases. Methods: The manuscript aims to present years of research dedicated to the application of nanotechnology in the field of TCM. Results: The manuscript discusses the formulation, characteristics and therapeutic effects of nano-TCM. Additionally, the formation of carrier-free nanomedicines through self-assembly between active ingredients of TCM is summarized. Finally, the paper discusses the safety behind the application of nano-TCM and proposes potential research directions. Discussion: Despite some achievements, the safety of nano-TCM still need special attention. Furthermore, exploring the substance basis of TCM formulas from the perspective of nanotechnology may provide direction for elucidating the scientific intension of TCM formulas.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Tian
- Department of Pharmacy, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
8
|
Wang F, Liang L, Yu M, Wang W, Badar IH, Bao Y, Zhu K, Li Y, Shafi S, Li D, Diao Y, Efferth T, Xue Z, Hua X. Advances in antitumor activity and mechanism of natural steroidal saponins: A review of advances, challenges, and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155432. [PMID: 38518645 DOI: 10.1016/j.phymed.2024.155432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Cancer, the second leading cause of death worldwide following cardiovascular diseases, presents a formidable challenge in clinical settings due to the extensive toxic side effects associated with primary chemotherapy drugs employed for cancer treatment. Furthermore, the emergence of drug resistance against specific chemotherapeutic agents has further complicated the situation. Consequently, there exists an urgent imperative to investigate novel anticancer drugs. Steroidal saponins, a class of natural compounds, have demonstrated notable antitumor efficacy. Nonetheless, their translation into clinical applications has remained unrealized thus far. In light of this, we conducted a comprehensive systematic review elucidating the antitumor activity, underlying mechanisms, and inherent limitations of steroidal saponins. Additionally, we propose a series of strategic approaches and recommendations to augment the antitumor potential of steroidal saponin compounds, thereby offering prospective insights for their eventual clinical implementation. PURPOSE This review summarizes steroidal saponins' antitumor activity, mechanisms, and limitations. METHODS The data included in this review are sourced from authoritative databases such as PubMed, Web of Science, ScienceDirect, and others. RESULTS A comprehensive summary of over 40 steroidal saponin compounds with proven antitumor activity, including their applicable tumor types and structural characteristics, has been compiled. These steroidal saponins can be primarily classified into five categories: spirostanol, isospirostanol, furostanol, steroidal alkaloids, and cholestanol. The isospirostanol and cholestanol saponins are found to have more potent antitumor activity. The primary antitumor mechanisms of these saponins include tumor cell apoptosis, autophagy induction, inhibition of tumor migration, overcoming drug resistance, and cell cycle arrest. However, steroidal saponins have limitations, such as higher cytotoxicity and lower bioavailability. Furthermore, strategies to address these drawbacks have been proposed. CONCLUSION In summary, isospirostanol and cholestanol steroidal saponins demonstrate notable antitumor activity and different structural categories of steroidal saponins exhibit variations in their antitumor signaling pathways. However, the clinical application of steroidal saponins in cancer treatment still faces limitations, and further research and development are necessary to advance their potential in tumor therapy.
Collapse
Affiliation(s)
- Fengge Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR, PR China
| | - Ma Yu
- School of Life Science and Engineering, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang, 621010, Sichuan, PR China
| | - Wenjie Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Iftikhar Hussain Badar
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, PR China; Department of Meat Science and Technology, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Kai Zhu
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yanlin Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Saba Shafi
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Dangdang Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yongchao Diao
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany.
| | - Zheyong Xue
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| | - Xin Hua
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| |
Collapse
|
9
|
Moghimipour E, Handali S. Functionalized liposomes as a potential drug delivery systems for colon cancer treatment: A systematic review. Int J Biol Macromol 2024; 269:132023. [PMID: 38697444 DOI: 10.1016/j.ijbiomac.2024.132023] [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: 07/14/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Colon cancer is one of the lethal diseases in the world with approximately 700,000 fatalities annually. Nowadays, due to the side effects of existing methods in the treatment of colon cancer such as radiotherapy and chemotherapy, the use of targeted nanocarriers in cancer treatment has received wide attention, and among them, especially liposomes have been studied a lot. Based on this, anti-tumor drugs hidden in targeted active liposomes can selectively act on cancer cells. In this systematic review, the use of various ligands such as folic acid, transferrin, aptamer, hyaluronic acid and cRGD for active targeting of liposomes to achieve improved drug delivery to colon cancer cells has been reviewed. The original articles published in English in the databases of Science Direct, PubMed and Google scholar from 2012 to 2022 were reviewed. From the total of 26,256 published articles, 19 studies met the inclusion criteria. The results of in vitro and in vivo studies have revealed that targeted liposomes lead to increasing the efficacy of anti-cancer agents on colon cancer cells with reducing side effects compared to free drugs and non-targeted liposomes. To the best of our knowledge, this is the first systematic review showing promising results for improvement treatment of colon cancer using targeted liposomes.
Collapse
Affiliation(s)
- Eskandar Moghimipour
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Marques AC, Costa PC, Velho S, Amaral MH. Analytical Techniques for Characterizing Tumor-Targeted Antibody-Functionalized Nanoparticles. Life (Basel) 2024; 14:489. [PMID: 38672759 PMCID: PMC11051252 DOI: 10.3390/life14040489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The specific interaction between cell surface receptors and corresponding antibodies has driven opportunities for developing targeted cancer therapies using nanoparticle systems. It is challenging to design and develop such targeted nanomedicines using antibody ligands, as the final nanoconjugate's specificity hinges on the cohesive functioning of its components. The multicomponent nature of antibody-conjugated nanoparticles also complicates the characterization process. Regardless of the type of nanoparticle, it is essential to perform physicochemical characterization to establish a solid foundation of knowledge and develop suitable preclinical studies. A meaningful physicochemical evaluation of antibody-conjugated nanoparticles should include determining the quantity and orientation of the antibodies, confirming the antibodies' integrity following attachment, and assessing the immunoreactivity of the obtained nanoconjugates. In this review, the authors describe the various techniques (electrophoresis, spectroscopy, colorimetric assays, immunoassays, etc.) used to analyze the physicochemical properties of nanoparticles functionalized with antibodies and discuss the main results.
Collapse
Affiliation(s)
- Ana Camila Marques
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paulo C. Costa
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Sérgia Velho
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Maria Helena Amaral
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
11
|
Shazleen Ibrahim I, Starlin Chellathurai M, Mahmood S, Hakim Azmi A, Harun N, Ulul Ilmie Ahmad Nazri M, Muzamir Mahat M, Mohamed Sofian Z. Engineered liposomes mediated approach for targeted colorectal cancer drug Delivery: A review. Int J Pharm 2024; 651:123735. [PMID: 38142874 DOI: 10.1016/j.ijpharm.2023.123735] [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: 08/18/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 12/26/2023]
Abstract
Colorectal cancer (CRC) continues to be one of the most prevalent and deadliest forms of cancer worldwide, despite notable advancements in its management. The prognosis for metastatic CRC remains discouraging, with a relative 5-year survival rate for stage IV CRC patients. Conventional treatments for advanced malignancies such as chemotherapy, often face limitations in effectively targeting cancer cells resulting in off-target distribution and significant side effects. In the quest for better strategies, researchers have explored numerous alternatives. Among these, nanoparticles (NPs) specifically liposomes have emerged as one of the most promising candidates in developing targeted delivery systems for cancer therapeutics. This review discusses the current approaches employing functionalised liposomes to overcome major biological barriers in therapeutics delivery for CRC treatment. We have also shared our perspectives on the technological development of liposomes for future clinical use and highlighted a few useful insights on the material choices for future research work in CRC.
Collapse
Affiliation(s)
- Intan Shazleen Ibrahim
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Melbha Starlin Chellathurai
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Syed Mahmood
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Amirul Hakim Azmi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Norsyifa Harun
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | | | - Mohd Muzamir Mahat
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor
| | - Zarif Mohamed Sofian
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
12
|
Bobik TV, Simonova MA, Rushkevich NU, Kostin NN, Skryabin GA, Knorre VD, Schulga AA, Konovalova EV, Proshkina GM, Gabibov AG, Deev SM. Immunoliposomes As a Promising Antiviral Agent against SARS-CoV-2. DOKL BIOCHEM BIOPHYS 2024; 514:6-10. [PMID: 38189883 PMCID: PMC11021331 DOI: 10.1134/s1607672923700618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 01/09/2024]
Abstract
According to the World Health Organization, as of January 3, 2020 to September 13, 2023, there were approximately 23 million confirmed cases of COVID-19 reported in the Russian Federation, about 400 thousand of which were fatal. Considering the high rate of mutation of the RNA-containing virus genome, which inevitably leads to the emergence of new infectious strains (Eris and Pyrola), the search for medicinal antiviral agents remains an urgent task. Moreover, taking into account the actively mutating receptor-binding domain, this task requires fundamentally new solutions. This study proposes a candidate immunoliposomal drug that targets the S protein of SARS-CoV-2 by the monoclonal neutralizing antibody P4A1 and ensures the penetration of a highly active ribonuclease into the virus-infected cell, which degrades, among cellular RNA, viral RNA too. We demonstrate a more than 40-fold increase in the neutralizing activity of the developed drug compared to the free monoclonal neutralizing antibody.
Collapse
Affiliation(s)
- T V Bobik
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | - M A Simonova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - N U Rushkevich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - N N Kostin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - G A Skryabin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - V D Knorre
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - A A Schulga
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - E V Konovalova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - G M Proshkina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - A G Gabibov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow State University, Moscow, Russia
| | - S M Deev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- National Research University Higher School of Economics, Moscow, Russia
| |
Collapse
|
13
|
Wang H, Wu D, Wang P, Gao C, Teng H, Liu D, Zhao Y, Du R. Albumin nanoparticles and their folate modified counterparts for delivery of a lupine derivative to hepatocellular carcinoma. Biomed Pharmacother 2023; 167:115485. [PMID: 37713994 DOI: 10.1016/j.biopha.2023.115485] [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: 07/20/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
In this study, folate polyethylene glycol CTr albumin nanoparticles (FA-PEG-CTr-NPs) targeting hepatocellular carcinoma (HCC) were prepared. The nanoparticle preparation method was optimized using single-factor and response surface analysis. The prepared nanoparticles were characterized for their particle size, zeta potential, and morphology. The particle size and zeta potential were also determined. Additionally, drug loading, encapsulation efficiency, and in vitro drug release of the nanoparticles were determined. Using the Cell Counting Kit-8 method, their cytotoxicity and their cell-targeted uptake were determined using confocal microscopy and flow cytometry. Finally, the in vivo antitumor impact and tumor-targeting ability of the nanoparticles were evaluated by determining tumor volume inhibition and drug biodistribution and performing hematoxylin-eosin (H&E) staining. It was found that CTr could be effectively encapsulated into albumin nanoparticles and functionalized. The drug loading of the two nanoparticles was 67.12 ± 2.4% and 69.33 ± 2.8%, respectively. Regarding drug release, FA-PEG-CTr-NPs (89.0%) exhibited a superior release rate to CTr-NPs (70.5%) in an acidic environment. The in vitro experiments confirmed that FA-PEG-CTr-NPs yielded better cytotoxicity and faster drug uptake results than CTr and CTr-NPs. In vivo experiments confirmed that FA-PEG-CTr-NPs exhibited markedly better tumor inhibitory activity (inhibition rate was 80.21%), drug safety, and targeting than CTr and CTr-NPs. In conclusion, functionalized nanoparticles (FA-PEG-CTr-NPs) can specifically inhibit the malignant proliferation of HCC cells and are thus a promising nanoagent for the treatment of HCC.
Collapse
Affiliation(s)
- Haohao Wang
- School of biological and pharmaceutical engineering, West Anhui University, Lu'an 237012, China
| | - Di Wu
- Department of Breast Surgery, General Surgery Center, First Hospital of Jilin University, Changchun 130118, China
| | - Pan Wang
- School of biological and pharmaceutical engineering, West Anhui University, Lu'an 237012, China
| | - Chunyu Gao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Hongbo Teng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Dong Liu
- School of biological and pharmaceutical engineering, West Anhui University, Lu'an 237012, China; Anhui Traditional Chinese Medicine Ecological Agricultural engineering Research Center, Lu'an 237012, China.
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
14
|
Suman SK, Mukherjee A, Sharma RK. A liposomal radionanoformulation for targeted drug delivery and real time monitoring by radionuclide imaging for HER2 overexpressing cancers. Drug Dev Res 2023; 84:1553-1563. [PMID: 37578143 DOI: 10.1002/ddr.22106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/15/2023]
Abstract
Liposomal formulations carrying chemotherapeutic drugs have demonstrated great potential as effective drug delivery systems. Smart nanoformulations decorated with targeting agents and probes are desired for site specific delivery of drugs and real time monitoring. In this study, we aimed to develop liposomal formulation loaded with doxorubicin and tagged with trastuzumab antibody (Ab) for targeting human epidermal growth factor receptor 2 (HER2) positive tumors. Liposomes were prepared by ethanol injection method using modified lipids to conjugate trastuzumab and radiolabel with Tc-99m radioisotope using DTPA for imaging by single photon emission computed tomography (SPECT). Doxorubicin was loaded using the active pH gradient method. The conjugation of Ab to liposomes was validated by SDS-PAGE and MALDI-MS. 99m Tc labeled liposomes encapsulating doxorubicin conjugated with antibody (99m Tc-Lip-Ab-Dox) and 99m Tc labeled liposomes encapsulating doxorubicin (99m Tc-Lip-Dox) were found to be stable in blood plasma and saline using chromatography method. The specificity of 99m Tc-Lip-Ab-Dox against HER2 receptor was evident from cell uptake and inhibition studies. Results also corroborated with confocal microscopy studies. In vivo studies in tumor bearing severe combined immunodeficient mice by SPECT imaging and biodistribution studies revealed higher uptake of 99m Tc-Lip-Ab-Dox in tumor and less accumulation in the liver compared to 99m Tc-Lip-Dox. In conclusion, liposomal nanoformulation for immunotargeting and monitoring of drug delivery was successfully formulated and evaluated. Encouraging results in preclinical studies were obtained with the radioformulation. Such smart radioformulations will not only serve the purpose of site-specific controlled release of drugs at the target site but also aid in optimizing the drug doses and schedule of cancer treatment by monitoring pharmacokinetics.
Collapse
Affiliation(s)
- Shishu Kant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rohit Kumar Sharma
- Department of Chemistry and Centre of Advanced Studies, Panjab University, Chandigarh, India
| |
Collapse
|
15
|
Liu Z, Cao Y, Guo X, Chen Z. The Potential Role of Timosaponin-AIII in Cancer Prevention and Treatment. Molecules 2023; 28:5500. [PMID: 37513375 PMCID: PMC10386027 DOI: 10.3390/molecules28145500] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer, as one of the leading causes of death worldwide, has challenged current chemotherapy drugs. Considering that treatments are expensive, alongside the resistance of tumor cells to anticancer drugs, the development of alternative medicines is necessary. Anemarrhena asphodeloides Bunge, a recognized and well-known medicinal plant for more than two thousand years, has demonstrated its effectiveness against cancer. Timosaponin-AIII (TSAIII), as a bioactive steroid saponin isolated from A. asphodeloides, has shown multiple pharmacological activities and has been developed as an anticancer agent. However, the molecular mechanisms of TSAIII in protecting against cancer development are still unclear. In this review article, we provide a comprehensive discussion on the anticancer effects of TSAIII, including proliferation inhibition, cell cycle arrest, apoptosis induction, autophagy mediation, migration and invasion suppression, anti-angiogenesis, anti-inflammation, and antioxidant effects. The pharmacokinetic profiles of TSAII are also discussed. TSAIII exhibits efficacy against cancer development. However, hydrophobicity and low bioavailability may limit the application of TSAIII. Effective delivery systems, particularly those with tissue/cell-targeted properties, can also significantly improve the anticancer effects of TSAIII.
Collapse
Affiliation(s)
- Zhaowen Liu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Yifan Cao
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Xiaohua Guo
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
16
|
Kansız S, Elçin YM. Advanced liposome and polymersome-based drug delivery systems: Considerations for physicochemical properties, targeting strategies and stimuli-sensitive approaches. Adv Colloid Interface Sci 2023; 317:102930. [PMID: 37290380 DOI: 10.1016/j.cis.2023.102930] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023]
Abstract
Liposomes and polymersomes are colloidal vesicles that are self-assembled from lipids and amphiphilic polymers, respectively. Because of their ability to encapsulate both hydrophilic and hydrophobic therapeutics, they are of great interest in drug delivery research. Today, the applications of liposomes and polymersomes have expanded to a wide variety of complex therapeutic molecules, including nucleic acids, proteins and enzymes. Thanks to their chemical versatility, they can be tailored to different drug delivery applications to achieve maximum therapeutic index. This review article evaluates liposomes and polymersomes from a perspective that takes into account the physical and biological barriers that reduce the efficiency of the drug delivery process. In this context, the design approaches of liposomes and polymersomes are discussed with representative examples in terms of their physicochemical properties (size, shape, charge, mechanical), targeting strategies (passive and active) and response to different stimuli (pH, redox, enzyme, temperature, light, magnetic field, ultrasound). Finally, the challenges limiting the transition from laboratory to practice, recent clinical developments, and future perspectives are addressed.
Collapse
Affiliation(s)
- Seyithan Kansız
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey; Biovalda Health Technologies, Inc., Ankara, Turkey.
| |
Collapse
|
17
|
Chen X, Yang H, Li C, Hu W, Cui H, Lin L. Enhancing the targeting performance and prolonging the antibacterial effects of clove essential oil liposomes to Campylobacter jejuni by antibody modification. Food Res Int 2023; 167:112736. [PMID: 37087219 DOI: 10.1016/j.foodres.2023.112736] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
The application of plant essential oil liposomes to prevent and control food safety risks caused by Campylobacter jejuni (C. jejuni) still faces challenges such as lack of targeting and low release rate. Here, a bacteria-targeted and protease-activated antibacterial liposome (ACCLPs) was successfully synthesized through encapsulation of clove essential oil (CEO) by film dispersion method, embedding of casein by freeze-thaw method, and conjugation of C. jejuni antibody on the liposome membrane by post-insertion method. The average particle size, the essential oil encapsulation rate, the casein mosaic rate, and the antibody coupling efficiency of ACCLPs were determined as185.87 nm,16.9%,70.1% and 87.5%, respectively. The modification with C. jejuni antibody could significantly improve the targeting of ACCLPs to C. jejuni. Controlled release experiments showed that the exocrine protease from C. jejuni could hydrolyze the embedded casein and perforation on the ACCLPs, thus leading to a bacteria-dependent CEO release and significant prolonging the antibacterial effects of ACCLPs. Application results of ACCLPs on C. jejuni-contaminated foods showed that ACCLPs could effectively inhibit C. jejuni in a variety of meat products, fruits and vegetables and extend their shelf life without significantly affecting food quality. The results above in this work would provide a new view for the development of high efficient liposome-based antibacterial system of plant essential oil.
Collapse
|
18
|
Mohammadi AH, Ghazvinian Z, Bagheri F, Harada M, Baghaei K. Modification of Extracellular Vesicle Surfaces: An Approach for Targeted Drug Delivery. BioDrugs 2023; 37:353-374. [PMID: 37093521 DOI: 10.1007/s40259-023-00595-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/25/2023]
Abstract
Extracellular vesicles (EVs) are a promising drug delivery vehicle candidate because of their natural origin and intrinsic function of transporting various molecules between different cells. Several advantages of the EV delivery platform include enhanced permeability and retention effect, efficient interaction with recipient cells, the ability to traverse biological barriers, high biocompatibility, high biodegradability, and low immunogenicity. Furthermore, EV membranes share approximately similar structures and contents to the cell membrane, which allows surface modification of EVs, an approach to enable specific targeting. Enhanced drug accumulation in intended sites and reduced adverse effects of chemotherapeutic drugs are the most prominent effects of targeted drug delivery. In order to improve the targeting ability of EVs, chemical modification and genetic engineering are the most adopted methods to date. Diverse chemical methods are employed to decorate EV surfaces with various ligands such as aptamers, carbohydrates, peptides, vitamins, and antibodies. In this review, we introduce the biogenesis, content, and cellular pathway of natural EVs and further discuss the genetic modification of EVs, and its challenges. Furthermore, we provide a comprehensive deliberation on the various chemical modification methods for improved drug delivery, which are directly related to increasing the therapeutic index.
Collapse
Affiliation(s)
- Amir Hossein Mohammadi
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Zeinab Ghazvinian
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran.
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Lahooti B, Akwii RG, Patel D, ShahbaziNia S, Lamprou M, Madadi M, Abbruscato TJ, Astrinidis A, Bickel U, Al-Ahmad A, German NA, Mattheolabakis G, Mikelis CM. Endothelial-Specific Targeting of RhoA Signaling via CD31 Antibody-Conjugated Nanoparticles. J Pharmacol Exp Ther 2023; 385:35-49. [PMID: 36746610 PMCID: PMC10029826 DOI: 10.1124/jpet.122.001384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/04/2023] [Accepted: 01/17/2023] [Indexed: 02/08/2023] Open
Abstract
Existing vascular endothelial growth factor-oriented antiangiogenic approaches are known for their high potency. However, significant side effects associated with their use drive the need for novel antiangiogenic strategies. The small GTPase RhoA is an established regulator of actin cytoskeletal dynamics. Previous studies have highlighted the impact of endothelial RhoA pathway on angiogenesis. Rho-associate kinase (ROCK), a direct RhoA effector, is potently inhibited by Fasudil, a clinically relevant ROCK inhibitor. Here, we aimed to target the RhoA signaling in endothelial cells by generating Fasudil-encapsulated CD31-targeting liposomes as a potential antiangiogenic therapy. The liposomes presented desirable characteristics, preferential binding to CD31-expressing HEK293T cells and to endothelial cells, inhibited stress fiber formation and cytoskeletal-related morphometric parameters, and inhibited in vitro angiogenic functions. Overall, this work shows that the nanodelivery-mediated endothelial targeting of RhoA signaling can offer a promising strategy for angiogenesis inhibition in vascular-related diseases. SIGNIFICANCE STATEMENT: Systemic administration of antiangiogenic therapeutics induces side effects to non-targeted tissues. This study, among others, has shown the impact of the RhoA signaling in the endothelial cells and their angiogenic functions. Here, to minimize potential toxicity, this study generated CD31-targeting liposomes with encapsulated Fasudil, a clinically relevant Rho kinase inhibitor, and successfully targeted endothelial cells. In this proof-of-principle study, the efficient Fasudil delivery, its impact on the endothelial signaling, morphometric alterations, and angiogenic functions verify the benefits of site-targeted antiangiogenic therapy.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Dhavalkumar Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Siavash ShahbaziNia
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Margarita Lamprou
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Mahboubeh Madadi
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Aristotelis Astrinidis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Ulrich Bickel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Nadezhda A German
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - George Mattheolabakis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| |
Collapse
|
20
|
Lee JH, Chapman DV, Saltzman WM. Nanoparticle Targeting with Antibodies in the Central Nervous System. BME FRONTIERS 2023; 4:0012. [PMID: 37849659 PMCID: PMC10085254 DOI: 10.34133/bmef.0012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/19/2023] [Indexed: 10/19/2023] Open
Abstract
Treatments for disease in the central nervous system (CNS) are limited because of difficulties in agent penetration through the blood-brain barrier, achieving optimal dosing, and mitigating off-target effects. The prospect of precision medicine in CNS treatment suggests an opportunity for therapeutic nanotechnology, which offers tunability and adaptability to address specific diseases as well as targetability when combined with antibodies (Abs). Here, we review the strategies to attach Abs to nanoparticles (NPs), including conventional approaches of chemisorption and physisorption as well as attempts to combine irreversible Ab immobilization with controlled orientation. We also summarize trends that have been observed through studies of systemically delivered Ab-NP conjugates in animals. Finally, we discuss the future outlook for Ab-NPs to deliver therapeutics into the CNS.
Collapse
Affiliation(s)
| | | | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
21
|
Recent Preclinical and Clinical Progress in Liposomal Doxorubicin. Pharmaceutics 2023; 15:pharmaceutics15030893. [PMID: 36986754 PMCID: PMC10054554 DOI: 10.3390/pharmaceutics15030893] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Doxorubicin (DOX) is a potent anti-cancer agent that has garnered great interest in research due to its high efficacy despite dose-limiting toxicities. Several strategies have been exploited to enhance the efficacy and safety profile of DOX. Liposomes are the most established approach. Despite the improvement in safety properties of liposomal encapsulated DOX (in Doxil and Myocet), the efficacy is not superior to conventional DOX. Functionalized (targeted) liposomes present a more effective system to deliver DOX to the tumor. Moreover, encapsulation of DOX in pH-sensitive liposomes (PSLs) or thermo-sensitive liposomes (TSLs) combined with local heating has improved DOX accumulation in the tumor. Lyso-thermosensitive liposomal DOX (LTLD), MM-302, and C225-immunoliposomal(IL)-DOX have reached clinical trials. Further functionalized PEGylated liposomal DOX (PLD), TSLs, and PSLs have been developed and evaluated in preclinical models. Most of these formulations improved the anti-tumor activity compared to the currently available liposomal DOX. However, the fast clearance, the optimization of ligand density, stability, and release rate need more investigations. Therefore, we reviewed the latest approaches applied to deliver DOX more efficiently to the tumor, preserving the benefits obtained from FDA-approved liposomes.
Collapse
|
22
|
Arslan FB, Öztürk K, Tavukçuoğlu E, Öztürk SC, Esendağlı G, Çalış S. A novel combination for the treatment of small cell lung cancer: Active targeted irinotecan and stattic co-loaded PLGA nanoparticles. Int J Pharm 2023; 632:122573. [PMID: 36592892 DOI: 10.1016/j.ijpharm.2022.122573] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022]
Abstract
Polymeric nanoparticles are widely used drug delivery systems for cancer treatment due to their properties such as ease of passing through biological membranes, opportunity to modify drug release, specifically targeting drugs to diseased areas, and potential of reducing side effects. Here, we formulated irinotecan and Stattic co-loaded PLGA nanoparticles targeted to small cell lung cancer. Nanoparticles were successfully conjugated with CD56 antibody with a conjugation efficiency of 84.39 ± 1.01%, and characterization of formulated nanoparticles was conducted with in-vitro and in-vivo studies. Formulated particles had sizes in the range of 130-180 nm with PDI values smaller than 0.3. Encapsulation and active targeting of irinotecan and Stattic resulted in increased cytotoxicity and anti-cancer efficiency in-vitro. Furthermore, it was shown with ex-vivo biodistribution studies that conjugated nanoparticles were successfully targeted to CD56-expressing SCLC cells and distributed mainly to tumor tissue and lungs. Compliant with our hypothesis and literature, the STAT3 pathway was successfully inhibited with Stattic solution and Stattic loaded nanoparticles. Additionally, intravenous injection of conjugated co-loaded nanoparticles resulted in decreased side effects and better anti-tumor activity than individual solutions of drugs in SCLC tumor-bearing mice. These results may indicate a new treatment option for clinically aggressive small cell lung cancer.
Collapse
Affiliation(s)
- Fatma Betül Arslan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkiye
| | - Kıvılcım Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkiye
| | - Ece Tavukçuoğlu
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkiye
| | - Süleyman Can Öztürk
- Centre for Laboratory Animals Research and Application, Hacettepe University, Ankara, Turkiye
| | - Güneş Esendağlı
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkiye
| | - Sema Çalış
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkiye.
| |
Collapse
|
23
|
Marques AC, Costa PC, Velho S, Amaral MH. Lipid Nanoparticles Functionalized with Antibodies for Anticancer Drug Therapy. Pharmaceutics 2023; 15:216. [PMID: 36678845 PMCID: PMC9864942 DOI: 10.3390/pharmaceutics15010216] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Nanotechnology takes the lead in providing new therapeutic options for cancer patients. In the last decades, lipid-based nanoparticles-solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), liposomes, and lipid-polymer hybrid nanoparticles-have received particular interest in anticancer drug delivery to solid tumors. To improve selectivity for target cells and, thus, therapeutic efficacy, lipid nanoparticles have been functionalized with antibodies that bind to receptors overexpressed in angiogenic endothelial cells or cancer cells. Most papers dealing with the preclinical results of antibody-conjugated nanoparticles claim low systemic toxicity and effective tumor inhibition, which have not been successfully translated into clinical use yet. This review aims to summarize the current "state-of-the-art" in anticancer drug delivery using antibody-functionalized lipid-based nanoparticles. It includes an update on promising candidates that entered clinical trials and some explanations for low translation success.
Collapse
Affiliation(s)
- Ana Camila Marques
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paulo C. Costa
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sérgia Velho
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - Maria Helena Amaral
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
24
|
Markowski A, Zaremba-Czogalla M, Jaromin A, Olczak E, Zygmunt A, Etezadi H, Boyd BJ, Gubernator J. Novel Liposomal Formulation of Baicalein for the Treatment of Pancreatic Ductal Adenocarcinoma: Design, Characterization, and Evaluation. Pharmaceutics 2023; 15:pharmaceutics15010179. [PMID: 36678808 PMCID: PMC9865389 DOI: 10.3390/pharmaceutics15010179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers so there is an urgent need to develop new drugs and therapies to treat it. Liposome-based formulations of naturally-derived bioactive compounds are promising anticancer candidates due to their potential for passive accumulation in tumor tissues, protection against payload degradation, and prevention of non-specific toxicity. We chose the naturally-derived flavonoid baicalein (BAI) due to its promising effect against pancreatic ductal adenocarcinoma (PDAC) and encapsulated it into a liposomal bilayer using the passive loading method, with an almost 90% efficiency. We performed a morphological and stability analysis of the obtained BAI liposomal formulation and evaluated its activity on two-dimensional and three-dimensional pancreatic cell models. As the result, we obtained a stable BAI-encapsulated liposomal suspension with a size of 100.9 nm ± 2.7 and homogeneity PDI = 0.124 ± 0.02, suitable for intravenous administration. Furthermore, this formulation showed high cytotoxic activity towards AsPC-1 and BxPC-3 PDAC cell lines (IC50 values ranging from 21 ± 3.6 µM to 27.6 ± 4.1 µM), with limited toxicity towards normal NHDF cells and a lack of hemolytic activity. Based on these results, this new BAI liposomal formulation is an excellent candidate for potential anti-PDAC therapy.
Collapse
Affiliation(s)
- Adam Markowski
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
- Correspondence: (A.M.); (A.J.)
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
- Correspondence: (A.M.); (A.J.)
| | - Ewa Olczak
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Adrianna Zygmunt
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Haniyeh Etezadi
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Ben J. Boyd
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| |
Collapse
|
25
|
Guo C, Su Y, Wang H, Cao M, Diao N, Liu Z, Chen D, Kong M. A novel saponin liposomes based on the couplet medicines of Platycodon grandiflorum-Glycyrrhiza uralensis for targeting lung cancer. Drug Deliv 2022; 29:2743-2750. [PMID: 35999702 PMCID: PMC9487977 DOI: 10.1080/10717544.2022.2112997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Liposomes have been widely used for targeted drug delivery, but the disadvantages caused by cholesterol limit the application of conventional liposomes in cancer treatment. The compatibility basis of couplet medicines and the compatibility principle of the traditional Chinese medicine principle of ‘monarch, minister, assistant and guide’ are the important theoretical basis of Chinese medicine in the treatment of tumor and the important method to solve the problem of high toxicity. In this study, the active ingredients of the couplet medicines Platycodon grandiflorum and Glycyrrhiza uralensis were innovatively utilized, and glycyrrhizic acid (GA) was encapsulated in liposomes constructed by mixing saponin and lecithin, and cholesterol was replaced by platycodin and ginsenoside to construct saponin liposomes (RP-lipo) for the drug delivery system of Chinese medicine. Compared with conventional liposomes, PR-lipo@GA has no significant difference in morphological characteristics and drug release behavior, and also shows stronger targeting of lung cancer cells and anti-tumor ability in vitro, which may be related to the pharmacological properties of saponins themselves. Thus, PR-lipo@GA not only innovatively challenges the status of cholesterol as a liposome component, but also provides another innovative potential system with multiple functions for the clinical application of TCM couplet medicines.
Collapse
Affiliation(s)
- Chunjing Guo
- College of Marine Life Science, Ocean University of China, Qingdao266003, P.R. China
| | - Yanguo Su
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Hui Wang
- Gynecology Department, Affiliated hospital of Weifang Medical University, Weifang261053, P.R. China
| | - Min Cao
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Ningning Diao
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Zhongxin Liu
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Daquan Chen
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Ming Kong
- College of Marine Life Science, Ocean University of China, Qingdao266003, P.R. China
| |
Collapse
|
26
|
Hybrid Ultrasound-Activated Nanoparticles Based on Graphene Quantum Dots for Cancer Treatment. Int J Pharm 2022; 629:122373. [DOI: 10.1016/j.ijpharm.2022.122373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/23/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
|
27
|
Chen L, Lan J, Li Z, Zeng R, Wang Y, Zhen L, Jin H, Ding Y, Zhang T. A Novel Diosgenin-Based Liposome Delivery System Combined with Doxorubicin for Liver Cancer Therapy. Pharmaceutics 2022; 14:1685. [PMID: 36015311 PMCID: PMC9416271 DOI: 10.3390/pharmaceutics14081685] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/02/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022] Open
Abstract
As a malignant tumor, liver cancer is mainly treated with chemotherapy, while chemotherapeutic drugs, such as doxorubicin (DOX), may lead to toxicity, drug resistance and poor prognosis. The targeted delivery systems of combining natural products and chemotherapeutic drugs are useful to eliminate cancers with reduced toxicity and increased efficiency. In this study, a diosgenin-based liposome loaded with DOX (Dios-DOX-LP) was developed for synergistic treatment of liver cancer, in which Dios not only replaced cholesterol as the membrane regulator to keep stability of liposomes, but also became the chemotherapy adjuvant of DOX for synergistic treatment. Dios-DOX-LP was characterized by particle size (99.4 ± 6.2 nm), zeta potential (-33.3 ± 2.5 mV), and entrapment efficiency (DOX: 98.77 ± 2.04%, Dios: 87.75 ± 2.93%), which had a good stability and slow-release effect. Compared with commercial DOX liposome (CHOL-DOX-LP), Dios-DOX-LP had an improved anti-tumor effect in vitro and in vivo by inducing the apoptosis and inhibiting the proliferation of the tumor cell, which was 1.6 times better than CHOL-DOX-LP in cytotoxicity, and had 78% of the tumor inhibition rate on tumor-bearing nude mice. Dios-DOX-LP provided a novel idea to achieve synergistic tumor treatment using diosgenin as a liposome material.
Collapse
Affiliation(s)
- Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Experiment Center of Teaching & Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhe Li
- Experiment Center of Teaching & Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruifeng Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lu Zhen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Haojieyin Jin
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Experiment Center of Teaching & Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
28
|
Correa VA, Portilho AI, De Gaspari E. Vaccines, Adjuvants and Key Factors for Mucosal Immune Response. Immunology 2022; 167:124-138. [PMID: 35751397 DOI: 10.1111/imm.13526] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/26/2022] [Indexed: 11/29/2022] Open
Abstract
Vaccines are the most effective tool to control infectious diseases, which provoke significant morbidity and mortality. Most vaccines are administered through the parenteral route and can elicit a robust systemic humoral response, but they induce a weak T-cell-mediated immunity and are poor inducers of mucosal protection. Considering that most pathogens enter the body through mucosal surfaces, a vaccine that elicits protection in the first site of contact between the host and the pathogen is promising. However, despite the advantages of mucosal vaccines as good options to confer protection on the mucosal surface, only a few mucosal vaccines are currently approved. In this review, we discuss the impact of vaccine administration in different mucosal surfaces; how appropriate adjuvants enhance the induction of protective mucosal immunity and other factors that can influence the mucosal immune response to vaccines. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Victor Araujo Correa
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| | - Amanda Izeli Portilho
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| | - Elizabeth De Gaspari
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| |
Collapse
|
29
|
Application of Non-Viral Vectors in Drug Delivery and Gene Therapy. Polymers (Basel) 2021; 13:polym13193307. [PMID: 34641123 PMCID: PMC8512075 DOI: 10.3390/polym13193307] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 12/13/2022] Open
Abstract
Vectors and carriers play an indispensable role in gene therapy and drug delivery. Non-viral vectors are widely developed and applied in clinical practice due to their low immunogenicity, good biocompatibility, easy synthesis and modification, and low cost of production. This review summarized a variety of non-viral vectors and carriers including polymers, liposomes, gold nanoparticles, mesoporous silica nanoparticles and carbon nanotubes from the aspects of physicochemical characteristics, synthesis methods, functional modifications, and research applications. Notably, non-viral vectors can enhance the absorption of cargos, prolong the circulation time, improve therapeutic effects, and provide targeted delivery. Additional studies focused on recent innovation of novel synthesis techniques for vector materials. We also elaborated on the problems and future research directions in the development of non-viral vectors, which provided a theoretical basis for their broad applications.
Collapse
|
30
|
Zhang L, Zhang S, Jiang M, Lu L, Ding Y, Ma N, Zhao Y, Xuchen S, Zhang N. Novel Timosaponin AIII-Based Multifunctional Liposomal Delivery System for Synergistic Therapy Against Hepatocellular Carcinoma Cancer. Int J Nanomedicine 2021; 16:5531-5550. [PMID: 34429598 PMCID: PMC8379713 DOI: 10.2147/ijn.s313759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction As high cholesterol level has been reported to be associated with cancer cell growth and cholesterol is vulnerable to oxidation, the conventional liposomes including cholesterol in the formulation seem to be challenged. Timosaponin AIII (TAIII), as a steroid saponin from Anemarrhena asphodeloides Bunge, possesses a similar structure with cholesterol and exhibits a wide range of antitumor activities, making it possible to develop a TAIII-based liposome where TAIII could potentially stabilize the phospholipid bilayer as a substitution of cholesterol and work as a chemotherapeutic drug as well. Meanwhile, TAIII could enhance the uptake of doxorubicin hydrochloride (DOX) in human hepatocellular carcinoma (HCC) cells and exhibit synergistic effect. Thus, we designed a novel thermally sensitive multifunctional liposomal system composed of TAIII and lipids to deliver DOX for enhanced HCC treatment. Methods The synergistic effects of DOX and TAIII were explored on HCC cells and the tumor inhibition rate of TAIII-based liposomes carrying DOX was evaluated on both subcutaneous and orthotopic transplantation tumor models. TAIII-based multifunctional liposomes were characterized. Results Synergistic HCC cytotoxicity was achieved at molar ratios of 1:1, 1:2 and 1:4 of DOX/TAIII. TAIII-based liposomes carrying a low DOX dose of 2 mg/kg exhibited significantly enhanced antitumor activity than 5 mg/kg of DOX without detected cardiotoxicity on both subcutaneous and orthotopic transplantation tumor models. TAIII-based liposomes were characterized with smaller size than cholesterol liposomes but exhibited favorable stability. Mild hyperthermia generated by laser irradiation accelerated the release of DOX and TAIII from liposomes at tumor site, and cell permeability of TAIII enhanced uptake of DOX in HCC cells. Conclusion The innovative application of TAIII working as bilayer stabilizer and chemotherapeutic drug affords a stable multifunctional liposomal delivery system for synergistic therapy against HCC, which may be referred for the development of other types of saponins with similar property.
Collapse
Affiliation(s)
- Lijuan Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shengan Zhang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Min Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Lu Lu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yue Ding
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Ninghui Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yuan Zhao
- Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Sihan Xuchen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Nailian Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
31
|
Moulahoum H, Ghorbanizamani F, Zihnioglu F, Timur S. Surface Biomodification of Liposomes and Polymersomes for Efficient Targeted Drug Delivery. Bioconjug Chem 2021; 32:1491-1502. [PMID: 34283580 DOI: 10.1021/acs.bioconjchem.1c00285] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chemotherapy has seen great progress in the development of performant treatment strategies. Nanovesicles such as liposomes and polymersomes demonstrated great potential in cancer therapy. However, these nanocarriers deliver their content passively, which faces a lot of constraints during blood circulation. The main challenge resides in degradation and random delivery to normal tissues. Hence, targeting drug delivery using specific molecules (such as antibodies) grafted over the surface of these nanocarriers came as the answer to overcome many problems faced before. The advantage of using antibodies is their antigen/antibody recognition, which provides a high level of specificity to reach treatment targets. This review discusses the many techniques of nanocarrier functionalization with antibodies. The aim is to recognize the various approaches by describing their advantages and deficiencies to create the most suitable drug delivery platform. Some methods are more suitable for other applications rather than drug delivery, which can explain the low success of some proposed targeted nanocarriers. In here, a critical analysis of how every method could impact the recognition and targeting capacity of some nanocarriers (liposomes and polymersomes) is discussed to make future research more impactful and advance the field of biomedicine further.
Collapse
Affiliation(s)
- Hichem Moulahoum
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Faezeh Ghorbanizamani
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Figen Zihnioglu
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Suna Timur
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey.,Central Research Testing and Analysis Laboratory Research and Application Center, Ege University, 35100, Bornova, Izmir, Turkey
| |
Collapse
|
32
|
Liao Y, Li Z, Zhou Q, Sheng M, Qu Q, Shi Y, Yang J, Lv L, Dai X, Shi X. Saponin surfactants used in drug delivery systems: A new application for natural medicine components. Int J Pharm 2021; 603:120709. [PMID: 33992714 DOI: 10.1016/j.ijpharm.2021.120709] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 12/16/2022]
Abstract
Saponins are a group of compounds widely distributed in the plant kingdom. Due to their amphiphilic characteristic structure, saponins have high surface activity and self-assembly property and can be used as natural biosurfactants. Therefore, saponin has become a potential drug delivery system (DDS) carrier and has attracted the attention of many researchers. Increasing studies have found that when drugs combining with saponins, their solubility or bioavailability are improved. This phenomenon may be due to a synergistic mechanism and provides a potentially novel concept for DDS: saponins may be also used for carrier materials. This review emphasized the molecular characteristics and mechanism of saponins as carriers and the research on the morphology of saponin carriers. Besides, the article also introduced the role and application of saponins in DDS. Although there are still some limitations with the application of saponins such as cost, applicability, and hemolysis, the development of technology and in-depth molecular mechanism research will provide saponins with greater application prospects as DDS carriers.
Collapse
Affiliation(s)
- Yuyao Liao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhixun Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qing Zhou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mengke Sheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qingsong Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanshuang Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaqi Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lijing Lv
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingxing Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science & Technology Commission, Beijing 102488, China.
| | - Xinyuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science & Technology Commission, Beijing 102488, China.
| |
Collapse
|
33
|
Targeting Chronic Myeloid Leukemia Stem/Progenitor Cells Using Venetoclax-Loaded Immunoliposome. Cancers (Basel) 2021; 13:cancers13061311. [PMID: 33804056 PMCID: PMC8000981 DOI: 10.3390/cancers13061311] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
CML is a hematopoietic stem-cell disorder emanating from breakpoint cluster region/Abelson murine leukemia 1 (BCR/ABL) translocation. Introduction of different TKIs revolutionized treatment outcome in CML patients, but CML LSCs seem insensitive to TKIs and are detectable in newly diagnosed and resistant CML patients and in patients who discontinued therapy. It has been reported that CML LSCs aberrantly express some CD markers such as CD26 that can be used for the diagnosis and for targeting. In this study, we confirmed the presence of CD26+ CML LSCs in newly diagnosed and resistant CML patients. To selectively target CML LSCs/progenitor cells that express CD26 and to spare normal HSCs/progenitor cells, we designed a venetoclax-loaded immunoliposome (IL-VX). Our results showed that by using this system we could selectively target CD26+ cells while sparing CD26- cells. The efficiency of venetoclax in targeting CML LSCs has been reported and our system demonstrated a higher potency in cell death induction in comparison to free venetoclax. Meanwhile, treatment of patient samples with IL-VX significantly reduced CD26+ cells in both stem cells and progenitor cells population. In conclusion, this approach showed that selective elimination of CD26+ CML LSCs/progenitor cells can be obtained in vitro, which might allow in vivo reduction of side effects and attainment of treatment-free, long-lasting remission in CML patients.
Collapse
|
34
|
Mahmoudi R, Ashraf Mirahmadi-Babaheidri S, Delaviz H, Fouani MH, Alipour M, Jafari Barmak M, Christiansen G, Bardania H. RGD peptide-mediated liposomal curcumin targeted delivery to breast cancer cells. J Biomater Appl 2020; 35:743-753. [PMID: 32807016 DOI: 10.1177/0885328220949367] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study, turmeric's active ingredient (Curcumin) was encapsulated into RGD modified Liposomes (RGD-Lip-Cur) its cytotoxic effect on the breast cancer cell line (MCF-7) was evaluated by MTT, flow cytometry and Caspase assay. Liposomes were characterized using transmission electron microscopy (TEM). Results demonstrated that the liposomes were spherical in shape, ranging from 70 to 100 nm. MTT assay revealed that RGD-Lip-Cur had a significant cytotoxic effect on MCF-7 cells at concentrations of 32, 16 and 4 μg/ml compared to Lip-Cur (P < 0.05) and curcumin (P < 0.01). The apoptosis assay demonstrated that RGD-Lip-Cur induces the apoptosis in MCF-7 cells (39.6% vs 40.2% for initial and secondary apoptosis) significantly more than Lip-Cur (67.7% vs 9.16% for initial and secondary apoptosis) and free curcumin (7.84% vs 38.8% for initial and secondary apoptosis). Moreover, caspase assay showed that RGD-Lip-Cur activates caspase 3/7 compared to Lip-Cur (P < 0.05) and free curcumin (P < 0.01). The RGD-Lip-Cur was similar to the control group and had no significant cytotoxicity effect. It is concluded that RGD-Lip-Cur as a novel carrier have high cytotoxicity effect on breast cancer cell line (MCF-7).
Collapse
Affiliation(s)
- Reza Mahmoudi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Hamdollah Delaviz
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohamad Hassan Fouani
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Alipour
- Department of Advanced Medical Sciences & Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mehrzad Jafari Barmak
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Medicinal Plant Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Clinical Research Development Unit, Imamsajad Hospital, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
35
|
Hepatosplenic phagocytic cells indirectly contribute to anti-PEG IgM production in the accelerated blood clearance (ABC) phenomenon against PEGylated liposomes: Appearance of an unexplained mechanism in the ABC phenomenon. J Control Release 2020; 323:102-109. [DOI: 10.1016/j.jconrel.2020.04.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 01/02/2023]
|
36
|
Lin Y, Zhao WR, Shi WT, Zhang J, Zhang KY, Ding Q, Chen XL, Tang JY, Zhou ZY. Pharmacological Activity, Pharmacokinetics, and Toxicity of Timosaponin AIII, a Natural Product Isolated From Anemarrhena asphodeloides Bunge: A Review. Front Pharmacol 2020; 11:764. [PMID: 32581782 PMCID: PMC7283383 DOI: 10.3389/fphar.2020.00764] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Anemarrhena asphodeloides Bunge is a famous Chinese Materia Medica and has been used in traditional Chinese medicine for more than two thousand years. Steroidal saponins are important active components isolated from A. asphodeloides Bunge. Among which, the accumulation of numerous experimental studies involved in Timosaponin AIII (Timo AIII) draws our attention in the recent decades. In this review, we searched all the scientific literatures using the key word "timosaponin AIII" in the PubMed database update to March 2020. We comprehensively summarized the pharmacological activity, pharmacokinetics, and toxicity of Timo AIII. We found that Timo AIII presents multiple-pharmacological activities, such as anti-cancer, anti-neuronal disorders, anti-inflammation, anti-coagulant, and so on. And the anti-cancer effect of Timo AIII in various cancers, especially hepatocellular cancer and breast cancer, is supposed as its most potential activity. The anti-inflammatory activity of Timo AIII is also beneficial to many diseases. Moreover, VEGFR, X-linked inhibitor of apoptosis protein (XIAP), B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1), thromboxane (Tx) A2 receptor, mTOR, NF-κB, COX-2, MMPs, acetylcholinesterase (AChE), and so on are identified as the crucial pharmacological targets of Timo AIII. Furthermore, the hepatotoxicity of Timo AIII was most concerned, and the pharmacokinetics and toxicity of Timo AIII need further studies in diverse animal models. In conclusion, Timo AIII is potent as a compound or leading compound for further drug development while still needs in-depth studies.
Collapse
Affiliation(s)
- Yan Lin
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Oncology, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wai-Rong Zhao
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Ting Shi
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhang
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai-Yu Zhang
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Ding
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xin-Lin Chen
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing-Yi Tang
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhong-Yan Zhou
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, Macau
| |
Collapse
|
37
|
Zhang J, Li X, Huang L. Anticancer activities of phytoconstituents and their liposomal targeting strategies against tumor cells and the microenvironment. Adv Drug Deliv Rev 2020; 154-155:245-273. [PMID: 32473991 PMCID: PMC7704676 DOI: 10.1016/j.addr.2020.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/07/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Various bioactive ingredients have been extracted from Chinese herbal medicines (CHMs) that affect tumor progression and metastasis. To further understand the mechanisms of CHMs in cancer therapy, this article summarizes the effects of five categories of CHMs and their active ingredients on tumor cells and the tumor microenvironment. Despite their treatment potential, the undesirable physicochemical properties (poor permeability, instability, high hydrophilicity or hydrophobicity, toxicity) and unwanted pharmacokinetic profiles (short half-life in blood and low bioavailability) restrict clinical studies of CHMs. Therefore, development of liposomes through relevant surface modifying techniques to achieve targeted CHM delivery for cancer cells, i.e., extracellular and intracellular targets and targets in tumor microenvironment or vasculature, have been reviewed. Current challenges of liposomal targeting of these phytoconstituents and future perspective of CHM applications are discussed to provide an informative reference for interested readers.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Xiang Li
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
38
|
Marques AC, Costa PJ, Velho S, Amaral MH. Functionalizing nanoparticles with cancer-targeting antibodies: A comparison of strategies. J Control Release 2020; 320:180-200. [PMID: 31978444 DOI: 10.1016/j.jconrel.2020.01.035] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 01/07/2023]
Abstract
Standard cancer therapies sometimes fail to deliver chemotherapeutic drugs to tumor cells in a safe and effective manner. Nanotechnology takes the lead in providing new therapeutic options for cancer due to major potential for selective targeting and controlled drug release. Antibodies and antibody fragments are attracting much attention as a source of targeting ligands to bind specific receptors that are overexpressed on cancer cells. Therefore, researchers are devoting time and effort to develop targeting strategies based on nanoparticles functionalized with antibodies, which hold great promise to enhance therapeutic efficacy and circumvent severe side effects. Several methods have been described to immobilize antibodies on the surface of nanoparticles. However, selecting the most appropriate for each application is challenging but also imperative to preserve antigen binding ability and yield stable antibody-conjugated nanoparticles. From this perspective, we aim to provide considerable knowledge on the most widely used methods of functionalization that can be helpful for decision-making and design of conjugation protocols as well. This review summarizes adsorption, covalent conjugation (carbodiimide, maleimide and "click" chemistries) and biotin-avidin interaction, while discussing the advantages, limitations and relevant therapeutic approaches currently under investigation.
Collapse
Affiliation(s)
- A C Marques
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - P J Costa
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - S Velho
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - M H Amaral
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
39
|
Li Y, Cong H, Wang S, Yu B, Shen Y. Liposomes modified with bio-substances for cancer treatment. Biomater Sci 2020; 8:6442-6468. [DOI: 10.1039/d0bm01531h] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In recent years, liposomes have been used in the field of biomedicine and have achieved many significant results.
Collapse
Affiliation(s)
- Yanan Li
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Song Wang
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Bing Yu
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering
- College of Chemistry and Chemical Engineering
- College of Materials Science and Engineering
- Affiliated Hospital of Qingdao University
- Qingdao University
| |
Collapse
|
40
|
Yan W, Leung SS, To KK. Updates on the use of liposomes for active tumor targeting in cancer therapy. Nanomedicine (Lond) 2019; 15:303-318. [PMID: 31802702 DOI: 10.2217/nnm-2019-0308] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In the development of cancer chemotherapy, besides the discovery of new anticancer drugs, a variety of nanocarrier systems for the delivery of previously developed and new chemotherapeutic drugs have currently been explored. Liposome is one of the most studied nanocarrier systems because of its biodegradability, simple preparation method, high efficacy and low toxicity. To make the best use of this vehicle, a number of multifunctionalized liposomal formulations have been investigated. The objective of this review is to summarize the current development of novel active targeting liposomal formulations, and to give insight into the challenges and future direction of the field. The recent studies in active targeting liposomes suggest the great potential of precise targeted anticancer drug delivery in cancer therapeutics.
Collapse
Affiliation(s)
- Wei Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Sharon Sy Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Kenneth Kw To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
41
|
Kim HY, Kang M, Choo YW, Go SH, Kwon SP, Song SY, Sohn HS, Hong J, Kim BS. Immunomodulatory Lipocomplex Functionalized with Photosensitizer-Embedded Cancer Cell Membrane Inhibits Tumor Growth and Metastasis. NANO LETTERS 2019; 19:5185-5193. [PMID: 31298024 DOI: 10.1021/acs.nanolett.9b01571] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Liposomes are clinically used as drug carriers for cancer therapy; however, unwanted leakage of the encapsulated anticancer drug and poor tumor-targeting efficiency of liposomes may generate toxic side effects on healthy cells and lead to failure of tumor eradication. To overcome these limitations, we functionalized liposomes with a photosensitizer (KillerRed, KR)-embedded cancer cell membrane (CCM). A lipid adjuvant was also embedded in the lipocomplex to promote the anticancer immune response. KR proteins were expressed on CCM and did not leak from the lipocomplex. Owing to the homotypic affinity of the CCM for the source cancer cells, the lipocomplex exhibited a 3.3-fold higher cancer-targeting efficiency in vivo than a control liposome. The liposome functionalized with KR-embedded CCM and lipid adjuvant generated cytotoxic reactive oxygen species in photodynamic therapy and effectively induced anticancer immune responses, inhibiting primary tumor growth and lung metastasis in homotypic tumor-bearing mice. Taken together, the lipocomplex technology may improve liposome-based cancer therapy.
Collapse
Affiliation(s)
- Han Young Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Yeon Woong Choo
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Seok-Hyeong Go
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
- Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| |
Collapse
|
42
|
Song XY, Han FY, Chen JJ, Wang W, Zhang Y, Yao GD, Song SJ. Timosaponin AIII, a steroidal saponin, exhibits anti-tumor effect on taxol-resistant cells in vitro and in vivo. Steroids 2019; 146:57-64. [PMID: 30951756 DOI: 10.1016/j.steroids.2019.03.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/14/2019] [Accepted: 03/28/2019] [Indexed: 12/29/2022]
Abstract
Timosaponin AIII (TAIII), a steroidal saponin isolated from the rhizome of Anemarrhena asphodeloides, exerted cytotoxic effect in many cancer cell lines. However, the effect of TAIII on resistant tumor cancer cells was unclear. In this study, MTT assay showed that TAIII exhibited significant cytotoxicity against A549/Taxol and A2780/Taxol cells in vitro. Annexin V-FITC/PI staining revealed that TAIII induced apoptosis in A549/T and A2780/T cells. Furthermore, Western blot analysis demonstrated that TAIII inhibited the expressions of phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT), mammalian target of rapamycin (mTOR) as well as Ras, Raf, mitogen-activated protein kinase (MEPK), extracellular regulated protein kinases (ERK) in two taxol-resistant cancer cell lines. Besides, in vivo studies demonstrated that TAIII inhibited tumor growth in a nude mouse xenograft model. Additionally, TAIII (2.5 and 5 mg/kg) also down-regulated the protein expressions of PI3K/AKT/mTOR and Ras/Raf/MEK/ERK pathways in vivo. Taken together, these findings demonstrated that TAIII exhibited significant anti-tumor effect on taxol-resistant cells.
Collapse
Affiliation(s)
- Xiao-Yu Song
- Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, People's Republic of China
| | - Feng-Ying Han
- Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, People's Republic of China
| | - Jing-Jie Chen
- Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, People's Republic of China
| | - Wei Wang
- Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, People's Republic of China
| | - Yan Zhang
- Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, People's Republic of China
| | - Guo-Dong Yao
- Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, People's Republic of China.
| | - Shao-Jiang Song
- Department of Natural Products Chemistry, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, People's Republic of China.
| |
Collapse
|
43
|
Saeed M, Zalba S, Seynhaeve ALB, Debets R, Ten Hagen TLM. Liposomes targeted to MHC-restricted antigen improve drug delivery and antimelanoma response. Int J Nanomedicine 2019; 14:2069-2089. [PMID: 30988609 PMCID: PMC6440454 DOI: 10.2147/ijn.s190736] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Purpose Melanoma is the most aggressive form of skin cancer. Chemotherapy at a late stage fails due to low accumulation in tumors, indicating the need for targeted therapy. Materials and methods To increase drug uptake by tumor cells, we have targeted doxorubicin-containing liposomes using a T-cell receptor (TCR)-like antibody (scFv G8 and Hyb3) directed against melanoma antigen A1 (MAGE-A1) presented by human leukocyte antigen A1 (M1/A1). With the use of flow cytometry and confocal microscopy, we have tested our formulation in vitro. In vivo pharmacokinetics was done in tumor-free nu/nu mice, while biodistribution and efficacy study was done in nu/nu mice xenograft. Results We demonstrated two to five times higher binding and internalization of these immunoliposomes by M1+/A1+ melanoma cells in vitro in comparison with nontargeted liposomes. Cytotoxicity assay showed significant tumor cell kill at 10 µM doxorubicin (DXR) for targeted vs nontargeted liposomes. In vivo pharmacokinetics of nontargeted and targeted liposomes were similar, while accumulation of targeted liposomes was 2- to 2.5-fold and 6.6-fold enhanced when compared with nontargeted liposomes and free drug, respectively. Notably, we showed a superior antitumor activity of MAGE-A1-targeted DXR liposomes toward M1+/A1+ expressing tumors in mice compared with the treatment of M1−/A1+ tumors. Our results indicate that targeted liposomes showed better cytotoxicity in vitro and pharmacokinetics in vivo. Conclusion Liposomes decorated with TCR-mimicking scFv antibodies effectively and selectively target antigen-positive melanoma. We showed that DXR-loaded liposomes coupled to anti-M1/-A1 scFv inflict a significant antitumor response. Targeting tumor cells specifically promotes internalization of drug-containing nanoparticles and may improve drug delivery and ultimately antitumor efficacy. Our data argue that targeting MAGE in A1 context, by nanosized carriers decorated with TCR-like antibodies mimicking scFv, can be used as a theragnostic platform for drug delivery, immunotherapy, and potentially imaging, and diagnosis of melanoma.
Collapse
Affiliation(s)
- Mesha Saeed
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| | - Sara Zalba
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| | - Ann L B Seynhaeve
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Timo L M Ten Hagen
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| |
Collapse
|
44
|
|
45
|
Mohamed M, Abu Lila AS, Shimizu T, Alaaeldin E, Hussein A, Sarhan HA, Szebeni J, Ishida T. PEGylated liposomes: immunological responses. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2019; 20:710-724. [PMID: 31275462 PMCID: PMC6598536 DOI: 10.1080/14686996.2019.1627174] [Citation(s) in RCA: 287] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 05/10/2023]
Abstract
A commonly held view is that nanocarriers conjugated to polyethylene glycol (PEG) are non-immunogenic. However, many studies have reported that unexpected immune responses have occurred against PEG-conjugated nanocarriers. One unanticipated response is the rapid clearance of PEGylated nanocarriers upon repeat administration, called the accelerated blood clearance (ABC) phenomenon. ABC involves the production of antibodies toward nanocarrier components, including PEG, which reduces the safety and effectiveness of encapsulated therapeutic agents. Another immune response is the hypersensitivity or infusion reaction referred to as complement (C) activation-related pseudoallergy (CARPA). Such immunogenicity and adverse reactivities of PEGylated nanocarriers may be of potential concern for the clinical use of PEGylated therapeutics. Accordingly, screening of the immunogenicity and CARPA reactogenicity of nanocarrier-based therapeutics should be a prerequisite before they can proceed into clinical studies. This review presents PEGylated liposomes, immunogenicity of PEG, the ABC phenomenon, C activation and lipid-induced CARPA from a toxicological point of view, and also addresses the factors that influence these adverse interactions with the immune system.
Collapse
Affiliation(s)
- Marwa Mohamed
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Department of Pharmaceutics, Minia University, Minia, Egypt
| | - Amr S. Abu Lila
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Department of Pharmaceutics, College of Pharmacy, Hail University, Hail, Saudi Arabia
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Eman Alaaeldin
- Department of Pharmaceutics, Minia University, Minia, Egypt
| | - Amal Hussein
- Department of Pharmaceutics, Minia University, Minia, Egypt
| | | | - Janos Szebeni
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
- SeroScience LCC., Cambridge, MA, USA
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
- CONTACT Tatsuhiro Ishida Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima770-8505, Japan
| |
Collapse
|
46
|
Sun Y, Zhao Y, Teng S, Hao F, Zhang H, Meng F, Zhao X, Zheng X, Bi Y, Yao Y, Lee RJ, Teng L. Folic acid receptor-targeted human serum albumin nanoparticle formulation of cabazitaxel for tumor therapy. Int J Nanomedicine 2018; 14:135-148. [PMID: 30613142 PMCID: PMC6306057 DOI: 10.2147/ijn.s181296] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background We previously developed cabazitaxel (CTX)-loaded human serum albumin nanoparticles (NPs-CTX) via a self-assembly method, and these NPs showed efficacy in prostate cancer therapy. Many studies have shown that the levels of folic acid (FA) receptor on the surface of various tumor cells are high. Therefore, FA-modified NPs-CTX may have enhanced antitumor effects compared with unmodified NPs-CTX. Methods NPs-CTX were first prepared via self-assembly, and FA was conjugated on the surface of NPs-CTX through the -NH2 groups of the NPs to produce FA-NPs-CTX. The FA-NPs-CTX were evaluated in tumor cells with high FA receptor (FR) expression in vitro and in vivo. Results Both NPs-CTX and FA-NPs-CTX exhibited good stability and morphology. Drug release from the NPs was not affected by FA conjugation. Compared with CTX dissolved in a mixture of Tween 80 and 13% ethanol (w/w) at a ratio of 1:4 (v/v) (Tween-CTX), the two nanoformulations had lower lytic activity against normal red blood cells. However, FA-NPs-CTX showed greater inhibition of tumor cells with overexpressed FR, compared with NPs-CTX, in the cytotoxicity experiments. Moreover, the cellular uptake of FA-NPs-CTX was enhanced through FR-mediated endocytosis in HeLa cells in vitro and HeLa xenograft tumors in vivo. Although Tween-CTX exhibited tumor growth inhibition similar to FA-NPs-CTX in vivo, this inhibition also caused adverse side effects; the median lethal dose (LD50) of Tween-CTX to mice was 5.68 mg/kg, while FA-NPs-CTX-treated mice survived at doses exceeding 400 mg/kg. Conclusion The results showed that FA-NPs-CTX caused inhibition of tumor growth in a manner similar to that of Tween-CTX; however, the safety and tolerability of CTX were greatly improved by FA conjugation compared with those of Tween-CTX. In summary, FA-NPs-CTX have great potential in CTX delivery, and this formulation is a promising candidate for the treatment of cancers with high FR levels.
Collapse
Affiliation(s)
- Yating Sun
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Yarong Zhao
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Shanshan Teng
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Fei Hao
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Huan Zhang
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Fanchao Meng
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Xiuting Zhao
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Xiaolong Zheng
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | - Ye Bi
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| | | | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, Jilin, China, .,Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, Jilin, China,
| |
Collapse
|