1
|
Azimizonuzi H, Ghayourvahdat A, Ahmed MH, Kareem RA, Zrzor AJ, Mansoor AS, Athab ZH, Kalavi S. A state-of-the-art review of the recent advances of theranostic liposome hybrid nanoparticles in cancer treatment and diagnosis. Cancer Cell Int 2025; 25:26. [PMID: 39871316 PMCID: PMC11773959 DOI: 10.1186/s12935-024-03610-z] [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: 08/19/2024] [Accepted: 12/10/2024] [Indexed: 01/29/2025] Open
Abstract
Theranostics is a way of treating illness that blends medicine with testing. Specific characteristics should be present in the best theranostic agents for cancer: (1) the drugs should be safe and non-toxic; (2) they should be able to treat cancer selectively; and (3) they should be able to build up only in the cancerous tissue. Liposomes (LPs) are one of the most efficient drug delivery methods based on nanotechnology. Stealth LPs and commercial LPs have recently had an impact on cancer treatment. Using the valuable information from each imaging technique, along with the multimodality imaging functionality of liposomal therapeutic agents, makes them very appealing for personalized monitoring of how well therapeutic drugs are working against cancer in vivo and for predicting how well therapies will work. On the other hand, their use as nanoparticle delivery systems is currently in the research and development phase. Nanoscale delivery system innovation has made LP-nanoparticle hybrid structures very useful for combining therapeutic and imaging methods. LP-hybrid nanoparticles are better at killing cancer cells than their LP counterparts, making them excellent options for in vivo and in vitro drug delivery applications. Hybrid liposomes (HLs) could be used in the future as theranostic carriers to find and treat cancer targets. This would combine the best features of synthetic and biological drug delivery systems. Overarchingly, this article provided a comprehensive overview of the many LP types used in cancer detection, therapy, and theranostic analysis. An evaluation of the pros and cons of the many HLs types used in cancer detection and treatment has also been conducted. The study also included recent and significant research on HLs for cancer theranostic applications. We conclude by outlining the potential benefits and drawbacks of this theranostic approach to the concurrent detection and treatment of different malignancies, as well as its prospects.
Collapse
Affiliation(s)
- Hannaneh Azimizonuzi
- Inventor Member of International Federation of Inventors Associations, Geneva, Switzerland
| | - Arman Ghayourvahdat
- Inventor Member of International Federation of Inventors Associations, Geneva, Switzerland
| | | | | | - Athmar Jaber Zrzor
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Shaylan Kalavi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Islamic Azad University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Fernández-García R, Fraguas-Sánchez AI. Nanomedicines for Pulmonary Drug Delivery: Overcoming Barriers in the Treatment of Respiratory Infections and Lung Cancer. Pharmaceutics 2024; 16:1584. [PMID: 39771562 PMCID: PMC11677881 DOI: 10.3390/pharmaceutics16121584] [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: 11/12/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The pulmonary route for drug administration has garnered a great deal of attention in therapeutics for treating respiratory disorders. It allows for the delivery of drugs directly to the lungs and, consequently, the maintenance of high concentrations at the action site and a reduction in systemic adverse effects compared to other routes, such as oral or intravenous. Nevertheless, the pulmonary administration of drugs is challenging, as the respiratory system tries to eliminate inhaled particles, being the main responsible mucociliary escalator. Nanomedicines represent a primary strategy to overcome the limitations of this route as they can be engineered to prolong pulmonary retention and avoid their clearance while reducing drug systemic distribution and, consequently, systemic adverse effects. This review analyses the use of pulmonary-administered nanomedicines to treat infectious diseases affecting the respiratory system and lung carcinoma, two pathologies that represent major health threats.
Collapse
Affiliation(s)
| | - Ana I. Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
3
|
Dejeu IL, Vicaș LG, Marian E, Ganea M, Frenț OD, Maghiar PB, Bodea FI, Dejeu GE. Innovative Approaches to Enhancing the Biomedical Properties of Liposomes. Pharmaceutics 2024; 16:1525. [PMID: 39771504 PMCID: PMC11728823 DOI: 10.3390/pharmaceutics16121525] [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: 10/13/2024] [Revised: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Liposomes represent a promising class of drug delivery systems that enhance the therapeutic efficacy and safety of various pharmaceutical agents. Also, they offer numerous advantages compared to traditional drug delivery methods, including targeted delivery to specific sites, controlled release, and fewer side effects. This review meticulously examines the methodologies employed in the preparation and characterization of liposomal formulations. With the rising incidence of adverse drug reactions, there is a pressing need for innovative delivery strategies that prioritize selectivity, specificity, and safety. Nanomedicine promises to revolutionize diagnostics and treatments, addressing current limitations and improving disease management, including cancer, which remains a major global health challenge. This paper aims to conduct a comprehensive study on the interest of biomedical research regarding nanotechnology and its implications for further applications.
Collapse
Affiliation(s)
- Ioana Lavinia Dejeu
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Mariana Ganea
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Olimpia Daniela Frenț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Paula Bianca Maghiar
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - Flaviu Ionut Bodea
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - George Emanuiel Dejeu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 Piata 1 Decembrie Street, 410073 Oradea, Romania;
| |
Collapse
|
4
|
Chen Y, Zhou Q, Jia Z, Cheng N, Zhang S, Chen W, Wang L. Enhancing cancer immunotherapy: Nanotechnology-mediated immunotherapy overcoming immunosuppression. Acta Pharm Sin B 2024; 14:3834-3854. [PMID: 39309502 PMCID: PMC11413684 DOI: 10.1016/j.apsb.2024.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/12/2024] [Accepted: 05/24/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapy is an important cancer treatment method that offers hope for curing cancer patients. While immunotherapy has achieved initial success, a major obstacle to its widespread adoption is the inability to benefit the majority of patients. The success or failure of immunotherapy is closely linked to the tumor's immune microenvironment. Recently, there has been significant attention on strategies to regulate the tumor immune microenvironment in order to stimulate anti-tumor immune responses in cancer immunotherapy. The distinctive physical properties and design flexibility of nanomedicines have been extensively utilized to target immune cells (including tumor-associated macrophages (TAMs), T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated fibroblasts (TAFs)), offering promising advancements in cancer immunotherapy. In this article, we have reviewed treatment strategies aimed at targeting various immune cells to regulate the tumor immune microenvironment. The focus is on cancer immunotherapy models that are based on nanomedicines, with the goal of inducing or enhancing anti-tumor immune responses to improve immunotherapy. It is worth noting that combining cancer immunotherapy with other treatments, such as chemotherapy, radiotherapy, and photodynamic therapy, can maximize the therapeutic effects. Finally, we have identified the challenges that nanotechnology-mediated immunotherapy needs to overcome in order to design more effective nanosystems.
Collapse
Affiliation(s)
- Yunna Chen
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qianqian Zhou
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Zongfang Jia
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Nuo Cheng
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Sheng Zhang
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Weidong Chen
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Lei Wang
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| |
Collapse
|
5
|
Radha R, Paul V, Anjum S, Bouakaz A, Pitt WG, Husseini GA. Enhancing Curcumin's therapeutic potential in cancer treatment through ultrasound mediated liposomal delivery. Sci Rep 2024; 14:10499. [PMID: 38714740 PMCID: PMC11076529 DOI: 10.1038/s41598-024-61278-x] [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: 02/09/2024] [Accepted: 05/03/2024] [Indexed: 05/10/2024] Open
Abstract
Improving the efficacy of chemotherapy remains a key challenge in cancer treatment, considering the low bioavailability, high cytotoxicity, and undesirable side effects of some clinical drugs. Targeted delivery and sustained release of therapeutic drugs to cancer cells can reduce the whole-body cytotoxicity of the agent and deliver a safe localized treatment to the patient. There is growing interest in herbal drugs, such as curcumin, which is highly noted as a promising anti-tumor drug, considering its wide range of bioactivities and therapeutic properties against various tumors. Conversely, the clinical efficacy of curcumin is limited because of poor oral bioavailability, low water solubility, instability in gastrointestinal fluids, and unsuitable pH stability. Drug-delivery colloid vehicles like liposomes and nanoparticles combined with microbubbles and ultrasound-mediated sustained release are currently being explored as effective delivery modes in such cases. This study aimed to synthesize and study the properties of curcumin liposomes (CLs) and optimize the high-frequency ultrasound release and uptake by a human breast cancer cell line (HCC 1954) through in vitro studies of culture viability and cytotoxicity. CLs were effectively prepared with particles sized at 81 ± 2 nm, demonstrating stability and controlled release of curcumin under ultrasound exposure. In vitro studies using HCC1954 cells, the combination of CLs, ultrasound, and Definity microbubbles significantly improved curcumin's anti-tumor effects, particularly under specific conditions: 15 s of continuous ultrasound at 0.12 W/cm2 power density with 0.6 × 107 microbubbles/mL. Furthermore, the study delved into curcumin liposomes' cytotoxic effects using an Annexin V/PI-based apoptosis assay. The treatment with CLs, particularly in conjunction with ultrasound and microbubbles, amplified cell apoptosis, mainly in the late apoptosis stage, which was attributed to heightened cellular uptake within cancer cells.
Collapse
Affiliation(s)
- Remya Radha
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Vinod Paul
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
- Material Science and Engineering PhD Program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - Shabana Anjum
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84604, USA
| | - Ghaleb A Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE.
- Material Science and Engineering PhD Program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE.
| |
Collapse
|
6
|
Yao R, Zhu M, Guo Z, Shen J. Refining nanoprobes for monitoring of inflammatory bowel disease. Acta Biomater 2024; 177:37-49. [PMID: 38364928 DOI: 10.1016/j.actbio.2024.02.014] [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: 10/19/2023] [Revised: 01/11/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
Inflammatory bowel disease (IBD) is a gastrointestinal immune disease that requires clear diagnosis, timely treatment, and lifelong monitoring. The diagnosis and monitoring methods of IBD mainly include endoscopy, imaging examination, and laboratory examination, which are constantly developed to achieve early definite diagnosis and accurate monitoring. In recent years, with the development of nanotechnology, the diagnosis and monitoring methods of IBD have been remarkably enriched. Nanomaterials, characterized by their minuscule dimensions that can be tailored, along with their distinctive optical, magnetic, and biodistribution properties, have emerged as valuable contrast agents for imaging and targeted agents for endoscopy. Through both active and passive targeting mechanisms, nanoparticles accumulate at the site of inflammation, thereby enhancing IBD detection. This review comprehensively outlines the existing IBD detection techniques, expounds upon the utilization of nanoparticles in IBD detection and diagnosis, and offers insights into the future potential of in vitro diagnostics. STATEMENT OF SIGNIFICANCE: Due to their small size and unique physical and chemical properties, nanomaterials are widely used in the biological and medical fields. In the area of oncology and inflammatory disease, an increasing number of nanomaterials are being developed for diagnostics and drug delivery. Here, we focus on inflammatory bowel disease, an autoimmune inflammatory disease that requires early diagnosis and lifelong monitoring. Nanomaterials can be used as contrast agents to visualize areas of inflammation by actively or passively targeting them through the intestinal mucosal epithelium where gaps exist due to inflammation stimulation. In this article, we summarize the utilization of nanoparticles in inflammatory bowel disease detection and diagnosis, and offers insights into the future potential of in vitro diagnostics.
Collapse
Affiliation(s)
- Ruchen Yao
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China; NHC Key Laboratory of Digestive Diseases, China
| | - Mingming Zhu
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China; NHC Key Laboratory of Digestive Diseases, China
| | - Zhiqian Guo
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Jun Shen
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China; NHC Key Laboratory of Digestive Diseases, China.
| |
Collapse
|
7
|
Shanmugavadivu A, Lekhavadhani S, Miranda PJ, Selvamurugan N. Current approaches in tissue engineering-based nanotherapeutics for osteosarcoma treatment. Biomed Mater 2024; 19:022003. [PMID: 38324905 DOI: 10.1088/1748-605x/ad270b] [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: 09/16/2023] [Accepted: 02/07/2024] [Indexed: 02/09/2024]
Abstract
Osteosarcoma (OS) is a malignant bone neoplasm plagued by poor prognosis. Major treatment strategies include chemotherapy, radiotherapy, and surgery. Chemotherapy to treat OS has severe adverse effects due to systemic toxicity to healthy cells. A possible way to overcome the limitation is to utilize nanotechnology. Nanotherapeutics is an emerging approach in treating OS using nanoparticulate drug delivery systems. Surgical resection of OS leaves a critical bone defect requiring medical intervention. Recently, tissue engineered scaffolds have been reported to provide physical support to bone defects and aid multimodal treatment of OS. These scaffolds loaded with nanoparticulate delivery systems could also actively repress tumor growth and aid new bone formation. The rapid developments in nanotherapeutics and bone tissue engineering have paved the way for improved treatment efficacy for OS-related bone defects. This review focuses on current bifunctional nanomaterials-based tissue engineered (NTE) scaffolds that use novel approaches such as magnetic hyperthermia, photodynamic therapy, photothermal therapy, bioceramic and polymeric nanotherapeutics against OS. With further optimization and screening, NTE scaffolds could meet clinical applications for treating OS patients.
Collapse
Affiliation(s)
- Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Sundaravadhanan Lekhavadhani
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | | | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| |
Collapse
|
8
|
Li X, Peng X, Zoulikha M, Boafo GF, Magar KT, Ju Y, He W. Multifunctional nanoparticle-mediated combining therapy for human diseases. Signal Transduct Target Ther 2024; 9:1. [PMID: 38161204 PMCID: PMC10758001 DOI: 10.1038/s41392-023-01668-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 09/14/2023] [Accepted: 10/10/2023] [Indexed: 01/03/2024] Open
Abstract
Combining existing drug therapy is essential in developing new therapeutic agents in disease prevention and treatment. In preclinical investigations, combined effect of certain known drugs has been well established in treating extensive human diseases. Attributed to synergistic effects by targeting various disease pathways and advantages, such as reduced administration dose, decreased toxicity, and alleviated drug resistance, combinatorial treatment is now being pursued by delivering therapeutic agents to combat major clinical illnesses, such as cancer, atherosclerosis, pulmonary hypertension, myocarditis, rheumatoid arthritis, inflammatory bowel disease, metabolic disorders and neurodegenerative diseases. Combinatorial therapy involves combining or co-delivering two or more drugs for treating a specific disease. Nanoparticle (NP)-mediated drug delivery systems, i.e., liposomal NPs, polymeric NPs and nanocrystals, are of great interest in combinatorial therapy for a wide range of disorders due to targeted drug delivery, extended drug release, and higher drug stability to avoid rapid clearance at infected areas. This review summarizes various targets of diseases, preclinical or clinically approved drug combinations and the development of multifunctional NPs for combining therapy and emphasizes combinatorial therapeutic strategies based on drug delivery for treating severe clinical diseases. Ultimately, we discuss the challenging of developing NP-codelivery and translation and provide potential approaches to address the limitations. This review offers a comprehensive overview for recent cutting-edge and challenging in developing NP-mediated combination therapy for human diseases.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Xiuju Peng
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Makhloufi Zoulikha
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - George Frimpong Boafo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, PR China
| | - Kosheli Thapa Magar
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Yanmin Ju
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
| |
Collapse
|
9
|
Dabke A, Ghosh S, Dabke P, Sawant K, Khopade A. Revisiting the in-vitro and in-vivo considerations for in-silico modelling of complex injectable drug products. J Control Release 2023; 360:185-211. [PMID: 37353161 DOI: 10.1016/j.jconrel.2023.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/24/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Complex injectable drug products (CIDPs) have often been developed to modulate the pharmacokinetics along with efficacy for therapeutic agents used for remediation of chronic disorders. The effective development of CIDPs has exhibited complex kinetics associated with multiphasic drug release from the prepared formulations. Consequently, predictability of pharmacokinetic modelling for such CIDPs has been difficult and there is need for advanced complex computational models for the establishment of accurate prediction models for in-vitro-in-vivo correlation (IVIVC). The computational modelling aims at supplementing the existing knowledge with mathematical equations to develop formulation strategies for generation of predictable and discriminatory IVIVC. Such an approach would help in reduction of the burden of effect of hidden factors on preclinical to clinical translations. Computational tools like physiologically based pharmacokinetics (PBPK) modelling have combined physicochemical and physiological properties along with IVIVC characteristics of clinically used formulations. Such techniques have helped in prediction and understanding of variability in pharmacodynamic parameters of potential generic products to clinically used formulations like Doxil®, Ambisome®, Abraxane® in healthy and diseased population using mathematical equations. The current review highlights the important formulation characteristics, in-vitro, preclinical in-vivo aspects which need to be considered while developing a stimulatory predictive PBPK model in establishment of an IVIVC and in-vitro-in-vivo relationship (IVIVR).
Collapse
Affiliation(s)
- Amit Dabke
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India; Formulation Research & Development- Biopharmaceutics, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat 390012, India
| | - Saikat Ghosh
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India
| | - Pallavi Dabke
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India
| | - Krutika Sawant
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India.
| | - Ajay Khopade
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India; Formulation Research & Development- Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat 390012, India.
| |
Collapse
|
10
|
Yang Y, Lin M, Sun M, Zhang GQ, Guo J, Li J. Nanotechnology boosts the efficiency of tumor diagnosis and therapy. Front Bioeng Biotechnol 2023; 11:1249875. [PMID: 37576984 PMCID: PMC10419217 DOI: 10.3389/fbioe.2023.1249875] [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: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023] Open
Abstract
The incidence and mortality of cancer are gradually increasing. The highly invasive and metastasis of tumor cells increase the difficulty of diagnosis and treatment, so people pay more and more attention to the diagnosis and treatment of cancer. Conventional treatment methods, including surgery, radiotherapy and chemotherapy, are difficult to eliminate tumor cells completely. And the emergence of nanotechnology has boosted the efficiency of tumor diagnosis and therapy. Herein, the research progress of nanotechnology used for tumor diagnosis and treatment is reviewed, and the emerging detection technology and the application of nanodrugs in clinic are summarized and prospected. The first part refers to the application of different nanomaterials for imaging in vivo and detection in vitro, which includes magnetic resonance imaging, fluorescence imaging, photoacoustic imaging and biomarker detection. The distinctive physical and chemical advantages of nanomaterials can improve the detection sensitivity and accuracy to achieve tumor detection in early stage. The second part is about the nanodrug used in clinic for tumor treatment. Nanomaterials have been widely used as drug carriers, including the albumin paclitaxel, liposome drugs, mRNA-LNP, protein nanocages, micelles, membrane nanocomplexes, microspheres et al., which could improve the drug accumulate in tumor tissue through enhanced permeability and retention effect to kill tumor cells with high efficiency. But there are still some challenges to revolutionize traditional tumor diagnosis and anti-drug resistance based on nanotechnology.
Collapse
Affiliation(s)
| | | | | | | | - Jianshuang Guo
- Pharmacology and Toxicology Research Laboratory, College of Pharmaceutical Science, Hebei University, Baoding, Hebei, China
| | - Jianheng Li
- Pharmacology and Toxicology Research Laboratory, College of Pharmaceutical Science, Hebei University, Baoding, Hebei, China
| |
Collapse
|
11
|
Fan X, Jiang K, Geng F, Lu W, Wei G. Ocular therapies with biomacromolecules: From local injection to eyedrop and emerging noninvasive delivery strategies. Adv Drug Deliv Rev 2023; 197:114864. [PMID: 37156266 DOI: 10.1016/j.addr.2023.114864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/15/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
The last two decades have witnessed a continuously increasing number of biomacromolecules approved for the treatment of ocular diseases. The eye possesses multiple protective mechanisms to resist the invasion of exogenous substances, but meanwhile these physiological defense systems also act as strong barriers, impeding absorption of most biomacromolecules into the eye. As a result, local injections play predominant roles for posterior ocular delivery of biomacromolecules in clinical practice. To achieve safe and convenient application of biomacromolecules, alternative strategies to realize noninvasive intraocular delivery are necessary. Various nanocarriers, novel penetration enhancers and physical strategies have been explored to facilitate delivery of biomacromolecules to both anterior and posterior ocular segments but still suffered difficulties in clinical translation. This review compares the anatomical and physiological characteristics of the eyes from those frequently adopted experimental species and profiles the well-established animal models of ocular diseases. We also summarize the ophthalmic biomacromolecules launched on the market and put emphasis on emerging noninvasive intraocular delivery strategies of peptides, proteins and genes.
Collapse
Affiliation(s)
- Xingyan Fan
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Kuan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China; Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200030, P.R. China
| | - Feiyang Geng
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China; The Institutes of Integrative Medicine of Fudan University, Shanghai, 200040, PR China
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China; The Institutes of Integrative Medicine of Fudan University, Shanghai, 200040, PR China; Shanghai Engineering Research Center of ImmunoTherapeutics, Shanghai, 201203, PR China.
| |
Collapse
|
12
|
DOPE/CHEMS-Based EGFR-Targeted Immunoliposomes for Docetaxel Delivery: Formulation Development, Physicochemical Characterization and Biological Evaluation on Prostate Cancer Cells. Pharmaceutics 2023; 15:pharmaceutics15030915. [PMID: 36986777 PMCID: PMC10052572 DOI: 10.3390/pharmaceutics15030915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Docetaxel (DTX) is a non-selective antineoplastic agent with low solubility and a series of side effects. The technology of pH-sensitive and anti-epidermal growth factor receptor (anti-EGFR) immunoliposomes aims to increase the selective delivery of the drug in the acidic tumor environment to cells with EFGR overexpression. Thus, the study aimed to develop pH-sensitive liposomes based on DOPE (dioleoylphosphatidylethanolamine) and CHEMS (cholesteryl hemisuccinate), using a Box–Behnken factorial design. Furthermore, we aimed to conjugate the monoclonal antibody cetuximab onto liposomal surface, as well as to thoroughly characterize the nanosystems and evaluate them on prostate cancer cells. The liposomes prepared by hydration of the lipid film and optimized by the Box–Behnken factorial design showed a particle size of 107.2 ± 2.9 nm, a PDI of 0.213 ± 0.005, zeta potential of −21.9 ± 1.8 mV and an encapsulation efficiency of 88.65 ± 20.3%. Together, FTIR, DSC and DRX characterization demonstrated that the drug was properly encapsulated, with reduced drug crystallinity. Drug release was higher in acidic pH. The liposome conjugation with the anti-EGFR antibody cetuximab preserved the physicochemical characteristics and was successful. The liposome containing DTX reached an IC50 at a concentration of 65.74 nM in the PC3 cell line and 28.28 nM in the DU145 cell line. Immunoliposome, in turn, for PC3 cells reached an IC50 of 152.1 nM, and for the DU145 cell line, 12.60 nM, a considerable enhancement of cytotoxicity for the EGFR-positive cell line. Finally, the immunoliposome internalization was faster and greater than that of liposome in the DU145 cell line, with a higher EGFR overexpression. Thus, based on these results, it was possible to obtain a formulation with adequate characteristics of nanometric size, a high encapsulation of DTX and liposomes and particularly immunoliposomes containing DTX, which caused, as expected, a reduction in the viability of prostate cells, with high cellular internalization in EGFR overexpressing cells.
Collapse
|
13
|
Hung J, Perez SM, Dasa SSK, Hall SP, Heckert DB, Murphy BP, Crawford HC, Kelly KA, Brinton LT. A Bitter Taste Receptor as a Novel Molecular Target on Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma. Pharmaceuticals (Basel) 2023; 16:389. [PMID: 36986488 PMCID: PMC10058050 DOI: 10.3390/ph16030389] [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: 12/03/2022] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) execute diverse and complex functions in cancer progression. While reprogramming the crosstalk between CAFs and cancer epithelial cells is a promising avenue to evade the adverse effects of stromal depletion, drugs are limited by their suboptimal pharmacokinetics and off-target effects. Thus, there is a need to elucidate CAF-selective cell surface markers that can improve drug delivery and efficacy. Here, functional proteomic pulldown with mass spectrometry was used to identify taste receptor type 2 member 9 (TAS2R9) as a CAF target. TAS2R9 target characterization included binding assays, immunofluorescence, flow cytometry, and database mining. Liposomes conjugated to a TAS2R9-specific peptide were generated, characterized, and compared to naked liposomes in a murine pancreatic xenograft model. Proof-of-concept drug delivery experiments demonstrate that TAS2R9-targeted liposomes bind with high specificity to TAS2R9 recombinant protein and exhibit stromal colocalization in a pancreatic cancer xenograft model. Furthermore, the delivery of a CXCR2 inhibitor by TAS2R9-targeted liposomes significantly reduced cancer cell proliferation and constrained tumor growth through the inhibition of the CXCL-CXCR2 axis. Taken together, TAS2R9 is a novel cell-surface CAF-selective target that can be leveraged to facilitate small-molecule drug delivery to CAFs, paving the way for new stromal therapies.
Collapse
Affiliation(s)
- Jessica Hung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Siva Sai Krishna Dasa
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | - Howard C. Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, MI 48202, USA
| | - Kimberly A. Kelly
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- ZielBio Inc., Charlottesville, VA 22902, USA
| | - Lindsey T. Brinton
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- ZielBio Inc., Charlottesville, VA 22902, USA
| |
Collapse
|
14
|
Huang H, Yi X, Wei Q, Li M, Cai X, Lv Y, Weng L, Mao Y, Fan W, Zhao M, Weng Z, Zhao Q, Zhao K, Cao M, Chen J, Cao P. Edible and cation-free kiwi fruit derived vesicles mediated EGFR-targeted siRNA delivery to inhibit multidrug resistant lung cancer. J Nanobiotechnology 2023; 21:41. [PMID: 36740689 PMCID: PMC9901103 DOI: 10.1186/s12951-023-01766-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/03/2023] [Indexed: 02/07/2023] Open
Abstract
Clinically, activated EGFR mutation associated chemo-drugs resistance has severely threaten NSCLC patients. Nanoparticle based small interfering RNA (siRNA) therapy representing another promising alternative by silencing specific gene while still suffered from charge associated toxicity, strong immunogenicity and poor targetability. Herein, we reported a novel EGFR-mutant NSCLC therapy relying on edible and cation-free kiwi-derived extracellular vesicles (KEVs), which showed sevenfold enhancement of safe dosage compared with widely used cationic liposomes and could be further loaded with Signal Transducer and Activator of Transcription 3 interfering RNA (siSTAT3). siSTAT3 loaded KEVs (STAT3/KEVs) could be easily endowed with EGFR targeting ability (STAT3/EKEVs) and fluorescence by surface modification with tailor-making aptamer through hydrophobic interaction. STAT3/EKEVs with a controlled size of 186 nm displayed excellent stability, high specificity and good cytotoxicity towards EGFR over-expressing and mutant PC9-GR4-AZD1 cells. Intriguingly, the systemic administration of STAT3/EKEVs significantly suppressed subcutaneous PC9-GR4-AZD1 tumor xenografts in nude mice by STAT3 mediated apoptosis. This safe and robust KEVs has emerged as the next generation of gene delivery platform for NSCLC therapy after multiple drug-resistance.
Collapse
Affiliation(s)
- Haoying Huang
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China ,grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028 Jiangsu China
| | - Xiaohan Yi
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China ,grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028 Jiangsu China
| | - Qingyun Wei
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China ,grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028 Jiangsu China
| | - Mengyuan Li
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China
| | - Xueting Cai
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China ,grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028 Jiangsu China
| | - Yan Lv
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China
| | - Ling Weng
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China
| | - Yujie Mao
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China
| | - Weiwei Fan
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028 Jiangsu China
| | - Mengmeng Zhao
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China
| | - Zhongpei Weng
- Gaoyou Hospital of Traditional Chinese Medicine, Yangzhou, 225600 Jiangsu China
| | - Qing Zhao
- grid.411866.c0000 0000 8848 7685Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378 China
| | - Kewei Zhao
- grid.411866.c0000 0000 8848 7685Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378 China
| | - Meng Cao
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China ,grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028 Jiangsu China
| | - Jing Chen
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China
| | - Peng Cao
- grid.410745.30000 0004 1765 1045School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023 China ,grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028 Jiangsu China ,Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212000 China ,Haihe Laboratory of Modern Chinese Medicine, Jinghai District, No.10 Poyanghu Road, 301617 Tianjin, China
| |
Collapse
|
15
|
Kim KR, Lee AS, Kim SM, Heo HR, Kim CS. Virus-like nanoparticles as a theranostic platform for cancer. Front Bioeng Biotechnol 2023; 10:1106767. [PMID: 36714624 PMCID: PMC9878189 DOI: 10.3389/fbioe.2022.1106767] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Virus-like nanoparticles (VLPs) are natural polymer-based nanomaterials that mimic viral structures through the hierarchical assembly of viral coat proteins, while lacking viral genomes. VLPs have received enormous attention in a wide range of nanotechnology-based medical diagnostics and therapies, including cancer therapy, imaging, and theranostics. VLPs are biocompatible and biodegradable and have a uniform structure and controllable assembly. They can encapsulate a wide range of therapeutic and diagnostic agents, and can be genetically or chemically modified. These properties have led to sophisticated multifunctional theranostic platforms. This article reviews the current progress in developing and applying engineered VLPs for molecular imaging, drug delivery, and multifunctional theranostics in cancer research.
Collapse
Affiliation(s)
- Kyeong Rok Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Ae Sol Lee
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Su Min Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Hye Ryoung Heo
- Senotherapy-Based Metabolic Disease Control Research Center, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| | - Chang Sup Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea,School of Chemistry and Biochemistry, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| |
Collapse
|
16
|
Rethi L, Mutalik C, Anurogo D, Lu LS, Chu HY, Yougbaré S, Kuo TR, Cheng TM, Chen FL. Lipid-Based Nanomaterials for Drug Delivery Systems in Breast Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2948. [PMID: 36079985 PMCID: PMC9458017 DOI: 10.3390/nano12172948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 06/15/2023]
Abstract
Globally, breast cancer is one of the most prevalent diseases, inducing critical intimidation to human health. Lipid-based nanomaterials have been successfully demonstrated as drug carriers for breast cancer treatment. To date, the development of a better drug delivery system based on lipid nanomaterials is still urgent to make the treatment and diagnosis easily accessible to breast cancer patients. In a drug delivery system, lipid nanomaterials have revealed distinctive features, including high biocompatibility and efficient drug delivery. Specifically, a targeted drug delivery system based on lipid nanomaterials has inherited the advantage of optimum dosage and low side effects. In this review, insights on currently used potential lipid-based nanomaterials are collected and introduced. The review sheds light on conjugation, targeting, diagnosis, treatment, and clinical significance of lipid-based nanomaterials to treat breast cancer. Furthermore, a brighter side of lipid-based nanomaterials as future potential drug delivery systems for breast cancer therapy is discussed.
Collapse
Affiliation(s)
- Lekshmi Rethi
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chinmaya Mutalik
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Dito Anurogo
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan or
- Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Makassar, Makassar City 90221, South Sulawesi, Indonesia
| | - Long-Sheng Lu
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Yi Chu
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Sibidou Yougbaré
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro BP 218, 11, Burkina Faso
| | - Tsung-Rong Kuo
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsai-Mu Cheng
- Graduate Institute of Translational Medicine, College of Medicine and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Fu-Lun Chen
- Department of Internal Medicine, Division of Infectious Diseases, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
17
|
Wei H, Chen F, Chen J, Lin H, Wang S, Wang Y, Wu C, Lin J, Zhong G. Mesenchymal Stem Cell Derived Exosomes as Nanodrug Carrier of Doxorubicin for Targeted Osteosarcoma Therapy via SDF1-CXCR4 Axis. Int J Nanomedicine 2022; 17:3483-3495. [PMID: 35959282 PMCID: PMC9359454 DOI: 10.2147/ijn.s372851] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose The objective of this study was to investigate the antitumor activity, targeting capability, and mechanism of the developed nanodrug consisting of doxorubicin and exosome (Exo-Dox) derived from mesenchymal stem cells in vitro and in vivo. Methods The exosomes were isolated with Exosome Isolation Kit, and the Exo-Dox was prepared by mixing exosome with Dox-HCl, desalinizing with triethylamine and then dialyzing against PBS overnight. The exosome and Exo-Dox were examined by nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM). The antitumor activity, targeting capability, and mechanism of the developed Exo-Dox were evaluated by cell viability assay, histological and immunofluorescence analysis and in vivo imaging system. Results NTA results showed the size of the exosomes had increased from 141.6 nm to 178.1 nm after loading with doxorubicin. Compared with free Dox, the Exo-Dox exhibited higher cytotoxicity against osteosarcoma MG63 cells, HOS cells, and 143B cells than free Dox, the half-maximal inhibitory concentrations (IC50) of Dox, Exo-Dox were calculated to be 0.178 and 0.078 μg mL−1 in MG63 cells, 0.294 and 0.109μg mL−1 in HOS cells, 0.315 and 0.123 μg mL−1 in 143B cells, respectively. The in vivo imaging showed that MSC derived Exo could serve as a highly efficient delivery vehicle for targeted drug delivery. The immunohistochemistry and histology analysis indicated that compared with the free Dox group, the Ki67-positive cells and cardiotoxicity in Exo-Dox group were significantly decreased. Conclusion Our results suggested that MSC-derived Exo could be excellent nanocarriers used to deliver chemotherapeutic drug Dox specifically and efficiently in osteosarcoma, resulting in enhanced toxicity against osteosarcoma and less toxicity in heart tissue. We further demonstrated the targeting capability of Exo was due to the chemotaxis of MSC-derived exosomes to osteosarcoma cells via SDF1-CXCR4 axis.
Collapse
Affiliation(s)
- Hongxiang Wei
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Fei Chen
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Jinyuan Chen
- The Centralab, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Huangfeng Lin
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Shenglin Wang
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Yunqing Wang
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Chaoyang Wu
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Jianhua Lin
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Guangxian Zhong
- Department of Orthopaedics, Fujian Institute of Orthopaedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Correspondence: Guangxian Zhong; Jianhua Lin, Tel/Fax +86 591 87981029, Email ;
| |
Collapse
|
18
|
Nano- and Crystal Engineering Approaches in the Development of Therapeutic Agents for Neoplastic Diseases. CRYSTALS 2022. [DOI: 10.3390/cryst12070926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer is a leading cause of death worldwide. It is a global quandary that requires the administration of many different active pharmaceutical ingredients (APIs) with different characteristics. As is the case with many APIs, cancer treatments exhibit poor aqueous solubility which can lead to low drug absorption, increased doses, and subsequently poor bioavailability and the occurrence of more adverse events. Several strategies have been envisaged to overcome this drawback, specifically for the treatment of neoplastic diseases. These include crystal engineering, in which new crystal structures are formed to improve drug physicochemical properties, and/or nanoengineering in which the reduction in particle size of the pristine crystal results in much improved physicochemical properties. Co-crystals, which are supramolecular complexes that comprise of an API and a co-crystal former (CCF) held together by non-covalent interactions in crystal lattice, have been developed to improve the performance of some anti-cancer drugs. Similarly, nanosizing through the formation of nanocrystals and, in some cases, the use of both crystal and nanoengineering to obtain nano co-crystals (NCC) have been used to increase the solubility as well as overall performance of many anticancer drugs. The formulation process of both micron and sub-micron crystalline formulations for the treatment of cancers makes use of relatively simple techniques and minimal amounts of excipients aside from stabilizers and co-formers. The flexibility of these crystalline formulations with regards to routes of administration and ability to target neoplastic tissue makes them ideal strategies for effectiveness of cancer treatments. In this review, we describe the use of crystalline formulations for the treatment of various neoplastic diseases. In addition, this review attempts to highlight the gaps in the current translation of these potential treatments into authorized medicines for use in clinical practice.
Collapse
|
19
|
Chavda VP, Vihol D, Mehta B, Shah D, Patel M, Vora LK, Pereira-Silva M, Paiva-Santos AC. Phytochemical-loaded liposomes for anticancer therapy: an updated review. Nanomedicine (Lond) 2022; 17:547-568. [PMID: 35259920 DOI: 10.2217/nnm-2021-0463] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The major obstacles observed in current chemotherapy are severe adverse effects, narrow therapeutic indexes and multidrug resistance. Anticancer phytochemicals are extracted and purified from natural plants, providing alternative therapeutic approaches with recognized biomedical benefits. However, poor bioavailability, high dose requirements and non-specific targeting have made those molecules less effective. To tackle those issues, liposomal nanovesicles for phytochemical delivery are taken into consideration for improving the therapeutic effectiveness by increasing transportation across cell barriers and conferring attractive cancer-specific targeting capabilities. In the present review, the liposomal approaches of anticancer phytochemicals are discussed, and recent advances in these formulations applied to cancer phytotherapy are further reviewed by an informed approach.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics & Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Disha Vihol
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Bhavya Mehta
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Dhruvil Shah
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Manan Patel
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| |
Collapse
|
20
|
Wang W, Joyce P, Bremmell K, Milne R, Prestidge CA. Liposomal 5-Fluorouracil Polymer Complexes Facilitate Tumor-Specific Delivery: Pharmaco-Distribution Kinetics Using Microdialysis. Pharmaceutics 2022; 14:pharmaceutics14020221. [PMID: 35213954 PMCID: PMC8878722 DOI: 10.3390/pharmaceutics14020221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Liposomes are widely used as carriers for anticancer drugs due to their ability to prolong the retention of encapsulated drugs in blood plasma while directing their distribution increasingly into tumor tissue. We report on the development of stealth liposomal formulations for the common chemotherapy drug 5-fluorouracil, where pharmacokinetic studies were undertaken using a microdialysis probe to specifically quantify drug accumulation in tumor, which was contrasted to drug exposure to healthy tissue. Greater accumulation of the drug into the tumor than into healthy subcutaneous tissue was observed for neutral and cationic liposomal 5-fluorouracil polymer complexes in comparison to the conventional delivery by an injected solution. Increased drug accumulation in tumor also correlated to reduced tumor growth. This research has generated new mechanistic insight into liposomal-specific delivery to tumors with potential to improve the efficacy and reduce the toxicity of chemotherapy.
Collapse
|
21
|
Spray drying: Inhalable powders for pulmonary gene therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 133:112601. [DOI: 10.1016/j.msec.2021.112601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/04/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022]
|
22
|
Safety and Efficacy Evaluation In Vivo of a Cationic Nucleolipid Nanosystem for the Nanodelivery of a Ruthenium(III) Complex with Superior Anticancer Bioactivity. Cancers (Basel) 2021; 13:cancers13205164. [PMID: 34680314 PMCID: PMC8534243 DOI: 10.3390/cancers13205164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary The availability of selective, effective, and safe anticancer agents is a major challenge in the field of cancer research. As part of a multidisciplinary research project, in recent years our group has proposed an original class of nanomaterials for the delivery of new anticancer drugs based on ruthenium(III) complexes. In cellular models, these nanosystems have been shown to be effective in counteracting growth and proliferation of human breast cancer cells. Compared to conventional metallochemotherapeutics such as platinum-based agents whose clinical practice is associated with serious undesirable effects, ruthenium complexes share improved biochemical profiles making them more selective towards cancer cells and less cytotoxic to healthy cells. Their combination with biocompatible nanocarriers further enhances these promising features, as here showcased by our research carried out in an animal model which underscores the efficacy and safety in vivo of one of our most promising ruthenium-based nanosystems. Abstract Selectivity and efficacy towards target cancer cells, as well as biocompatibility, are current challenges of advanced chemotherapy powering the discovery of unconventional metal-based drugs and the search for novel therapeutic approaches. Among second-generation metal-based chemotherapeutics, ruthenium complexes have demonstrated promising anticancer activity coupled to minimal toxicity profiles and peculiar biochemical features. In this context, our research group has recently focused on a bioactive Ru(III) complex—named AziRu—incorporated into a suite of ad hoc designed nucleolipid nanosystems to ensure its chemical stability and delivery. Indeed, we proved that the structure and properties of decorated nucleolipids can have a major impact on the anticancer activity of the ruthenium core. Moving in this direction, here we describe a preclinical study performed by a mouse xenograft model of human breast cancer to establish safety and efficacy in vivo of a cationic Ru(III)-based nucleolipid formulation, named HoThyRu/DOTAP, endowed with superior antiproliferative activity. The results show a remarkable reduction in tumour with no evidence of animal suffering. Blood diagnostics, as well as biochemical analysis in both acute and chronic treated animal groups, demonstrate a good tolerability profile at the therapeutic regimen, with 100% of mice survival and no indication of toxicity. In addition, ruthenium plasma concentration analysis and tissue bioaccumulation were determined via appropriate sampling and ICP-MS analysis. Overall, this study supports both the efficacy of our Ru-containing nanosystem versus a human breast cancer model and its safety in vivo through well-tolerated animal biological responses, envisaging a possible forthcoming use in clinical trials.
Collapse
|
23
|
Hani U, M. YB, Wahab S, Siddiqua A, Osmani RAM, Rahamathulla M. A Comprehensive Review of Current Perspectives on Novel Drug Delivery Systems and Approaches for Lung Cancer Management. J Pharm Innov 2021. [DOI: 10.1007/s12247-021-09582-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Nakhaei P, Margiana R, Bokov DO, Abdelbasset WK, Jadidi Kouhbanani MA, Varma RS, Marofi F, Jarahian M, Beheshtkhoo N. Liposomes: Structure, Biomedical Applications, and Stability Parameters With Emphasis on Cholesterol. Front Bioeng Biotechnol 2021; 9:705886. [PMID: 34568298 PMCID: PMC8459376 DOI: 10.3389/fbioe.2021.705886] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Liposomes are essentially a subtype of nanoparticles comprising a hydrophobic tail and a hydrophilic head constituting a phospholipid membrane. The spherical or multilayered spherical structures of liposomes are highly rich in lipid contents with numerous criteria for their classification, including structural features, structural parameters, and size, synthesis methods, preparation, and drug loading. Despite various liposomal applications, such as drug, vaccine/gene delivery, biosensors fabrication, diagnosis, and food products applications, their use encounters many limitations due to physico-chemical instability as their stability is vigorously affected by the constituting ingredients wherein cholesterol performs a vital role in the stability of the liposomal membrane. It has well established that cholesterol exerts its impact by controlling fluidity, permeability, membrane strength, elasticity and stiffness, transition temperature (Tm), drug retention, phospholipid packing, and plasma stability. Although the undetermined optimum amount of cholesterol for preparing a stable and controlled release vehicle has been the downside, but researchers are still focused on cholesterol as a promising material for the stability of liposomes necessitating explanation for the stability promotion of liposomes. Herein, the prior art pertaining to the liposomal appliances, especially for drug delivery in cancer therapy, and their stability emphasizing the roles of cholesterol.
Collapse
Affiliation(s)
- Pooria Nakhaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Depok, Indonesia
- Cipto Mangunkusumo Hospital, The National Referral Hospital, Central Jakarta, Indonesia
- Master’s Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Depok, Indonesia
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology, and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Czechia
| | - Rajender S. Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University in Olomouc, Olomouc, Czechia
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Czechia
| |
Collapse
|
25
|
Kashapov R, Ibragimova A, Pavlov R, Gabdrakhmanov D, Kashapova N, Burilova E, Zakharova L, Sinyashin O. Nanocarriers for Biomedicine: From Lipid Formulations to Inorganic and Hybrid Nanoparticles. Int J Mol Sci 2021; 22:7055. [PMID: 34209023 PMCID: PMC8269010 DOI: 10.3390/ijms22137055] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Encapsulation of cargoes in nanocontainers is widely used in different fields to solve the problems of their solubility, homogeneity, stability, protection from unwanted chemical and biological destructive effects, and functional activity improvement. This approach is of special importance in biomedicine, since this makes it possible to reduce the limitations of drug delivery related to the toxicity and side effects of therapeutics, their low bioavailability and biocompatibility. This review highlights current progress in the use of lipid systems to deliver active substances to the human body. Various lipid compositions modified with amphiphilic open-chain and macrocyclic compounds, peptide molecules and alternative target ligands are discussed. Liposome modification also evolves by creating new hybrid structures consisting of organic and inorganic parts. Such nanohybrid platforms include cerasomes, which are considered as alternative nanocarriers allowing to reduce inherent limitations of lipid nanoparticles. Compositions based on mesoporous silica are beginning to acquire no less relevance due to their unique features, such as advanced porous properties, well-proven drug delivery efficiency and their versatility for creating highly efficient nanomaterials. The types of silica nanoparticles, their efficacy in biomedical applications and hybrid inorganic-polymer platforms are the subject of discussion in this review, with current challenges emphasized.
Collapse
Affiliation(s)
- Ruslan Kashapov
- A.E. Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Street 8, 420088 Kazan, Russia; (A.I.); (R.P.); (D.G.); (N.K.); (E.B.); (L.Z.); (O.S.)
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Wu Q, Ou H, Shang Y, Zhang X, Wu J, Fan F. Nanoscale Formulations: Incorporating Curcumin into Combination Strategies for the Treatment of Lung Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2695-2709. [PMID: 34188448 PMCID: PMC8232383 DOI: 10.2147/dddt.s311107] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer remains the most common cancer worldwide. Although significant advances in screening have been made and early diagnosis strategies and therapeutic regimens have been developed, the overall survival rate remains bleak. Curcumin is extracted from the rhizomes of turmeric and exhibits a wide range of biological activities. In lung cancer, evidence has shown that curcumin can markedly inhibit tumor growth, invasion and metastasis, overcome resistance to therapy, and even eliminate cancer stem cells (CSCs). Herein, the underlying molecular mechanisms of curcumin were summarized by distinct biological processes. To solve the limiting factors that curtail the clinical applications of curcumin, nanoformulations encapsulating curcumin were surveyed in detail. Nanoparticles, including liposomes, micelles, carbon nanotubes (CNTs), solid lipid nanoparticles (SLNs), nanosuspensions, and nanoemulsions, were explored as proper carriers of curcumin. Moreover, it was firmly verified that curcumin has the ability to sensitize lung cancer cells to chemotherapeutic drugs, such as cisplatin and docetaxel, and to various targeted therapies. Regarding the advantages and drawbacks of curcumin, we concluded that combination therapy based on nanoparticles would be the optimal approach to broaden the application of curcumin in the clinic in the near future.
Collapse
Affiliation(s)
- Quhui Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Huiping Ou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Yan Shang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Xi Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Junyong Wu
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Fuyuan Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| |
Collapse
|
27
|
de Oliveira TL, Bavia L, Fontana PD, Cruz LS, Paludo KS, Crisma AR, Messias-Reason IJ, Beltrame FL. Immunomodulatory and cytotoxic activities of euphol. Life Sci 2021; 280:119700. [PMID: 34111465 DOI: 10.1016/j.lfs.2021.119700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 12/01/2022]
Abstract
AIMS This study evaluated the effect of euphol isolated from Euphorbia umbellata (Pax) Bruyns latex on the activation of complement pathways (classical (CP), alternative (AP) and lectin (LP)), neutrophil chemotaxis, cytotoxic activity, cell morphology and death in HRT-18 and 3T3 cells lines. MAIN METHODS CP and AP were assessed using hemolytic assays and ELISA for LP; neutrophil chemotaxis was performed using Boyden's chamber; cytotoxicity was evaluated by neutral red methodology and characteristics of cell death were assessed by cell morphology with hematological staining. KEY FINDINGS Although euphol increased CP activation (38% at a concentration of 976.1 μM), an inhibitory effect on AP, LP (31% and 32% reduction in the concentration of 976.1 μM) and neutrophil chemotaxis (inhibit 84% of neutrophil migration at a concentration 292.9 μM) was observed. In addiction euphol was able to induce significant cell death in a time-dependent manner, presenting an IC50 of 70.8 μM and 39.2 μM for HRT-18 and 3T3 cell lines respectively and it was also observed apoptotic characteristics as cellular rounding, chromatin condensation and blebs formation for both cell lines. SIGNIFICANCE Euphol has a potential use for the treatment of complement-related inflammatory diseases due to its ability to downregulate inflammation. On the other hand, the controlled activation of CP can contribute to complement-dependent cytotoxicity in the context of monoclonal antibody-based cancer treatment.
Collapse
Affiliation(s)
- Thais Latansio de Oliveira
- Laboratory of Molecular Immunopathology, Clinical Hospital, Federal University of Paraná, Curitiba, Brazil.
| | - Lorena Bavia
- Laboratory of Molecular Immunopathology, Clinical Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Pâmela Dias Fontana
- Laboratory of Molecular Immunopathology, Clinical Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Luiza Stolz Cruz
- Laboratory of Phytotherapy, Phytotherapy Technology and Chemistry of Natural Products, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Katia Sabrina Paludo
- Multidisciplinary Laboratory of Biological Sciences and Health, State University of Ponta Grossa, Ponta Grossa, Brazil
| | | | - Iara Jose Messias-Reason
- Laboratory of Molecular Immunopathology, Clinical Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Flávio Luís Beltrame
- Laboratory of Phytotherapy, Phytotherapy Technology and Chemistry of Natural Products, State University of Ponta Grossa, Ponta Grossa, Brazil
| |
Collapse
|
28
|
Roy S, Mukherjee P, Das PK, Ghosh PR, Datta P, Kundu B, Nandi SK. Local delivery systems of morphogens/biomolecules in orthopedic surgical challenges. MATERIALS TODAY COMMUNICATIONS 2021; 27:102424. [DOI: 10.1016/j.mtcomm.2021.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
29
|
Fu L, Zhou X, He C. Polymeric Nanosystems for Immunogenic Cell Death-Based Cancer Immunotherapy. Macromol Biosci 2021; 21:e2100075. [PMID: 33885225 DOI: 10.1002/mabi.202100075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Immunotherapy has pointed out a scientific and promising direction for cancer treatment through the rouse of immunosurveillance and the decrease of possible side effects in recent years. In immunotherapy, immunogenic cancer cell death (ICD) plays a critical role in regulating anti-cancer immune system in vivo via the release of damage-associated molecular patterns. ICD can not only induce in situ cancer cells apoptosis, but also arouse the immune response against metastatic tumors, which is of great clinical significance to eradicate tumors. In cancer immunotherapy, polymer nanoparticles have drawn increasing attention as an important component of ICD-based immunotherapy attributing to their controllable size, excellent biocompatibility, promising ability of protecting cargo from surrounding environment, which delivers the antigens or immune inducers to antigen-presenting cells, and further triggers sinnvoll T cell response. In this review, the recent advances in the development of polymeric material-based nanosystems for ICD-mediated cancer immunotherapy are summarized. The mechanism of ICD and some current restrictions inhibiting the efficiency of immunotherapy and future prospects are also discussed.
Collapse
Affiliation(s)
- Liwen Fu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| |
Collapse
|
30
|
Liu Z, Chu W, Sun Q, Zhao L, Tan X, Zhang Y, Yin T, He H, Gou J, Tang X. Micelle-contained and PEGylated hybrid liposomes of combined gemcitabine and cisplatin delivery for enhancing antitumor activity. Int J Pharm 2021; 602:120619. [PMID: 33887396 DOI: 10.1016/j.ijpharm.2021.120619] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/30/2021] [Accepted: 04/15/2021] [Indexed: 12/25/2022]
Abstract
Combination, synergistic chemotherapy with gemcitabine (GEM) and cisplatin (CDDP) is a common strategy, and has been recommended for tumor treatment due to its promoted therapeutic effect and reduced systemic toxicity. However, this process involves the intravenous infusion of GEM prior to that of CDDP, which is inconvenient for patients and staff. Here, a novel hybrid nano-carrier system comprised of micelles encapsulated within PEGylated liposomes is proposed, in order to combine the unique strengths of each component. CDDP was bonded with PLG-PEG, and then the formed CDDP@PLG-PEG micelles and GEM were co-loaded inside PEGylated liposomes. The hybrid liposomes with the optimized GEM/CDDP ratio (1:0.6) showed a roughly spherical morphology, appropriate drug loading, and sustained release behavior. In vitro, the hybrid liposomes had 1.72-fold increased cellular uptake, and 57.42%-fold decreased IC50 value. In vivo, pharmacokinetic studies showed increased t1/2 values (125.64%- and 128.57%-folds for GEM and CDDP), decreased clearance (41.90%- and 2.37%-folds), and promoted AUC (262.76%- and 4577.24%-folds). Finally, an in vivo antitumor study showed effective activity in regards to lung tumor size and weight, which were 40.48%- and 33.11%-folds that of GEM/CDDP solution. In summary, we demonstrated the development of an effective micelle-containing PEGylated hybrid liposomes for combined GEM/CDDP delivery.
Collapse
Affiliation(s)
- Zixu Liu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Wei Chu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Qianhe Sun
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Linxuan Zhao
- Department of Pharmaceutics, College of Pharmacy Sciences, Jilin University, Changchun 130021, China
| | - Xinyi Tan
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Yu Zhang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Tian Yin
- Department of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Haibing He
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Jingxin Gou
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China.
| | - Xing Tang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China.
| |
Collapse
|
31
|
Quadros M, Momin M, Verma G. Design strategies and evolving role of biomaterial assisted treatment of osteosarcoma. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111875. [PMID: 33579498 DOI: 10.1016/j.msec.2021.111875] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/16/2022]
Abstract
Osteosarcoma is the most commonly diagnosed form of bone cancer. It is characterized by a high risk of developing lung metastasis as the disease progresses. Standard treatment includes combination of surgical intervention, chemotherapy and radiotherapy. However, the non-specificity of potent chemotherapeutic agents often leads to major side effects. In this review, we discuss the role of various classes of biomaterials, including both organic as well as inorganic in realizing the local and systemic delivery of therapeutic agents like drugs, radioisotopes and even gene silencing agents to treat osteosarcoma. Biomaterial assisted unconventional therapies such as targeted therapy, nanotherapy, magnetic hyperthermia, gene therapy, photothermal and photodynamic therapies are also being explored. A wide variety of biomaterials including lipids, carbon-based materials, polymers, silica, bioactive glass, hydroxyapatite and metals are designed as delivery systems with the desired loading efficiency, release profile, and on-demand delivery. Among others, liposomal carriers have attracted a great deal of attention due to their capability to encapsulate both hydrophobic and hydrophilic drugs. Polymeric systems have high drug loading efficiency and stability and can even be tailored to achieve desired size and physiochemical properties. Carbon-based systems can also be seen as an upcoming class of therapeutics with great potential in treating different types of cancer. Inorganic materials like silica nanoparticles have high drug payload owing to their mesoporous structure. On the other hand, ceramic materials like bioactive glass and hydroxyapatite not only act as excellent delivery vectors but also participate in osteo-regeneration activity. These multifunctional biomaterials are also being investigated for their theranostic abilities to monitor cancer ablation. This review systematically discusses the vast landscape of biomaterials along with their challenges and respective opportunities for osteosarcoma therapy.
Collapse
Affiliation(s)
- Mural Quadros
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, First floor, V M Road, Vile Parle West, Mumbai, Maharashtra 400 056, India; Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, First floor, V M Road, Vile Parle West, Mumbai, Maharashtra 400 056, India.
| | - Gunjan Verma
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar 400 094, India.
| |
Collapse
|
32
|
Ye H, Chu X, Cao Z, Hu X, Wang Z, Li M, Wan L, Li Y, Cao Y, Diao Z, Peng F, Liu J, Xu L. A Novel Targeted Therapy System for Cervical Cancer: Co-Delivery System of Antisense LncRNA of MDC1 and Oxaliplatin Magnetic Thermosensitive Cationic Liposome Drug Carrier. Int J Nanomedicine 2021; 16:1051-1066. [PMID: 33603368 PMCID: PMC7886386 DOI: 10.2147/ijn.s258316] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND This study was aimed to prepare a novel magnetic thermosensitive cationic liposome drug carrier for the codelivery of Oxaliplatin (OXA) and antisense lncRNA of MDC1 (MDC1-AS) to Cervical cancer cells and evaluate the efficiency of this drug carrier and its antitumor effects on Cervical cancer. METHODS Thermosensitive magnetic cationic liposomes were prepared using thin-film hydration method. The OXA and MDC1-AS vectors were loaded into the codelivery system, and the in vitro OXA thermosensitive release activity, efficiency of MDC1-AS regulating MDC1, in vitro cytotoxicity, and in vivo antitumor activity were determined. RESULTS The codelivery system had desirable targeted delivery efficacy, OXA thermosensitive release, and MDC1-AS regulating MDC1. Codelivery of OXA and MDC1-AS enhanced the inhibition of cervical cancer cell growth in vitro and in vivo, compared with single drug delivery. CONCLUSION The novel codelivery of OXA and MDC1-AS magnetic thermosensitive cationic liposome drug carrier can be applied in the combined chemotherapy and gene therapy for cervical cancer.
Collapse
Affiliation(s)
- Hui Ye
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Xiaoying Chu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Zhensheng Cao
- School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Xuanxuan Hu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Zihan Wang
- School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Meiqi Li
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Leyu Wan
- School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Yongping Li
- Department of Surgery, Chengdu Shuangliu District Maternal and Child Health Hospital, ChengDu, Sichuan, 610200, People’s Republic of China
| | - Yongge Cao
- Department of Stomatology, Haiyuan College, Kunming, Yunnan, 650106, People’s Republic of China
| | - Zhanqiu Diao
- School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Fengting Peng
- School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Jinsong Liu
- School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Lihua Xu
- Department of General Medicine, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| |
Collapse
|
33
|
Liu D, Zhang Q, Wang J, Guan S, Cai D, Liu J. Inhibition of growth and metastasis of breast cancer by targeted delivery of 17-hydroxy-jolkinolide B via hyaluronic acid-coated liposomes. Carbohydr Polym 2021; 257:117572. [PMID: 33541631 DOI: 10.1016/j.carbpol.2020.117572] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/11/2020] [Accepted: 12/25/2020] [Indexed: 01/01/2023]
Abstract
Hyaluronic acid (HA)-coated liposomes were designed for the targeted delivery of 17-hydroxy-jolkinolide B (HA-Lip-HJB). HA-Lip-HJB had a particle size of 130.8 ± 1.9 nm, zeta potential of -52.36 ± 1.91 mV, and encapsulation efficiency of 89.2 ± 1.5 %. In vitro cell experiments indicated that modification of HA-Lip-HJB increased its cytotoxicity and cellular uptake via CD44 receptor-mediated endocytosis pathway. Of particular importance is that HA-Lip-HJB suppressed cell migration and invasion by inhibiting epithelial-mesenchymal transition (EMT) process. Moreover, the HA-Lip-HJB displayed notable growth inhibition on tumor spheroids. Furthermore, in vivo tissue distribution and anti-tumor experiments carried on BALB/C mice bearing 4T1 tumor indicated that HA-Lip-HJB had strong tumor targeting and tumor suppression abilities. The results also demonstrated that HA-Lip-HJB inhibited tumor cells migration and colonization on the lung. Therefore, HA-Lip-HJB is a promising formulation for metastatic breast cancer therapy.
Collapse
Affiliation(s)
- Dan Liu
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Qi Zhang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Jing Wang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Shuang Guan
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Defu Cai
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Jicheng Liu
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| |
Collapse
|
34
|
Jiménez-López J, Bravo-Caparrós I, Cabeza L, Nieto FR, Ortiz R, Perazzoli G, Fernández-Segura E, Cañizares FJ, Baeyens JM, Melguizo C, Prados J. Paclitaxel antitumor effect improvement in lung cancer and prevention of the painful neuropathy using large pegylated cationic liposomes. Biomed Pharmacother 2021; 133:111059. [PMID: 33378963 DOI: 10.1016/j.biopha.2020.111059] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Paclitaxel (PTX), a drug widely used in lung cancer, has serious limitations including the development of peripheral neurotoxicity, which may lead to treatment discontinuation and therapy failure. The transport of PTX in large cationic liposomes could avoid this undesirable effect, improving the patient's prognosis. PTX was encapsulated in cationic liposomes with two different sizes, MLV (180-200 nm) and SUV (80-100 nm). In both cases, excellent biocompatibility and improved internalization and antitumor effect of PTX were observed in human and mice lung cancer cells in culture, multicellular spheroids and cancer stem cells (CSCs). In addition, both MLV and SUV with a polyethylene glycol (PEG) shell, induced a greater tumor volume reduction than PTX (56.4 % and 57.1 % vs. 36.7 %, respectively) in mice. Interestingly, MLV-PEG-PTX did not induce either mechanical or heat hypersensitivity whereas SUV-PEG-PTX produced a similar response to free PTX. Analysis of PTX distribution showed a very low concentration of the drug in the dorsal root ganglia (DRG) with MLV-PEG-PTX, but not with SUV-PEG-PTX or free PTX. These results support the hypothesis that PTX induces peripheral neuropathy by penetrating the endothelial fenestrations of the DRG (80-100 nm, measured in mice). In conclusion, our larger liposomes (MLV-PEG-PTX) not only showed biocompatibility, antitumor activity against CSCs, and in vitro and in vivo antitumor effect that improved PTX free activity, but also protected from PTX-induced painful peripheral neuropathy. These advantages could be used as a new strategy of lung cancer chemotherapy to increase the PTX activity and reduce its side effects.
Collapse
Affiliation(s)
- Julia Jiménez-López
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Inmaculada Bravo-Caparrós
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Francisco R Nieto
- Department of Pharmacology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - Raúl Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain
| | - Eduardo Fernández-Segura
- Department of Histology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - Francisco J Cañizares
- Department of Histology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - José M Baeyens
- Department of Pharmacology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain.
| | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| |
Collapse
|
35
|
Rucins M, Smits R, Sipola A, Vigante B, Domracheva I, Turovska B, Muhamadejev R, Pajuste K, Plotniece M, Sobolev A, Duburs G, Plotniece A. Pleiotropic Properties of Amphiphilic Dihydropyridines, Dihydropyridones, and Aminovinylcarbonyl Compounds. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8413713. [PMID: 33488932 PMCID: PMC7790557 DOI: 10.1155/2020/8413713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/09/2020] [Accepted: 11/10/2020] [Indexed: 11/18/2022]
Abstract
Three groups of synthetic lipids are chosen for studies: (1) 1,4-dihydropyridines (1,4-DHPs) containing two cationic moieties and their analogues; (2) 3,4-dihydro-2(1H)-pyridones containing a cationic moiety; and (3) acyclic, open-chain analogues, i.e., 2-amino-3-alkoxycarbonylalkylammonium derivatives. 1,4-DHPs possessing dodecyl alkyl chains in the ester groups in positions 3 and 5 and cationic nitrogen-containing groups in positions 2 and 6 have high cytotoxicity in cancer cells HT-1080 (human lung fibrosarcoma) and MH-22A (mouse hepatoma), but low cytotoxicity in the noncancerous NIH3T3 cells (mouse embryonic fibroblast). On the contrary, similar compounds having short (methyl, ethyl, or propoxyethyl) chains in the ester groups in positions 3 and 5 lack cytotoxicity in the cancer cells HT-1080 and MH-22A even at high doses. Inclusion of fluorine atoms in the alkyl chains in positions 3 and 5 of the DHP cycle decreases the cytotoxicity of the mentioned compounds. Structurally related dihydropyridones with a polar head group are substantially more toxic to normal and cancerous cells than the DHP analogues. Open-chain analogues of DHP lipids comprise the same conjugated aminovinylcarbonyl moiety and possess anticancer activity, but they also have high basal cytotoxicity. Electrochemical oxidation data demonstrate that oxidation potentials of selected compounds are in the range of 1.6-1.7 V for cationic 1,4-DHP, 2.0-2.4 V for cationic 3,4-dihydropyridones, and 1.2-1.5 V for 2-amino-3-alkoxycarbonylalkylammonium derivatives. Furthermore, the tested cationic 1,4-DHP amphiphiles possess antiradical activity. Molecular topological polar surface area values for the tested compounds were defined in accordance with the main fragments of compound structures. The determined logP values were highest for dodecyl ester groups in positions 3 and 5 of the 1,4-DHP and lowest for short alkyl chain-containing amphiphiles.
Collapse
Affiliation(s)
- Martins Rucins
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Rufus Smits
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Anda Sipola
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Brigita Vigante
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Ilona Domracheva
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Baiba Turovska
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Ruslan Muhamadejev
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Karlis Pajuste
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Mara Plotniece
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Riga Stradiņš University, Dzirciema 16, Riga LV-1007, Latvia
| | - Arkadij Sobolev
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Gunars Duburs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Aiva Plotniece
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| |
Collapse
|
36
|
Liu J, Zhang Y, Chen T, Chen H, He H, Jin T, Wang J, Ke Y. Environmentally Self-Adaptative Nanocarriers Suppress Glioma Proliferation and Stemness via Codelivery of shCD163 and Doxorubicin. ACS APPLIED MATERIALS & INTERFACES 2020; 12:52354-52369. [PMID: 33196179 DOI: 10.1021/acsami.0c14288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Gliomas-devastating intracranial tumors with a dismal outcome-are in dire need of innovative treatment. Although nanodrugs have been utilized as a target therapy for certain types of solid tumors, their therapeutic effects in gliomas are limited due to the complications of the systemic circulation, blood-brain barrier (BBB), and specific glioma environment. Thus, we aimed to establish a nanoliposome adaptable to different environments by codelivery of shCD163 and doxorubicin (DOX) to treat gliomas. In this study, we first synthesized pH-sensitive DSPE-cRGD-Hz-PEG2000 to form an environmentally self-adaptative nanoliposome (cRGD-DDD Lip) via a thin film method. We used in vitro BBB models, in vitro cell uptake experiments, and in vivo biodistribution assays to confirm the long circulation time and low cell uptake of the cRGD-DDD Lip as a result of the poly(ethylene glycol) (PEG) shell of cRGD-DDD Lip in the neutral pH systemic circulation. Moreover, the cRGD-DDD Lip bypassed the BBB and attached to the intracranial glioma following the removal of the PEG shell and the exposure of cRGD to the weakly acidic tumor microenvironment. We further assembled the shCD163/DOX@cRGD-DDD Lip through cRGD-DDD Lip loading of shCD163 and DOX. In vitro, cell proliferation and self-renewal of glioma cells were inhibited by the shCD163/DOX@cRGD-DDD Lip due to the toxicity of DOX and the suppression of shCD163 via the CD163 pathway. In vivo, the shCD163/DOX@cRGD-DDD Lip disturbed the progression of in situ gliomas by inhibiting the growth and stemness of glioma cells and prevented the recurrence of gliomas after resection. In conclusion, the cRGD-DDD Lip may be a promising nanodrug-loading platform to cope with different environments and the shCD163/DOX@cRGD-DDD Lip may potentially be a novel nanodrug for glioma therapy.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/therapeutic use
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/metabolism
- Brain Neoplasms/drug therapy
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Glioma/drug therapy
- Glioma/mortality
- Glioma/pathology
- Humans
- Liposomes/chemistry
- Mice
- Mice, Nude
- Nanoparticles/chemistry
- Nanoparticles/metabolism
- Oligopeptides/chemistry
- Polyethylene Glycols/chemistry
- RNA Interference
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/metabolism
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Survival Rate
- Tissue Distribution
Collapse
Affiliation(s)
- Jie Liu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yuxuan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Taoliang Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Huajian Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Haoqi He
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jihui Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yiquan Ke
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
37
|
Barrera-Martínez CL, Padilla-Vaca F, Liakos I, Meléndez-Ortiz HI, Cortez-Mazatan GY, Peralta-Rodríguez RD. Chitosan microparticles as entrapment system for trans- cinnamaldehyde: Synthesis, drug loading, and in vitro cytotoxicity evaluation. Int J Biol Macromol 2020; 166:322-332. [PMID: 33127551 DOI: 10.1016/j.ijbiomac.2020.10.188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022]
Abstract
The ionic gelation method was used to study the effect of the crosslinking agent, sodium tripolyphosphate on average particle size (Dp) and zeta potential (ζp) of chitosan microparticles (CSMP) unloaded and loaded with trans-cinnamaldehyde (TCIN). The obtained values of Dp and ζp trend as 117.6 ± 0.4 ≤ Dp ≤ 478.5 ± 3.5 nm and +27.8 ± 1.3 ≤ ζp ≤ +103.5 ± 4.2 mV, respectively. The entrapment efficiency of TCIN in CSMP was 9.1 ± 2.0% and 71.5 ± 2.9% was released after 360 min (pH = 6.5) which reveals a potential anti-cancer activity in acidic environment. Cytotoxicity of TCIN in DMSO (0-50 μM) was evaluated on MDCK and HeLa cell lines and exhibited low effect at either 24 or 48 h of exposure; whereas TCIN-loaded CSMP (0-50 μM) showed, after 24 h of exposure, 67.6 ± 7.0 and 64.5 ± 3.9% cytotoxicity for MDCK and HeLa cell lines, respectively. At 48 h of exposure, TCIN-loaded CSMP achieved 81.1 ± 0.26 and 77.9 ± 4.2% cytotoxicity for MDCK and HeLa cell lines, respectively.
Collapse
Affiliation(s)
- Cynthia Lizeth Barrera-Martínez
- Centro de Investigación en Química Aplicada (CIQA), Blvd. Enrique Reyna Hermosillo 140, Colonia San Jose de los Cerritos, C.P. 25294 Saltillo, Coahuila, Mexico
| | - Felipe Padilla-Vaca
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Noria Alta s/n Zona Universitaria, C.P. 36050 Guanajuato, Mexico
| | - Ioannis Liakos
- Center for Micro-BioRobotics, Istituto Italiano di Tecnologia (IIT), Viale Rinaldo Piaggio 34, 56025 Pontedera (Pisa), Italy
| | - Héctor Iván Meléndez-Ortiz
- CONACyT-Centro de Investigación en Química Aplicada (CIQA), Blvd. Enrique Reyna Hermosillo 140, Colonia San Jose de los Cerritos, C.P. 25294 Saltillo, Coahuila, Mexico
| | - Gladis Y Cortez-Mazatan
- Centro de Investigación en Química Aplicada (CIQA), Blvd. Enrique Reyna Hermosillo 140, Colonia San Jose de los Cerritos, C.P. 25294 Saltillo, Coahuila, Mexico
| | - René Darío Peralta-Rodríguez
- Centro de Investigación en Química Aplicada (CIQA), Blvd. Enrique Reyna Hermosillo 140, Colonia San Jose de los Cerritos, C.P. 25294 Saltillo, Coahuila, Mexico.
| |
Collapse
|
38
|
Witika BA, Makoni PA, Matafwali SK, Chabalenge B, Mwila C, Kalungia AC, Nkanga CI, Bapolisi AM, Walker RB. Biocompatibility of Biomaterials for Nanoencapsulation: Current Approaches. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1649. [PMID: 32842562 PMCID: PMC7557593 DOI: 10.3390/nano10091649] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/05/2020] [Accepted: 08/09/2020] [Indexed: 12/12/2022]
Abstract
Nanoencapsulation is an approach to circumvent shortcomings such as reduced bioavailability, undesirable side effects, frequent dosing and unpleasant organoleptic properties of conventional drug delivery systems. The process of nanoencapsulation involves the use of biomaterials such as surfactants and/or polymers, often in combination with charge inducers and/or ligands for targeting. The biomaterials selected for nanoencapsulation processes must be as biocompatible as possible. The type(s) of biomaterials used for different nanoencapsulation approaches are highlighted and their use and applicability with regard to haemo- and, histocompatibility, cytotoxicity, genotoxicity and carcinogenesis are discussed.
Collapse
Affiliation(s)
- Bwalya A. Witika
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa; (B.A.W.); (P.A.M.)
| | - Pedzisai A. Makoni
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa; (B.A.W.); (P.A.M.)
| | - Scott K. Matafwali
- Department of Basic Sciences, School of Medicine, Copperbelt University, Ndola 10101, Zambia;
| | - Billy Chabalenge
- Department of Market Authorization, Zambia Medicines Regulatory Authority, Lusaka 10101, Zambia;
| | - Chiluba Mwila
- Department of Pharmacy, School of Health Sciences, University of Zambia, Lusaka 10101, Zambia; (C.M.); (A.C.K.)
| | - Aubrey C. Kalungia
- Department of Pharmacy, School of Health Sciences, University of Zambia, Lusaka 10101, Zambia; (C.M.); (A.C.K.)
| | - Christian I. Nkanga
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, P.O. Box 212, Kinshasa XI, Democratic Republic of the Congo;
| | - Alain M. Bapolisi
- Department of Chemistry, Faculty of Science, Rhodes University, Makhanda 6140, South Africa;
| | - Roderick B. Walker
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa; (B.A.W.); (P.A.M.)
| |
Collapse
|
39
|
Cong X, Tian H, Liu S, Mao K, Chen H, Xin Y, Liu F, Wang X, Meng X, Zhu G, Wang J, Gao X, Tan H, Yang YG, Sun T. Cationic Liposome/DNA Complexes Mediate Antitumor Immunotherapy by Promoting Immunogenic Tumor Cell Death and Dendritic Cell Activation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:28047-28056. [PMID: 32478501 DOI: 10.1021/acsami.0c08112] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Immunotherapy has been successfully used in the treatment of multiple malignancies, but clinical studies revealed low response rates. Thus, the development of new effective immunotherapeutic modalities is urgently needed. Successfully inducing tumor cell death with enhanced antigenicity is important for the expansion and differentiation of tumor-specific CD8+ cytotoxic T lymphocytes. Cationic liposome/DNA complexes (CLN/DNA), which usually have obvious cytotoxic effects, may improve the antitumor immunity through enhancing the immunogenicity of dying tumor cells. Herein, we report that a plasmid DNA-encapsulated cationic lipid nanoparticle formulated with cholesterol, DOTAP, and DSPE-mPEG2000 significantly increases the tumor cell death with high antigenicity in vitro. Furthermore, the cationic liposome/DNA complex (CLN/DNA) treatment promotes the activation of dendritic cells (DCs). We also find that the intratumorally injected CLN/DNA successfully promoted the activation of DCs in the tumor-draining lymph node. Importantly, both local tumor growth and distant tumor formation were significantly inhibited by T cell-dependent antitumor immune responses after intratumoral injection of CLN/DNA. This study presents a simple and effective strategy for improving the cancer immunotherapy.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Huimin Tian
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
| | - Hongmei Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Feiqi Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
| | - Xin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Xue Gao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Huizhu Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
40
|
Liposome-based drug delivery of various anticancer agents of synthetic and natural product origin: a patent overview. Pharm Pat Anal 2020; 9:87-116. [DOI: 10.4155/ppa-2019-0020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Phospholipid-based liposomal vesicles are among the most effective delivery options currently available for various classes of anticancer drugs. The patents granted to inventions disclosing details on liposomal delivery module by the US Patent and Trademark Office, European Patent Office, and world patent holdings through WIPO (World Intellectual Property Organization) patenting have been sorted based upon liposome, and anticancer keywords within the abstract and claims sections of the patents for the period between 2000 and 2019, thereby disclosing novel liposome formulations encapsulating single, or combination of chemotherapeutic agents that have been far more chemically and physiologically stable, therapeutically efficacious, and comparatively less toxic than their nonliposomal free-drug counterparts. The added stability, site-specific transport, and payload delivery, enhanced bioavailability, fast body clearance, and biocompatibility together with the controlled and sustained delivery-related benefits claimed in the patent literature have been exclusively discussed with a focus on the last 5-year period.
Collapse
|
41
|
Affiliation(s)
- Xianxun Sun
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of Sciences Wuhan 430071 China
- College of Life ScienceJiang Han University Wuhan 430056 China
| | - Zongqiang Cui
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of Sciences Wuhan 430071 China
| |
Collapse
|
42
|
Wang SY, Hu HZ, Qing XC, Zhang ZC, Shao ZW. Recent advances of drug delivery nanocarriers in osteosarcoma treatment. J Cancer 2020; 11:69-82. [PMID: 31892974 PMCID: PMC6930408 DOI: 10.7150/jca.36588] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor mainly occurred in children and adolescence, and chemotherapy is limited for the side effects and development of drug resistance. Advances in nanotechnology and knowledge of cancer biology have led to significant improvements in developing tumor-targeted drug delivery nanocarriers, and some have even entered clinically application. Delivery of chemotherapeutic agents by functionalized smart nanocarriers could protect the drugs from rapid clearance, prolong the circulating time, and increase the drug concentration at tumor sites, thus enhancing the therapeutic efficacy and reducing side effects. Various drug delivery nanocarriers have been designed and tested for osteosarcoma treatment, but most of them are still at experimental stage, and more further studies are needed before clinical application. In this present review, we briefly describe the types of commonly used nanocarriers in osteosarcoma treatment, and discuss the strategies for osteosarcoma-targeted delivery and controlled release of drugs. The application of nanoparticles in the management of metastatic osteosarcoma is also briefly discussed. The purpose of this article is to present an overview of recent progress of nanoscale drug delivery platforms in osteosarcoma, and inspire new ideas to develop more effective therapeutic options.
Collapse
Affiliation(s)
- Shang-Yu Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hong-Zhi Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiang-Cheng Qing
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhi-Cai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zeng-Wu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
43
|
Di Mascolo D, Varesano S, Benelli R, Mollica H, Salis A, Zocchi MR, Decuzzi P, Poggi A. Nanoformulated Zoledronic Acid Boosts the Vδ2 T Cell Immunotherapeutic Potential in Colorectal Cancer. Cancers (Basel) 2019; 12:E104. [PMID: 31906080 PMCID: PMC7017311 DOI: 10.3390/cancers12010104] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/19/2019] [Accepted: 12/29/2019] [Indexed: 12/11/2022] Open
Abstract
Aminobisphosphonates, such as zoledronic acid (ZA), have shown potential in the treatment of different malignancies, including colorectal carcinoma (CRC). Yet, their clinical exploitation is limited by their high bone affinity and modest bioavailability. Here, ZA is encapsulated into the aqueous core of spherical polymeric nanoparticles (SPNs), whose size and architecture resemble that of biological vesicles. On Vδ2 T cells, derived from the peripheral blood of healthy donors and CRC patients, ZA-SPNs induce proliferation and trigger activation up to three orders of magnitude more efficiently than soluble ZA. These activated Vδ2 T cells kill CRC cells and tumor spheroids, and are able to migrate toward CRC cells in a microfluidic system. Notably, ZA-SPNs can also stimulate the proliferation of Vδ2 T cells from the tumor-infiltrating lymphocytes of CRC patients and boost their cytotoxic activity against patients' autologous tumor organoids. These data represent a first step toward the use of nanoformulated ZA for immunotherapy in CRC patients.
Collapse
Affiliation(s)
- Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy; (D.D.M.); (H.M.); (P.D.)
| | - Serena Varesano
- Molecular Oncology and Angiogenesis Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy;
| | - Roberto Benelli
- Immunology Unit, Ospedale Policlinico San Martino, University of Genoa, 16132 Genoa, Italy;
| | - Hilaria Mollica
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy; (D.D.M.); (H.M.); (P.D.)
| | | | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy; (D.D.M.); (H.M.); (P.D.)
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy;
| |
Collapse
|
44
|
Pashirova TN, Burilova EA, Lukashenko SS, Gaysin NK, Gnezdilov OI, Sapunova AS, Fernandes AR, Voloshina AD, Souto EB, Zhiltsova EP, Zakharova LY. Nontoxic antimicrobial micellar systems based on mono- and dicationic Dabco-surfactants and furazolidone: Structure-solubilization properties relationships. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2019.112062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
45
|
Complement Activation-Related Pathophysiological Changes in Anesthetized Rats: Activator-Dependent Variations of Symptoms and Mediators of Pseudoallergy. Molecules 2019; 24:molecules24183283. [PMID: 31505853 PMCID: PMC6767111 DOI: 10.3390/molecules24183283] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
Complement (C) activation can underlie the infusion reactions to liposomes and other nanoparticle-based medicines, a hypersensitivity syndrome that can be partially reproduced in animal models. However, the sensitivities and manifestations substantially differ in different species, and C activation may not be the only cause of pathophysiological changes. In order to map the species variation of C-dependent and -independent pseudoallergy (CARPA/CIPA), here we used known C activators and C activator liposomes to compare their acute hemodynamic, hematological, and biochemical effects in rats. These C activators were cobra venom factor (CVF), zymosan, AmBisome (at 2 doses), its amphotericin B-free vehicle (AmBisombo), and a PEGylated cholesterol-containing liposome (PEG-2000-chol), all having different powers to activate C in rat blood. The pathophysiological endpoints measured were blood pressure, leukocyte and platelet counts, and plasma thromboxane B2, while C activation was assessed by C3 consumption using the Pan-Specific C3 assay. The results showed strong linear correlation between C activation and systemic hypotension, pointing to a causal role of C activation in the hemodynamic changes. The observed thrombocytopenia and leukopenia followed by leukocytosis also correlated with C3 conversion in case of C activators, but not necessarily with C activation by liposomes. These findings are consistent with the double hit hypothesis of hypersensitivity reactions (HSRs), inasmuch as strong C activation can fully account for all symptoms of HSRs, but in case of no-, or weak C activators, the pathophysiological response, if any, is likely to involve other activation pathways.
Collapse
|
46
|
de Araújo DR, Ribeiro LNDM, de Paula E. Lipid-based carriers for the delivery of local anesthetics. Expert Opin Drug Deliv 2019; 16:701-714. [PMID: 31172838 DOI: 10.1080/17425247.2019.1629415] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION There is a clinical need for pharmaceutical dosage forms devised to prolong the acting time of local anesthetic (LA) agents or to reduce their toxicity. Encapsulation of LA in drug delivery systems (DDSs) can provide long-term anesthesia for inpatients (e.g. in immediate postsurgical pain control, avoiding the side effects from systemic analgesia) and diminished systemic toxicity for outpatients (in ambulatory/dentistry procedures). The lipid-based formulations described here, such as liposomes, microemulsions, and lipid nanoparticles, have provided several nanotechnological advances and therapeutic alternatives despite some inherent limitations associated with the fabrication processes, costs, and preclinical evaluation models. AREAS COVERED A description of the currently promising lipid-based carriers, including liposomes, microemulsions, and nanostructured lipid carriers, followed by a systematic review of the existing lipid-based formulations proposed for LA. Trends in the research of these LA-in-DDS are then exposed, from the point of view of administration route and alternatives for non-traditionally administered LA molecules. EXPERT OPINION Considering the current state and potential future developments in the field, we discuss the reasons for why dozens of formulations published every year fail to reach clinical trials; only one lipid-based formulation for the delivery of local anesthetic (Exparel®) has been approved so far.
Collapse
Affiliation(s)
| | - Lígia Nunes de Morais Ribeiro
- b Department of Biochemistry and Tissue Biology , Institute of Biology, University of Campinas - UNICAMP , Campinas, São Paulo , Brazil
| | - Eneida de Paula
- b Department of Biochemistry and Tissue Biology , Institute of Biology, University of Campinas - UNICAMP , Campinas, São Paulo , Brazil
| |
Collapse
|
47
|
Galassi VV, Villarreal MA, Montich GG. Relevance of the protein macrodipole in the membrane-binding process. Interactions of fatty-acid binding proteins with cationic lipid membranes. PLoS One 2018. [PMID: 29518146 PMCID: PMC5843346 DOI: 10.1371/journal.pone.0194154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The fatty acid-binding proteins L-BABP and Rep1-NCXSQ bind to anionic lipid membranes by electrostatic interactions. According to Molecular Dynamics (MD) simulations, the interaction of the protein macrodipole with the membrane electric field is a driving force for protein binding and orientation in the interface. To further explore this hypothesis, we studied the interactions of these proteins with cationic lipid membranes. As in the case of anionic lipid membranes, we found that both proteins, carrying a negative as well as a positive net charge, were bound to the positively charged membrane. Their major axis, those connecting the bottom of the β-barrel with the α-helix portal domain, were rotated about 180 degrees as compared with their orientations in the anionic lipid membranes. Fourier transform infrared (FTIR) spectroscopy of the proteins showed that the positively charged membranes were also able to induce conformational changes with a reduction of the β-strand proportion and an increase in α-helix secondary structure. Fatty acid-binding proteins (FABPs) are involved in several cell processes, such as maintaining lipid homeostasis in cells. They transport hydrophobic molecules in aqueous medium and deliver them into lipid membranes. Therefore, the interfacial orientation and conformation, both shown herein to be electrostatically determined, have a strong correlation with the specific mechanism by which each particular FABP exerts its biological function.
Collapse
Affiliation(s)
- Vanesa V. Galassi
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Biológica “Ranwel Caputto”, Córdoba, Argentina
- CONICET, Universidad Nacional de Córdoba, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Córdoba, Argentina
| | - Marcos A. Villarreal
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Teórica y Computacional, Córdoba, Argentina
- CONICET, Universidad Nacional de Córdoba. Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Córdoba, Argentina
| | - Guillermo G. Montich
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Biológica “Ranwel Caputto”, Córdoba, Argentina
- CONICET, Universidad Nacional de Córdoba, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Córdoba, Argentina
- * E-mail:
| |
Collapse
|