1
|
Wang T, Cao J, Sun L, Zhao J, Fu Y. Cellulose-based molecularly imprinted core-shell microspheres with dual environment responsiveness for specific recognition and enrichment of paclitaxel. Int J Biol Macromol 2025; 309:143164. [PMID: 40239786 DOI: 10.1016/j.ijbiomac.2025.143164] [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: 12/29/2024] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Paclitaxel (PTX), a natural diterpene alkaloid with potent anticancer activity, has been a focus of global research due to its complex structure, significant biological effects, unique mechanism of action, and limited natural availability. In this study, cellulose was used as the raw material to synthesize functionalized pH-responsive modified cellulose porous hydrogel microspheres (MCC/SA/MA-POSS-PTX) via inverse suspension polymerization, using sodium alginate (SA) as the pH-responsive material. Based on this using PTX as the template molecule, 4-VP as the functional monomer, N-isopropylacrylamide (NIPAM) as the thermosensitive monomer, cellulose-based PTX molecularly imprinted hydrogel microspheres (CDLMIHM-PTX) with dual pH and temperature responsiveness were prepared. The adsorption mechanism of the novel material was elucidated through a series of characterization and adsorption experiments. The CDLMIHM-PTX exhibited exceptional adsorption capacity (35.12 mg·g-1) and selectivity, as well as good reusability (6 of cycles). Specific enrichment of PTX from the crude extract of Taxus × media branches was achieved, increasing its content from 7.59 % to 55.98 %. This highlights the capability of CDLMIHM-PTX for targeted enrichment and separation of PTX from complex plant matrices, suggesting promising industrial applications. This work explores the construction of pH/temperature dual responsive cellulose-based molecularly imprinted hydrogel microspheres for the selective capture of target compounds, with potential for practical applications.
Collapse
Affiliation(s)
- Tao Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Foresty University, 150040 Harbin, PR China; The College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 150040 Harbin, PR China
| | - Jingsong Cao
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Foresty University, 150040 Harbin, PR China; The College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 150040 Harbin, PR China
| | - Linan Sun
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Foresty University, 150040 Harbin, PR China; The College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 150040 Harbin, PR China
| | - Jingru Zhao
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Foresty University, 150040 Harbin, PR China; The College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, 150040 Harbin, PR China
| | - Yujie Fu
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Forestry University, 100083 Beijing, PR China; The College of Forestry, Beijing Forestry University, 100083 Beijing, PR China.
| |
Collapse
|
2
|
Mohammadi M, Ahmed Qadir S, Mahmood Faraj A, Hamid Shareef O, Mahmoodi H, Mahmoudi F, Moradi S. Navigating the future: Microfluidics charting new routes in drug delivery. Int J Pharm 2024:124142. [PMID: 38648941 DOI: 10.1016/j.ijpharm.2024.124142] [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: 10/12/2023] [Revised: 03/30/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Microfluidics has emerged as a transformative force in the field of drug delivery, offering innovative avenues to produce a diverse range of nano drug delivery systems. Thanks to its precise manipulation of small fluid volumes and its exceptional command over the physicochemical characteristics of nanoparticles, this technology is notably able to enhance the pharmacokinetics of drugs. It has initiated a revolutionary phase in the domain of drug delivery, presenting a multitude of compelling advantages when it comes to developing nanocarriers tailored for the delivery of poorly soluble medications. These advantages represent a substantial departure from conventional drug delivery methodologies, marking a paradigm shift in pharmaceutical research and development. Furthermore, microfluidic platformsmay be strategically devised to facilitate targeted drug delivery with the objective of enhancing the localized bioavailability of pharmaceutical substances. In this paper, we have comprehensively investigated a range of significant microfluidic techniques used in the production of nanoscale drug delivery systems. This comprehensive review can serve as a valuable reference and offer insightful guidance for the development and optimization of numerous microfluidics-fabricated nanocarriers.
Collapse
Affiliation(s)
- Mohammad Mohammadi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Syamand Ahmed Qadir
- Department of Medical Laboratory Techniques, Halabja Technical Institute, Research Center, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Aryan Mahmood Faraj
- Department of Medical Laboratory Sciences, Halabja Technical College of Applied Sciences, Sulaimani Polytechnic University, Halabja, Iraq
| | - Osama Hamid Shareef
- Department of Medical Laboratory Techniques, Halabja Technical Institute, Research Center, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Hassan Mahmoodi
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mahmoudi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Ebrahimnejad P, Mohammadi Z, Babaei A, Ahmadi M, Amirkhanloo S, Asare-Addo K, Nokhodchid A. Novel Strategies Using Sagacious Targeting for Site-Specific Drug Delivery in Breast Cancer Treatment: Clinical Potential and Applications. Crit Rev Ther Drug Carrier Syst 2024; 41:35-84. [PMID: 37824418 DOI: 10.1615/critrevtherdrugcarriersyst.v41.i1.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
For more than a decade, researchers have been working to achieve new strategies and smart targeting drug delivery techniques and technologies to treat breast cancer (BC). Nanotechnology presents a hopeful strategy for targeted drug delivery into the building of new therapeutics using the properties of nanomaterials. Nanoparticles are of high regard in the field of diagnosis and the treatment of cancer. The use of these nanoparticles as an encouraging approach in the treatment of various cancers has drawn the interest of researchers in recent years. In order to achieve the maximum therapeutic effectiveness in the treatment of BC, combination therapy has also been adopted, leading to minimal side effects and thus an enhancement in the quality of life for patients. This review article compares, discusses and criticizes the approaches to treat BC using novel design strategies and smart targeting of site-specific drug delivery systems.
Collapse
Affiliation(s)
- Pedram Ebrahimnejad
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Mohammadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amirhossein Babaei
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Melika Ahmadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shervin Amirkhanloo
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kofi Asare-Addo
- Department of Pharmacy, University of Huddersfield, Huddersfield, UK
| | - Ali Nokhodchid
- Lupin Pharmaceutical Research Center, Coral Springs, Florida, USA; Pharmaceutics Research Lab, Arundel Building, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
4
|
Pei Q, Jiang B, Hao D, Xie Z. Self-assembled nanoformulations of paclitaxel for enhanced cancer theranostics. Acta Pharm Sin B 2023; 13:3252-3276. [PMID: 37655323 PMCID: PMC10465968 DOI: 10.1016/j.apsb.2023.02.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/15/2023] [Accepted: 01/23/2023] [Indexed: 03/07/2023] Open
Abstract
Chemotherapy has occupied the critical position in cancer therapy, especially towards the post-operative, advanced, recurrent, and metastatic tumors. Paclitaxel (PTX)-based formulations have been widely used in clinical practice, while the therapeutic effect is far from satisfied due to off-target toxicity and drug resistance. The caseless multi-components make the preparation technology complicated and aggravate the concerns with the excipients-associated toxicity. The self-assembled PTX nanoparticles possess a high drug content and could incorporate various functional molecules for enhancing the therapeutic index. In this work, we summarize the self-assembly strategy for diverse nanodrugs of PTX. Then, the advancement of nanodrugs for tumor therapy, especially emphasis on mono-chemotherapy, combinational therapy, and theranostics, have been outlined. Finally, the challenges and potential improvements have been briefly spotlighted.
Collapse
Affiliation(s)
- Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Bowen Jiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Dengyuan Hao
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
5
|
Zhang H, Yang J, Sun R, Han S, Yang Z, Teng L. Microfluidics for nano-drug delivery systems: From fundamentals to industrialization. Acta Pharm Sin B 2023; 13:3277-3299. [PMID: 37655333 PMCID: PMC10466004 DOI: 10.1016/j.apsb.2023.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/10/2022] [Accepted: 12/15/2022] [Indexed: 01/27/2023] Open
Abstract
In recent years, owing to the miniaturization of the fluidic environment, microfluidic technology offers unique opportunities for the implementation of nano drug delivery systems (NDDSs) production processes. Compared with traditional methods, microfluidics improves the controllability and uniformity of NDDSs. The fast mixing and laminar flow properties achieved in the microchannels can tune the physicochemical properties of NDDSs, including particle size, distribution and morphology, resulting in narrow particle size distribution and high drug-loading capacity. The success of lipid nanoparticles encapsulated mRNA vaccines against coronavirus disease 2019 by microfluidics also confirmed its feasibility for scaling up the preparation of NDDSs via parallelization or numbering-up. In this review, we provide a comprehensive summary of microfluidics-based NDDSs, including the fundamentals of microfluidics, microfluidic synthesis of NDDSs, and their industrialization. The challenges of microfluidics-based NDDSs in the current status and the prospects for future development are also discussed. We believe that this review will provide good guidance for microfluidics-based NDDSs.
Collapse
Affiliation(s)
- Huan Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jie Yang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Rongze Sun
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Songren Han
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
6
|
Liu Y, Hong G, Mao L, Su Z, Liu T, Liu H. A Novel Paclitaxel Derivative for Triple-Negative Breast Cancer Chemotherapy. Molecules 2023; 28:molecules28093662. [PMID: 37175072 PMCID: PMC10180349 DOI: 10.3390/molecules28093662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023] Open
Abstract
Paclitaxel-triethylenetetramine hexaacetic acid conjugate (PTX-TTHA), a novel semi-synthetic taxane, is designed to improve the water solubility and cosolvent toxicity of paclitaxel in several aminopolycarboxylic acid groups. In this study, the in vitro and in vivo antitumor effects and mechanisms of PTX-TTHA against triple-negative breast cancer (TNBC) and its intravenous toxicity were evaluated. Results showed the water solubility of PTX-TTHA was greater than 5 mg/mL, which was about 7140-fold higher than that of paclitaxel (<0.7 µg/mL). PTX-TTHA (10-105 nmol/L) could significantly inhibit breast cancer proliferation and induce apoptosis by stabilizing microtubules and arresting the cell cycle in the G2/M phase in vitro, with its therapeutic effect and mechanism similar to paclitaxel. However, when the MDA-MB-231 cell-derived xenograft (CDX) tumor model received PTX-TTHA (13.73 mg/kg) treatment once every 3 days for 21 days, the tumor inhibition rate was up to 77.32%. Furthermore, PTX-TTHA could inhibit tumor proliferation by downregulating Ki-67, and induce apoptosis by increasing pro-apoptotic proteins (Bax, cleaved caspase-3) and TdT-mediated dUTP nick end labeling (TUNEL) positive apoptotic cells, and reducing anti-apoptotic protein (Bcl-2). Moreover, PTX-TTHA demonstrated no sign of acute toxicity on vital organs, hematological, and biochemical parameters at the limit dose (138.6 mg/kg, i.v.). Our study indicated that PTX-TTHA showed better water solubility than paclitaxel, as well as comparable in vitro and in vivo antitumor activity in TNBC models. In addition, the antitumor mechanism of PTX-TTHA was related to microtubule regulation and apoptosis signaling pathway activation.
Collapse
Affiliation(s)
- Yuetong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ge Hong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, China
| | - Lina Mao
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, China
| | - Zhe Su
- Tianjin Institute for Drug Control, Tianjin 300070, China
| | - Tianjun Liu
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
7
|
Chauhan N, Cabrera M, Chowdhury P, Nagesh PK, Dhasmana A, Pranav, Jaggi M, Chauhan SC, Yallapu MM. Indocyanine Green-based Glow Nanoparticles Probe for Cancer Imaging. Nanotheranostics 2023; 7:353-367. [PMID: 37151801 PMCID: PMC10161388 DOI: 10.7150/ntno.78405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 03/22/2023] [Indexed: 08/31/2023] Open
Abstract
Indocyanine green (ICG) is one of the FDA-approved near infra-red fluorescent (NIRF) probes for cancer imaging and image-guided surgery in the clinical setting. However, the limitations of ICG include poor photostability, high concentration toxicity, short circulation time, and poor cancer cell specificity. To overcome these hurdles, we engineered a nanoconstruct composed of poly (vinyl pyrrolidone) (PVP)-indocyanine green that is cloaked self-assembled with tannic acid (termed as indocyanine green-based glow nanoparticles probe, ICG-Glow NPs) for the cancer cell/tissue-specific targeting. The self-assembled ICG-Glow NPs were confirmed by spherical nanoparticles formation (DLS and TEM) and spectral analyses. The NIRF imaging characteristic of ICG-Glow NPs was established by superior fluorescence counts on filter paper and chicken tissue. The ICG-Glow NPs exhibited excellent hemo and cellular compatibility with human red blood cells, kidney normal, pancreatic normal, and other cancer cell lines. An enhanced cancer-specific NIRF binding and imaging capability of ICG-Glow NPs was confirmed using different human cancer cell lines and human tumor tissues. Additionally, tumor-specific binding/accumulation of ICG-Glow NPs was confirmed in MDA-MB-231 xenograft mouse model. Collectively, these findings suggest that ICG-Glow NPs have great potential as a novel and safe NIRF imaging probe for cancer cell/tumor imaging. This can lead to a quicker cancer diagnosis facilitating precise disease detection and management.
Collapse
Affiliation(s)
- Neeraj Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Marco Cabrera
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Prashanth K.B. Nagesh
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Pranav
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Murali M. Yallapu
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| |
Collapse
|
8
|
Chowdhury P, Bhusetty Nagesh PK, Hollingsworth TJ, Jaggi M, Chauhan SC, Yallapu MM. Coating a Self-Assembly Nanoconstruct with a Neutrophil Cell Membrane Enables High Specificity for Triple Negative Breast Cancer Treatment. ACS APPLIED BIO MATERIALS 2022; 5:4554-4566. [PMID: 35976626 DOI: 10.1021/acsabm.2c00614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Breast cancer is one of the most commonly diagnosed cancers in American women. Triple negative breast cancer is among the most advanced and aggressive forms of breast cancer. Treatment options are limited for such cancers, making chemotherapy a convenient and effective treatment. Although these therapies can reduce morbidity and mortality, it is often followed by systemic side effects or relapse. Nanoparticles (NPs) have been considered for drug delivery approaches due to their ability to target various disease sites. Herein, we aim to develop a biomimetic NP construct (cell membrane-cloaked NPs) that exhibits specific affinity with triple negative breast cancer cells. In this regard, we designed biomimetic supramolecular nanoconstructs composed of a poly(vinyl pyrrolidone)-tannic acid (PVP-TA NPs/ PVT NPs) core and biofunctionalized with neutrophil cell membranes (PVT-NEU NPs). In this study, we have synthesized a PVT-NEU NP construct, characterized it, and evaluated it for improved targeting and therapeutic benefits in in vitro and in vivo models. Analysis of PVT-NEU NPs confirms the presence of the core of PVP-TA NPs coated with activated human neutrophil membranes. The study results confirmed that PVT-NEU NPs demonstrated an enhanced interaction and targeting with the tumor cells, thus improving the therapeutic activity of a model therapeutic agent (paclitaxel). Altogether, this study suggests the potential of biomimetic NPs as a promising therapeutic option for targeted drug delivery for advanced-stage breast cancer and other similar diseased conditions.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Prashanth Kumar Bhusetty Nagesh
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - T J Hollingsworth
- Department of Ophthalmology, Hamilton Eye Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Subhash Chand Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Murali Mohan Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| |
Collapse
|
9
|
Salari N, Faraji F, Torghabeh FM, Faraji F, Mansouri K, Abam F, Shohaimi S, Akbari H, Mohammadi M. Polymer-based drug delivery systems for anticancer drugs: A systematic review. Cancer Treat Res Commun 2022; 32:100605. [PMID: 35816909 DOI: 10.1016/j.ctarc.2022.100605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/21/2022] [Accepted: 07/02/2022] [Indexed: 06/15/2023]
Abstract
Recent advances in nanotechnology sciences lead to the development of new treatment approaches for various diseases such as cancer. Nanotechnology advances can potentially minimize the side effects of drugs through the employment of effective and controlled drug delivery systems (DDSs). Polymers are optimal tools providing drug delivery mechanisms through the unique features of pharmacokinetics, circulation time, biocompatibility, and biodegradability. This systematic review aimed to evaluate polymer-based DDSs for anticancer drugs and their various therapeutic applications in cancer treatment. This study was conducted with no time limitation by November 2021. Related articles were collected through a deep search in English and Persian databases of SID, MagIran, Scopus, Web Of Science (WoS), PubMed, Science Direct, and Google Scholar. Keywords included drug delivery system, anticancer agent, polymeric nanostructure-based drug delivery, polymer-based drug delivery, and polymeric system. As the results showed, polymeric nanoparticles (PNPs) have influential roles in cancer treatment than conventional chemotherapy procedures. PNPs can reduce cytotoxicity following chemotherapy drug administration, improve the solubility characteristics of these therapeutic agents and inhibit the rate of tumor growth.
Collapse
Affiliation(s)
- Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Mansouri Torghabeh
- Department of Physiology Sciences, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Faraji
- Student research committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzaneh Abam
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shamarina Shohaimi
- Department of Biology, Faculty of Science, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hakimeh Akbari
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Masoud Mohammadi
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran.
| |
Collapse
|
10
|
Hatami E, Nagesh PKB, Chauhan N, Jaggi M, Chauhan SC, Yallapu MM. In Situ Nanoparticle Self-Assembly for Combination Delivery of Therapeutics to Non-Small Cell Lung Cancer. ACS APPLIED BIO MATERIALS 2022; 5:1104-1119. [PMID: 35179871 DOI: 10.1021/acsabm.1c01158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chemotherapy often experiences several challenges including severe systemic toxicity and adverse effects. The combination chemotherapy arose as an effective clinical practice aimed at reducing doses of drugs to achieve synergistic actions with low toxicity. Our recent efforts demonstrated a synergistic therapeutic benefit of gambogic acid (GA) and gemcitabine (Gem) against lung cancer. However, simultaneous delivery of these two drugs at the tumor site is highly challenging. Therefore, the development of an injectable formulation that can effectively deliver both hydrophobic (GA) and hydrophilic (Gem) drugs in one formulation is a clinically unmet need. Herein, this study reports an in situ human serum albumin (HSA)- and tannic acid (TA)-mediated complexed GA and Gem nanoparticles (G-G@HTA NPs). G-G@HTA NP formation was confirmed by the particle size, Fourier transform infrared spectroscopy, and 1H NMR spectroscopy. The superior therapeutic activity of G-G@HTA NPs was demonstrated by multiple in vitro functional assays. Additionally, G-G@HTA NPs revealed an obvious and precise targeting of tumors in vivo. The promoted and more synergistic anti-tumor efficacy of G-G@HTA NPs was attained than that of combined treatments and single drug treatments. These events have resulted in no apparent systemic and organ toxicities. Together, this study suggests that in situ HSA-TA-based combinatorial treatment strategy is a suitable approach for application in lung cancer treatment.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Neeraj Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| |
Collapse
|
11
|
Liu Y, Lu X, Zhang Z, Jiang S, Lv H. mPEG-Cholic acid/TPGS mixed micelles for combined delivery of paclitaxel and bufalin to treat hepatocellular carcinoma. Pharm Dev Technol 2022; 27:215-227. [PMID: 35105263 DOI: 10.1080/10837450.2022.2037140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In this study, amphiphilic block copolymer mPEG-cholic acid was synthesized in conjunction with TPGS as stabilizer to prepare multifunctional micelles. The formed polymeric micelles (PCTm) were used for the delivery of paclitaxel (PTX) and bufalin (BF). PEG group could enhance solubility and extend circulation time, while cholic acid groups achieved the liver targeted function. Combinations of these approaches could realize a synergistic therapeutic effect in the treatment of advanced hepatocellular carcinoma. CLSM in vitro results demonstrated that drug capsulation into PCTm could enhance cellular uptake. FCM results confirmed the uptake amount of C6/PCTm was 7.5-fold higher than that of free C6 after incubation for 2 h. Competitive inhibition test proved the Na+-taurocholate co-transporting polypeptide (NTCP) involved in the uptake mechanism of PCTm. Meanwhile, in vivo imaging assays demonstrated that the fluorescence intensity of Cy5.5/PCTm was higher than that of free Cy5.5 on liver and tumor with extended circulation time to 48 h. In addition, in vivo studies confirmed that the combined therapy exhibited the strongest tumor inhibition rate of 82.29% with lower systemic toxicity. Hence, these results indicated that PCTm could provide a promising strategy for targeting hepatocellular carcinoma and achieve the goal of the synergism and attenuation.
Collapse
Affiliation(s)
- Yujia Liu
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaoyu Lu
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenhai Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining No. 1 People's Hospital, Jining, Shandong 272000, China
| | - Huixia Lv
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
12
|
Abdelrasoul A, Shoker A. Induced hemocompatibility of polyethersulfone (PES) hemodialysis membrane using polyvinylpyrrolidone: Investigation on human serum fibrinogen adsorption and inflammatory biomarkers released. Chem Eng Res Des 2022. [DOI: 10.1016/j.cherd.2021.11.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Pereira P, Serra AC, Coelho JF. Vinyl Polymer-based technologies towards the efficient delivery of chemotherapeutic drugs. Prog Polym Sci 2021. [DOI: 10.1016/j.progpolymsci.2021.101432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
14
|
Kuskov AN, Luss AL, Gritskova IA, Shtilman MI, Motyakin MV, Levina II, Nechaeva AM, Sizova OY, Tsatsakis AM, Mezhuev YO. Kinetics and Mechanism of Synthesis of Carboxyl-Containing N-Vinyl-2-Pyrrolidone Telehelics for Pharmacological Use. Polymers (Basel) 2021; 13:polym13152569. [PMID: 34372172 PMCID: PMC8347008 DOI: 10.3390/polym13152569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
It was found that sulfanylethanoic and 3-sulfanylpropanoic acids are effective regulators of molecular weight with chain transfer constants of 0.441 and 0.317, respectively, and show an unexpected acceleration effect on the radical polymerization of N-vinyl-2-pyrrolidone, initiated by 2,2’-azobisisobutyronitrile. It was determined for the first time that the thiolate anions of mercapto acids form a high-temperature redox initiating system with 2,2’-azobisisobutyronitrile during the radical polymerization of N-vinyl-2-pyrrolidone in 1,4-dioxane. Considering the peculiarities of initiation, a kinetic model of the polymerization of N-vinyl-2-pyrrolidone is proposed, and it is shown that the theoretical orders of the reaction rate, with respect to the monomer, initiator, and chain transfer agent, are 1, 0.75, 0.25, and are close to their experimentally determined values. Carboxyl-containing techelics of N-vinyl-2-pyrrolidone were synthesized so that it can slow down the release of the anticancer drug, doxorubicin, from aqueous solutions, which can find its application in the pharmacological field.
Collapse
Affiliation(s)
- Andrey N. Kuskov
- Department of Biomaterials, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (A.L.L.); (M.I.S.); (A.M.N.); (O.Y.S.)
- Correspondence: (A.N.K.); (Y.O.M.)
| | - Anna L. Luss
- Department of Biomaterials, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (A.L.L.); (M.I.S.); (A.M.N.); (O.Y.S.)
| | - Inessa A. Gritskova
- Department of Chemistry and Technology of Macromolecular Compounds, MIREA—Russian Technological University (RTU MIREA), 119454 Moscow, Russia;
| | - Mikhail I. Shtilman
- Department of Biomaterials, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (A.L.L.); (M.I.S.); (A.M.N.); (O.Y.S.)
| | - Mikhail V. Motyakin
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.M.); (I.I.L.)
- Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Irina I. Levina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.M.); (I.I.L.)
| | - Anna M. Nechaeva
- Department of Biomaterials, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (A.L.L.); (M.I.S.); (A.M.N.); (O.Y.S.)
| | - Oksana Yu. Sizova
- Department of Biomaterials, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (A.L.L.); (M.I.S.); (A.M.N.); (O.Y.S.)
| | - Aristidis M. Tsatsakis
- Center of Toxicology Science & Research, Division of Morphology, Medical School, Voutes Campus, University of Crete, 71003 Heraklion, Greece;
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119991 Moscow, Russia
| | - Yaroslav O. Mezhuev
- Department of Biomaterials, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (A.L.L.); (M.I.S.); (A.M.N.); (O.Y.S.)
- Correspondence: (A.N.K.); (Y.O.M.)
| |
Collapse
|
15
|
Wang Z, Zhang Q, Shen H, Yang P, Zhou X. Optimized MALDI-TOF MS Strategy for Characterizing Polymers. Front Chem 2021; 9:698297. [PMID: 34249867 PMCID: PMC8264446 DOI: 10.3389/fchem.2021.698297] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/04/2021] [Indexed: 11/13/2022] Open
Abstract
In recent years, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) plays an essential role in the analysis of polymers. To acquire a more reliable strategy for polymer profiling, we characterized four representative polymers including polyethylene glycol 6000, polyvinylpyrrolidone K12, polymer polyol KPOP-5040, and polyether polyol DL-4000. The preparation methods of these four polymer samples have been optimized from six aspects, including matrix, cationization reagent, solvent, mixing ratio of cationization reagent to polymer, mixing ratio of matrix to polymer, and laser intensity. After investigating the effects of seven commonly used matrices on the ionization efficiency of four polymers, trans-2-[3-(4-tert-butylphenyl)-2-methyl-2-propenylidene] malononitrile (DCTB) was found to be the only matrix suitable for the analysis of all the four polymers. Our experimental results suggested that different polymers showed a certain preference for different cationization reagents. For example, the polymer polyol KPOP-5040 was suitable for sodium iodide as the cationization reagent, while polyvinylpyrrolidone K12 was more suitable for silver trifluoroacetate (AgTFA). For the choice of solvent, tetrahydrofuran is a reagent with rapid evaporation and a wide range of dissolution which can achieve the best results for the analysis of four polymers. The optimized method was successfully applied to the identification of DSPE-PEG-NH2 with different polymerized degrees. This MALDI-TOF strategy potentially provided the supplementary function through the polymer's application in biomedical and visible probing.
Collapse
Affiliation(s)
- Zhenxin Wang
- Institutes of Biomedical Sciences of Shanghai Medical School and Laboratory Medicine of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Quanqing Zhang
- Department of Chemistry, University of California, Riverside, CA, United States
| | - Huali Shen
- Institutes of Biomedical Sciences of Shanghai Medical School and Minhang Hospital, Fudan University, Shanghai, China
| | - Pengyuan Yang
- Institutes of Biomedical Sciences of Shanghai Medical School and Department of Chemistry, Fudan University, Shanghai, China
| | - Xinwen Zhou
- Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Voci S, Gagliardi A, Molinaro R, Fresta M, Cosco D. Recent Advances of Taxol-Loaded Biocompatible Nanocarriers Embedded in Natural Polymer-Based Hydrogels. Gels 2021; 7:33. [PMID: 33804970 PMCID: PMC8103278 DOI: 10.3390/gels7020033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
The discovery of paclitaxel (PTX) has been a milestone in anti-cancer therapy and has promoted the development and marketing of various formulations that have revolutionized the therapeutic approach towards several malignancies. Despite its peculiar anti-cancer activity, the physico-chemical properties of PTX compromise the administration of the compound in polar media. Because of this, since the development of the first Food and Drug Administration (FDA)-approved formulation (Taxol®), consistent efforts have been made to obtain suitable delivery systems able to preserve/increase PTX efficacy and to overcome the side effects correlated to the presence of some excipients. The exploitation of natural polymers as potential materials for drug delivery purposes has favored the modulation of the bioavailability and the pharmacokinetic profiles of the drug, and in this regard, several formulations have been developed that allow the controlled release of the active compound. In this mini-review, the recent advances concerning the design and applications of natural polymer-based hydrogels containing PTX-loaded biocompatible nanocarriers are discussed. The technological features of these formulations as well as the therapeutic outcome achieved following their administration will be described, demonstrating their potential role as innovative systems to be used in anti-tumor therapy.
Collapse
Affiliation(s)
- Silvia Voci
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S.Venuta”, I-88100 Catanzaro, Italy; (S.V.); (A.G.); (M.F.)
| | - Agnese Gagliardi
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S.Venuta”, I-88100 Catanzaro, Italy; (S.V.); (A.G.); (M.F.)
| | | | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S.Venuta”, I-88100 Catanzaro, Italy; (S.V.); (A.G.); (M.F.)
| | - Donato Cosco
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S.Venuta”, I-88100 Catanzaro, Italy; (S.V.); (A.G.); (M.F.)
| |
Collapse
|
17
|
Chen Y, Pan Y, Hu D, Peng J, Hao Y, Pan M, Yuan L, Yu Y, Qian Z. Recent progress in nanoformulations of cabazitaxel. Biomed Mater 2021; 16:032002. [PMID: 33545700 DOI: 10.1088/1748-605x/abe396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The antitumor efficacy of various paclitaxel (PTX) and docetaxel (DTX) formulations in clinical applications is seriously affected by drug resistance. Cabazitaxel, a second-generation taxane, exhibits greater anticancer activity than paclitaxel and docetaxel and has low affinity for the P-glycoprotein (P-gp) efflux pump because of its structure. Therefore, cabazitaxel has the potential to overcome taxane resistance. However, owing to the high systemic toxicity and hydrophobicity of cabazitaxel and the instability of its commercial preparation, Jevtana®, the clinical use of cabazitaxel is restricted to patients with metastatic castration-resistant prostate cancer (mCRPC) who show progression after docetaxel-based chemotherapy. Nanomedicine is expected to overcome the limitations associated with cabazitaxel application and surmount taxane resistance. This review outlines the drug delivery systems of cabazitaxel published in recent years, summarizes the challenges faced in the development of cabazitaxel nanoformulations, and proposes strategies to overcome these challenges.
Collapse
Affiliation(s)
- Yu Chen
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Yue Pan
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Danrong Hu
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Jinrong Peng
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Ying Hao
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Meng Pan
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Liping Yuan
- Sichuan University, Sichuan University, Chengdu, 610065, CHINA
| | - Yongyang Yu
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Zhiyong Qian
- West China Hospital West China Medical School, Sichuan University, Sichuan University, Chengdu, 610041, CHINA
| |
Collapse
|
18
|
Chang X, Sun D, Shi D, Wang G, Chen Y, Zhang K, Tan H, Liu J, Liu B, Ouyang L. Design, synthesis, and biological evaluation of quinazolin-4(3 H)-one derivatives co-targeting poly(ADP-ribose) polymerase-1 and bromodomain containing protein 4 for breast cancer therapy. Acta Pharm Sin B 2021; 11:156-180. [PMID: 33532187 PMCID: PMC7838034 DOI: 10.1016/j.apsb.2020.06.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/08/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
This study was aimed to design the first dual-target small-molecule inhibitor co-targeting poly (ADP-ribose) polymerase-1 (PARP1) and bromodomain containing protein 4 (BRD4), which had important cross relation in the global network of breast cancer, reflecting the synthetic lethal effect. A series of new BRD4 and PARP1 dual-target inhibitors were discovered and synthesized by fragment-based combinatorial screening and activity assays that together led to the chemical optimization. Among these compounds, 19d was selected and exhibited micromole enzymatic potencies against BRD4 and PARP1, respectively. Compound 19d was further shown to efficiently modulate the expression of BRD4 and PARP1. Subsequently, compound 19d was found to induce breast cancer cell apoptosis and stimulate cell cycle arrest at G1 phase. Following pharmacokinetic studies, compound 19d showed its antitumor activity in breast cancer susceptibility gene 1/2 (BRCA1/2) wild-type MDA-MB-468 and MCF-7 xenograft models without apparent toxicity and loss of body weight. These results together demonstrated that a highly potent dual-targeted inhibitor was successfully synthesized and indicated that co-targeting of BRD4 and PARP1 based on the concept of synthetic lethality would be a promising therapeutic strategy for breast cancer.
Collapse
Key Words
- BC, breast cancer
- BET, bromodomain and extra-terminal domain
- BRCA1/2, breast cancer susceptibility gene 1/2
- BRD4
- BRD4, bromodomain 4
- CDK4/6, cyclin-dependent kinase 4/6
- DSB, DNA double-strand break
- Dual-target inhibitor
- EGFR, epidermal growth factor receptor
- ELISA, enzyme linked immunosorbent assay
- ER, estrogen receptor
- ESI-HR-MS, high-resolution mass spectra
- FDA, U.S. Food and Drug Administration
- FITC, fluorescein isothiocyanate isomer I
- HE, hematoxylin-eosin
- HPLC, high-performance liquid chromatography
- HR, homologous recombination
- HRD, homologous recombination deficiency
- IHC, immunohistochemistry
- NHEJ, nonhomologous end-joining
- PARP1
- PARP1, poly(ADP-ribose) polymerase-1
- PI, propidium iodide
- PK, pharmacokinetics
- PPI, protein−protein interaction
- Quinazolin-4(3H)-one derivatives
- SAR, structure–activity relationship
- SOP, standard operation process
- Synthetic lethality
- TCGA, the cancer genome atlas
- TGI, tumor growth inhibition
- TLC, thin-layer chromatography
- TNBC, triple-negative breast cancer
- TR-FRET, time-resolved fluorescence resonance energy transfer.
- shRNA, short hairpin RNA
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jie Liu
- Corresponding authors. Tel./fax: +86 28 85503817 (Jie Liu), +86 28 85164063 (Bo Liu), +86 28 85503817 (Liang Ouyang).
| | - Bo Liu
- Corresponding authors. Tel./fax: +86 28 85503817 (Jie Liu), +86 28 85164063 (Bo Liu), +86 28 85503817 (Liang Ouyang).
| | - Liang Ouyang
- Corresponding authors. Tel./fax: +86 28 85503817 (Jie Liu), +86 28 85164063 (Bo Liu), +86 28 85503817 (Liang Ouyang).
| |
Collapse
|
19
|
Liu M, Fu M, Yang X, Jia G, Shi X, Ji J, Liu X, Zhai G. Paclitaxel and quercetin co-loaded functional mesoporous silica nanoparticles overcoming multidrug resistance in breast cancer. Colloids Surf B Biointerfaces 2020; 196:111284. [DOI: 10.1016/j.colsurfb.2020.111284] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/16/2020] [Accepted: 07/26/2020] [Indexed: 12/24/2022]
|
20
|
Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B 2020; 10:2110-2124. [PMID: 33304781 PMCID: PMC7714990 DOI: 10.1016/j.apsb.2020.05.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
The complex tumor microenvironment is a most important factor in cancer development. The biological microenvironment is composed of a variety of barriers including the extracellular matrix and associated cells such as endothelia cells, pericytes, and cancer-associated fibroblasts. Different strategies can be utilized to enhance nanoparticle-based drug delivery and distribution into tumor tissues addressing the extracellular matrix or cellular components. In addition to the biological microenvironment, the immunological conditions around the tumor tissue can be very complicated and cancer cells have various ways of evading immune surveillance. Nanoparticle drug delivery systems can enhance cancer immunotherapy by tuning the immunological response and memory of various immune cells such as T cells, B cells, macrophages, and dendritic cells. In this review, the main components in the tumor biological and immunological environment are discussed. The focus is on recent advances in nanoparticle-based drug delivery systems towards targets within the tumor microenvironment to improve cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| | - Diane J. Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
21
|
Liu Y, Wang Q, Lu Y, Deng H, Zhou X. Synergistic enhancement of cytotoxicity against cancer cells by incorporation of rectorite into the paclitaxel immobilized cellulose acetate nanofibers. Int J Biol Macromol 2020; 152:672-680. [DOI: 10.1016/j.ijbiomac.2020.02.184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/15/2020] [Accepted: 02/16/2020] [Indexed: 12/18/2022]
|
22
|
Fan GH, Zhu TY, Huang J. FNDC5 promotes paclitaxel sensitivity of non-small cell lung cancers via inhibiting MDR1. Cell Signal 2020; 72:109665. [PMID: 32353410 DOI: 10.1016/j.cellsig.2020.109665] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/18/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022]
Abstract
Therapeutic benefits and clinical application of paclitaxel for treating non-small cell lung cancers (NSCLCs) are extremely hampered due to the chemoresistance. A recent study found that fibronectin type III domain-containing protein 5 (FNDC5) was downregulated in NSCLCs cells and negatively correlated with the clinicopathological characteristics in patients with NSCLCs. However, the role and potential molecular basis for FNDC5 in paclitaxel sensitivity of NSCLCs remain unclear. Paclitaxel-sensitive or resistant NSCLCs cell lines were exposed to small interfering RNA against FNDC5 (siFndc5) or recombinant irisin in the presence or absence of paclitaxel. NSCLCs cell lines have decreased FNDC5 expression compared with the normal human lung epithelial cells, which was further downregulated in paclitaxel-resistant cells. Irisin treatment suppressed, whereas Fndc5 silence promoted NSCLCs cells proliferation under basal conditions. Besides, we found that FNDC5 increased paclitaxel chemosensitivity in paclitaxel-sensitive or resistant NSCLCs cell lines via downregulating multidrug resistance protein 1 (MDR1). Further studies revealed that FNDC5 inhibited MDR1 expression via blocking nuclear factor-κB (NF-κB) activation. FNDC5 promotes paclitaxel sensitivity of NSCLCs cells via inhibiting NF-κB/MDR1 signaling, and FNDC5 might be a novel therapeutic target for the treatment of NSCLCs.
Collapse
Affiliation(s)
- Guo-Hua Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tie-Yuan Zhu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Jie Huang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
23
|
Shetty A, Nagesh PK, Setua S, Hafeez BB, Jaggi M, Yallapu MM, Chauhan SC. Novel Paclitaxel Nanoformulation Impairs De Novo Lipid Synthesis in Pancreatic Cancer Cells and Enhances Gemcitabine Efficacy. ACS OMEGA 2020; 5:8982-8991. [PMID: 32337462 PMCID: PMC7178800 DOI: 10.1021/acsomega.0c00793] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/01/2020] [Indexed: 05/08/2023]
Abstract
Pancreatic cancer (PanCa) is a highly lethal disease with a poor 5 year survival rate, less than 7%. It has a dismal prognosis, and more than 50% of cases are detected at an advanced and metastatic stage. Gemcitabine (GEM) is a gold standard chemotherapy used for PanCa treatment. However, GEM-acquired resistance in cancer cells is considered as a major setback for its continued clinical implementation. This phenomenon is evidently linked to de novo lipid synthesis. PanCa cells rely on de novo lipid synthesis, which is a prime event in survival and one of the key drivers for tumorigenesis, cancer progression, and drug resistance. Thus, the depletion of lipogenesis or lipid metabolism can not only improve treatment outcomes but also overcome chemoresistance, which is an unmet clinical need. Toward this effort, our study reports a unique paclitaxel-poly(lactic-co-glycolic acid) (PLGA) nanoparticles (PPNPs) formulation which can target lipid metabolism and improve anticancer efficacy of GEM in PanCa cells. PPNPs inhibit excessive lipid formation and alter membrane stability with compromised membrane integrity, which was confirmed by Fourier transform infrared and zeta potential measurements. The effective interference of PPNPs in lipid metabolic signaling was determined by reduction in the expression of FASN, ACC, lipin, and Cox-2 proteins. This molecular action profoundly enhances efficacy of GEM as evident through enhanced inhibitory effects on the tumorigenic and metastasis assays in PanCa cells. These data clearly suggest that the ablation of lipid metabolism might offer an innovative approach for the improved therapeutic outcome in PanCa patients.
Collapse
Affiliation(s)
- Advait Shetty
- Department
of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
| | - Prashanth K.B. Nagesh
- Department
of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
| | - Saini Setua
- Department
of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
| | - Bilal B. Hafeez
- Department
of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
| | - Meena Jaggi
- Department
of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
| | - Murali M. Yallapu
- Department
of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
| | - Subhash C. Chauhan
- Department
of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, 78539 Texas, United States
| |
Collapse
|
24
|
Wang H, Li X, Yang H, Wang J, Li Q, Qu R, Wu X. Nanocomplexes based polyvinylpyrrolidone K-17PF for ocular drug delivery of naringenin. Int J Pharm 2020; 578:119133. [DOI: 10.1016/j.ijpharm.2020.119133] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/17/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023]
|
25
|
d'Avanzo N, Celia C, Barone A, Carafa M, Di Marzio L, Santos HA, Fresta M. Immunogenicity of Polyethylene Glycol Based Nanomedicines: Mechanisms, Clinical Implications and Systematic Approach. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900170] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Nicola d'Avanzo
- Department of Health SciencesUniversity of Catanzaro “Magna Græcia” Campus Universitario “S. Venuta”, Viale Europa I‐88100 Catanzaro Italy
| | - Christian Celia
- Department of PharmacyUniversity of Chieti‐Pescara “G. d'Annunzio” Via dei Vestini 31 I‐66100 Chieti Italy
| | - Antonella Barone
- Department of Health SciencesUniversity of Catanzaro “Magna Græcia” Campus Universitario “S. Venuta”, Viale Europa I‐88100 Catanzaro Italy
| | - Maria Carafa
- Department of Drug Chemistry and TechnologyUniversity of Rome “Sapienza” 00185 Rome Italy
| | - Luisa Di Marzio
- Department of PharmacyUniversity of Chieti‐Pescara “G. d'Annunzio” Via dei Vestini 31 I‐66100 Chieti Italy
| | - Hélder A. Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy; and Helsinki Institute of Life Science (HiLIFE)University of Helsinki FI‐00014 Helsinki Finland
| | - Massimo Fresta
- Department of Health SciencesUniversity of Catanzaro “Magna Græcia” Campus Universitario “S. Venuta”, Viale Europa I‐88100 Catanzaro Italy
| |
Collapse
|
26
|
Nagesh PKB, Chowdhury P, Hatami E, Jain S, Dan N, Kashyap VK, Chauhan SC, Jaggi M, Yallapu MM. Tannic acid inhibits lipid metabolism and induce ROS in prostate cancer cells. Sci Rep 2020; 10:980. [PMID: 31969643 PMCID: PMC6976712 DOI: 10.1038/s41598-020-57932-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/29/2019] [Indexed: 12/27/2022] Open
Abstract
Prostate cancer (PCa) cells exploit the aberrant lipid signaling and metabolism as their survival advantage. Also, intracellular storage lipids act as fuel for the PCa proliferation. However, few studies were available that addressed the topic of targeting lipid metabolism in PCa. Here, we assessed the tannic acid (TA) lipid-targeting ability and its capability to induce endoplasmic reticulum (ER) stress by reactive oxygen species (ROS) in PCa cells. TA exhibited dual effects by inhibiting lipogenic signaling and suppression of lipid metabolic pathways. The expression of proteins responsible for lipogenesis was down regulated. The membrane permeability and functionality of PCa were severely affected and caused nuclear disorganization during drug exposure. Finally, these consolidated events shifted the cell's survival balance towards apoptosis. These results suggest that TA distinctly interferes with the lipid signaling and metabolism of PCa cells.
Collapse
Affiliation(s)
- Prashanth K B Nagesh
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Elham Hatami
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Shashi Jain
- Tumor Initiation and Maintenance, Sanford-Burnham Medical Research Institute, La Jolla, California, 92037, USA
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Nirnoy Dan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Vivek Kumar Kashyap
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Subhash C Chauhan
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Meena Jaggi
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA.
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
27
|
Kashyap VK, Dan N, Chauhan N, Wang Q, Setua S, Nagesh PKB, Malik S, Batra V, Yallapu MM, Miller DD, Li W, Hafeez BB, Jaggi M, Chauhan SC. VERU-111 suppresses tumor growth and metastatic phenotypes of cervical cancer cells through the activation of p53 signaling pathway. Cancer Lett 2019; 470:64-74. [PMID: 31809801 DOI: 10.1016/j.canlet.2019.11.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/19/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
In this study, we investigated the therapeutic efficacy of VERU-111 in vitro and in vivo model systems of cervical cancer. VERU-111 treatment inhibited cell proliferation and, clonogenic potential, induce accumulation of p53 and down regulated the expression of HPV E6/E7 expression in cervical cancer cells. In addition, VERU-111 treatment also decreased the phosphorylation of Jak2(Tyr1007/1008) and STAT3 at Tyr705 and Ser727. VERU-111 treatment arrested cell cycle in the G2/M phase and modulated cell cycle regulatory proteins (cyclin B1, p21, p34cdc2 and pcdk1). Moreover, VERU-111 treatment induced apoptosis and modulated the expression of Bid, Bcl-xl, Survivin, Bax, Bcl2 and cleavage in PARP. In functional assays, VERU-111 markedly reduced the migratory and invasive potential of cervical cancer cells via modulations of MMPs. VERU-111 treatment also showed significant (P < 0.05) inhibition of orthotopic xenograft tumor growth in athymic nude mice. Taken together, our results demonstrate the potent anti-cancer efficacy of VERU-111 in experimental cervical cancer models.Thus, VERU-111 can be explored as a promising therapeutic agent for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Vivek K Kashyap
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nirnoy Dan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Neeraj Chauhan
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Qinghui Wang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Saini Setua
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Prashanth K B Nagesh
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Shabnam Malik
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Vivek Batra
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Bilal B Hafeez
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, The University of Texas Rio Grande Valley, McAllen, TX, 78504, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
28
|
Gjuroski I, Girousi E, Meyer C, Hertig D, Stojkov D, Fux M, Schnidrig N, Bucher J, Pfister S, Sauser L, Simon HU, Vermathen P, Furrer J, Vermathen M. Evaluation of polyvinylpyrrolidone and block copolymer micelle encapsulation of serine chlorin e6 and chlorin e4 on their reactivity towards albumin and transferrin and their cell uptake. J Control Release 2019; 316:150-167. [PMID: 31689463 DOI: 10.1016/j.jconrel.2019.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/02/2019] [Accepted: 10/02/2019] [Indexed: 01/10/2023]
Abstract
Encapsulation of porphyrinic photosensitizers (PSs) into polymeric carriers plays an important role in enhancing their efficiency as drugs in photodynamic therapy (PDT). Porphyrin aggregation and low solubility as well as the preservation of the advantageous photophysical properties pose a challenge on the design of efficient PS-carrier systems. Block copolymer micelles (BCMs) and polyvinylpyrrolidone (PVP) are promising drug delivery vehicles for physical entrapment of PSs. BCMs exhibit enhanced dynamics as compared to the less flexible PVP network. In the current work the question is addressed how these different dynamics affect PS encapsulation, release from the carrier, reaction with serum proteins, and cellular uptake. The porphyrinic compounds serine-amide of chlorin e6 (SerCE) and chlorin e4 (CE4) were used as model PSs with different lipophilicity and aggregation properties. 1H NMR and fluorescence spectroscopy were applied to study their interactions with PVP and BCMs consisting of Kolliphor P188 (KP). Both chlorins were well encapsulated by the carriers and had improved photophysical properties. Compared to SerCE, the more lipophilic CE4 exhibited stronger hydrophobic interactions with the BCM core, stabilizing the system and preventing exchange with the surrounding medium as was shown by NMR NOESY and DOSY experiments. PVP and BCMs protected the encapsulated chlorins against interaction with human transferrin (Tf). However, SerCE and CE4 were released from BCMs in favor of binding to human serum albumin (HSA) while PVP prevented interaction with HSA. Fluorescence spectroscopic studies revealed that HSA binds to the surface of PVP forming a protein corona. PVP and BCMs reduced cellular uptake of the chlorins. However, encapsulation into BCMs resulted in more efficient cell internalization for CE4 than for SerCE. HSA significantly lowered both, free and carrier-mediated cell uptake for CE4 and SerCE. In conclusion, PVP appears as the more universal delivery system covering a broad range of host molecules with respect to polarity, whereas BCMs require a higher drug-carrier compatibility. Poorly soluble hydrophobic PSs benefit stronger from BCM-type carriers due to enhanced bioavailability through disaggregation and solubilization allowing for more efficient cell uptake. In addition, increased PS-carrier hydrophobic interactions have a stabilizing effect. For more hydrophilic PSs, the main advantage of polymeric carriers like PVP or poloxamer micelles lies in their protection during the transport through the bloodstream. HSA binding plays an important role for drug release and cell uptake in carrier-mediated delivery to the target tissue.
Collapse
Affiliation(s)
- Ilche Gjuroski
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland.
| | - Eleftheria Girousi
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Christoph Meyer
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Damian Hertig
- Department of BioMedical Research and Radiology, University of Bern and Inselspital, sitem-insel AG, Freiburgstrasse 3, CH-3010, Bern, Switzerland; Institute of Clinical Chemistry, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Michaela Fux
- Institute of Clinical Chemistry, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Nicolas Schnidrig
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Jan Bucher
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Sara Pfister
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Luca Sauser
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Peter Vermathen
- Department of BioMedical Research and Radiology, University of Bern and Inselspital, sitem-insel AG, Freiburgstrasse 3, CH-3010, Bern, Switzerland
| | - Julien Furrer
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Martina Vermathen
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland.
| |
Collapse
|
29
|
Nagesh PK, Chowdhury P, Hatami E, Kumari S, Kashyap VK, Tripathi MK, Wagh S, Meibohm B, Chauhan SC, Jaggi M, Yallapu MM. Cross-Linked Polyphenol-Based Drug Nano-Self-Assemblies Engineered to Blockade Prostate Cancer Senescence. ACS APPLIED MATERIALS & INTERFACES 2019; 11:38537-38554. [PMID: 31553876 PMCID: PMC8020616 DOI: 10.1021/acsami.9b14738] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Cellular senescence is one of the prevailing issues in cancer therapeutics that promotes cancer relapse, chemoresistance, and recurrence. Patients undergoing persistent chemotherapy often develop drug-induced senescence. Docetaxel, an FDA-approved treatment for prostate cancer, is known to induce cellular senescence which often limits the overall survival of patients. Strategic therapies that counter the cellular and drug-induced senescence are an unmet clinical need. Towards this an effort was made to develop a novel therapeutic strategy that targets and removes senescent cells from the tumors, we developed a nanoformulation of tannic acid-docetaxel self-assemblies (DSAs). The construction of DSAs was confirmed through particle size measurements, spectroscopy, thermal, and biocompatibility studies. This formulation exhibited enhanced in vitro therapeutic activity in various biological functional assays with respect to native docetaxel treatments. Microarray and immunoblot analysis results demonstrated that DSAs exposure selectively deregulated senescence associated TGFβR1/FOXO1/p21 signaling. Decrease in β-galactosidase staining further suggested reversion of drug-induced senescence after DSAs exposure. Additionally, DSAs induced profound cell death by activation of apoptotic signaling through bypassing senescence. Furthermore, in vivo and ex vivo imaging analysis demonstrated the tumor targeting behavior of DSAs in mice bearing PC-3 xenograft tumors. The antisenescence and anticancer activity of DSAs was further shown in vivo by inhibiting TGFβR1 proteins and regressing tumor growth through apoptotic induction in the PC-3 xenograft mouse model. Overall, DSAs exhibited such advanced features due to a natural compound in the formulation as a matrix/binder for docetaxel. Overall, DSAs showed superior tumor targeting and improved cellular internalization, promoting docetaxel efficacy. These findings may have great implications in prostate cancer therapy.
Collapse
Affiliation(s)
- Prashanth K.B. Nagesh
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Elham Hatami
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Sonam Kumari
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Vivek Kumar Kashyap
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Manish K. Tripathi
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Santosh Wagh
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Subhash C. Chauhan
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Meena Jaggi
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Murali M. Yallapu
- Department of Microbiology and Immunology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Corresponding Author Mailing address: Department of Immunology and Microbiology, 5300 North L Street, Room 2.249, McAllen, TX 78504. Phone: (956) 296-1705. Fax No: (956)-296-1325.
| |
Collapse
|
30
|
Wang D, Wang Y, Zhao G, Zhuang J, Wu W. Improving systemic circulation of paclitaxel nanocrystals by surface hybridization of DSPE-PEG2000. Colloids Surf B Biointerfaces 2019; 182:110337. [DOI: 10.1016/j.colsurfb.2019.06.066] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/16/2019] [Accepted: 06/28/2019] [Indexed: 01/15/2023]
|
31
|
Amanzadeh E, Esmaeili A, Rahgozar S, Nourbakhshnia M. Application of quercetin in neurological disorders: from nutrition to nanomedicine. Rev Neurosci 2019; 30:555-572. [DOI: 10.1515/revneuro-2018-0080] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022]
Abstract
Abstract
Quercetin is a polyphenolic flavonoid, which is frequently found in fruits and vegetables. The antioxidant potential of quercetin has been studied from subcellular compartments, that is, mitochondria to tissue levels in the brain. The neurodegeneration process initiates alongside aging of the neurons. It appears in different parts of the brain as Aβ plaques, neurofibrillary tangles, Lewy bodies, Pick bodies, and others, which leads to Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and other diseases. So far, no specific treatment has been identified for these diseases. Despite common treatments that help to prevent the development of disease, the condition of patients with progressive neurodegenerative diseases usually do not completely improve. Currently, the use of flavonoids, especially quercetin for the treatment of neurodegenerative diseases, has been expanded in animal models. It has also been used to treat animal models of neurodegenerative diseases. In addition, improvements in behavioral levels, as well as in cellular and molecular levels, decreased activity of antioxidant and apoptotic proteins, and increased levels of antiapoptotic proteins have been observed. Low bioavailability of quercetin has also led researchers to construct various quercetin-involved nanoparticles. The treatment of animal models of neurodegeneration using quercetin-involved nanoparticles has shown that improvements are observed in shorter periods and with use of lower concentrations. Indeed, intranasal administration of quercetin-involved nanoparticles, constructing superparamagnetic nanoparticles, and combinational treatment using nanoparticles such as quercetin and other drugs are suggested for future studies.
Collapse
|
32
|
Lu J, Li J, Hu Y, Guo Z, Sun D, Wang P, Guo K, Duan DD, Gao S, Jiang J, Wang J, Liu P. Chrysophanol protects against doxorubicin-induced cardiotoxicity by suppressing cellular PARylation. Acta Pharm Sin B 2019; 9:782-793. [PMID: 31384538 PMCID: PMC6663922 DOI: 10.1016/j.apsb.2018.10.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/26/2018] [Accepted: 10/09/2018] [Indexed: 01/13/2023] Open
Abstract
The clinical application of doxorubicin (DOX) in cancer chemotherapy is limited by its life-threatening cardiotoxic effects. Chrysophanol (CHR), an anthraquinone compound isolated from the rhizome of Rheum palmatum L., is considered to play a broad role in a variety of biological processes. However, the effects of CHR׳s cardioprotection in DOX-induced cardiomyopathy is poorly understood. In this study, we found that the cardiac apoptosis, mitochondrial injury and cellular PARylation levels were significantly increased in H9C2 cells treated by Dox, while these effects were suppressed by CHR. Similar results were observed when PARP1 activity was suppressed by its inhibitors 3-aminobenzamide (3AB) and ABT888. Ectopic expression of PARP1 effectively blocked this CHR׳s cardioprotection against DOX-induced cardiomyocyte injury in H9C2 cells. Furthermore, pre-administration with both CHR and 3AB relieved DOX-induced cardiac apoptosis, mitochondrial impairment and heart dysfunction in Sprague-Dawley rat model. These results revealed that CHR protects against DOX-induced cardiotoxicity by suppressing cellular PARylation and provided critical evidence that PARylation may be a novel target for DOX-induced cardiomyopathy.
Collapse
Key Words
- 3AB, 3-aminobenzamide
- ADR, adriamycin
- ANOVA, one-way analysis of variance
- Apoptosis
- CHR, chrysophanol
- CMC-Na, sodium carboxymethyl
- CO, cardiac output
- Cardiotoxicity
- Chrysophanol
- Cyt c, Cytochrome c
- DOX, doxorubicin
- Doxorubicin
- EF, ejection fraction
- FBS, fetal bovine serum
- FS, fractional shortening
- HE, hematoxylin-eosin
- HR, heart rate
- IVSd, end-diastolic interventricular septum
- IVSs, end-systolic interventricular septum
- LV, end-systolic volume
- LVEDV, LV end-diastolic volume
- LVIDd, LV end-diastolic internal diameter
- LVIDs, LV end-systolic internal diameter
- LVPWd, LV end-diastolic posterior wall thickness
- LVPWs, LV end-systolic posterior wall thickness
- Mitochondria
- NS, normal saline
- PAR, polymers of ADP-ribose
- PARP1, poly(ADP-ribose) polymerase 1
- PARylated, poly(ADP-ribosyl)ated
- PARylation
- PARylation, poly(ADP-ribosyl)ation
- PBS, phosphate-buffered saline
- RCR, respiratory control ratio
- ROS, reactive oxygen species
- Rh123, rhodamine 123
- SD, Sprague–Dawley
- TUNEL, TdT-mediated dUTP nick end labeling
- VDAC1, voltage dependent anion channel 1
Collapse
|
33
|
Vinothini K, Jeyaraj M, Kumar SK, Rajan M. Dual Role of Lanthanum Oxide Nanoparticles Functionalized Co‐Polymeric Micelle for Extended Anti‐Cancer Drug Delivery. ChemistrySelect 2019. [DOI: 10.1002/slct.201803339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Kandasamy Vinothini
- Biomaterials in Medicinal Chemistry LaboratoryDepartment of Natural Products ChemistrySchool of ChemistryMadurai Kamaraj University Madurai - 625021 India
| | - Murugaraj Jeyaraj
- National Centre for Nanoscience and NanotechnologyUniversity of Madras, Guindy Campus Chennai- 25 India
| | | | - Mariappan Rajan
- Biomaterials in Medicinal Chemistry LaboratoryDepartment of Natural Products ChemistrySchool of ChemistryMadurai Kamaraj University Madurai - 625021 India
| |
Collapse
|
34
|
Kim D, Le QV, Kim YB, Oh YK. Safety and photochemotherapeutic application of poly( γ-glutamic acid)-based biopolymeric nanoparticle. Acta Pharm Sin B 2019; 9:565-574. [PMID: 31193800 PMCID: PMC6543094 DOI: 10.1016/j.apsb.2019.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/09/2018] [Accepted: 11/15/2018] [Indexed: 12/17/2022] Open
Abstract
The safety of nanomaterials, a crucial consideration for clinical translation, is enhanced by using building blocks that are biologically nontoxic. Here, we used poly(γ-glutamic acid) (γ-PGA) and dopamine as building blocks of polymeric nanomaterials for carrying hydrophobic anticancer drugs. The introduction of phenylalanine onto γ-PGA enabled the resulting amphiphilic derivative of γ-PGA acid to self-assemble in the presence of the anticancer drug paclitaxel (PTX) to form PTX-encapsulated micelles. The surfaces of PTX-loaded micelles were then coated with polymerized dopamine (PDA). The PDA-coated, amphiphilic γ-PGA-based micelles (AM) carrying PTX (PDA/AM/P) exerted near-infrared-responsive photothermal effects. Near-infrared irradiation of cancer cells treated with PDA/AM/P nanoparticles produced a greater anticancer effect than that observed in other treatment groups, indicating a synergistic effect. Intravenous administration of PDA/AM/P completely ablated tumors and prevented their recurrence. Notably, the in vivo safety profile of PDA/AM/P nanoparticles allowed PTX to be delivered at a 3.6-fold higher dose than was possible with PTX solubilized in surfactant, and circumvented the side effects of the surfactant. These results support the multifunctional potential of PDA/AM for the delivery of various hydrophobic drugs and imaging dyes for safe translation of nanomaterials into the clinic.
Collapse
|
35
|
Hatami E, Bhusetty Nagesh PK, Chowdhury P, Elliot S, Shields D, Chand Chauhan S, Jaggi M, Yallapu MM. Development of Zoledronic Acid-Based Nanoassemblies for Bone-Targeted Anticancer Therapy. ACS Biomater Sci Eng 2019; 5:2343-2354. [PMID: 33405784 DOI: 10.1021/acsbiomaterials.9b00362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Bone metastasis occurs in the majority of cancer patients, which hampers quality of life and significantly decreases survival. Aggressive chemotherapy is a traditional treatment regimen that induces severe systemic toxicities. Therefore, bone-directed therapies are highly warranted. We report a novel nanoparticle formulation that is composed of poly(vinylpyrrolidone) and tannic acid core nanoparticles (PVT NPs) that forms self-assembly with zoledronic acid (ZA@PVT NPs). The construction of ZA@PVT NPs was confirmed by particle size, zeta potential, transmission electron microscopy, and spectral analyses. An optimized bone-targeted ZA@PVT NPs formulation showed greater binding and internalization in in vitro with metastasis prostate and breast cancer cells. ZA@PVT NPs were able to deliver ZA more efficiently to tumor cells, which inhibited proliferation of human prostate and breast cancer cells. In addition, ZA@PVT NPs were capable of targeting mouse bones and prostate tumor microarray tissues (ex vivo) while sparing all other vital organs. More importantly, ZA@PVT NPs induce chemo sensitization to docetaxel treatment in cancer cells. Overall, the study results confirm that ZA-based, bone-targeted NPs have great potential for the treatment of bone metastasis in the near future.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Prashanth Kumar Bhusetty Nagesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Stacie Elliot
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Deanna Shields
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Subhash Chand Chauhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Murali Mohan Yallapu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| |
Collapse
|
36
|
Redox-sensitive prodrug nanoassemblies based on linoleic acid-modified docetaxel to resist breast cancers. Acta Pharm Sin B 2019; 9:421-432. [PMID: 30972286 PMCID: PMC6437471 DOI: 10.1016/j.apsb.2018.08.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/12/2018] [Accepted: 07/30/2018] [Indexed: 12/21/2022] Open
Abstract
Prodrug nanoassemblies, which can refrain from large excipients, achieve higher drug loading and control drug release, have been placed as the priority in drug delivery system. Reasoning that glutathione (GSH) and reactive oxygen species (ROS) are highly upgraded in tumor tissues which makes them attractive targets for drug delivery system, we designed and synthetized a novel prodrug which utilized mono thioether bond as a linker to bridge linoleic acid (LA) and docetaxel (DTX). This mono thioether-linked conjugates (DTX-S-LA) could self-assemble into nanoparticles without the aid of much excipients. The mono thioether endowed the nanoparticles redox sensitivity resulting in specific release at the tumor tissue. Our studies demonstrated that the nanoassemblies had uniform particle size, high stability and fast release behavior. DTX-S-LA nanoassemblies outperformed DTX solution in pharmacokinetic profiles for it had longer circulation time and higher area under curve (AUC). Compared with DTX solution, the redox dual-responsive nanoassemblies had comparable cytotoxic activity. Besides, the antitumor efficacy was evaluated in mice bearing 4T1 xenograft. It turned out this nanoassemblies could enhance anticancer efficacy by increasing the dose because of higher tolerance. Overall, these results indicated that the redox sensitivity nanoassemblies may have a great potential to cancer therapy.
Collapse
Key Words
- ALT, alanine transaminase
- AST, aspartate transaminase
- AUC, area under the curve
- Antitumor efficacy
- BUN, blood urea nitrogen
- C-6, coumarin-6
- CREA, creatinine
- DDS, drug delivery system
- DMSO, dimethyl sulfoxide
- DSPE-PEG2K, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethyleneglycol)-2000]
- DTT, d,l-dithiothreitol
- DTX, docetaxel
- Docetaxel
- EDCI, N-(3-dimethylaminopropyl)-N′-ethyl carbodiimide hydrochloride
- FBS, fetal bovine serum
- GSH, glutathione
- H2O2, hydrogen peroxide
- HOBt, 1-hydroxybenzotriazole monohydrate
- HPLC, high-performance liquid chromatography
- IC50, half maximal inhibitory concentration
- LA, linoleic acid
- Linoleic acid
- MTT, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide
- Mono thioether bond
- Nanoassemblies
- PBS, phosphate buffer saline
- PDI, polydispersity index
- PTX, paclitaxel
- Pharmacokinetics
- ROS, reactive oxygen species
- SD, standard deviation
- TLC, thin layer chromatography
Collapse
|
37
|
Hatami E, Mu Y, Shields DN, Chauhan SC, Kumar S, Cory TJ, Yallapu MM. Mannose-decorated hybrid nanoparticles for enhanced macrophage targeting. Biochem Biophys Rep 2019; 17:197-207. [PMID: 30723809 PMCID: PMC6351286 DOI: 10.1016/j.bbrep.2019.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
Our goal was to design nanocarriers that specifically target and deliver therapeutics to polarized macrophages. Mannose receptors are highly overexpressed on polarized macrophages. In this study, we constructed Pluronic® -F127 polymer and tannic acid (TA) based nanoparticles (F127-TA core nanoparticles) with varying mannose densities. The particle size of the optimized mannose-decorated F127-TA hybrid nanoparticles (MDNPs) was found to be ~ 265 nm with a negative zeta potential of ~ - 4.5 mV. No significant changes in the size and zeta potentials of nanoparticles were observed, which demonstrated structural integrity and stability of the nanoformulation. Physicochemical characteristics of MDNPs were evaluated by FTIR and TGA and demonstrated the presence of mannose units on surface nanoparticles. A mannose-dependent cellular targeting and uptake of MDNPs was found in U937 macrophages. The uptake process was found to vary directly with time and volume of MDNPs nanoparticles. The uptake pattern is higher in M2 than M1. This behavior was also evident from the instantaneous and superior binding profile of M2 macrophage lysate protein with MDNPs over that of M1 macrophage lysate protein. These results demonstrated that an appropriate mannose ligand density was confirmed, suggesting efficient targeting of M2. Altogether, these data support that the MDNPs formulation could serve as a targeted therapeutic guide in the generation of nanomedicine to treat various conditions as an anti-inflammation therapy.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ying Mu
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | - Subhash C. Chauhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Theodore J. Cory
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Murali M. Yallapu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
38
|
Chowdhury P, Nagesh PKB, Hatami E, Wagh S, Dan N, Tripathi MK, Khan S, Hafeez BB, Meibohm B, Chauhan SC, Jaggi M, Yallapu MM. Tannic acid-inspired paclitaxel nanoparticles for enhanced anticancer effects in breast cancer cells. J Colloid Interface Sci 2019; 535:133-148. [PMID: 30292104 PMCID: PMC6992213 DOI: 10.1016/j.jcis.2018.09.072] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022]
Abstract
Paclitaxel (PTX) is a gold standard chemotherapeutic agent for breast, ovarian, pancreatic and non-small cell lung carcinoma. However, in clinical use PTX can have adverse side effects or inadequate pharmacodynamic parameters, limiting its use. Nanotechnology is often employed to reduce the therapeutic dosage required for effective therapy, while also minimizing the systemic side effects of chemotherapy drugs. However, there is no nanoformulation of paclitaxel with chemosensitization motifs built in. With this objective, we screened eleven pharmaceutical excipients to develop an alternative paclitaxel nanoformulation using a self-assembly method. Based on the screening results, we observed tannic acid possesses unique properties to produce a paclitaxel nanoparticle formulation, i.e., tannic acid-paclitaxel nanoparticles. This stable TAP nanoformulation, referred to as TAP nanoparticles (TAP NPs), showed a spherical shape of ~ 102 nm and negative zeta potential of ~ -8.85. The presence of PTX in TAP NPs was confirmed by Fourier Transform Infrared (FTIR) spectra, thermogravimetric analyzer (TGA), and X-ray diffraction (XRD). Encapsulation efficiency of PTX in TAP NPs was determined to be ≥96%. Intracellular drug uptake of plain drug PTX on breast cancer cells (MDA-MB-231) shows more or less constant drug levels in 2 to 6 h, suggesting drug efflux by the P-gp transporters, over TAP NPs, in which PTX uptake was more than 95.52 ± 11.01% in 6 h, as analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Various biological assays such as proliferation, clonogenic formation, invasion, and migration confirm superior anticancer effects of TAP NPs over plain PTX at all tested concentrations. P-gp expression, beta-tubulin stabilization, Western blot, and microarray analysis further confirm the improved therapeutic potential of TAP NPs. These results suggest that the TAP nanoformulation provides an important reference for developing a therapeutic nanoformulation affording pronounced, enhanced effects in breast cancer therapy.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Elham Hatami
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Santosh Wagh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nirnoy Dan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Manish K Tripathi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sheema Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bilal B Hafeez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
39
|
Yu M, Su D, Yang Y, Qin L, Hu C, Liu R, Zhou Y, Yang C, Yang X, Wang G, Gao H. D-T7 Peptide-Modified PEGylated Bilirubin Nanoparticles Loaded with Cediranib and Paclitaxel for Antiangiogenesis and Chemotherapy of Glioma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:176-186. [PMID: 30525386 DOI: 10.1021/acsami.8b16219] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The blood-brain tumor barrier (BTB) and blood-brain barrier (BBB) have always been the major barriers in glioma therapy. In this report, we proposed D-T7 peptide-modified nanoparticles actively targeted glioma by overcoming the BBB and BTB to improve the antiglioma efficacy. Glioma-targeting experiments showed that the penetration effect of the D-T7 peptide-modified nanoparticles was 7.89-fold higher than that of unmodified nanoparticles. Furthermore, cediranib (CD) and paclitaxel (PTX) were used for the combination of the antiangiogenesis and chemotherapy for glioma. PEGylated bilirubin nanoparticles (BRNPs) were selected as a suitable drug delivery system (CD&PTX@TBRBPs) owing to the antioxidant, anti-inflammatory, and reactive oxygen species-responsive ability. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and apoptosis assays showed that CD&PTX@TBRBPs had the highest cytotoxicity and the median survival time of the CD&PTX@TBRNP group was 3.31-fold and 1.23-fold longer than that of the saline and CD&PTX@BRNP groups, respectively. All the results showed that we constructed a novel and accessible peptide-modified dual drug carrier with an enhanced antiglioma effect.
Collapse
Affiliation(s)
- Meinan Yu
- Faculty of Life Science and Technology , Kunming University of Science and Technology , 727 South Jing Ming Road , Chenggong County, Kunming 650500 , P. R. China
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Dunyan Su
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Yuanyuan Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Lin Qin
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Chuan Hu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Rui Liu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Yang Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Chuanyao Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Xiaotong Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| | - Guanlin Wang
- Faculty of Life Science and Technology , Kunming University of Science and Technology , 727 South Jing Ming Road , Chenggong County, Kunming 650500 , P. R. China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , No. 17, Block 3, Southern Renmin Road , Chengdu 610041 , P. R. China
| |
Collapse
|
40
|
Fu JJ, Zhang JY, Li SP, Zhang LM, Lin ZX, Liang L, Qin AP, Yu XY. CuS Nanodot-Loaded Thermosensitive Hydrogel for Anticancer Photothermal Therapy. Mol Pharm 2018; 15:4621-4631. [PMID: 30179511 DOI: 10.1021/acs.molpharmaceut.8b00624] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ji-jun Fu
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
| | - Jian-ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
| | - Song-pei Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
| | - Ling-min Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhong-xiao Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
| | - Lu Liang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
| | - Ai-ping Qin
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
| | - Xi-yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 511436, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
| |
Collapse
|
41
|
Nagesh PKB, Chowdhury P, Hatami E, Boya VKN, Kashyap VK, Khan S, Hafeez BB, Chauhan SC, Jaggi M, Yallapu MM. miRNA-205 Nanoformulation Sensitizes Prostate Cancer Cells to Chemotherapy. Cancers (Basel) 2018; 10:E289. [PMID: 30149628 PMCID: PMC6162422 DOI: 10.3390/cancers10090289] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/07/2018] [Accepted: 08/21/2018] [Indexed: 02/07/2023] Open
Abstract
The therapeutic application of microRNA(s) in the field of cancer has generated significant attention in research. Previous studies have shown that miR-205 negatively regulates prostate cancer cell proliferation, metastasis, and drug resistance. However, the delivery of miR-205 is an unmet clinical need. Thus, the development of a viable nanoparticle platform to deliver miR-205 is highly sought. A novel magnetic nanoparticle (MNP)-based nanoplatform composed of an iron oxide core with poly(ethyleneimine)-poly(ethylene glycol) layer(s) was developed. An optimized nanoplatform composition was confirmed by examining the binding profiles of MNPs with miR-205 using agarose gel and fluorescence methods. The novel formulation was applied to prostate cancer cells for evaluating cellular uptake, miR-205 delivery, and anticancer, antimetastasis, and chemosensitization potentials against docetaxel treatment. The improved uptake and efficacy of formulations were studied with confocal imaging, flow cytometry, proliferation, clonogenicity, Western blot, q-RT-PCR, and chemosensitization assays. Our findings demonstrated that the miR-205 nanoplatform induces significant apoptosis and enhancing chemotherapeutic effects in prostate cancer cells. Overall, these study results provide a strong proof-of-concept for a novel nonviral-based nanoparticle protocol for effective microRNA delivery to prostate cancer cells.
Collapse
Affiliation(s)
- Prashanth K B Nagesh
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Elham Hatami
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Vijaya K N Boya
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Vivek K Kashyap
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sheema Khan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Bilal B Hafeez
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
42
|
Hatami E, Nagesh PKB, Chowdhury P, Chauhan SC, Jaggi M, Samarasinghe AE, Yallapu MM. Tannic Acid-Lung Fluid Assemblies Promote Interaction and Delivery of Drugs to Lung Cancer Cells. Pharmaceutics 2018; 10:E111. [PMID: 30071698 PMCID: PMC6161105 DOI: 10.3390/pharmaceutics10030111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/21/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023] Open
Abstract
Lung cancer (LC) is one of the leading causes of death in both men and women in the United States. Tannic acid (TA), a water-soluble polyphenol, exhibits a wide range of biological activities. TA has received much attention as a promising compound in the biomaterial and drug delivery fields. Lung fluid (LF) is a major barrier for distribution of drugs to the lungs. Therefore, the purpose of this study was to examine TA interaction with LF for effective delivery of anti-cancer drug molecules via pulmonary delivery. The extent of adsorption of LF proteins by TA was revealed by fluorescence quenching in fluorescence spectroscopy. The presence of LF in TA-LF complexes was noticed by the presence of protein peaks at 1653 cm-1. Both protein dot and SDS-PAGE analysis confirmed LF protein complexation at all TA concentrations employed. A stable particle TA-LF complex formation was observed through transmission electron microscopy (TEM) analysis. The complexation pattern measured by dynamic light scattering (DLS) indicated that it varies depending on the pH of the solutions. The degree of LF presence in TA-LF complexes signifies its interactive behavior in LC cell lines. Such superior interaction offered an enhanced anti-cancer activity of drugs encapsulated in TA-LF complex nanoformulations. Our results indicate that TA binds to LF and forms self-assemblies, which profoundly enhance interaction with LC cells. This study demonstrated that TA is a novel carrier for pharmaceutical drugs such as gemcitabine, carboplatin, and irinotecan.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - Meena Jaggi
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - Amali E Samarasinghe
- Department of Paediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| |
Collapse
|
43
|
|