1
|
Laura Soriano Pérez M, Montironi I, Alejandro Funes J, Margineda C, Campra N, Noelia Cariddi L, José Garrido J, Molina M, Alustiza F. Nanogel-Mediated antigen delivery: Biocompatibility, immunogenicity, and potential for tailored vaccine design across species. Vaccine 2024; 42:3721-3732. [PMID: 38719694 DOI: 10.1016/j.vaccine.2024.04.086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/27/2024] [Accepted: 04/28/2024] [Indexed: 06/07/2024]
Abstract
Nanotechnology has emerged as a promising avenue for enhancing the efficacy of vaccine delivery systems. This study investigates the utilization of nanogels as carriers for the model antigen ovalbumin, with a focus on in vivo assessments in equine and murine models. Nanogels, owing to their biocompatibility and tunable physicochemical properties, offer a versatile platform for efficient antigen encapsulation and controlled release. The encapsulation efficiency and physicochemical characteristics of ovalbumin-loaded nanogels were comprehensively characterized. In vitro biocompatibility was evaluated, finding excellent properties of these nanogels. In vivo evaluations were conducted on both equine and murine subjects, assessing immunogenicity through antibody and splenic cell response. Furthermore, the study propose the potential use of nanogels in tailoring immune responses through the modulation of antigen release kinetics. The results obtained in the in vitro assays showed an increase in the uptake of nanogels by APCs compared to free antigen (OVA). In mice, an absence of inflammatory response in the inoculation site was observed, without systemic damage in the evaluated organs. In addition, non-significant humoral response was found nor cellular proliferation and proinflammatory cytokine production, compared with a traditional adjuvant as aluminum hydroxide, in both animal models. These findings allow further insights into nanogel-based delivery systems and offer valuable insights into their application in various animal models. In conclusion, this research establishes the utility of nanogels as effective carriers for antigens-based vaccines, with interesting biocompatibility properties and highly taken affinity by antigen-presenting cells, without inducing inflammation at the injection site. The study underscores the potential of nanogel technology in revolutionizing vaccine design and highlights the importance of tailored approaches for diverse target species.
Collapse
Affiliation(s)
| | - Ivana Montironi
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | | | | | - Noelia Campra
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Laura Noelia Cariddi
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Juan José Garrido
- Laboratorio de Inmunogenómica y Patogénesis Molecular, Departamento de Genética, Facultad de Medicina Veterinaria, Universidad de Córdoba, España
| | - Maria Molina
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina.
| | | |
Collapse
|
2
|
Foster D, Cakley A, Larsen J. Optimizing enzyme-responsive polymersomes for protein-based therapies. Nanomedicine (Lond) 2024; 19:213-229. [PMID: 38271081 DOI: 10.2217/nnm-2023-0300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
Aims: Stimuli-responsive polymersomes are promising tools for protein-based therapies, but require deeper understanding and optimization of their pathology-responsive behavior. Materials & methods: Hyaluronic acid (HA)-poly(b-lactic acid) (PLA) polymersomes self-assembled from block copolymers of varying molecular weights of HA were compared for their physical properties, degradation and intracellular behavior. Results: Major results showed increasing enzyme-responsivity associated with decreasing molecular weight. The major formulation differences were as follows: the HA(5 kDa)-PLA formulation exhibited the most pronounced release of encapsulated proteins, while the HA(7 kDa)-PLA formulation showed the most different release behavior from neutral. Conclusion: We have discovered design rules for HA-PLA polymersomes for protein delivery, with lower molecular weight leading to higher encapsulation efficiency, greater release and greater intracellular uptake.
Collapse
Affiliation(s)
- Dorian Foster
- Department of Chemical & Biomolecular Engineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
| | - Alaura Cakley
- Department of Chemical & Biomolecular Engineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
| | - Jessica Larsen
- Department of Chemical & Biomolecular Engineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
- Department of Bioengineering, Center for Nanotherapeutic Strategies in the Central Nervous System, Clemson University, Clemson, SC 29631, USA
| |
Collapse
|
3
|
Kubeil M, Suzuki Y, Casulli MA, Kamal R, Hashimoto T, Bachmann M, Hayashita T, Stephan H. Exploring the Potential of Nanogels: From Drug Carriers to Radiopharmaceutical Agents. Adv Healthc Mater 2024; 13:e2301404. [PMID: 37717209 PMCID: PMC11468994 DOI: 10.1002/adhm.202301404] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/21/2023] [Indexed: 09/18/2023]
Abstract
Nanogels open up access to a wide range of applications and offer among others hopeful approaches for use in the field of biomedicine. This review provides a brief overview of current developments of nanogels in general, particularly in the fields of drug delivery, therapeutic applications, tissue engineering, and sensor systems. Specifically, cyclodextrin (CD)-based nanogels are important because they have exceptional complexation properties and are highly biocompatible. Nanogels as a whole and CD-based nanogels in particular can be customized in a wide range of sizes and equipped with a desired surface charge as well as containing additional molecules inside and outside, such as dyes, solubility-mediating groups or even biological vector molecules for pharmaceutical targeting. Currently, biological investigations are mainly carried out in vitro, but more and more in vivo applications are gaining importance. Modern molecular imaging methods are increasingly being used for the latter. Due to an extremely high sensitivity and the possibility of obtaining quantitative data on pharmacokinetic and pharmacodynamic properties, nuclear methods such as single photon emission computed tomography (SPECT) and positron emission tomography (PET) using radiolabeled compounds are particularly suitable here. The use of radiolabeled nanogels for imaging, but also for therapy, is being discussed.
Collapse
Affiliation(s)
- Manja Kubeil
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Radiopharmaceutical Cancer Research Bautzner Landstraße 40001328DresdenGermany
| | - Yota Suzuki
- Graduate School of Science and EngineeringSaitama University255 Shimo‐OkuboSakura‐KuSaitama338‐8570Japan
- Faculty of Science & TechnologySophia University7‐1 Kioi‐cho, Chiyoda‐kuTokyo102‐8554Japan
| | | | - Rozy Kamal
- Department of Nuclear MedicineManipal College of Health ProfessionsManipal Academy of Higher EducationManipalKarnataka576104India
| | - Takeshi Hashimoto
- Faculty of Science & TechnologySophia University7‐1 Kioi‐cho, Chiyoda‐kuTokyo102‐8554Japan
| | - Michael Bachmann
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Radiopharmaceutical Cancer Research Bautzner Landstraße 40001328DresdenGermany
| | - Takashi Hayashita
- Faculty of Science & TechnologySophia University7‐1 Kioi‐cho, Chiyoda‐kuTokyo102‐8554Japan
| | - Holger Stephan
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Radiopharmaceutical Cancer Research Bautzner Landstraße 40001328DresdenGermany
| |
Collapse
|
4
|
Myint SS, Laomeephol C, Thamnium S, Chamni S, Luckanagul JA. Hyaluronic Acid Nanogels: A Promising Platform for Therapeutic and Theranostic Applications. Pharmaceutics 2023; 15:2671. [PMID: 38140012 PMCID: PMC10747897 DOI: 10.3390/pharmaceutics15122671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/18/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Hyaluronic acid (HA) nanogels are a versatile class of nanomaterials with specific properties, such as biocompatibility, hygroscopicity, and biodegradability. HA nanogels exhibit excellent colloidal stability and high encapsulation capacity, making them promising tools for a wide range of biomedical applications. HA nanogels can be fabricated using various methods, including polyelectrolyte complexation, self-assembly, and chemical crosslinking. The fabrication parameters can be tailored to control the physicochemical properties of HA nanogels, such as size, shape, surface charge, and porosity, enabling the rational design of HA nanogels for specific applications. Stimulus-responsive nanogels are a type of HA nanogels that can respond to external stimuli, such as pH, temperature, enzyme, and redox potential. This property allows the controlled release of encapsulated therapeutic agents in response to specific physiological conditions. HA nanogels can be engineered to encapsulate a variety of therapeutic agents, such as conventional drugs, genes, and proteins. They can then be delivered to target tissues with high efficiency. HA nanogels are still under development, but they have the potential to become powerful tools for a wide range of theranostic or solely therapeutic applications, including anticancer therapy, gene therapy, drug delivery, and bioimaging.
Collapse
Affiliation(s)
- Su Sundee Myint
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.M.); (S.C.)
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Chavee Laomeephol
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Biomaterial Engineering in Medical and Health, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sirikool Thamnium
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.M.); (S.C.)
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok 10330, Thailand
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Biomaterial Engineering in Medical and Health, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
5
|
Umar AK, Limpikirati PK, Luckanagul JA. From Linear to Nets: Multiconfiguration Polymer Structure Generation with PolyFlin. J Chem Inf Model 2023; 63:6717-6726. [PMID: 37851376 DOI: 10.1021/acs.jcim.3c01221] [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: 10/19/2023]
Abstract
Molecular modeling and simulations are essential tools in polymer science and engineering, enabling researchers to predict and understand the properties of macromolecules, including their structure, dynamics, thermodynamics, and overall material characteristics. However, one of the key challenges in polymer simulation and modeling lies in the initial topology design, as existing programs often lack the capability to generate all types of polymer forms. In this study, we present PolyFlin, a powerful Python module that addresses this limitation by allowing the generation of a wide range of polymer structures, from simple homopolymers to complex copolymers, including grafts, cyclic, star, dendrimers, and nets. PolyFlin offers a versatile and efficient tool for exploring and creating diverse polymer architectures, facilitating advancements in various fields that require precise polymer modeling and simulation.
Collapse
Affiliation(s)
- Abd Kakhar Umar
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
- Medical Informatics Laboratory, ETFLIN, Palu 94225, Indonesia
| | - Patanachai K Limpikirati
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
6
|
Yuan M, Niu J, Li F, Ya H, Liu X, Li K, Fan Y, Zhang Q. Dipeptide-1 modified nanostructured lipid carrier-based hydrogel with enhanced skin retention and topical efficacy of curcumin. RSC Adv 2023; 13:29152-29162. [PMID: 37800130 PMCID: PMC10549242 DOI: 10.1039/d3ra04739c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
Topical administration of curcumin (CUR), a natural polyphenol with potent anti-inflammation and analgesic activities, provides a potential approach for local skin diseases. However, the drug delivery efficiency is highly limited by skin barriers and poor bioavailability of CUR. Herein, we propose hydrogel containing CUR-encapsulated dipeptide-1-modified nanostructured lipid carriers (CUR-DP-NLCs gel) to enhance topical drug delivery, and improve the topical therapeutic effect. The prepared CUR-DP-NLCs were characterized and were suitably dispersed into the Pluronic F127 hydrogel for topical application. The optimized CUR-DP-NLCs had a particle size of 152.6 ± 3.47 nm, a zeta potential of -33.1 ± 1.46 mV, an entrapment efficiency of 99.83 ± 0.14%, and a spherical morphology. X-ray diffraction (XRD) studies confirmed that CUR was successfully entrapped by the NLCs in an amorphous form. CUR-DP-NLCs gel exhibited sustained release over 48 h and significantly increased the skin retention of CUR. In vitro skin retention of CUR with CUR-DP-NLCs gel was 2.14 and 2.85 times higher than that of unmodified NLCs gel and free CUR, respectively. Fluorescence microscopy imaging revealed the formed nanoparticles accumulated in the hair follicles with prolonged retention time to form a drug reservoir. The hematoxylin-eosin staining showed that CUR-DP-NLCs gel could change the microstructure of skin layers and disturb the skin barriers. After topical administration to mice, CUR-DP-NLCs gel showed better analgesic and anti-inflammatory activities with no potentially hazardous skin irritation. These results concluded that CUR-DP-NLCs gel is a promising strategy to increase topical drug delivery of CUR in the treatment of local skin diseases.
Collapse
Affiliation(s)
- Ming Yuan
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| | - Jiangxiu Niu
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| | - Fei Li
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| | - Huiyuan Ya
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| | - Xianghui Liu
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| | - Keying Li
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| | - Yanli Fan
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| | - Qiuyan Zhang
- College of Food and Drug, Luoyang Normal University Luoyang Henan 471934 People's Republic of China
| |
Collapse
|
7
|
Shi W, Mirza S, Kuss M, Liu B, Hartin A, Wan S, Kong Y, Mohapatra B, Krishnan M, Band H, Band V, Duan B. Embedded Bioprinting of Breast Tumor Cells and Organoids Using Low-Concentration Collagen-Based Bioinks. Adv Healthc Mater 2023; 12:e2300905. [PMID: 37422447 PMCID: PMC10592394 DOI: 10.1002/adhm.202300905] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
Bioinks for 3D bioprinting of tumor models should not only meet printability requirements but also accurately maintain and support phenotypes of tumor surrounding cells to recapitulate key tumor hallmarks. Collagen is a major extracellular matrix protein for solid tumors, but low viscosity of collagen solution has made 3D bioprinted cancer models challenging. This work produces embedded, bioprinted breast cancer cells and tumor organoid models using low-concentration collagen I based bioinks. The biocompatible and physically crosslinked silk fibroin hydrogel is used to generate the support bath for the embedded 3D printing. The composition of the collagen I based bioink is optimized with a thermoresponsive hyaluronic acid-based polymer to maintain the phenotypes of both the noninvasive epithelial and invasive breast cancer cells, as well as cancer-associated fibroblasts. Mouse breast tumor organoids are bioprinted using optimized collagen bioink to mimic in vivo tumor morphology. A vascularized tumor model is also created using a similar strategy, with significantly enhanced vasculature formation under hypoxia. This study shows the great potential of embedded bioprinted breast tumor models utilizing a low-concentration collagen-based bioink for advancing the understanding of tumor cell biology and facilitating drug discovery research.
Collapse
Affiliation(s)
- Wen Shi
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Sameer Mirza
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of ChemistryCollege of ScienceUnited Arab Emirates UniversityAbu DhabiUnited Arab Emirates
| | - Mitchell Kuss
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bo Liu
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Andrew Hartin
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Shibiao Wan
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Yunfan Kong
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bhopal Mohapatra
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Mena Krishnan
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Hamid Band
- Eppley InstituteUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Vimla Band
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of SurgeryUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of Mechanical EngineeringUniversity of Nebraska–LincolnLincolnNE68588USA
| |
Collapse
|
8
|
Shelash Al-Hawary SI, Abdalkareem Jasim S, M Kadhim M, Jaafar Saadoon S, Ahmad I, Romero Parra RM, Hasan Hammoodi S, Abulkassim R, M Hameed N, K Alkhafaje W, Mustafa YF, Javed Ansari M. Curcumin in the treatment of liver cancer: From mechanisms of action to nanoformulations. Phytother Res 2023; 37:1624-1639. [PMID: 36883769 DOI: 10.1002/ptr.7757] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/10/2023] [Accepted: 01/22/2023] [Indexed: 03/09/2023]
Abstract
Liver cancer is the sixth most prevalent cancer and ranks third in cancer-related death, after lung and colorectal cancer. Various natural products have been discovered as alternatives to conventional cancer therapy strategies, including radiotherapy, chemotherapy, and surgery. Curcumin (CUR) with antiinflammatory, antioxidant, and antitumor activities has been associated with therapeutic benefits against various cancers. It can regulate multiple signaling pathways, such as PI3K/Akt, Wnt/β-catenin, JAK/STAT, p53, MAPKs, and NF-ĸB, which are involved in cancer cell proliferation, metastasis, apoptosis, angiogenesis, and autophagy. Due to its rapid metabolism, poor oral bioavailability, and low solubility in water, CUR application in clinical practices is restricted. To overcome these limitations, nanotechnology-based delivery systems have been applied to use CUR nanoformulations with added benefits, such as reducing toxicity, improving cellular uptake, and targeting tumor sites. Besides the anticancer activities of CUR in combating various cancers, especially liver cancer, here we focused on the CUR nanoformulations, such as micelles, liposomes, polymeric, metal, and solid lipid nanoparticles, and others, in the treatment of liver cancer.
Collapse
Affiliation(s)
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Al-Anbar-Ramadi, Iraq
| | - Mustafa M Kadhim
- Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, Iraq.,Medical Laboratory Techniques Department, Al-Turath University College, Baghdad, Iraq
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | | | | | - Noora M Hameed
- Anesthesia Techniques, Al-Nisour University College, Baghdad, Iraq
| | - Waleed K Alkhafaje
- Anesthesia Techniques Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
| |
Collapse
|
9
|
Jongjitphisut N, Thitikornpong W, Wichitnithad W, Thanusuwannasak T, Vajragupta O, Rojsitthisak P. A Stability-Indicating Assay for Tetrahydrocurcumin-Diglutaric Acid and Its Applications to Evaluate Bioaccessibility in an In Vitro Digestive Model. Molecules 2023; 28:molecules28041678. [PMID: 36838664 PMCID: PMC9966976 DOI: 10.3390/molecules28041678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
A simple and reliable ultra-high-performance liquid chromatographic (UHPLC) method was developed and validated for determination of tetrahydrocurcumin diglutaric acid (TDG) and applied for evaluation of its bioaccessibility. The analytical method was validated to demonstrate as a stability-indicating assay (SIA) according to the ICH Q2(R1) guidelines under various force degradation conditions including thermal degradation, moisture, acid and base hydrolysis, oxidation, and photolysis. The developed chromatographic condition could completely separate all degradants from the analyte of interest. The method linearity was verified in the range of 0.4-12 μg/mL with the coefficient of determination (r2) > 0.995. The accuracy and precision of the method provided %recovery in the range of 98.9-104.2% and %RSD lower than 4.97%, respectively. The limit of detection and quantitation were found to be 0.25 μg/mL and 0.40 μg/mL, respectively. This method has been successfully applied for the bioaccessibility assessment of TDG with the bioaccessibility of TDG approximately four fold greater than THC in simulated gastrointestinal fluid. The validated SIA method can also benefit the quality control of TDG raw materials in pharmaceutical and nutraceutical development.
Collapse
Affiliation(s)
- Nattapong Jongjitphisut
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Government Pharmaceutical Organization, Bangkok 10400, Thailand
| | - Worathat Thitikornpong
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: ; Tel.: +66-2-218-8315; Fax: +66-2-254-5195
| | - Wisut Wichitnithad
- Department of Analytical and Clinical Development, Pharma Nueva Co., Ltd., Bangkok 10900, Thailand
| | - Thanundorn Thanusuwannasak
- CU Drug and Health Products Innovation Promotion Center, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Opa Vajragupta
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand
- Molecular Probes for Imaging Research Network, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pornchai Rojsitthisak
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
10
|
Bhaladhare S, Bhattacharjee S. Chemical, physical, and biological stimuli-responsive nanogels for biomedical applications (mechanisms, concepts, and advancements): A review. Int J Biol Macromol 2023; 226:535-553. [PMID: 36521697 DOI: 10.1016/j.ijbiomac.2022.12.076] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The development of nanotechnology has influenced the advancements in biomedical and pharmaceutical fields. The design and formulation of stimuli-responsive nano-drug delivery systems, also called smart drug delivery systems, have attracted significant research worldwide and have been seen as a breakthrough in nanomedicines. The ability of these nanocarriers to respond to external and internal stimuli, such as pH, temperature, redox, electric and magnetic fields, enzymes, etc., has allowed them to deliver the cargo at targeted sites in a controlled fashion. The targeted drug delivery systems limit the harmful side effects on healthy tissue by toxic drugs and furnish spatial and temporal control drug delivery, improved patient compliance, and treatment efficiency. The polymeric nanogels (hydrogel nanoparticles) with stimuli-responsive characteristics have shown great potential in various biomedical, tissue engineering, and pharmaceutical fields. It is primarily because of their small size, biocompatibility, biodegradability, stimuli-triggered drug deliverability, high payload capacity, and tailored functionality. This comprehensive review deals distinctively with polymeric nanogels, their chemical, physical, and biological stimuli, the concepts of nanogels response to different stimuli, and recent advancements. This document will further improve the current understanding of stimuli-responsive materials and drug delivery systems and assist in exploring advanced potential applications of these intelligent materials.
Collapse
Affiliation(s)
- Sachin Bhaladhare
- Chemical and Polymer Engineering, Tripura University, Suryamaninagar, Tripura 799022, India.
| | - Sulagna Bhattacharjee
- Chemical and Polymer Engineering, Tripura University, Suryamaninagar, Tripura 799022, India
| |
Collapse
|
11
|
Chen R, Funnell JL, Quinones GB, Bentley M, Capadona JR, Gilbert RJ, Palermo EF. Poly(pro-curcumin) Materials Exhibit Dual Release Rates and Prolonged Antioxidant Activity as Thin Films and Self-Assembled Particles. Biomacromolecules 2023; 24:294-307. [PMID: 36512693 DOI: 10.1021/acs.biomac.2c01135] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Curcumin is a natural polyphenol that exhibits remarkable antioxidant and anti-inflammatory activities; however, its clinical application is limited in part by its physiological instability. Here, we report the synthesis of curcumin-derived polyesters that release curcumin upon hydrolytic degradation to improve curcumin stability and solubility in physiological conditions. Curcumin was incorporated in the polymer backbone by a one-pot condensation polymerization in the presence of sebacoyl chloride and polyethylene glycol (PEG, Mn = 1 kDa). The thermal and mechanical properties, surface wettability, self-assembly behavior, and drug-release kinetics all depend sensitively on the mole percentage of curcumin incorporated in these statistical copolymers. Curcumin release was triggered by the hydrolysis of phenolic esters on the polymer backbone, which was confirmed using a PEGylated curcumin model compound, which represented a putative repeating unit within the polymer. The release rate of curcumin was controlled by the hydrophilicity of the polymers. Burst release (2 days) and extended release (>8 weeks) can be achieved from the same polymer depending on curcumin content in the copolymer. The materials can quench free radicals for at least 8 weeks and protect primary neurons from oxidative stress in vitro. Further, these copolymer materials could be processed into both thin films and self-assembled particles, depending on the solvent-based casting conditions. Finally, we envision that these materials may have potential for neural tissue engineering application, where antioxidant release can mitigate oxidative stress and the inflammatory response following neural injury.
Collapse
Affiliation(s)
- Ruiwen Chen
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Jessica L Funnell
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Geraldine B Quinones
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Marvin Bentley
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States.,Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, Cleveland, Ohio 44106, United States
| | - Ryan J Gilbert
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Albany Stratton Veteran Affairs Medical Center, Albany, New York 12208, United States
| | - Edmund F Palermo
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| |
Collapse
|
12
|
Kaewruethai T, Lin Y, Wang Q, Luckanagul JA. The Dual Modification of PNIPAM and β-Cyclodextrin Grafted on Hyaluronic Acid as Self-Assembled Nanogel for Curcumin Delivery. Polymers (Basel) 2022; 15:polym15010116. [PMID: 36616466 PMCID: PMC9824384 DOI: 10.3390/polym15010116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Curcumin is an extract of turmeric (Curcuma longa) which possesses anti-inflammatory, anti-cancer and wound-healing effects and has been used as an active compound in biomedical research for many years. However, its poor solubility presents challenges for its use in drug delivery systems. A modified nanogel delivery system, with PNIPAM and β-cyclodextrin grafted onto hyaluronic acid (PNCDHA), was utilized to enhance the solubility. The polymer was characterized by NMR, and the inclusion complex between curcumin and β-cyclodextrin was confirmed by FTIR. The potential of this PNCDHA polymer complex as a drug delivery vehicle was supported by a curcumin encapsulation efficiency of 93.14 ± 5.6% and the release of encapsulated curcumin at 37 °C. At a concentration of 0.5% w/v in water, PNCDHA nanogels were biocompatible with fibroblast cell line (L929) up to a curcumin concentration of 50 µM. There was a direct concentration between curcumin loading and cellular internalization. A more detailed study of the cellular internalization of PNCDHA nanogel should be considered in order to clarify cellular delivery mechanisms and to assess how its viability as a carrier may be optimized.
Collapse
Affiliation(s)
- Tisana Kaewruethai
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Phayathai Road, Bangkok 10330, Thailand
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Phayathai Road, Bangkok 10330, Thailand
| | - Yuan Lin
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Changchun 130022, China
| | - Qian Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Changchun 130022, China
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Phayathai Road, Bangkok 10330, Thailand
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Phayathai Road, Bangkok 10330, Thailand
- Correspondence: ; Tel.: +66-2-218-8400; Fax: +66-2-218-8401
| |
Collapse
|
13
|
Development and biological evaluation of pNIPAM-based nanogels as vaccine carriers. Int J Pharm 2022; 630:122435. [PMID: 36442723 DOI: 10.1016/j.ijpharm.2022.122435] [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] [Received: 07/05/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
"Smart" nanogels are an attractive tool for the development of new strategies of immunization in veterinary medicine. Here, we reported the synthesis and physicochemical characterization of thermoresponsive nanogels based on poly(N-isopropylacrylamide) (pNIPAM) and theirin vitro, ex vivoand in vivo (mice model) performance. Smart nanogels of ca. 250 nm, with a transition temperature of 32 °C were obtained by precipitation polymerization. Assays to evaluatepNIPAM nanogels cytotoxicity were performed in different cell lines showing high biocompatibility (>70 %). The efficient internalization of the system was studied by confocal microscopy as well as flow cytometry. The ability to protect and deliver antigens was analyzed using the outer membrane lipoprotein A (OmlA), an important virulence factor ofActinobacillus pleuropneumoniae(App)cause of porcine pleuropneumonia. This lipoprotein was synthesized by recombinant technology and its technique was also described. The biodistribution ofpNIPAM nanogels administered intranasally was performedinvivo and ex vivo through Pearl Imaging System, which showed that nanogels were kept mostly in the lungs during the evaluated time. Besides, the efficacy of the proposal nanogel-based vaccine was studiedin vivoby measuring the antibody titers of BALB/c mice inoculated with OmlA encapsulated intopNIPAM nanogels compared to OmlA plus aluminum hydroxide adjuvant. The results proved the ability of nanogels to stimulate a humoral immune response. Therefore, we have demonstrated thatpNIPAM nanogels can be used as an efficient platform for vaccine nanocarriers.
Collapse
|
14
|
Electrospun Core–Sheath Nanofibers with Variable Shell Thickness for Modifying Curcumin Release to Achieve a Better Antibacterial Performance. Biomolecules 2022; 12:biom12081057. [PMID: 36008951 PMCID: PMC9406017 DOI: 10.3390/biom12081057] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
The inefficient use of water-insoluble drugs is a major challenge in drug delivery systems. Core–sheath fibers with various shell thicknesses based on cellulose acetate (CA) were prepared by the modified triaxial electrospinning for the controlled and sustained release of the water-insoluble Chinese herbal active ingredient curcumin. The superficial morphology and internal structure of core–sheath fibers were optimized by increasing the flow rate of the middle working fluid. Although the prepared fibers were hydrophobic initially, the core–sheath structure endowed fibers with better water retention property than monolithic fibers. Core–sheath fibers had flatter sustained-release profiles than monolithic fibers, especially for thick shell layers, which had almost zero-order release for almost 60 h. The shell thickness and sustained release of drugs brought about a good antibacterial effect to materials. The control of flow rate during fiber preparation is directly related to the shell thickness of core–sheath fibers, and the shell thickness directly affects the controlled release of drugs. The fiber preparation strategy for the precise control of core–sheath structure in this work has remarkable potential for modifying water-insoluble drug release and improving its antibacterial performance.
Collapse
|
15
|
Electrospun Coaxial Fibers to Optimize the Release of Poorly Water-Soluble Drug. Polymers (Basel) 2022; 14:polym14030469. [PMID: 35160459 PMCID: PMC8839822 DOI: 10.3390/polym14030469] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
In a drug delivery system, the physicochemical properties of the polymeric matrix have a positive impact on the bioavailability of poorly water-soluble drugs. In this work, monolithic F1 fibers and coaxial F2 fibers were successfully prepared using polyvinylpyrrolidone as the main polymer matrix for drug loading and the poorly water-soluble curcumin (Cur) as a model drug. The hydrophobic poly (3-hydroxybutyric acid-co-3-hydroxyvaleric acid) (PHBV) was designed as a blank layer to change the hydrophilicity of the fiber and restrain the drug dissolution rate. The curved linear morphology without beads of F1 fibers and the straight linear morphology with few spindles of F2 fibers were characterized using field-emission environmental scanning electron microscopy. The amorphous forms of the drug and its good compatibility with polymeric matrix were verified by X-ray diffraction and attenuated total reflectance Fourier transformed infrared spectroscopy. Surface wettability and drug dissolution data showed that the weaker hydrophilicity F2 fibers (31.42° ± 3.07°) had 24 h for Cur dissolution, which was much longer than the better hydrophilic F1 fibers (15.31° ± 2.79°) that dissolved the drug in 4 h.
Collapse
|
16
|
Yang J, Jin RM, Wang SY, Xie XT, Hu W, Tang HF, Liu B. Co-delivery of paclitaxel and doxorubicin using polypeptide-engineered nanogels for combination therapy of tumor. NANOTECHNOLOGY 2022; 33:155101. [PMID: 34963110 DOI: 10.1088/1361-6528/ac46b4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/27/2021] [Indexed: 05/24/2023]
Abstract
Loading of chemotherapeutic agents into nanoparticles has been demonstrated to be an effective strategy for cancer therapy. However, simultaneous delivery of different functional drugs to tumor sites for chemotherapy still remains challenging. In this study, nanogels formed by an engineered coiled-coil polypeptide PC10A were designed and prepared as a carrier for co-delivery of paclitaxel (PTX) and doxorubicin (DOX) through ultrasonic treatment and electrostatic adsorption. The drug loading content and encapsulation efficiency of PTX and DOX in the PC10A/PTX/DOX nanogels were 5.98 wt%, 70 wt%, and 8.55 wt%, 83 wt%, respectively. Because the polypeptide PC10A was non-toxic and biodegradable, the PC10A/PTX/DOX nanogels exhibited good biocompatibility. Thein vitroandin vivoantitumor experiments showed that the PC10A/PTX/DOX nanogels possessed obviously synergistic therapy effect of tumors and lower side effects compared with free PTX/DOX. Therefore, the PC10A/PTX/DOX nanogels are promising to provide a new strategy for combination therapy of different functional drugs.
Collapse
Affiliation(s)
- Jie Yang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, People's Republic of China
| | - Rui-Mei Jin
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, People's Republic of China
| | - Shen-Yan Wang
- Innovation Institute for Biomedical Materials, College of Nursing and Health Management & College of Life Science and Chemistry, Wuhan Donghu University, Wuhan 430212, People's Republic of China
| | - Xiao-Ting Xie
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, People's Republic of China
| | - Wei Hu
- Innovation Institute for Biomedical Materials, College of Nursing and Health Management & College of Life Science and Chemistry, Wuhan Donghu University, Wuhan 430212, People's Republic of China
| | - Hong-Feng Tang
- Innovation Institute for Biomedical Materials, College of Nursing and Health Management & College of Life Science and Chemistry, Wuhan Donghu University, Wuhan 430212, People's Republic of China
| | - Bo Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, People's Republic of China
| |
Collapse
|
17
|
In Vivo Biocompatible Self-Assembled Nanogel Based on Hyaluronic Acid for Aqueous Solubility and Stability Enhancement of Asiatic Acid. Polymers (Basel) 2021; 13:polym13234071. [PMID: 34883575 PMCID: PMC8659171 DOI: 10.3390/polym13234071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 12/17/2022] Open
Abstract
Asiatic acid (AA), a natural triterpene found in Centalla asiatica, possesses polypharmacological properties that can contribute to the treatment and prophylaxis of various diseases. However, its hydrophobic nature and rapid metabolic rate lead to poor bioavailability. The aim of this research was to develop a thermoresponsive nanogel from hyaluronic acid (HA) for solubility and stability enhancement of AA. Poly(N-isopropylacrylamide) (pNIPAM) was conjugated onto HA using a carbodiimide reaction followed by 1H NMR characterization. pNIPAM-grafted HA (HA-pNIPAM) nanogels were prepared with three concentrations of polymer, 0.1, 0.15 and 0.25% w/v, in water by the sonication method. AA was loaded into the nanogel by the incubation method. Size, morphology, AA loading capacity and encapsulation efficiency (EE) were analyzed. In vitro cytocompatibility was evaluated in fibroblast L-929 cells using the PrestoBlue assay. Single-dose toxicity was studied using rats. HA-pNIPAM nanogels at a 4.88% grafting degree showed reversible thermo-responsive behavior. All nanogel formulations could significantly increase AA water solubility and the stability was higher in nanogels prepared with high polymer concentrations over 180 days. The cell culture study showed that 12.5 µM AA in nanogel formulations was considered non-toxic to the L-929 cells; however, a dose-dependent cytotoxic effect was observed at higher AA-loaded concentrations. In vivo study proved the non-toxic effect of AA loaded in HA-pNIPAM nanogels compared with the control. Taken together, HA-pNIPAM nanogel is a promising biocompatible delivery system both in vitro and in vivo for hydrophobic AA molecules.
Collapse
|
18
|
Ayar Z, Shafieian M, Mahmoodi N, Sabzevari O, Hassannejad Z. A rechargeable drug delivery system based on
pNIPAM
hydrogel for the local release of curcumin. J Appl Polym Sci 2021. [DOI: 10.1002/app.51167] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Zahra Ayar
- Department of Biomedical Engineering Amirkabir University of Technology (Tehran Polytechnic) Tehran Iran
| | - Mehdi Shafieian
- Department of Biomedical Engineering Amirkabir University of Technology (Tehran Polytechnic) Tehran Iran
| | - Narges Mahmoodi
- Sina Trauma and Surgery Research Center Tehran University of Medical Sciences Tehran Iran
| | - Omid Sabzevari
- Toxicology and Poisoning Research Centre Tehran University of Medical Sciences Tehran Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy Tehran University of Medical Sciences Tehran Iran
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
19
|
A novel 'smart' PNIPAM-based copolymer for breast cancer targeted therapy: Synthesis, and characterization of dual pH/temperature-responsive lactoferrin-targeted PNIPAM-co-AA. Colloids Surf B Biointerfaces 2021; 202:111694. [PMID: 33740633 DOI: 10.1016/j.colsurfb.2021.111694] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/02/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022]
Abstract
Despite the active research towards introducing novel anticancer agents, the long-term sequelae and side effects of chemotherapy remain the major obstacle to achieving clinical success. Recent cancer research is now utilizing the medicinal chemistry toolbox to tailor novel 'smart' carrier systems that can reduce the major limitations of chemotherapy ranging from non-specificity and ubiquitous biodistribution to systemic toxicity. In this aspect, various stimuli-responsive polymers have gained considerable interest due to their intrinsic tumor targeting properties. Among these polymers, poly(N-isopropylacrylamide (PNIPAM) has been chemically modified to tune its thermoresponsivity or even copolymerized to endow new stimulus responsiveness for enhancing tumor targeting. Herein, we set our design rationale to impart additional active targeting entity to pH/temperature-responsive PNIPAM-based polymer for more efficient controlled payloads accumulation at the tumor through cellular internalization via synthesizing novel "super intelligent" lactoferrin conjugated PNIPAM-acrylic acid (LF-PNIPAM-co-AA) copolymer. The synthesized copolymer was physicochemically characterized and evaluated as a smart nanocarrier for targeting breast cancer. In this regard, Honokiol (HK) was utilized as a model anticancer drug and encapsulated in the nanoparticles to overcome its lipophilic nature and allow its parenteral administration, for achieving sustainable drug release with targeting action. Results showed that the developed HK-loaded LF-PNIPAM-co-AA nanohydrogels displayed high drug loading capacity reaching to 18.65 wt.% with excellent physical and serum stability. Moreover, the prepared HK-loaded nanohydrogels exhibited efficient in vitro and in vivo antitumor activities. In vivo, HK-loaded nanohydrogels demonstrated suppression of VEGF-1 and Ki-67 expression levels, besides inducing apoptosis through upregulating the expression level of active caspase-3 in breast cancer-bearing mice. Overall, the developed nanohydrogels (NGs) with pH and temperature responsivity provide a promising nanocarrier for anticancer treatment.
Collapse
|