1
|
Kumar A, Vaiphei KK, Gulbake A. A nanotechnology driven effectual localized lung cancer targeting approaches using tyrosine kinases inhibitors: Recent progress, preclinical assessment, challenges, and future perspectives. Int J Pharm 2024; 666:124745. [PMID: 39321904 DOI: 10.1016/j.ijpharm.2024.124745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
The higher incidence and mortality rate among all populations worldwide explains the unmet solutions in the treatment of lung cancer. The evolution of targeted therapies using tyrosine kinase inhibitors (TKI) has encouraged anticancer therapies. However, on-target and off-target effects and the development of drug resistance limited the anticancer potential of such targeted biologics. The advances in nanotechnology-driven-TKI embedded carriers that offered a new path toward lung cancer treatment. It is the inhalation route of administration known for its specific, precise, and efficient drug delivery to the lungs. The development of numerous TKI-nanocarriers through inhalation is proof of TKI growth. The future scopes involve using potential lung cancer biomarkers to achieve localized active cancer-targeting strategies. The adequate knowledge of in vitro absorption models usually helps establish better in vitro - in vivo correlation/extrapolation (IVIVC/E) to successfully evaluate inhalable drugs and drug products. The advanced in vitro and ex vivo lung tissue/ organ models offered better tumor heterogeneity, etiology, and microenvironment heterogeneity. The involvement of lung cancer organoids (LCOs), human organ chip models, and genetically modified mouse models (GEMMs) has resolved the challenges associated with conventional in vitro and in vivo models. To access potential inhalation-based drugtherapies, biological barriers, drug delivery, device-based challenges, and regulatory challenges must be encountered associated with their development. A proper understanding of material toxicity, size-based particle deposition at active disease sites, mucociliary clearance, phagocytosis, and the presence of enzymes and surfactants are required to achieve successful inhalational drug delivery (IDD). This article summarizes the future of lung cancer therapy using targeted drug-mediated inhalation using TKI.
Collapse
Affiliation(s)
- Ankaj Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Klaudi K Vaiphei
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India.
| |
Collapse
|
2
|
Al-Taie A, Özcan Bülbül E. A paradigm use of monoclonal antibodies-conjugated nanoparticles in breast cancer treatment: current status and potential approaches. J Drug Target 2024; 32:45-56. [PMID: 38096045 DOI: 10.1080/1061186x.2023.2295803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/12/2023] [Indexed: 12/30/2023]
Abstract
Monoclonal antibodies (mAbs) are integral to cancer treatment over conventional non-specific therapy methods. This study provides a scoping review of the clinically approved mAbs, focusing on the current application of different nanocarrier technologies as drug delivery targets for mAb-conjugated nanoparticles (NPs) as potential features for breast cancer (BC) treatment. An extensive literature search was conducted between the years 2000 and 2023 using various sources of databases. The first part covered mAb classification, types, and mechanisms of action, pharmacokinetics and clinical applications in BC. The second part covered polymeric, lipid and inorganic-based NPs, which are a variety of mAb-conjugated NPs targeting BC. A total of 20 relevant studies were enrolled indicating there are three different types of nanoparticular systems (polymeric NPs, inorganic NPs and lipid-based NPs) that can be used for BC treatment by being loaded with various active substances and conjugated with these antibodies. While mAbs have altered the way in cancer treatment due to targeting cancer cells specifically, the delivery of mAbs with nanoparticulate systems is important in the treatment of BC, as NPs are still being investigated as distinctive and promising drug delivery methods that can be employed for effective treatment of BC.
Collapse
Affiliation(s)
- Anmar Al-Taie
- Clinical Pharmacy Department, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| | - Ece Özcan Bülbül
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| |
Collapse
|
3
|
Zhao Z, Song H, Qi M, Liu Y, Zhang Y, Li S, Zhang H, Sun Y, Sun Y, Gao Z. Brain targeted polymeric micelles as drug carriers for ischaemic stroke treatment. J Drug Target 2024:1-17. [PMID: 39403962 DOI: 10.1080/1061186x.2024.2417190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Ischaemic stroke is a central nervous system disease with high morbidity, recurrence and mortality rates. Thrombolytic and neuroprotective therapies are the main therapeutic strategies for ischaemic stroke, however, the poor delivery efficiency of thrombolytic and neuroprotective drugs to the brain limits their clinical application. So far, the development of nanomedicine has brought opportunities for the above challenges, which can not only realise the effective accumulation of drugs in the target site, but also improve the pharmacokinetic behaviour of the drugs. Among the most rapidly developing nanoparticles, micelles gradually emerging as an effective strategy for ischaemic stroke treatment due to their own unique advantages. This review provided an overview of targeted and response-release micelles based on the physicochemical properties of the ischaemic stroke microenvironment, summarised the targeting strategies for delivering micellar formulations to the thrombus, blood-brain barrier, and brain parenchyma, and finally described the potentials and challenges of polymeric micelles in the treatment of ischaemic stroke.
Collapse
Affiliation(s)
- Zirui Zhao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huijia Song
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Mengge Qi
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yurong Liu
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanchao Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Shuo Li
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huimin Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yongjun Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanping Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Zibin Gao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| |
Collapse
|
4
|
Bu Y, Chen X, Wu T, Zhang R, Yan H, Lin Q. Synthesis, Optimization and Molecular Self-Assembly Behavior of Alginate-g-Oleylamine Derivatives Based on Ugi Reaction for Hydrophobic Drug Delivery. Int J Mol Sci 2024; 25:8551. [PMID: 39126119 PMCID: PMC11313573 DOI: 10.3390/ijms25158551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024] Open
Abstract
To achieve the optimal alginate-based oral formulation for delivery of hydrophobic drugs, on the basis of previous research, we further optimized the synthesis process parameters of alginate-g-oleylamine derivatives (Ugi-FOlT) and explored the effects of different degrees of substitution (DSs) on the molecular self-assembly properties of Ugi-FOlT, as well as the in vitro cytotoxicity and drug release behavior of Ugi-FOlT. The resultant Ugi-FOlT exhibited good amphiphilic properties with the critical micelle concentration (CMC) ranging from 0.043 mg/mL to 0.091 mg/mL, which decreased with the increase in the DS of Ugi-FOlT. Furthermore, Ugi-FOlT was able to self-assemble into spherical micellar aggregates in aqueous solution, whose sizes and zeta potentials with various DSs measured by dynamic light scattering (DLS) were in the range of 653 ± 25~710 ± 40 nm and -58.2 ± 1.92~-48.9 ± 2.86 mV, respectively. In addition, RAW 264.7 macrophages were used for MTT assay to evaluate the in vitro cytotoxicity of Ugi-FOlT in the range of 100~500 μg/mL, and the results indicated good cytocompatibility for Ugi-FOlT. Ugi-FOlT micellar aggregates with favorable stability also showed a certain sustained and pH-responsive release behavior for the hydrophobic drug ibuprofen (IBU). Meanwhile, it is feasible to control the drug release rate by regulating the DS of Ugi-FOlT. The influence of different DSs on the properties of Ugi-FOlT is helpful to fully understand the relationship between the micromolecular structure of Ugi-FOlT and its macroscopic properties.
Collapse
Affiliation(s)
- Yanan Bu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (Y.B.); (X.C.); (T.W.); (R.Z.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xiuqiong Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (Y.B.); (X.C.); (T.W.); (R.Z.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Ting Wu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (Y.B.); (X.C.); (T.W.); (R.Z.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Ruolin Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (Y.B.); (X.C.); (T.W.); (R.Z.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Huiqiong Yan
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (Y.B.); (X.C.); (T.W.); (R.Z.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Qiang Lin
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (Y.B.); (X.C.); (T.W.); (R.Z.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| |
Collapse
|
5
|
Tian M, Cheng J, Guo M. Stability, Digestion, and Cellular Transport of Soy Isoflavones Nanoparticles Stabilized by Polymerized Goat Milk Whey Protein. Antioxidants (Basel) 2024; 13:567. [PMID: 38790672 PMCID: PMC11117734 DOI: 10.3390/antiox13050567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Soy isoflavones (SIF) are bioactive compounds with low bioavailability due to their poor water solubility. In this study, we utilized polymerized goat milk whey protein (PGWP) as a carrier to encapsulate SIF with encapsulation efficiency of 89%, particle size of 135.53 nm, and zeta potential of -35.16 mV. The PGWP-SIF nanoparticles were evaluated for their stability and in vitro digestion properties, and their ability to transport SIF was assessed using a Caco-2 cell monolayer model. The nanoparticles were resistant to aggregation when subjected to pH changes (pH 2.0 to 8.0), sodium chloride addition (0-200 mM), temperature fluctuations (4 °C, 25 °C, and 37 °C), and long-term storage (4 °C, 25 °C, and 37 °C for 30 days), which was mainly attributed to the repulsion generated by steric hindrance effects. During gastric digestion, only 5.93% of encapsulated SIF was released, highlighting the nanoparticles' resistance to enzymatic digestion in the stomach. However, a significant increase in SIF release to 56.61% was observed during intestinal digestion, indicating the efficient transport of SIF into the small intestine for absorption. Cytotoxicity assessments via the MTT assay showed no adverse effects on Caco-2 cell lines after encapsulation. The PGWP-stabilized SIF nanoparticles improved the apparent permeability coefficient (Papp) of Caco-2 cells for SIF by 11.8-fold. The results indicated that using PGWP to encapsulate SIF was an effective approach for delivering SIF, while enhancing its bioavailability and transcellular transport.
Collapse
Affiliation(s)
- Mu Tian
- College of Food Science and Technology, Southwest Minzu University, Chengdu 610041, China;
- Key Laboratory of Dairy Science, Northeast Agricultural University, Harbin 150030, China;
| | - Jianjun Cheng
- Key Laboratory of Dairy Science, Northeast Agricultural University, Harbin 150030, China;
| | - Mingruo Guo
- Department of Nutrition and Food Sciences, College of Agriculture and Life Sciences, University of Vermont, Burlington, VT 05405, USA
| |
Collapse
|
6
|
Peng Y, Zhang P, Mei W, Zeng C. Exploring FGFR signaling inhibition as a promising approach in breast cancer treatment. Int J Biol Macromol 2024; 267:131524. [PMID: 38608977 DOI: 10.1016/j.ijbiomac.2024.131524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/18/2023] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
As our grasp of cancer genomics deepens, we are steadily progressing towards the domain of precision medicine, where targeted therapy stands out as a revolutionary breakthrough in the landscape of cancer therapeutics. The fibroblast growth factor receptors (FGFR) pathway has been unveiled as a fundamental instigator in the pathophysiological mechanisms underlying breast carcinoma, paving the way for the exhilarating development of precision-targeted therapeutics. In the pursuit of exploring inhibitors that specifically target the FGFR signaling pathways, a multitude of kinase inhibitors targeting FGFR has been assiduously engineered to address the heterogeneous landscape of human malignancies. This review offers an exhaustive exploration of aberrations within the FGFR pathway and their functional implications in breast cancer. Additionally, we delve into cutting-edge therapeutic approaches for the treatment of breast cancer patients bearing FGFR alterations and the management of toxicity associated with FGFR inhibitors. Furthermore, our contemplation of the evolution of cutting-edge FGFR inhibitors foresees their potential to spearhead innovative therapeutic approaches in the ongoing combat against cancer.
Collapse
Affiliation(s)
- Yan Peng
- Department of Obstetrics, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Pengfei Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Wuxuan Mei
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China; Department of General Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China.
| |
Collapse
|
7
|
Li C, Xu Y, Zhang Y, Shen Y, Deng X, Wang F. Novel bigels based on walnut oil oleogel and chitosan hydrogel: Preparation, characterization, and application as food spread. Int J Biol Macromol 2024; 260:129530. [PMID: 38296666 DOI: 10.1016/j.ijbiomac.2024.129530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 02/02/2024]
Abstract
This study developed new biphasic gel systems containing a walnut oil-based oleogel and a chitosan hydrogel and evaluated the application on food spread. The effects of different oleogelators [γ-oryzanol/β-sitosterol (γ-ORY/β-SIT), candelilla wax/span 65 (CW/SA), and mono- and diglycerides of fatty acids] were explored. Rheological analysis showed that γ-ORY/β-SIT-based bigel had the strongest gel strength, but XRD confirmed that β' crystal form (d = 3.72 Å, 4.12 Å) was predominantly in the CW/SA-based bigel, which was more appropriate for application as spread. The characteristics of CW/SA-based bigel with different oleogel fractions (40-80 wt%) were investigated. The microscopic images indicated that the hydrogels were dispersed as small droplets in the oleogels after oleogel fraction reaching 60 %. The highest crystallinity was achieved when the oleogel fraction was 60 %, and its oil binding capacity was 96.49 %. Textural analysis showed that the CW/SA-based bigel (OG-60 %) had similar properties with commercial spread B, and can be used as a partial replacement for spread B. Replacing 75 % of the commercial spread B with the bigel was found to be optimal and displayed acceptable sensory features. This study developed a healthy bigel based on walnut oil and provided the in-depth information for bigels as an alternative to plastic fats.
Collapse
Affiliation(s)
- Chang Li
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Forest Processing and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Yuanyuan Xu
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Forest Processing and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Yu Zhang
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Forest Processing and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Yijie Shen
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Forest Processing and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Xinyue Deng
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Forest Processing and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Fengjun Wang
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Forest Processing and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
8
|
Ilhan-Ayisigi E, Saglam-Metiner P, Sanci E, Bakan B, Yildirim Y, Buhur A, Yavasoglu A, Yavasoglu NUK, Yesil-Celiktas O. Receptor mediated targeting of EGF-conjugated alginate-PAMAM nanoparticles to lung adenocarcinoma: 2D/3D in vitro and in vivo evaluation. Int J Biol Macromol 2024; 261:129758. [PMID: 38286366 DOI: 10.1016/j.ijbiomac.2024.129758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/06/2024] [Accepted: 01/24/2024] [Indexed: 01/31/2024]
Abstract
Carboplatin (cis-diamine (1,1-cyclobutandicarboxylaso)‑platinum (II)) is a second-generation antineoplastic drug, which is widely used for chemotherapy of lung, colon, breast, cervix, testicular and digestive system cancers. Although preferred over cisplatin due to the lower incidence of nephrotoxicity and ototoxicity, efficient carboplatin delivery remains as a major challenge. In this study, carboplatin loaded alginate- poly(amidoamine) (PAMAM) hybrid nanoparticles (CAPs) with mean sizes of 192.13 ± 4.15 nm were synthesized using a microfluidic platform, then EGF was conjugated to the surface of CAPs (EGF-CAPs) for the receptor-targeted delivery. Hence, increased FITC+ cell counts were observed in A549 spheroids after EGF-CAP treatment compared to CAP in the 3D cellular uptake study. As such, the cytotoxicity of EGF-CAP was approximately 2-fold higher with an IC50 value of 35.89 ± 10.37 μg/mL compared to the CAPs in A549 spheroids. Based on in vivo experimental animal model, anti-tumor activities of the group treated with CAP decreased by 61 %, whereas the group treated with EGF-CAP completely recovered. Additionally, EGF-CAP application was shown to induce apoptotic cell death. Our study provided a new strategy for designing a hybrid nanoparticle for EGFR targeted carboplatin delivery with improved efficacy both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Esra Ilhan-Ayisigi
- Department of Genetic and Bioengineering, Faculty of Engineering and Architecture, Kirsehir Ahi Evran University, Kirsehir, Turkey; Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey
| | - Pelin Saglam-Metiner
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey; Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Turkey
| | - Ebru Sanci
- Center for Drug Research and Pharmacokinetic Applications (ARGEFAR), Ege University, Izmir, Turkey
| | - Buket Bakan
- Department of Molecular Biology and Genetics, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Yeliz Yildirim
- Department of Chemistry, Faculty of Science, Ege University, Izmir, Turkey
| | - Aylin Buhur
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Altug Yavasoglu
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - N Ulku Karabay Yavasoglu
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Turkey; Center for Drug Research and Pharmacokinetic Applications (ARGEFAR), Ege University, Izmir, Turkey; Department of Biology, Faculty of Science, Ege University, Izmir, Turkey.
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey; Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Turkey.
| |
Collapse
|
9
|
Kilicay E, Erdal E, Elci P, Hazer B, Denkbas EB. Tumour-specific hybrid nanoparticles in therapy of breast cancer. J Microencapsul 2024; 41:45-65. [PMID: 38095892 DOI: 10.1080/02652048.2023.2292226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 12/04/2023] [Indexed: 12/29/2023]
Abstract
In this study, salicylic acid (SA) dopped into poly(3-hydroxybutyrate) (PHB) and prepared nanoparticles (NPs) to increase encapsulation efficiency, anti-cancer activity of caffeic acid (Caff), and folic acid (FA) for breast cancer treatment. NPs were prepared by solvent evaporation method and characterised by FTIR, DSC, SEM, and entrapment-loading efficiencies. The mean diameter, polydispersity index (PDI), and zeta potential (ZP) were evaluated by dynamic light scattering (DLS). In vitro release and stability studies were done via eppendorf method. The cytotoxicity, cell dead and internalisation of NPs were shown by MTT, fluorescein and confocal microscopy. The diameter and ZP of NPs were 172 ± 7 nm and -29 ± 0.38 mV. The entrapment efficiencies of 5 and 10 Caff NPs were 79 ± 0.23% and 70 ± 0.42%. NPs showed good stability within 30 d and sustained release over 25 d. FA-5Caff NPs showed 37 ± 0.3% viability on MCF-7. FA-Caff NPs were identified as promising carrier system for breast cancer therapy.
Collapse
Affiliation(s)
- E Kilicay
- Vocational High School of Eldivan Health Care Services, Karatekin University, Cankiri, Turkey
| | - E Erdal
- Faculty of Medicine, Advanced Technologies Application and Research Center, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - P Elci
- Molecular Laboratory, University of Health Sciences Gülhane Health Institute, Ankara, Turkey
| | - B Hazer
- Department of Aircraft Airframe Engine Maintenance, Kapadokya University, Nevşehir, Turkey
- Department of Chemistry, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| | - E B Denkbas
- Department of Biomedical Engineering, Başkent University, Ankara, Turkey
| |
Collapse
|
10
|
Bandyopadhyay A, Das T, Nandy S, Sahib S, Preetam S, Gopalakrishnan AV, Dey A. Ligand-based active targeting strategies for cancer theranostics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3417-3441. [PMID: 37466702 DOI: 10.1007/s00210-023-02612-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/04/2023] [Indexed: 07/20/2023]
Abstract
In the past decades, for the intermediate or advanced cancerous stages, preclinical and clinical applications of nanomedicines in cancer theranostics have been extensively studied. Nevertheless, decreased specificity and poor targeting efficiency with low target concentration of theranostic are the major drawbacks of nanomedicine in employing clinical substitution over conventional systemic therapy. Consequently, ligand decorated nanocarrier-mediated targeted drug delivery system can transcend the obstructions through their enhanced retention activity and increased permeability with effective targeting. The highly efficient and specific nanocarrier-mediated ligand-based active therapy is one of the novel and promising approaches for delivery of the therapeutics for different cancers in recent years to restrict various cancer growth in vivo without harming healthy cells. The article encapsulates the features of nanocarrier-mediated ligands in augmentation of active targeting approaches of various cancers and summarizes ligand-based targeted delivery systems in treatment of cancer as plausible theranostics.
Collapse
Affiliation(s)
- Anupriya Bandyopadhyay
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India
| | - Tuyelee Das
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India
| | - Samapika Nandy
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India
- School of Pharmacy, Graphic Era Hill University, Bell Road, Clement Town, Dehradun, 248002, Uttarakhand, India
| | - Synudeen Sahib
- S.S. Cottage, Njarackal,, P.O.: Perinad, Kollam, 691601, Kerala, India
| | - Subham Preetam
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, 59053, Ulrika, Sweden
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India.
| |
Collapse
|
11
|
Deepak P, Kumar P, Pandey P, Arya DK, Jaiswal S, Kumar A, Sonkar AB, Ali D, Alarifi S, Ramar M, Rajinikanth PS. Pentapeptide cRGDfK-Surface Engineered Nanostructured Lipid Carriers as an Efficient Tool for Targeted Delivery of Tyrosine Kinase Inhibitor for Battling Hepatocellular Carcinoma. Int J Nanomedicine 2023; 18:7021-7046. [PMID: 38046236 PMCID: PMC10693281 DOI: 10.2147/ijn.s438307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/07/2023] [Indexed: 12/05/2023] Open
Abstract
Background Antitumor research aims to efficiently target hepatocarcinoma cells (HCC) for drug delivery. Nanostructured lipid carriers (NLCs) are promising for active tumour targeting. Cell-penetrating peptides are feasible ligands for targeted cancer treatment. Methods In this study, we optimized gefitinib-loaded NLCs (GF-NLC) for HCC treatment. The NLCs contained cholesterol, oleic acid, Pluronic F-68, and Phospholipon 90G. The NLC surface was functionalized to enhance targeting with the cRGDfK-pentapeptide, which binds to the αvβ3 integrin receptor overexpressed on hepatocarcinoma cells. Results GF-NLC formulation was thoroughly characterized for various parameters using differential scanning calorimetry and X-ray diffraction analysis. In-vitro and in-vivo studies on the HepG2 cell line showed cRGDfK@GF-NLC's superiority over GF-NLC and free gefitinib. cRGDfK@GF-NLC exhibited significantly higher cytotoxicity, growth inhibition, and cellular internalization. Biodistribution studies demonstrated enhanced tumour site accumulation without organ toxicity. The findings highlight cRGDfK@GF-NLC as a highly efficient carrier for targeted drug delivery, surpassing non-functionalized NLCs. These functionalized NLCs offer promising prospects for improving hepatocarcinoma therapy outcomes by specifically targeting HCC cells. Conclusion Based on these findings, cRGDfK@GF-NLC holds immense potential as a highly efficient carrier for targeted drug delivery of anticancer agents, surpassing the capabilities of non-functionalized NLCs. This research opens up new avenues for effective treatment strategies in hepatocarcinoma.
Collapse
Affiliation(s)
- Payal Deepak
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Praveen Kumar
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India
- S.D College of Pharmacy and Vocational Studies, Muzaffarnagar, Uttar Pradesh, India
| | - Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Dilip Kumar Arya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Shweta Jaiswal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Archana Bharti Sonkar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Daoud Ali
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Saud Alarifi
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohankumar Ramar
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Connecticut, Storrs, CT, 02903, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
12
|
Diniz F, Lamas S, Osório H, Aguiar P, Freitas D, Gärtner F, Sarmento B, Reis CA, Gomes J. Nanoparticles targeting Sialyl-Tn for efficient tyrosine kinase inhibitor delivery in gastric cancer. Acta Biomater 2023; 170:142-154. [PMID: 37586448 DOI: 10.1016/j.actbio.2023.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
Gastric cancer (GC) is the fourth leading cause of cancer-related deaths worldwide and, therefore, it is urgent to develop new and more efficient therapeutic approaches. Foretinib (FRT) is an oral multikinase inhibitor targeting MET (hepatocyte growth factor receptor) and RON (recepteur d'origine nantais) receptor tyrosine kinases (RTKs) that has been used in clinical trials for several solid tumors. Targeted uptake of therapeutic polymeric nanoparticles (NPs) represents a powerful approach in cancer cell drug delivery. Previously, a nanodelivery system composed of polymeric NPs functionalized with B72.3 antibody, which targets the tumor-associated antigen Sialyl-Tn (STn), has been developed. Herein, these NPs were loaded with FRT to evaluate its capacity in delivering the drug to multicellular tumors spheroids (MCTS) and mouse models. The data indicated that B72.3 functionalized FRT-loaded PLGA-PEG-COOH NPs (NFB72.3) specifically target gastric MCTS expressing the STn glycan (MKN45 SimpleCell (SC) cells), leading to a decrease in phospho-RTKs activation and reduced cell viability. In vivo evaluation using MKN45 SC xenograft mice revealed that NFB72.3 were able to decrease tumor growth, reduce cell proliferation and tumor necrosis. NFB72.3-treated tumors also showed inactivation of phospho-MET and phospho-RON. This study demonstrates the value of using NPs targeting STn for FRT delivery, highlighting its potential as a therapeutic application in GC. STATEMENT OF SIGNIFICANCE: Despite the advances in gastric cancer therapeutics, it remains one of the diseases with the highest incidence and mortality in the world. Combining targeted therapies with a controlled drug release is an attractive strategy to reduce drug cytotoxic effects and improve specific drug delivery efficiency to the cancer cells. Thus, we developed nanoparticles loaded with a tyrosine kinase inhibitor and targeting a specific tumor glycan exclusive of cancer cells. In in vivo gastric cancer xenograft mice models, these nanoparticles efficiently reduced tumor growth, cell proliferation and tumor necrosis area and inactivated phosphorylation of targeting receptors. This approach represents an innovative therapeutic strategy with high impact in gastric cancer.
Collapse
Affiliation(s)
- Francisca Diniz
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Sofia Lamas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Portugal
| | - Hugo Osório
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Paulo Aguiar
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Daniela Freitas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Fátima Gärtner
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; CESPU-IUCS, 4585-116 Gandra, Portugal
| | - Celso A Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal; FMUP - Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.
| | - Joana Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
13
|
Rezaei S, Gharapapagh E, Dabiri S, Heidari P, Aghanejad A. Theranostics in targeting fibroblast activation protein bearing cells: Progress and challenges. Life Sci 2023; 329:121970. [PMID: 37481033 PMCID: PMC10773987 DOI: 10.1016/j.lfs.2023.121970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/03/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Cancer cells are surrounded by a complex and highly dynamic tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs), a critical component of TME, contribute to cancer cell proliferation as well as metastatic spread. CAFs express a variety of biomarkers, which can be targeted for detection and therapy. Most importantly, CAFs express high levels of fibroblast activation protein (FAP) which contributes to progression of cancer, invasion, metastasis, migration, immunosuppression, and drug resistance. As a consequence, FAP is an attractive theranostic target. In this review, we discuss the latest advancement in targeting FAP in oncology using theranostic biomarkers and imaging modalities such as single-photon emission computed tomography (SPECT), positron emission tomography (PET), computed tomography (CT), fluorescence imaging, and magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Sahar Rezaei
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esmaeil Gharapapagh
- Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Dabiri
- Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pedram Heidari
- Departments of Radiology, Massachusetts General Hospital, Boston, United States
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
14
|
Wu X, Huang S, He W, Song M. Emerging insights into mechanisms of trastuzumab resistance in HER2-positive cancers. Int Immunopharmacol 2023; 122:110602. [PMID: 37437432 DOI: 10.1016/j.intimp.2023.110602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/19/2023] [Accepted: 07/02/2023] [Indexed: 07/14/2023]
Abstract
HER2 is an established therapeutic target in breast, gastric, and gastroesophageal junction carcinomas with HER2 overexpression or genomic alterations. The humanized monoclonal antibody trastuzumab targeting HER2 has substantially improved the clinical outcomes of HER2-positive patients, yet the inevitable intrinsic or acquired resistance to trastuzumab limits its clinical benefit, necessitating the elucidation of resistance mechanisms to develop alternate therapeutic strategies. This review presents an overview of trastuzumab resistance mechanisms involving signaling pathways, cellular metabolism, cell plasticity, and tumor microenvironment, particularly discussing the prospects of developing rational combinations to improve patient outcomes.
Collapse
Affiliation(s)
- Xiaoxue Wu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shuting Huang
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China.
| | - Mei Song
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
15
|
Fatima Qizilbash F, Sartaj A, Qamar Z, Kumar S, Imran M, Mohammed Y, Ali J, Baboota S, Ali A. Nanotechnology revolutionises breast cancer treatment: harnessing lipid-based nanocarriers to combat cancer cells. J Drug Target 2023; 31:794-816. [PMID: 37525966 DOI: 10.1080/1061186x.2023.2243403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023]
Abstract
One of the most common cancers that occur in females is breast cancer. Despite the significant leaps and bounds that have been made in treatment of breast cancer, the disease remains one of the leading causes of death among women and a major public health challenge. The therapeutic efficacy of chemotherapeutics is hindered by chemoresistance and toxicity. Nano-based lipid drug delivery systems offer controlled drug release, nanometric size and site-specific targeting. Breast cancer treatment includes surgery, chemotherapy and radiotherapy. Despite this, no single method of treatment for the condition is currently effective due to cancer stem cell metastasis and chemo-resistance. Therefore, the employment of nanocarrier systems is necessary in order to target breast cancer stem cells. This article addresses breast cancer treatment options, including modern treatment procedures such as chemotherapy, etc. and some innovative therapeutic options highlighting the role of lipidic nanocarriers loaded with chemotherapeutic drugs such as nanoemulsion, solid-lipid nanoparticles, nanostructured lipid carriers and liposomes, and their investigations have demonstrated that they can limit cancer cell growth, reduce the risk of recurrence, as well as minimise post-chemotherapy metastasis. This article also explores FDA-approved lipid-based nanocarriers, commercially available formulations, and ligand-based formulations that are being considered for further research.
Collapse
Affiliation(s)
| | - Ali Sartaj
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
- Lloyd School of Pharmacy, Greater Noida, India
| | - Zufika Qamar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology (MIET), Meerut, India
| | - Mohammad Imran
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Yousuf Mohammed
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Pharmacy, The University of Queensland, Brisbane, Australia
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Asgar Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| |
Collapse
|
16
|
Hu Y, Fan Y, Chen B, Li H, Zhang G, Su J. Stimulus-responsive peptide hydrogels: a safe and least invasive administration approach for tumor treatment. J Drug Target 2023:1-17. [PMID: 37469142 DOI: 10.1080/1061186x.2023.2236332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023]
Abstract
Tumours, with increasing mortality around the world, have bothered human beings for decades. Enhancing the targeting of antitumor drugs to tumour tissues is the key to enhancing their antitumor effects. The tumour microenvironment is characterised by a relatively low pH, overexpression of certain enzymes, redox imbalance, etc. Therefore, smart drug delivery systems that respond to the tumour microenvironment have been proposed to selectively release antitumor drugs. Among them, peptide hydrogels as a local drug delivery system have received much attention due to advantages such as high biocompatibility, degradability and high water-absorbing capacity. The combination of peptide segments with different physiological functions allows for tumour targeting, self-aggregation, responsiveness, etc. Morphological and microstructural changes in peptide hydrogels can occur when utilising the inherent pathological microenvironment of tumours to trigger drug release, which endows such systems with limited adverse effects and improved therapeutic efficiency. Herein, this review outlined the driving forces, impact factors, and sequence design in peptide hydrogels. We also discussed the triggers to induce the transformation of peptide-based hydrogels in the tumour microenvironment and described the advancements of peptide-based hydrogels for local drug delivery in tumour treatment. Finally, we gave a brief perspective on the prospects and challenges in this field.
Collapse
Affiliation(s)
- Yuchen Hu
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, China
| | - Ying Fan
- Chongqing University Jiangjin Hospital, Chongqing, P.R. China
| | - Ban Chen
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, China
| | - Hong Li
- School of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Gang Zhang
- Hubei Provincial Key Laboratory of Chemical Equipment Intensification and Intrinsic Safety, School of Mechanical and Electrical Engineering, Wuhan Institute of Technology, Wuhan, P.R. China
| | - Jiangtao Su
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, China
| |
Collapse
|
17
|
Abdouss H, Pourmadadi M, Zahedi P, Abdouss M, Yazdian F, Rahdar A, Díez-Pascual AM. Green synthesis of chitosan/polyacrylic acid/graphitic carbon nitride nanocarrier as a potential pH-sensitive system for curcumin delivery to MCF-7 breast cancer cells. Int J Biol Macromol 2023; 242:125134. [PMID: 37257532 DOI: 10.1016/j.ijbiomac.2023.125134] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
A novel pH-sensitive nanocarrier containing chitosan (CS), polyacrylic acid (PAA), and graphitic carbon nitride (g-C3N4) was designed via water/oil/water (W/O/W) emulsification to administer curcumin (CUR) drug. g-C3N4 nanosheets with a high surface area and porous structure were produced via simple one-step pyrolysis process using thiourea as precursor, and incorporated into CS/PAA hydrogel. X-ray diffraction (XRD) and Fourier-transform infrared spectroscopy (FT-IR) were used to assess the crystalline structure of the nanocarrier and the interactions between its components, respectively. Scanning electron microscopy (SEM) images revealed a spherical structure and confirmed the g-C3N4 impregnation into the CS/PAA matrix. Zeta potential and dynamic light scattering (DLS) provided information about the surface charge and average size distribution. High CUR loading and entrapment efficiencies were obtained, which were further improved upon addition of g-C3N4. The release kinetics of drug-loaded CS/PAA/g-C3N4 nanocomposites were investigated at pH = 5.4 and pH = 7.4, and the results showed an excellent controlled pH-sensitive release profile. Cell apoptosis and in vitro cytotoxicity were investigated using flow cytometry and MTT analyses. CS/PAA/g-C3N4/CUR resulted in the highest rate of apoptosis in MCF-7 breast cancer cells, demonstrating the excellent nanocomposite efficacy in eliminating cancerous cells. CS/PAA hydrogel coated with g-C3N4 shows great potential for pH-sensitive controlled drug release.
Collapse
Affiliation(s)
- Hamidreza Abdouss
- Department of Polymer, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrab Pourmadadi
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Payam Zahedi
- Department of Polymer, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Majid Abdouss
- Department of Chemistry, Amirkabir University of Technology, Tehran, Iran.
| | - Fatemeh Yazdian
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, Iran.
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran.
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
18
|
Liu A, Wu H, Naeem A, Du Q, Ni B, Liu H, Li Z, Ming L. Cellulose nanocrystalline from biomass wastes: An overview of extraction, functionalization and applications in drug delivery. Int J Biol Macromol 2023; 241:124557. [PMID: 37094644 DOI: 10.1016/j.ijbiomac.2023.124557] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/08/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023]
Abstract
Cellulose nanocrystals (CNC) have been extensively used in various fields due to their renewability, excellent biocompatibility, large specific surface area, and high tensile strength. Most biomass wastes contain significant amounts of cellulose, which forms the basis of CNC. Biomass wastes are generally made up of agricultural waste, and forest residues, etc. CNC can be produced from biomass wastes by removing the non-cellulosic components through acid hydrolysis, enzymatic hydrolysis, oxidation hydrolysis, and other mechanical methods. However, biomass wastes are generally disposed of or burned in a random manner, resulting in adverse environmental consequences. Hence, using biomass wastes to develop CNC-based carrier materials is an effective strategy to promote the high value-added application of biomass wastes. This review summarizes the advantages of CNC applications, the extraction process, and recent advances in CNC-based composites, such as aerogels, hydrogels, films, and metal complexes. Furthermore, the drug release characteristics of CNC-based material are discussed in detail. Additionally, we discuss some gaps in our understanding of the current state of knowledge and potential future directions of CNC-based materials.
Collapse
Affiliation(s)
- Ao Liu
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Research Center for Differentiation and Department of TCM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Hailian Wu
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Research Center for Differentiation and Department of TCM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Abid Naeem
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Research Center for Differentiation and Department of TCM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Qing Du
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Research Center for Differentiation and Department of TCM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Bin Ni
- First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou 341000, China
| | - Hongning Liu
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Research Center for Differentiation and Department of TCM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China
| | - Zhe Li
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Research Center for Differentiation and Department of TCM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China.
| | - Liangshan Ming
- Institute for Advanced Study, Key Laboratory of Modern Preparation of TCM, Ministry of Education, Research Center for Differentiation and Department of TCM Basic Theory, Jiangxi University of Chinese Medicine, Jiangxi, Nanchang 330004, China.
| |
Collapse
|
19
|
Li X, An S, Wang C, Jiang Q, Gao D, Wang L. Protein-polysaccharides based nanoparticles for loading with Malus baccata polyphenols and their digestibility in vitro. Int J Biol Macromol 2023; 228:783-793. [PMID: 36581037 DOI: 10.1016/j.ijbiomac.2022.12.236] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
The poor solubility, instability and low absorption rate obstruct the bioavailability of polyphenols isolated from Malus baccata (MBP) during gastrointestinal digestion. In order to solve the limitable problems, the food-grade nanoparticles were fabricated by mucin (MC) and Hohenbuehelia serotina polysaccharides (HSP) for delivery of MBP (MBP-NPs). The physicochemical properties and morphology of MBP-NPs prepared by different condition were respectively characterized. During gastrointestinal digestion in vitro, the release characteristic and variation in phenolic composition of MBP-NPs were evaluated. The results showed that MBP-NPs formed by hydrogen bonding and hydrophobic interaction possessed the regularly spherical shapes and smooth surfaces and semi-crystalline properties. Moreover, MBP-NPs presented the excellent physicochemical stability. During simulated gastrointestinal digestion in vitro, MBP-NPs exhibited the sustained release characteristics of phenolic compounds, which were confirmed by SDS-PAGE measurement. Compared with that of unencapsulated MBP, the significant variation was occurred in the phenolic composition of MBP-NPs, indicating that MBP-NPs could prevent the degradation and transformation of phenolic compounds. This study provides a novel strategy to improve the bioavailability of polyphenols.
Collapse
Affiliation(s)
- Xiaoyu Li
- Nano-biotechnology Key Lab of Hebei Province, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China; State Key Laboratory of Metastable Materials Science and Technology, Yanshan University, Qinhuangdao 066004, PR China
| | - Siying An
- Nano-biotechnology Key Lab of Hebei Province, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China
| | - Cheng Wang
- Nano-biotechnology Key Lab of Hebei Province, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China
| | - Qianyu Jiang
- Nano-biotechnology Key Lab of Hebei Province, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China
| | - Dawei Gao
- Nano-biotechnology Key Lab of Hebei Province, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China
| | - Lu Wang
- Nano-biotechnology Key Lab of Hebei Province, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China.
| |
Collapse
|
20
|
Torabi M, Aghanejad A, Savadi P, Barzegari A, Omidi Y, Barar J. Fabrication of mesoporous silica nanoparticles for targeted delivery of sunitinib to ovarian cancer cells. BIOIMPACTS : BI 2023; 13:255-267. [PMID: 37431477 PMCID: PMC10329750 DOI: 10.34172/bi.2023.25298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/06/2022] [Accepted: 07/23/2022] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Mesoporous silica nanoparticles (MSNPs) are considered innovative multifunctional structures for targeted drug delivery owing to their outstanding physicochemical characteristics. METHODS MSNPs were fabricated using the sol-gel method, and polyethylene glycol-600 (PEG600) was used for MSNPs modification. Subsequently, sunitinib (SUN) was loaded into the MSNPs, MSNP-PEG and MSNP-PEG/SUN were grafted with mucin 16 (MUC16) aptamers. The nanosystems (NSs) were characterized using FT-IR, TEM, SEM, DLS, XRD, BJH, and BET. Furthermore, the biological impacts of MSNPs were evaluated on the ovarian cancer cells by MTT assay and flow cytometry analysis. RESULTS The results revealed that the MSNPs have a spherical shape with an average dimension, pore size, and surface area of 56.10 nm, 2.488 nm, and 148.08 m2g-1, respectively. The cell viability results showed higher toxicity of targeted MSNPs in MUC16 overexpressing OVCAR-3 cells as compared to the SK-OV-3 cells; that was further confirmed by the cellular uptake results. The cell cycle analysis exhibited that the induction of sub-G1 phase arrest mostly occurred in MSNP-PEG/SUN-MUC16 treated OVCAR-3 cells and MSNP-PEG/SUN treated SK-OV-3 cells. DAPI staining showed apoptosis induction upon exposure to targeted MSNP in MUC16 positive OVCAR-3 cells. CONCLUSION According to our results, the engineered NSs could be considered an effective multifunctional targeted drug delivery platform for the mucin 16 overexpressing cells.
Collapse
Affiliation(s)
- Mitra Torabi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouria Savadi
- Di.S.T.A.Bi.F., University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
21
|
Indermun S, Kumar P, Govender M, Choonara YE. Can Nanomedicinal Approaches Provide an Edge to the Efficacy of Tyrosine Kinase Inhibitors? Curr Med Chem 2023; 30:1482-1501. [PMID: 35726410 DOI: 10.2174/0929867329666220618162303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/10/2022] [Accepted: 04/04/2022] [Indexed: 11/22/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are effective drug molecules for the treatment of various cancers. Nanomedicinal interventions and approaches may not only provide carrying capacities for TKIs but also potentially target tumor-specific environments and even cellular compartments. Nano-inspired drug delivery systems may hence enhance the efficacy of the drugs through enhanced tumour-availability resulting in greater efficacy and decreased side effects. A variety of nanosystems have been developed for the delivery of TKIs for the enhanced treatment of cancers, each with their own preparation methods and physicochemical properties. This review will therefore discuss the applicability of nano-interventions towards combination therapies, dose reduction, and greater potential treatment outcomes. The individual nanosystems have been highlighted with emphasis on the developed systems and their efficacy against various cancer cell lines and models.
Collapse
Affiliation(s)
- Sunaina Indermun
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Mershen Govender
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Yahya E Choonara
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| |
Collapse
|
22
|
Tyrosine Kinase Inhibitors for Glioblastoma Multiforme: Challenges and Opportunities for Drug Delivery. Pharmaceutics 2022; 15:pharmaceutics15010059. [PMID: 36678688 PMCID: PMC9863099 DOI: 10.3390/pharmaceutics15010059] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor with high mortality rates. Due to its invasiveness, heterogeneity, and incomplete resection, the treatment is very challenging. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have great potential for GBM treatment, however, their efficacy is primarily limited by poor brain distribution due to the presence of the blood-brain barrier (BBB). This review focuses on the potential of TKIs in GBM therapy and provides an insight into the reasons behind unsuccessful clinical trials of TKIs in GBM despite the success in treating other cancer types. The main section is dedicated to the use of promising drug delivery strategies for targeted delivery to brain tumors. Use of brain targeted delivery strategies can help enhance the efficacy of TKIs in GBM. Among various drug delivery approaches used to bypass or cross BBB, utilizing nanocarriers is a promising strategy to augment the pharmacokinetic properties of TKIs and overcome their limitations. This is because of their advantages such as the ability to cross BBB, chemical stabilization of drug in circulation, passive or active targeting of tumor, modulation of drug release from the carrier, and the possibility to be delivered via non-invasive intranasal route.
Collapse
|
23
|
Alsaab HO, Alharbi FD, Alhibs AS, Alanazi NB, Alshehri BY, Saleh MA, Alshehri FS, Algarni MA, Almugaiteeb T, Uddin MN, Alzhrani RM. PLGA-Based Nanomedicine: History of Advancement and Development in Clinical Applications of Multiple Diseases. Pharmaceutics 2022; 14:pharmaceutics14122728. [PMID: 36559223 PMCID: PMC9786338 DOI: 10.3390/pharmaceutics14122728] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Research on the use of biodegradable polymers for drug delivery has been ongoing since they were first used as bioresorbable surgical devices in the 1980s. For tissue engineering and drug delivery, biodegradable polymer poly-lactic-co-glycolic acid (PLGA) has shown enormous promise among all biomaterials. PLGA are a family of FDA-approved biodegradable polymers that are physically strong and highly biocompatible and have been extensively studied as delivery vehicles of drugs, proteins, and macromolecules such as DNA and RNA. PLGA has a wide range of erosion times and mechanical properties that can be modified. Many innovative platforms have been widely studied and created for the development of methods for the controlled delivery of PLGA. In this paper, the various manufacturing processes and characteristics that impact their breakdown and drug release are explored in depth. Besides different PLGA-based nanoparticles, preclinical and clinical applications for different diseases and the PLGA platform types and their scale-up issues will be discussed.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
- Correspondence: ; Tel.: +966-556047523
| | - Fatima D. Alharbi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alanoud S. Alhibs
- Department of Pharmacy, King Fahad Medical City, Riyadh 11564, Saudi Arabia
| | - Nouf B. Alanazi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Bayan Y. Alshehri
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marwa A. Saleh
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | - Fahad S. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Majed A. Algarni
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Turki Almugaiteeb
- Taqnia-Research Products Development Company, Riyadh 13244, Saudi Arabia
| | | | - Rami M. Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| |
Collapse
|
24
|
Jampilek J, Kralova K. Insights into Lipid-Based Delivery Nanosystems of Protein-Tyrosine Kinase Inhibitors for Cancer Therapy. Pharmaceutics 2022; 14:2706. [PMID: 36559200 PMCID: PMC9783038 DOI: 10.3390/pharmaceutics14122706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022] Open
Abstract
According to the WHO, cancer caused almost 10 million deaths worldwide in 2020, i.e., almost one in six deaths. Among the most common are breast, lung, colon and rectal and prostate cancers. Although the diagnosis is more perfect and spectrum of available drugs is large, there is a clear trend of an increase in cancer that ends fatally. A major advance in treatment was the introduction of gentler antineoplastics for targeted therapy-tyrosine kinase inhibitors (TKIs). Although they have undoubtedly revolutionized oncology and hematology, they have significant side effects and limited efficacy. In addition to the design of new TKIs with improved pharmacokinetic and safety profiles, and being more resistant to the development of drug resistance, high expectations are placed on the reformulation of TKIs into various drug delivery lipid-based nanosystems. This review provides an insight into the history of chemotherapy, a brief overview of the development of TKIs for the treatment of cancer and their mechanism of action and summarizes the results of the applications of self-nanoemulsifying drug delivery systems, nanoemulsions, liposomes, solid lipid nanoparticles, lipid-polymer hybrid nanoparticles and nanostructured lipid carriers used as drug delivery systems of TKIs obtained in vitro and in vivo.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| | - Katarina Kralova
- Institute of Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
25
|
Diniz F, Azevedo M, Sousa F, Osório H, Campos D, Sampaio P, Gomes J, Sarmento B, Reis CA. Polymeric nanoparticles targeting Sialyl-Tn in gastric cancer: A live tracking under flow conditions. Mater Today Bio 2022; 16:100417. [PMID: 36105678 PMCID: PMC9465339 DOI: 10.1016/j.mtbio.2022.100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022]
Abstract
Drug delivery using nanoparticles (NPs) represents a potential approach for therapy in cancer, such gastric cancer (GC) due to their targeting ability and controlled release properties. The use of advanced nanosystems that deliver anti-cancer drugs specifically to tumor cells may strongly rely on the expression of cancer-associated targets. Glycans aberrantly expressed by cancer cells are attractive targets for such delivery strategy. Sialylated glycans, such as Sialyl-Tn (STn) are aberrantly expressed in several epithelial tumors, including GC, being a potential target for a delivery nanosystem. The aim of this study was the development of NPs surface-functionalized with a specific antibody targeting the STn glycan and further evaluate this nanosystem effectiveness regarding its specificity and recognition capacity. Our results showed that the NPs surface-functionalized with anti-STn antibody efficiently are recognized by cells displaying the cancer-associated STn antigen under static and live cell monitoring flow conditions. This uncovers the potential use of such NPs for drug delivery in cancer. However, flow exposure was disclosed as an important biomechanical parameter to be taken into consideration. Here we presented an innovative and successful methodology to live track the NPs targeting STn antigen under shear stress, simulating the physiological flow. We demonstrate that unspecific binding of NPs agglomerates did not occur under flow conditions, in contrast with static assays. This robust approach can be applied for in vitro drug studies, giving valuable insights for in vivo studies.
Collapse
Affiliation(s)
- Francisca Diniz
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Maria Azevedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Flávia Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- CESPU-IUCS, 4585-116 Gandra, Portugal
| | - Hugo Osório
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Diana Campos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Paula Sampaio
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- CESPU-IUCS, 4585-116 Gandra, Portugal
| | - Celso A. Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- FMUP- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Corresponding author. i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
26
|
Lee HW, Seo HS, Yeom SY, Kim SN, Kim CR, Park DH, Park W, Choy YB, Park CG, Seo SI. Cabozantinib-Loaded PLGA Nanoparticles: A Potential Adjuvant Strategy for Surgically Resected High-Risk Non-Metastatic Renal Cell Carcinoma. Int J Mol Sci 2022; 23:12634. [PMID: 36293494 PMCID: PMC9604013 DOI: 10.3390/ijms232012634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Patients with high-risk non-metastatic renal cell carcinoma (RCC) are at risk of metastatic relapse following nephrectomy. Cabozantinib (CZ), a potent multitarget tyrosine kinase inhibitor, interferes with angiogenesis and immunosuppression associated with surgery-induced metastasis. Here, we explored the therapeutic potential of CZ-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (CZ-PLGA-NPs) as an adjuvant strategy for targeting post-nephrectomy metastasis. A clinically relevant subline recapitulating post-nephrectomy lung metastasis of high-risk human RCC, namely Renca-SRLu5-Luc, was established through in vivo serial selection of luciferase-expressing murine RCC Renca-Luc cells. CZ was encapsulated into PLGA-NPs via the conventional single emulsion technique. The multifaceted preclinical antimetastatic efficacy of CZ-PLGA-NPs was assessed in Renca-SRLu5-Luc cells. CZ-PLGA-NPs with a smooth surface displayed desirable physicochemical properties, good CZ encapsulation efficiency, as well as controlled and sustained CZ release. CZ-PLGA-NPs exhibited remarkable dose-dependent toxicity against Renca-SRLu5-Luc cells by inducing G2/M cell cycle arrest and apoptosis. CZ-PLGA-NPs attenuated in vitro colony formation, migration, and invasion by abrogating AKT and ERK1/2 activation. An intravenous injection of CZ-PLGA-NPs markedly reduced lung metastatic burden and prolonged lifespan with favorable safety in the Renca-SRLu5-Luc experimental lung metastasis model. The novel CZ-PLGA-NPs system with multifaceted antimetastatic effects and alleviating off-target toxicity potential is a promising adjunctive agent for patients with surgically resected high-risk RCC.
Collapse
Affiliation(s)
- Hye Won Lee
- Department of Urology, Center for Urologic Cancer, National Cancer Center, Goyang 10408, Korea
| | - Hee Seung Seo
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
| | - Seon-Yong Yeom
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Se-Na Kim
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Cho Rim Kim
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Korea
| | - Dae-Hwan Park
- Department of Engineering Chemistry, College of Engineering, Chungbuk National University, Cheongju 28644, Korea
- Department of Industrial Cosmetic Science and Department of Synchrotron Radiation Science and Technology, College of Bio-Health University System, Chungbuk National University, Cheongju 28644, Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Young Bin Choy
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul 03080, Korea
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Korea
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| | - Seong Il Seo
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| |
Collapse
|
27
|
Rajkumar M, Vimala K, Tamiliniyan DD, Thangaraj R, Jaganathan R, Kumaradhas P, Kannan S. Gelatin/polyvinyl alcohol loaded magnesium hydroxide nanocomposite attenuates neurotoxicity and oxidative stress in Alzheimer's disease induced rats. Int J Biol Macromol 2022; 222:2122-2143. [DOI: 10.1016/j.ijbiomac.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/05/2022]
|
28
|
Mohaghegh S, Tarighatnia A, Omidi Y, Barar J, Aghanejad A, Adibkia K. Multifunctional Magnetic Nanoparticles for MRI-guided Co-delivery of Erlotinib and L-Asparaginase to Ovarian Cancer. J Microencapsul 2022; 39:394-408. [PMID: 35748819 DOI: 10.1080/02652048.2022.2094487] [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: 01/19/2023]
Abstract
AIM(S) The use of magnetic nanoparticles (MNPs) in biomedical applications has been wildly opted due to their unique properties. The objective of this study was to evaluate the effects of aptamer-armed MNPs in ovarian cancer treatment and as T2 weighted MRI contrast agent. METHODS Here, we designed MNPs loaded with erlotinib (ERL/SPION-Val-PEG) and conjugated them with anti-mucin16 (MUC16) aptamer to introduce new image-guided nanoparticles (NPs) for targeted drug delivery as well as non-invasive magnetic resonance imaging (MRI) contrast agents. Also, the combination of our nanosystem (NS) along with L-Asparaginase (L-ASPN) led to synergistic effects in terms of reducing cell viability in ovarian cancer cells, which could suggest a novel combination therapy. RESULTS The mean size of our NS was about 63.4 ± 3.4 nm evaluated by DLS analysis and its morphology was confirmed using TEM. Moreover, the functional groups, as well as magnetic properties of our NS, were examined by FT-IR and VSM tests, respectively. The loading efficacy of erlotinib on MNPs was about 80% and its release reached 70.85% over 7 days in the pH value of 5.4. The MR images and flow cytometry results revealed that the cellular uptake of ERL/SPION-Val-PEG-MUC16 NPs in cells with MUC16 overexpression was considerably higher than unarmed NPs. In addition, T2-weight MR images of ovarian cancer-bearing mice indicated significant signal intensity changes at the tumor site 4 h after intravenous injection compared to the non-target MNPs. CONCLUSIONS Our data suggest ERL/SPION-Val-PEG NPs as an image-guided co-drug delivery system for ovarian cancer.
Collapse
Affiliation(s)
- Seraj Mohaghegh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Tarighatnia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Florida, USA
| | - Jaleh Barar
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Kadkhoda J, Tarighatnia A, Tohidkia MR, Nader ND, Aghanejad A. Photothermal therapy-mediated autophagy in breast cancer treatment: Progress and trends. Life Sci 2022; 298:120499. [DOI: 10.1016/j.lfs.2022.120499] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 12/17/2022]
|
30
|
Foroughi‐Nia B, Aghanejad A, Kadkhoda J, Barar J, Nosrati H, Davaran S. AS1411 conjugated magnetic‐based poly N‐isopropyl acrylamide nanoparticles for delivery of erlotinib to prostate cancer cells. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Behrouz Foroughi‐Nia
- Research Center for Pharmaceutical Nanotechnology Tabriz University of Medical Sciences Tabriz Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy Tabriz University of Medical Sciences Tabriz Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology Tabriz University of Medical Sciences Tabriz Iran
| | - Jamileh Kadkhoda
- Research Center for Pharmaceutical Nanotechnology Tabriz University of Medical Sciences Tabriz Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy Tabriz University of Medical Sciences Tabriz Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology Tabriz University of Medical Sciences Tabriz Iran
| | - Hamed Nosrati
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Soudabeh Davaran
- Research Center for Pharmaceutical Nanotechnology Tabriz University of Medical Sciences Tabriz Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy Tabriz University of Medical Sciences Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
31
|
One-Pot Synthesis of Amphiphilic Biopolymers from Oxidized Alginate and Self-Assembly as a Carrier for Sustained Release of Hydrophobic Drugs. Polymers (Basel) 2022; 14:polym14040694. [PMID: 35215606 PMCID: PMC8879484 DOI: 10.3390/polym14040694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 02/01/2023] Open
Abstract
In this paper, we developed an organic solvent-free, eco-friendly, simple and efficient one-pot approach for the preparation of amphiphilic conjugates (Ugi-OSAOcT) by grafting octylamine (OCA) to oxidized sodium alginate (OSA). The optimum reaction parameters that were obtained based on the degree of substitution (DS) of Ugi-OSAOcT were a reaction time of 12 h, a reaction temperature of 25 °C and a molar ratio of 1:2.4:3:3.3 (OSA:OCA:HAc:TOSMIC), respectively. The chemical structure and composition were characterized by Fourier transform infrared spectroscopy (FTIR), 1H nuclear magnetic resonance (1H NMR), X-ray diffraction (XRD), thermogravimetry analyser (TGA), gel permeation chromatography (GPC) and elemental analysis (EA). It was found that the Ugi-OSAOcT conjugates with a CMC value in the range of 0.30–0.085 mg/mL could self-assemble into stable and spherical micelles with a particle size of 135.7 ± 2.4–196.5 ± 3.8 nm and negative surface potentials of −32.8 ± 0.4–−38.2 ± 0.8 mV. Furthermore, ibuprofen (IBU), which served as a model poorly water-soluble drug, was successfully incorporated into the Ugi-OSAOcT micelles by dialysis method. The drug loading capacity (%DL) and encapsulation efficiency (%EE) of the IBU-loaded Ugi-OSAOcT micelles (IBU/Ugi-OSAOcT = 3:10) reached as much as 10.9 ± 0.4–14.6 ± 0.3% and 40.8 ± 1.6–57.2 ± 1.3%, respectively. The in vitro release study demonstrated that the IBU-loaded micelles had a sustained and pH-responsive drug release behavior. In addition, the DS of the hydrophobic segment on an OSA backbone was demonstrated to have an important effect on IBU loading and drug release behavior. Finally, the in vitro cytotoxicity assay demonstrated that the Ugi-OSAOcT conjugates exhibited no significant cytotoxicity against RAW 264.7 cells up to 1000 µg/mL. Therefore, the amphiphilic Ugi-OSAOcT conjugates synthesized by the green method exhibited great potential to load hydrophobic drugs, acting as a promising nanocarrier capable of responding to pH for sustained release of hydrophobic drugs.
Collapse
|
32
|
Recent advances and trends in nanoparticles based photothermal and photodynamic therapy. Photodiagnosis Photodyn Ther 2021; 37:102697. [PMID: 34936918 DOI: 10.1016/j.pdpdt.2021.102697] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022]
Abstract
Light-mediated therapies, including photodynamic therapy (PDT) and photothermal therapy (PTT) have been exploited as minimally invasive techniques for ablation of various tumors., Both modalities may eradicate tumors with minimal side effects to normal tissues and organs. Moreover, developments of light-mediated approaches using nanoparticles (NPs) and photosensitizer (PS) as diagnostic and therapeutic agents may have a crucial role in achieving successful cancer treatment. In recent years, novel nanoplatforms and strategies have been investigated to boost the therapeutic effect.. In this regard, gold, iron oxide, graphene oxide nanoparticles and hybrid nanocomposites have attracted attention.. Moreover, the combination of these materials with PS, in the form of hybrid NPs, reduces in vitro and in vivo normal tissue cytotoxicity, improves their solubility property in the biological environment and enhances the therapeutic effects. In this review, we look into the basic principles of PTT and PDT with their strengths and limitations to treat cancers. We also will discuss light-based nanoparticles and their PTT and PDT applications in the preclinical and clinical translation. Also, recent advances and trends in this field will be discussed along with the clinical challenges of PTT and PDT.
Collapse
|
33
|
A comprehensive review on immuno-nanomedicine for breast cancer therapy: Technical challenges and troubleshooting measures. Int Immunopharmacol 2021; 103:108433. [PMID: 34922248 DOI: 10.1016/j.intimp.2021.108433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022]
Abstract
Nanosized drug carriers have received a major attention in cancer therapeutics and theranostics. The immuno-nanomedicine is a combination of monoclonal antibody (mAb)/mAb-drug-nanoparticles. The immuno-nanomedicine offers a promising strategy to target cancer cells. However, the understating of nanotechnology, cancer biology, immunomedicine, and nanoparticle surface chemistry has provided a better clue to prepare the effective immuno-nanomedicine for cancer therapy. Moreover, the selection of nanoparticles type and its composition is essential for development of efficient drug delivery system (DDS) to target the cancer cell site. Immuno-nanomedicine works in the ligand-receptor binding mechanism through the interaction of mAb conjugated nanoparticles and specific antigen over expressed on target cancer cells. Therefore, the selection of specific receptors in the cancer cell and their ligand is important to prepare the active immuno-nanomedicines. Moreover, the factors such as drug loading, entrapment efficiency, size, shape, and ligand conjugation of a nanocarrier are considered as major factors for a better cancer cell, internalization, drug release, and cancer cell ablation. The target-based over-expression of antigen, mAb is engineered and conjugated with nanoparticles for successful targeting of the cancer cells without causing adverse effects to normal cells. Therefore, this review analyzed the fundamental factors in the immuno-nanomedicine for breast cancer and its technical challenges in the fabrication of the antibody alone/and drug conjugated nanoparticles.
Collapse
|
34
|
Mou L, Tian X, Zhou B, Zhan Y, Chen J, Lu Y, Deng J, Deng Y, Wu Z, Li Q, Song Y, Zhang H, Chen J, Tian K, Ni Y, Pu Z. Improving Outcomes of Tyrosine Kinase Inhibitors in Hepatocellular Carcinoma: New Data and Ongoing Trials. Front Oncol 2021; 11:752725. [PMID: 34707994 PMCID: PMC8543014 DOI: 10.3389/fonc.2021.752725] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022] Open
Abstract
Targeted therapies such as oral tyrosine kinase inhibitors (TKIs) are the main therapeutic strategy effective for advanced hepatocellular carcinoma (HCC). Currently six tyrosine kinase inhibitors for HCC therapy have been approved. The newly approved first-line drug donafenib represent the major milestones in HCC therapeutics in recent years. However, drug resistance in HCC remains challenging due to random mutations in target receptors as well as downstream pathways. TKIs-based combinatorial therapies with immune checkpoint inhibitors such as PD-1/PD-L1 antibodies afford a promising strategy to further clinical application. Recent developments of nanoparticle-based TKI delivery techniques improve drug absorption and bioavailability, enhance efficient targeting delivery, prolonged circulation time, and reduce harmful side effects on normal tissues, which may improve the therapeutic efficacy of the TKIs. In this review, we summarize the milestones and recent progress in clinical trials of TKIs for HCC therapy. We also provide an overview of the novel nanoparticle-based TKI delivery techniques that enable efficient therapy.
Collapse
Affiliation(s)
- Lisha Mou
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xiaohe Tian
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- Rausser College of Natural Resources, University of California, Berkeley, Berkeley, CA, United States
| | - Bo Zhou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- College of Engineering, Boston University, Boston, MA, United States
| | - Yongqiang Zhan
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jiao Chen
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Ying Lu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jing Deng
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Ying Deng
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Zijing Wu
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Qi Li
- Imaging Department, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Yi’an Song
- Imaging Department, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Hongyuan Zhang
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- The Faculty of Arts and Sciences, The University of British Columbia, Kelowna, BC, Canada
| | - Jinjun Chen
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Kuifeng Tian
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Yong Ni
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Zuhui Pu
- Imaging Department, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
35
|
Tarighatnia A, Abdkarimi MH, Nader ND, Mehdipour T, Fouladi MR, Aghanejad A, Ghadiri H. Mucin-16 targeted mesoporous nano-system for evaluation of cervical cancer via dual-modal computed tomography and ultrasonography. NEW J CHEM 2021. [DOI: 10.1039/d1nj04123a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mesoporous silica-coated bismuth nanoparticles (NPs) are dual-modal contrast agents that enable detection and quantification of cervical cancers at early stages using computed tomography (CT) and ultrasonography (US).
Collapse
Affiliation(s)
- Ali Tarighatnia
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Center for molecular and cellular imaging (RCMCI), Advanced Medical Technologies and Equipment Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nader D. Nader
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Tayebeh Mehdipour
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Fouladi
- Research Center for molecular and cellular imaging (RCMCI), Advanced Medical Technologies and Equipment Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Ghadiri
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for molecular and cellular imaging (RCMCI), Advanced Medical Technologies and Equipment Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|