1
|
Khateeb S, Hassan AI. Insights into nanostructured lipid carriers of etoricoxib for mitigating radiation-induced lung inflammation and exploring anti-inflammatory mechanisms in rats. Fundam Clin Pharmacol 2025; 39:e70014. [PMID: 40325971 DOI: 10.1111/fcp.70014] [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: 09/11/2024] [Revised: 04/03/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Radiation exposure can cause inflammation, which etoricoxib (ET), an anti-inflammatory drug, could potentially mitigate. OBJECTIVE This study aimed to evaluate the potential effectiveness of etoricoxib-loaded nanostructured lipid carriers (ET-NLCs) in mitigating radiation-induced acute lung inflammation in rats. METHODS Thirty-six rats were divided into six groups. Group 1 (C): control; group 2 (ET): normal rats given ET (10 mg/kg) orally for 14 days; group 3 (ET-NLC): normal rats administered ET-NLCs orally (10 mg/kg) for 14 days. Group 4 (R): rats exposed to 6 Gy whole-body gamma radiation, untreated thereafter to induce lung inflammation and injury. Group 5 (ET-R), irradiated rats, were administered ET (10 mg/kg) orally daily for 14 days. Group 6 (ET-NLC-R), irradiated rats, were administered ET-NLCs (10 mg/kg) orally daily for 14 days. Molecular, biochemical, and histopathological analyses were performed to assess inflammation, apoptosis, oxidative stress, and lung tissue architecture. RESULTS Radiation exposure led to a 1053% increase in Bax expression and an 81.5% decrease in Bcl-2, indicating heightened apoptosis. ET-NLCs treatment reversed these effects, reducing Bax by 59.9% and increasing Bcl-2 by 337.4%. Additionally, ET-NLCs reduced caspase-3 and caspase-8 activation by 54.5% and 62.9%, respectively, compared to radiation exposure alone. Furthermore, ET-NLCs demonstrated potent anti-inflammatory effects by reducing interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) levels by 49% and 39%, respectively, compared to the irradiated group. Radiation increased malondialdehyde (MDA) levels by 388%, indicating oxidative damage, and suppressed antioxidant enzymes such as catalase (CAT), glutathione peroxidase (GPX), and superoxide dismutase (SOD). ET-NLC treatment decreased MDA levels and increased CAT, GPX, and SOD by 35.7%, 4766.7%, and 765.9%, respectively, restoring antioxidant balance. Radiation reduced surfactant protein (SP-D) levels to 4.9% of control values, but ET-NLCs treatment restored them to 14%. Histopathological analysis revealed that radiation-exposed lungs showed thickened inter-alveolar septa, emphysematous areas, and inflammatory infiltration. ET-NLCs treatment exhibited only mild thickening and limited inflammatory cell infiltration, suggesting significant improvement in lung architecture. CONCLUSIONS Based on these results, NLCs are one of the most promising ways to deliver anti-inflammatory drugs for treating lung damage caused by radiation.
Collapse
Affiliation(s)
- Sahar Khateeb
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
- Biochemistry Division, Department of Chemistry, Faculty of Science, Fayoum University, Fayoum, Egypt
| | - Amal I Hassan
- Radioisotopes Department, Nuclear Research Centre, Egyptian Atomic Energy Authority, Giza, Egypt
| |
Collapse
|
2
|
Hu J, Arvejeh PM, Bone S, Hett E, Marincola FM, Roh KH. Nanocarriers for cutting-edge cancer immunotherapies. J Transl Med 2025; 23:447. [PMID: 40234928 PMCID: PMC12001629 DOI: 10.1186/s12967-025-06435-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Cancer immunotherapy aims to harness the body's own immune system for effective and long-lasting elimination of malignant neoplastic tissues. Owing to the advance in understanding of cancer pathology and immunology, many novel strategies for enhancing immunological responses against various cancers have been successfully developed, and some have translated into excellent clinical outcomes. As one promising strategy for the next generation of immunotherapies, activating the multi-cellular network (MCN) within the tumor microenvironment (TME) to deploy multiple mechanisms of action (MOAs) has attracted significant attention. To achieve this effectively and safely, delivering multiple or pleiotropic therapeutic cargoes to the targeted sites of cancerous tissues, cells, and intracellular organelles is critical, for which numerous nanocarriers have been developed and leveraged. In this review, we first introduce therapeutic payloads categorized according to their predicted functions in cancer immunotherapy and their physicochemical structures and forms. Then, various nanocarriers, along with their unique characteristics, properties, advantages, and limitations, are introduced with notable recent applications in cancer immunotherapy. Following discussions on targeting strategies, a summary of each nanocarrier matching with suitable therapeutic cargoes is provided with comprehensive background information for designing cancer immunotherapy regimens.
Collapse
Affiliation(s)
- Joyce Hu
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | - Pooria M Arvejeh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sydney Bone
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Erik Hett
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | | | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
- Biotechnology Science and Engineering Program, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
| |
Collapse
|
3
|
Morsy HM, Zaky MY, Yassin NYS, Khalifa AYZ. Nanoparticle-based flavonoid therapeutics: Pioneering biomedical applications in antioxidants, cancer treatment, cardiovascular health, neuroprotection, and cosmeceuticals. Int J Pharm 2025; 670:125135. [PMID: 39732216 DOI: 10.1016/j.ijpharm.2024.125135] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Flavonoids, a type of natural polyphenolic molecule, have garnered significant research interest due to their ubiquitous nature and diverse biological activities, including antioxidant, anti-inflammatory, and anticancer effects, making them appealing to various scientific disciplines. In this regard, the use of a flavonoid nanoparticle delivery system is to overcome low bioavailability, bioactivity, poor aqueous solubility, systemic absorption, and intensive metabolism. Therefore, this review summarizes the classification of nanoparticles (liposomes, polymeric, and solid lipid nanoparticles) and the advantages of using nanoparticle-flavonoid formulations to boost flavonoid bioavailability. Moreover, this review illustrated the pioneering biomedical applications of nanoparticle-based flavonoid therapeutics, as well as safety and toxicity considerations of using a flavonoid nanoparticle delivery system.
Collapse
Affiliation(s)
- Hadeer M Morsy
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Mohamed Y Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt.
| | - Nour Y S Yassin
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Ashraf Y Z Khalifa
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
4
|
Dejeu IL, Vicaș LG, Marian E, Ganea M, Frenț OD, Maghiar PB, Bodea FI, Dejeu GE. Innovative Approaches to Enhancing the Biomedical Properties of Liposomes. Pharmaceutics 2024; 16:1525. [PMID: 39771504 PMCID: PMC11728823 DOI: 10.3390/pharmaceutics16121525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Liposomes represent a promising class of drug delivery systems that enhance the therapeutic efficacy and safety of various pharmaceutical agents. Also, they offer numerous advantages compared to traditional drug delivery methods, including targeted delivery to specific sites, controlled release, and fewer side effects. This review meticulously examines the methodologies employed in the preparation and characterization of liposomal formulations. With the rising incidence of adverse drug reactions, there is a pressing need for innovative delivery strategies that prioritize selectivity, specificity, and safety. Nanomedicine promises to revolutionize diagnostics and treatments, addressing current limitations and improving disease management, including cancer, which remains a major global health challenge. This paper aims to conduct a comprehensive study on the interest of biomedical research regarding nanotechnology and its implications for further applications.
Collapse
Affiliation(s)
- Ioana Lavinia Dejeu
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Mariana Ganea
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Olimpia Daniela Frenț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Paula Bianca Maghiar
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - Flaviu Ionut Bodea
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - George Emanuiel Dejeu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 Piata 1 Decembrie Street, 410073 Oradea, Romania;
| |
Collapse
|
5
|
van Staden D, Gerber M, Lemmer HJR. The Application of Nano Drug Delivery Systems in Female Upper Genital Tract Disorders. Pharmaceutics 2024; 16:1475. [PMID: 39598598 PMCID: PMC11597179 DOI: 10.3390/pharmaceutics16111475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
The prevalence of female reproductive system disorders is increasing, especially among women of reproductive age, significantly impacting their quality of life and overall health. Managing these diseases effectively is challenging due to the complex nature of the female reproductive system, characterized by dynamic physiological environments and intricate anatomical structures. Innovative drug delivery approaches are necessary to facilitate the precise regulation and manipulation of biological tissues. Nanotechnology is increasingly considered to manage reproductive system disorders, for example, nanomaterial imaging allows for early detection and enhances diagnostic precision to determine disease severity and progression. Additionally, nano drug delivery systems are gaining attention for their ability to target the reproductive system successfully, thereby increasing therapeutic efficacy and decreasing side effects. This comprehensive review outlines the anatomy of the female upper genital tract by highlighting the complex mucosal barriers and their impact on systemic and local drug delivery. Advances in nano drug delivery are described for their sustainable therapeutic action and increased biocompatibility to highlight the potential of nano drug delivery strategies in managing female upper genital tract disorders.
Collapse
Affiliation(s)
| | | | - Hendrik J. R. Lemmer
- Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), North-West University, Potchefstroom 2531, South Africa; (D.v.S.); (M.G.)
| |
Collapse
|
6
|
Wang Y, Su L, Hu Z, Peng S, Li N, Fu H, Wang B, Wu H. Resveratrol suppresses liver cancer progression by downregulating AKR1C3: targeting HCC with HSA nanomaterial as a carrier to enhance therapeutic efficacy. Apoptosis 2024; 29:1429-1453. [PMID: 39023830 DOI: 10.1007/s10495-024-01995-w] [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] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
The enzyme AKR1C3 plays a crucial role in hormone and drug metabolism and is associated with abnormal expression in liver cancer, leading to tumor progression and poor prognosis. Nanoparticles modified with HSA can modulate the tumor microenvironment by enhancing photodynamic therapy to induce apoptosis in tumor cells and alleviate hypoxia. Therefore, exploring the potential regulatory mechanisms of resveratrol on AKR1C3 through the construction of HSA-RSV NPs carriers holds significant theoretical and clinical implications for the treatment of liver cancer. The aim of this study is to investigate the targeted regulation of AKR1C3 expression through the loading of resveratrol (RSV) on nanomaterials HSA-RSV NPs (Nanoparticles) in order to alleviate tumor hypoxia and inhibit the progression of hepatocellular carcinoma (HCC), and to explore its molecular mechanism. PubChem database and PharmMapper server were used to screen the target genes of RSV. HCC-related differentially expressed genes (DEGs) were analyzed through the GEO dataset, and relevant genes were retrieved from the GeneCards database, resulting in the intersection of the three to obtain candidate DEGs. GO and KEGG enrichment analyses were performed on the candidate DEGs to analyze the potential cellular functions and molecular signaling pathways affected by the main target genes. The cytohubba plugin was used to screen the top 10 target genes ranked by Degree and further intersected the results of LASSO and Random Forest (RF) to obtain hub genes. The expression analysis of hub genes and the prediction of malignant tumor prognosis were conducted. Furthermore, a pharmacophore model was constructed using PharmMapper. Molecular docking simulations were performed using AutoDockTools 1.5.6 software, and ROC curve analysis was performed to determine the core target. In vitro cell experiments were carried out by selecting appropriate HCC cell lines, treating HCC cells with different concentrations of RSV, or silencing or overexpressing AKR1C3 using lentivirus. CCK-8, clone formation, flow cytometry, scratch experiment, and Transwell were used to measure cancer cell viability, proliferation, migration, invasion, and apoptosis, respectively. Cellular oxygen consumption rate was analyzed using the Seahorse XF24 analyzer. HSA-RSV NPs were prepared, and their characterization and cytotoxicity were evaluated. The biological functional changes of HCC cells after treatment were detected. An HCC subcutaneous xenograft model was established in mice using HepG2 cell lines. HSA-RSV NPs were injected via the tail vein, with a control group set, to observe changes in tumor growth, tumor targeting of NPs, and biological safety. TUNEL, Ki67, and APC-hypoxia probe staining were performed on excised tumor tissue to detect tumor cell proliferation, apoptosis, and hypoxia. Lentivirus was used to silence or overexpress AKR1C3 simultaneously with the injection of HSA-RSV NPs via the tail vein to assess the impact of AKR1C3 on the regulation of HSA-RSV NPs in HCC progression. Bioinformatics analysis revealed that AKR1C3 is an important target gene involved in the regulation of HCC by RSV, which is associated with the prognosis of HCC patients and upregulated in expression. In vitro cell experiments showed that RSV significantly inhibits the respiratory metabolism of HCC cells, suppressing their proliferation, migration, and invasion and promoting apoptosis. Silencing AKR1C3 further enhances the toxicity of RSV towards HCC cells. The characterization and cytotoxicity experiments of nanomaterials demonstrated the successful construction of HSA-RSV NPs, which exhibited stronger inhibitory effects on HCC cells. In vivo, animal experiments further confirmed that targeted downregulation of AKR1C3 by HSA-RSV NPs suppresses the progression of HCC and tumor hypoxia while exhibiting tumor targeting and biological safety. Targeted downregulation of AKR1C3 by HSA-RSV NPs can alleviate HCC tumor hypoxia and inhibit the progression of HCC.
Collapse
Affiliation(s)
- Ying Wang
- Operating Room, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Longxiang Su
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Zhansheng Hu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Shuang Peng
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Na Li
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Haiyan Fu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Baoquan Wang
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Huiping Wu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China.
| |
Collapse
|
7
|
Chaudhary AA, Fareed M, Khan SUD, Alneghery LM, Aslam M, Alex A, Rizwanullah M. Exploring the therapeutic potential of lipid-based nanoparticles in the management of oral squamous cell carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1223-1246. [PMID: 39465011 PMCID: PMC11502080 DOI: 10.37349/etat.2024.00272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a highly malignant and invasive tumor with significant mortality and morbidity. Current treatment modalities such as surgery, radiotherapy, and chemotherapy encounter significant limitations, such as poor targeting, systemic toxicity, and drug resistance. There is an urgent need for novel therapeutic strategies that offer targeted delivery, enhanced efficacy, and reduced side effects. The advent of lipid-based nanoparticles (LNPs) offers a promising tool for OSCC therapy, potentially overcoming the limitations of current therapeutic approaches. LNPs are composed of biodegradable and biocompatible lipids, which minimize the risk of toxicity and adverse effects. LNPs can encapsulate hydrophobic drugs, improving their solubility and stability in the biological environment, thereby enhancing their bioavailability. LNPs demonstrate significantly higher ability to encapsulate lipophilic drugs than other nanoparticle types. LNPs offer excellent storage stability, minimal drug leakage, and controlled drug release, making them highly effective nanoplatforms for the delivery of chemotherapeutic agents. Additionally, LNPs can be modified by complexing them with specific target ligands on their surface. This surface modification allows the active targeting of LNPs to the tumors in addition to the passive targeting mechanism. Furthermore, the PEGylation of LNPs improves their hydrophilicity and enhances their biological half-life by reducing clearance by the reticuloendothelial system. This review aims to discuss current treatment approaches and their limitations, as well as recent advancements in LNPs for better management of OSCC.
Collapse
Affiliation(s)
- Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammad Fareed
- College of Medicine, AlMaarefa University, Diriyah, Riyadh 11597, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001, Kurdistan Region, Iraq
| | - Arockia Alex
- Molecular and Nanobiotechnology Laboratory (MNBL), Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Md Rizwanullah
- Drug Delivery and Nanomedicine Unit, Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| |
Collapse
|
8
|
Rajendran AT, Vadakkepushpakath AN. Natural Food Components as Biocompatible Carriers: A Novel Approach to Glioblastoma Drug Delivery. Foods 2024; 13:2812. [PMID: 39272576 PMCID: PMC11394703 DOI: 10.3390/foods13172812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/15/2024] Open
Abstract
Efficient drug delivery methods are crucial in modern pharmacotherapy to enhance treatment efficacy, minimize adverse effects, and improve patient compliance. Particularly in the context of glioblastoma treatment, there has been a recent surge in interest in using natural dietary components as innovative carriers for drug delivery. These food-derived carriers, known for their safety, biocompatibility, and multifunctional properties, offer significant potential in overcoming the limitations of conventional drug delivery systems. This article thoroughly overviews numerous natural dietary components, such as polysaccharides, proteins, and lipids, used as drug carriers. Their mechanisms of action, applications in different drug delivery systems, and specific benefits in targeting glioblastoma are examined. Additionally, the safety, biocompatibility, and regulatory considerations of employing food components in drug formulations are discussed, highlighting their viability and future prospects in the pharmaceutical field.
Collapse
Affiliation(s)
- Arunraj Tharamelveliyil Rajendran
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Anoop Narayanan Vadakkepushpakath
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore 575018, Karnataka, India
| |
Collapse
|
9
|
Yu G, Ye Z, Yuan Y, Wang X, Li T, Wang Y, Wang Y, Yan J. Recent Advancements in Biomaterials for Chimeric Antigen Receptor T Cell Immunotherapy. Biomater Res 2024; 28:0045. [PMID: 39011521 PMCID: PMC11246982 DOI: 10.34133/bmr.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/13/2024] [Indexed: 07/17/2024] Open
Abstract
Cellular immunotherapy is an innovative cancer treatment method that utilizes the patient's own immune system to combat tumor cells effectively. Currently, the mainstream therapeutic approaches include chimeric antigen receptor T cell (CAR-T) therapy, T cell receptor gene-modified T cell therapy and chimeric antigen receptor natural killer-cell therapy with CAR-T therapy mostly advanced. Nonetheless, the conventional manufacturing process of this therapy has shortcomings in each step that call for improvement. Marked efforts have been invested for its enhancement while notable progresses achieved in the realm of biomaterials application. With CAR-T therapy as a prime example, the aim of this review is to comprehensively discuss the various biomaterials used in cell immunotherapy, their roles in regulating immune cells, and their potential for breakthroughs in cancer treatment from gene transduction to efficacy enhancement. This article additionally addressed widely adopted animal models for efficacy evaluating.
Collapse
Affiliation(s)
- Gaoyu Yu
- School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Zhichao Ye
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Yuyang Yuan
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
- Department of Translational Medicine & Clinical Research, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Xiaofeng Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang Province, China
| | - Tianyu Li
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
- Department of Translational Medicine & Clinical Research, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Yi Wang
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Yifan Wang
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
- Department of Translational Medicine & Clinical Research, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Jianing Yan
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| |
Collapse
|
10
|
Meng Y, Chen S, Li P, Wang C, Ni X. Tumor Cell Membrane-Encapsulated MLA Solid Lipid Nanoparticles for Targeted Diagnosis and Radiosensitization Therapy of Cutaneous Squamous Cell Carcinoma. Mol Pharm 2024; 21:3218-3232. [PMID: 38885477 DOI: 10.1021/acs.molpharmaceut.3c01247] [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] [Indexed: 06/20/2024]
Abstract
Squamous cell carcinoma (SCC) is a common nonmelanoma skin cancer. Radiotherapy plays an integral role in treating SCC due to its characteristics, such as diminished intercellular adhesion, heightened cell migration and invasion capabilities, and immune evasion. These problems lead to inaccurate tumor boundary positioning and radiotherapy tolerance in SCC treatment. Thus, accurate localization and enhanced radiotherapy sensitivity are imperative for effective SCC treatment. To address the existing limitations in SCC therapy, we developed monoglyceride solid lipid nanoparticles (MG SLNs) and enveloped them with the A431 cell membrane (A431 CM) to create A431@MG. The characterization results showed that A431@MG was spherical. Furthermore, A431@MG had specific targeting for A431 cells. In A431 tumor-bearing mice, A431@MG demonstrated prolonged accumulation within tumors, ensuring precise boundary localization of SCC. We further advanced the approach by preparing MG SLNs encapsulating 5-aminolevulinic acid methyl ester (MLA) and desferrioxamine (DFO) with an A431 CM coating to yield A431@MG-MLA/DFO. Several studies have revealed that DFO effectively reduced iron content, impeding protoporphyrin IX (PpIX) biotransformation and promoting PpIX accumulation. Simultaneously, MLA was metabolized into PpIX upon cellular entry. During radiotherapy, the heightened PpIX levels enhanced reactive oxygen species (ROS) generation, inducing DNA and mitochondrial damage and leading to cell apoptosis. In A431 tumor-bearing mice, the A431@MG-MLA/DFO group exhibited notable radiotherapy sensitization, displaying superior tumor growth inhibition. Combining A431@MG-MLA/DFO with radiotherapy significantly improved anticancer efficacy, highlighting its potential to serve as an integrated diagnostic and therapeutic strategy for SCC.
Collapse
Affiliation(s)
- Yanyan Meng
- School of Pharmacy, Changzhou University, Changzhou 213164, China
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou 213003, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou 213003, China
- Changzhou Key Laboratory of Medical Physics, Changzhou 213003, China
| | - Shaoqing Chen
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou 213003, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou 213003, China
- Changzhou Key Laboratory of Medical Physics, Changzhou 213003, China
| | - Pengyin Li
- School of Pharmacy, Changzhou University, Changzhou 213164, China
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou 213003, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou 213003, China
- Changzhou Key Laboratory of Medical Physics, Changzhou 213003, China
| | - Cheli Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Xinye Ni
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou 213003, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou 213003, China
- Changzhou Key Laboratory of Medical Physics, Changzhou 213003, China
| |
Collapse
|
11
|
Rajoriya V, Gupta R, Vengurlekar S, Surendra Singh U. Nanostructured lipid carriers (NLCs): A promising candidate for lung cancer targeting. Int J Pharm 2024; 655:123986. [PMID: 38493842 DOI: 10.1016/j.ijpharm.2024.123986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/24/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
Lung cancer stands as the foremost health issue and the principal reason for mortality worldwide. It is projected that India will see over 1.73 million new cases and more than 880,000 deaths related to cancer, with lung cancer being a significant contributor. The efficiency of existing chemotherapy procedures is not optimal because of less soluble nature and short half-life of anticancer substances. More precipitated toxicity and non-existence of targeting propensity can lead to severe side effects, non-compliance, and inconvenience for patients. Nonetheless, the domain of nanomedicine has undergone a revolution in the past few years with the advent of novel drug delivery mechanisms that tackle the drawbacks of conventional approaches. Diverse nanoparticle-based drug delivery methods, including liposomes, nanoparticles, nanostructured lipid carrier and solid lipid nanoparticle that encapsulated chemotherapy drugs, are currently employed for efficient lung cancer therapy. NLCs, recognized as the second-generation lipid nanocarriers, are a focused drug delivery mechanism that has garnered significant interest owing to their multitude of advantages such as increased stability, minimal toxicity, prolonged shelf life, superior encapsulation capability, and biocompatible nature. This review focuses on the NLCs carrier system, discussing its preparation methods, types, characterization, applications, and future prospects in lung cancer treatment.
Collapse
Affiliation(s)
- Vaibhav Rajoriya
- University Institute of Pharmacy, Oriental University, Indore, Madhya Pradesh 453555 India.
| | - Ravikant Gupta
- Faculty, University Institute of Pharmacy, Oriental University, Indore, Madhya Pradesh 453555 India
| | - Sudha Vengurlekar
- Faculty, University Institute of Pharmacy, Oriental University, Indore, Madhya Pradesh 453555 India
| | - Upama Surendra Singh
- University Institute of Pharmacy, Oriental University, Indore, Madhya Pradesh 453555 India
| |
Collapse
|
12
|
Shukla R, Singh A, Singh KK. Vincristine-based nanoformulations: a preclinical and clinical studies overview. Drug Deliv Transl Res 2024; 14:1-16. [PMID: 37552393 PMCID: PMC10746576 DOI: 10.1007/s13346-023-01389-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2023] [Indexed: 08/09/2023]
Abstract
Vincristine (VCR) is a chemotherapeutic agent obtained from natural alkaloid plant source Catharanthus roseus. VCR has been significantly useful in treatments of lung cancer, lymphocyte-based leukaemia, glioblastomas and acute myeloid leukaemia. VCR attaches to tubulin fibrils and prevents filament polymerization that permanently led to mitosis inhibition in cancer cells. Clinically, VCR is administered to patients in multidrug combination to reduce adverse drug effects and potential blockage of bone marrow inhibition due to prescribed monotherapy. However, VCR possesses low cancer tissue affinity and at higher dose often led to irreversible neurotoxicity. Conventional VCR injectables are successfully used in clinics, but lack of controlled release, non-specific biodistribution and consequent off-target side effects are still major challenges. Currently, nanotechnological drug delivery systems are being explored for improvement of VCR pharmacokinetic profile and tumour-specific targeting. Various nanomedicine formulations such as liposomes, lipid nanoparticles, and polymeric nanocarriers of VCR have been studied under various in vitro and in vivo models. In this review, we have summarised the chemotherapeutic role of VCR, evaluated the mechanism of action, pharmacokinetics and challenges associated with VCR delivery. Moreover, application of VCR in nanomedicine and effect on anticancer efficacy in preclinical and clinical setting are also being discussed.
Collapse
Affiliation(s)
- Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, U.P, 226002, Lucknow, India.
| | - Ajit Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, U.P, 226002, Lucknow, India
| | - Kamalinder K Singh
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK.
| |
Collapse
|
13
|
Nayak V, Patra S, Rout S, Jena AB, Sharma R, Pattanaik KP, Singh J, Pandey SS, Singh RP, Majhi S, Singh KR, Kerry RG. Regulation of neuroinflammation in Alzheimer's disease via nanoparticle-loaded phytocompounds with anti-inflammatory and autophagy-inducing properties. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155150. [PMID: 37944239 DOI: 10.1016/j.phymed.2023.155150] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/23/2023] [Accepted: 10/14/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by neuroinflammation linked to amyloid β (Aβ) aggregation and phosphorylated tau (τ) protein in neurofibrillary tangles (NFTs). Key elements in Aβ production and NFT assembly, like γ-secretase and p38 mitogen-activated protein kinase (p38MAPK), contribute to neuroinflammation. In addition, impaired proteosomal and autophagic pathways increase Aβ and τ aggregation, leading to neuronal damage. Conventional neuroinflammation drugs have limitations due to unidirectional therapeutic approaches and challenges in crossing the Blood-Brain Barrier (BBB). Clinical trials for non-steroidal anti-inflammatory drugs (NSAIDs) and other therapeutics remain uncertain. Novel strategies addressing the complex pathogenesis and BBB translocation are needed to effectively tackle AD-related neuroinflammation. PURPOSE The current scenario demands for a much-sophisticated theranostic measures which could be achieved via customized engineering and designing of novel nanotherapeutics. As, these therapeutics functions as a double edge sword, having the efficiency of unambiguous targeting, multiple drug delivery and ability to cross BBB proficiently. METHODS Inclusion criteria involve selecting recent, English-language studies from the past decade (2013-2023) that explore the regulation of neuroinflammation in neuroinflammation, Alzheimer's disease, amyloid β, tau protein, nanoparticles, autophagy, and phytocompounds. Various study types, including clinical trials, experiments, and reviews, were considered. Exclusion criteria comprised non-relevant publication types, studies unrelated to Alzheimer's disease or phytocompounds, those with methodological flaws, duplicates, and studies with inaccessible data. RESULTS In this study, polymeric nanoparticles loaded with specific phytocompounds and coated with an antibody targeting the transferrin receptor (anti-TfR) present on BBB. Thereafter, the engineered nanoparticles with the ability to efficiently traverse the BBB and interact with target molecules within the brain, could induce autophagy, a cellular process crucial for neuronal health, and exhibit potent anti-inflammatory effects. Henceforth, the proposed combination of desired phytocompounds, polymeric nanoparticles, and anti-TfR coating presents a promising approach for targeted drug delivery to the brain, with potential implications in neuroinflammatory conditions such as Alzheimer's disease.
Collapse
Affiliation(s)
- Vinayak Nayak
- ICAR- National Institute on Foot and Mouth Disease-International Centre for Foot and Mouth Disease, Arugul, Bhubaneswar, Odisha (752050), India
| | - Sushmita Patra
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra (410210), India
| | - Shrushti Rout
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha (751004), India
| | - Atala Bihari Jena
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (02115), United States of America
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh (221005), India
| | - Kali Prasad Pattanaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India
| | - Jay Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh (221005), India
| | - Shyam S Pandey
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu (8080196), Japan
| | - Ravindra Pratap Singh
- Department of Biotechnology, Faculty of Science, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh 484887, India
| | - Sanatan Majhi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (02115), United States of America
| | - Kshitij Rb Singh
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu (8080196), Japan.
| | - Rout George Kerry
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha (751004), India.
| |
Collapse
|