1
|
Le Meur M, Pignatelli J, Blasi P, Palomo V. Nanoparticles targeting the central circadian clock: Potential applications for neurological disorders. Adv Drug Deliv Rev 2025; 220:115561. [PMID: 40120723 DOI: 10.1016/j.addr.2025.115561] [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: 11/20/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Circadian rhythms and their involvement with various human diseases, including neurological disorders, have become an intense area of research for the development of new pharmacological treatments. The location of the circadian clock machinery in the central nervous system makes it challenging to reach molecular targets at therapeutic concentrations. In addition, a timely administration of the therapeutic agents is necessary to efficiently modulate the circadian clock. Thus, the use of nanoparticles in circadian clock dysfunctions may accelerate their clinical translation by addressing these two key challenges: enhancing brain penetration and/or enabling their formulation in chronodelivery systems. This review describes the implications of the circadian clock in neurological pathologies, reviews potential molecular targets and their modulators and suggests how the use of nanoparticle-based formulations could improve their clinical success. Finally, the potential integration of nanoparticles into chronopharmaceutical drug delivery systems will be described.
Collapse
Affiliation(s)
- Marion Le Meur
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain; Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum - Università di Bologna, 40127 Bologna, Italy
| | - Jaime Pignatelli
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Paolo Blasi
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum - Università di Bologna, 40127 Bologna, Italy.
| | - Valle Palomo
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain; Unidad de Nanobiotecnología asociada al Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
| |
Collapse
|
2
|
Zhou S, Zhang M, Wang J, Chen X, Xu Z, Yan Y, Li Y. Nanofibers in Glioma Therapy: Advances, Applications, and Overcoming Challenges. Int J Nanomedicine 2025; 20:4677-4703. [PMID: 40255668 PMCID: PMC12008729 DOI: 10.2147/ijn.s510363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/31/2025] [Indexed: 04/22/2025] Open
Abstract
Despite relentless effort to study glioma treatment, the prognosis for glioma patients remains poor. The main obstacles include the high rate of recurrence and the difficulty of passing the blood-brain barrier (BBB) for therapeutic drugs. Nanomaterials owing to their special physicochemical properties have been used in a wide range of fields thus far. The nanodrug delivery system (NDDS) with the ability of crossing the BBB, targeting glioma site, maintaining drug stability and controlling drug release, has significantly enhanced the anti-tumor therapeutic effect, improving the prognosis of glioma patients. Aligned nanofibers (NFs) are ideal materials to establish in vitro models of glioma microenvironment (GME), enabling the exploration of the mechanism of glioma cell migration and invasion to discover novel therapeutic targets. Moreover, NFs are now widely used in glioma applications such as radiotherapy, phototherapy, thermotherapy and immunotherapy. Despite the absolute dominance of NFs in anti-glioma applications, there are still some problems such as the further optimization of NDDS, and the impact of interactions between nanofibers and the protein corona (PC) on glioma therapy. This paper will shed light on the latest glioma applications of NFs in drug delivery systems and mimicking the tumor microenvironment (TME), and discuss how to further optimize the NDDS and eliminate or utilize the nanomedicine-PC interactions.
Collapse
Affiliation(s)
- Shangjun Zhou
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, People’s Republic of China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Mingcheng Zhang
- Center of Endoscopy, The Second Affiliated Hospital of Shandong First Medical University Tai’an, Shandong, People’s Republic of China
| | - Jiayu Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yong Li
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
3
|
Bates AC, Klugh KL, Galaeva AO, Patch RA, Manganaro JF, Markham SA, Scurek E, Levina A, Lay PA, Crans DC. Optimizing Therapeutics for Intratumoral Cancer Treatments: Antiproliferative Vanadium Complexes in Glioblastoma. Int J Mol Sci 2025; 26:994. [PMID: 39940763 PMCID: PMC11817060 DOI: 10.3390/ijms26030994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Glioblastoma, an aggressive cancer, is difficult to treat due to its location, late detection, drug resistance, and poor absorption of chemotherapeutics. Intratumoral drug administration offers a promising potential treatment alternative with localized delivery and minimal systemic toxicity. Vanadium(V) coordination complexes, incorporating Schiff base and catecholate ligands, have shown effects as antiproliferative agents with tunable efficacy and reactivity, stability, steric bulk, hydrophobicity, uptake, and toxicity optimized for the intratumoral administration vehicle. A new series of oxovanadium(V) Schiff base-catecholate complexes were synthesized and characterized using nuclear magnetic resonance (NMR), UV-Vis, and infrared spectroscopy and mass spectrometry. Stability under physiological conditions was assessed via UV-Vis spectroscopy, and the antiproliferative activity was evaluated in T98G glioblastoma and SVG p12 normal glial cells using viability assays. The newly synthesized [VO(3-tBuHSHED)(TIPCAT)] complex was more stable (t1/2 ~4.5 h) and had strong antiproliferative activity (IC50 ~1.5 µM), comparing favorably with the current lead compound, [VO(HSHED)(DTB)]. The structural modifications enhanced stability, hydrophobicity, and steric bulk through substitution with iso-propyl and tert-butyl groups. The improved properties were attributed to steric hindrance associated with the new Schiff base and catecholato ligands, as well as the formation of non-toxic byproducts upon degradation. The [VO(3-tBuHSHED)(TIPCAT)] complex emerges as a promising candidate for glioblastoma therapy by demonstrating enhanced stability and a greater selectivity, which highlights the role of strategic ligand design in developing localized therapies for the treatment of resistant cancers. In reporting the new class of compounds effective against T98G glioblastoma cells, we describe the generally desirable properties that potential drugs being developed for intratumoral administration should have.
Collapse
Affiliation(s)
- Andrew C. Bates
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
| | - Kameron L. Klugh
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
| | - Anna O. Galaeva
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
| | - Raley A. Patch
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
| | - John F. Manganaro
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
| | - Skyler A. Markham
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
| | - Emma Scurek
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Peter A. Lay
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Debbie C. Crans
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (A.C.B.); (K.L.K.); (A.O.G.); (R.A.P.); (J.F.M.); (S.A.M.); (E.S.)
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
4
|
Zhang S, Zhong R, Younis MR, He H, Xu H, Li G, Yang R, Lui S, Wang Y, Wu M. Hydrogel Applications in the Diagnosis and Treatment of Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:65754-65778. [PMID: 39366948 DOI: 10.1021/acsami.4c11855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Glioblastoma multiforme (GBM), a common malignant neurological tumor, has boundaries indistinguishable from those of normal tissue, making complete surgical removal ineffective. The blood-brain barrier (BBB) further impedes the efficacy of radiotherapy and chemotherapy, leading to suboptimal treatment outcomes and a heightened probability of recurrence. Hydrogels offer multiple advantages for GBM diagnosis and treatment, including overcoming the BBB for improved drug delivery, controlled drug release for long-term efficacy, and enhanced relaxation properties of magnetic resonance imaging (MRI) contrast agents. Hydrogels, with their excellent biocompatibility and customizability, can mimic the in vivo microenvironment, support tumor cell culture, enable drug screening, and facilitate the study of tumor invasion and metastasis. This paper reviews the classification of hydrogels and recent research for the diagnosis and treatment of GBM, including their applications as cell culture platforms and drugs including imaging contrast agents carriers. The mechanisms of drug release from hydrogels and methods to monitor the activity of hydrogel-loaded drugs are also discussed. This review is intended to facilitate a more comprehensive understanding of the current state of GBM research. It offers insights into the design of integrated hydrogel-based GBM diagnosis and treatment with the objective of achieving the desired therapeutic effect and improving the prognosis of GBM.
Collapse
Affiliation(s)
- Shuaimei Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Renming Zhong
- Radiotherapy Physics & Technology Center, Cancer Center, West China Hospital, Chengdu, Sichuan 610041, P. R. China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Hualong He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ruiyan Yang
- Department of Biology, Macalester College, Saint Paul, Minnesota 55105, United States
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
5
|
Bostanci A, Doganlar O. MELATONIN ENHANCES TEMOZOLOMIDE-INDUCED APOPTOSIS IN GLIOBLASTOMA AND NEUROBLASTOMA CELLS. Exp Oncol 2024; 46:87-100. [PMID: 39396175 DOI: 10.15407/exp-oncology.2024.02.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND The combination of temozolomide (TMZ) and paclitaxel (PTX) is the most commonly used chemotherapy regimen for glioblastoma, but there is no specific treatment for neuroblastoma due to the acquired multidrug resistance. Approximately half of treated glioblastoma patients develop resistance to TMZ and experience serious side effects. Melatonin (MEL), a multifunctional hormone long known for its antitumor effects, has a great advantage in combination cancer therapy thanks to its ability to affect tumors differently than normal cells. AIM This study aims to evaluate the in vitro inhibitory effects of MEL in combination with TMZ on cancer cell viability and to elucidate the underlying mechanisms in the glioblastoma and neuroblastoma cell lines. MATERIALS AND METHODS C6 (Rattus norvegicus) and N1E-115 (Mus musculus) cancer cell lines and C8-D1A (mice) healthy cell lines were used. Cell proliferation was evaluated using the MTT test. IC50 values were determined by probit analysis. Two concentrations of TMZ (IC50 and 1/2 IC50) were used to induce cytotoxicity in the C6 and N1E-115 cell lines, both alone and in combination with PXT and MEL (all at IC50). The viable, dead, and apoptotic cells were determined by image-based cytometry using Annexin V/PI staining. The gene expression related to signaling pathways was assessed by the quantitative reverse transcription polymerase chain reaction (qRT-PCR), and key proteins were identified by the Western blot analysis. RESULTS MTT assay showed that the combination of TMZ and MEL significantly reduces the viability of both glioblastoma and neuroblastoma cells compared to the vehicle-treated controls. Notably, MEL combined with 1/2 IC50 TMZ showed a significant death rate of cancer cells compared to controls and PTX. According to qRT-PCR data, the TMZ + MEL combination resulted in the upregulation of the genes of antioxidative enzymes (Sod1 and Sod2) and DNA repair genes (Mlh1, Exo1, and Rad18) in both cell lines. Moreover, the levels of Nfkb1 and Pik3cg were significantly reduced following the TMZ + MEL treatment. The combination of MEL with TMZ also enhanced the cell cycle arrest and increased the expression of p53 and pro-apoptotic proteins (Bax and caspase-3), while significantly decreasing the expression of anti-apoptotic protein Bcl-2. CONCLUSIONS Our findings indicate that the combination of MEL with a low dose of TMZ may serve as an upstream inducer of apoptosis. This suggests the potential development of a novel selective therapeutic strategy as an alternative to TMZ for the treatment of both glioblastoma and neuroblastoma.
Collapse
Affiliation(s)
- A Bostanci
- Department of Genetics and Bioengineering, Trakya University, Edirne, Turkey
| | - O Doganlar
- Department of Medical Biology, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
6
|
Thomas RG, Kim S, Tran TAT, Kim YH, Nagareddy R, Jung TY, Kim SK, Jeong YY. Magnet-Guided Temozolomide and Ferucarbotran Loaded Nanoparticles to Enhance Therapeutic Efficacy in Glioma Model. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:939. [PMID: 38869565 PMCID: PMC11173836 DOI: 10.3390/nano14110939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024]
Abstract
Background. The aim of the study was to synthesize liposomal nanoparticles loaded with temozolomide and ferucarbotran (LTF) and to evaluate the theranostic effect of LTF in the glioma model. Methods. We synthesized an LTF that could pass through the Blood Brain Barrier (BBB) and localize in brain tumor tissue with the help of magnet guidance. We examined the chemical characteristics. Cellular uptake and cytotoxicity studies were conducted in vitro. A biodistribution and tumor inhibition study was conduted using an in vivo glioma model. Results. The particle size and surface charge of LTF show 108 nm and -38 mV, respectively. Additionally, the presence of ferucarbotran significantly increased the contrast agent effect of glioma compared to the control group in MR imaging. Magnet-guided LTF significantly reduced the tumor size compared to control and other groups. Furthermore, compared to the control group, our results demonstrate a significant inhibition in brain tumor size and an increase in lifespan. Conclusions. These findings suggest that the LTF with magnetic guidance represents a novel approach to address current obstacles, such as BBB penetration of nanoparticles and drug resistance. Magnet-guided LTF is able to enhance therapeutic efficacy in mouse brain glioma.
Collapse
Affiliation(s)
- Reju George Thomas
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
| | - Subin Kim
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea;
| | - Thi-Anh-Thuy Tran
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea
- Brain Tumor Research Laboratory, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea (T.-Y.J.)
| | - Young Hee Kim
- Brain Tumor Research Laboratory, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea (T.-Y.J.)
| | - Raveena Nagareddy
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
| | - Tae-Young Jung
- Brain Tumor Research Laboratory, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea (T.-Y.J.)
- Department of Neurosurgery, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea
| | - Seul Kee Kim
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
- Department of Radiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun 58128, Republic of Korea; (R.G.T.)
- Department of Radiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
7
|
Shahid N, Erum A, Hanif S, Malik NS, Tulain UR, Syed MA. Nanocomposite Hydrogels-A Promising Approach towards Enhanced Bioavailability and Controlled Drug Delivery. Curr Pharm Des 2024; 30:48-62. [PMID: 38155469 DOI: 10.2174/0113816128283466231219071151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/06/2023] [Indexed: 12/30/2023]
Abstract
Nanotechnology has emerged as the eminent focus of today's research to overcome challenges related to conventional drug delivery systems. A wide spectrum of novel delivery systems has been investigated to improve the therapeutic outcomes of drugs. The polymer-based nanocomposite hydrogels (NCHs) that have evolved as efficient carriers for controlled drug delivery are of particular interest in this regard. Nanocomposites amalgamate the properties of both nanoparticles (NPs) as well as hydrogels, exhibiting superior functionalities over conventional hydrogels. This multiple functionality is based upon advanced mechanical, electrical, optical as well as magnetic properties. Here is a brief overview of the various types of nanocomposites, such as NCHs based on Carbon-bearing nanomaterials, polymeric nanoparticles, inorganic nanoparticles, and metal and metal-oxide NPs. Accordingly, this article will review numerous ways of preparing these NCHs with particular emphasis on the vast biomedical applications displayed by them in numerous fields such as tissue engineering, drug delivery, wound healing, bioprinting, biosensing, imaging and gene silencing, cancer therapy, antibacterial therapy, etc. Moreover, various features can be tuned, based on the final application, by controlling the chemical composition of hydrogel network, which may also influence the released conduct. Subsequently, the recent work and future prospects of this newly emerging class of drug delivery system have been enlisted.
Collapse
Affiliation(s)
- Nariman Shahid
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Alia Erum
- Faculty of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Sana Hanif
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Nadia Shamshad Malik
- Faculty of Pharmacy, Capital University of Science and Technology, Islamabad, Pakistan
| | | | - Muhammad Ali Syed
- Department of Pharmaceutical Sciences, Faculty of Chemistry & Life Sciences, GC University Lahore, Lahore, Pakistan
| |
Collapse
|
8
|
Chiariello M, Inzalaco G, Barone V, Gherardini L. Overcoming challenges in glioblastoma treatment: targeting infiltrating cancer cells and harnessing the tumor microenvironment. Front Cell Neurosci 2023; 17:1327621. [PMID: 38188666 PMCID: PMC10767996 DOI: 10.3389/fncel.2023.1327621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Glioblastoma (GB) is a highly malignant primary brain tumor with limited treatment options and poor prognosis. Despite current treatment approaches, including surgical resection, radiation therapy, and chemotherapy with temozolomide (TMZ), GB remains mostly incurable due to its invasive growth pattern, limited drug penetration beyond the blood-brain barrier (BBB), and resistance to conventional therapies. One of the main challenges in GB treatment is effectively eliminating infiltrating cancer cells that remain in the brain parenchyma after primary tumor resection. We've reviewed the most recent challenges and surveyed the potential strategies aimed at enhancing local treatment outcomes.
Collapse
Affiliation(s)
- Mario Chiariello
- Institute of Clinical Physiology, Consiglio Nazionale delle Ricerche, Via Fiorentina, Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina, Siena, Italy
| | - Giovanni Inzalaco
- Institute of Clinical Physiology, Consiglio Nazionale delle Ricerche, Via Fiorentina, Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina, Siena, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Virginia Barone
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Lisa Gherardini
- Institute of Clinical Physiology, Consiglio Nazionale delle Ricerche, Via Fiorentina, Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina, Siena, Italy
| |
Collapse
|
9
|
Nie Q, Chen W, Zhang T, Ye S, Ren Z, Zhang P, Wen J. Iron oxide nanoparticles induce ferroptosis via the autophagic pathway by synergistic bundling with paclitaxel. Mol Med Rep 2023; 28:198. [PMID: 37681444 PMCID: PMC10510030 DOI: 10.3892/mmr.2023.13085] [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: 02/09/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
In recent years, inhibiting tumor cell activity by triggering cell ferroptosis has become a research hotspot. The development of generic targeted nanotherapeutics might bring new ideas for non‑invasive applications. Currently, the potential mechanism underlying the universal application of paclitaxel (PTX)‑loaded iron oxide nanoparticles (IONP@PTX) to different types of tumors is unclear. The present study aimed to prepare IONP@PTX for targeted cancer therapy and further explore the potential mechanisms underlying the inhibitory effects of this material on the NCI‑H446 human small cell lung cancer and brain M059K malignant glioblastoma cell lines. First, a CCK‑8 assay was performed to determine cell viability, and then the combination index for evaluating drug combination interaction effect was evaluated. Intracellular reactive oxygen species (ROS) and lipid peroxidation levels were monitored using a DCFH‑DA fluorescent probe and a C11‑BODIPY™ fluorescent probe, respectively. Furthermore, western blotting assay was performed to determine the expression of autophagy‑ and iron death‑related proteins. The experimental results showed that, compared with either IONP monotherapy, PTX monotherapy, or IONP + PTX, IONP@PTX exerted a synergistic effect on the viability of both cell types, with significantly increased total iron ion concentration, ROS levels and lipid peroxidation levels. IONP@PTX significantly increased the expression of autophagy‑related proteins Beclin 1 and histone deacetylase 6 (HDAC6) in both cell lines (P<0.05), increased the expression of light chain 3 (LC3)‑II/I in NCI‑H446 cells (P<0.05) and decreased that of sequestosome1 (p62) in M059K cells (P<0.05). Moreover, the addition of rapamycin enhanced the IONP@PTX‑induced the upregulation of Beclin 1, LC3‑II/I and HDAC6 and the downregulation of mTORC1 protein in both cell lines (P<0.05). Moreover, rapamycin enhanced the IONP@PTX‑induced downregulation of p62 protein in NCI‑H446 cells (P<0.05), suggesting that IONP@PTX induces ferroptosis, most likely through autophagy. Collectively, the present findings show that IONP works synergistically with PTX to induce ferroptosis via the autophagic pathway.
Collapse
Affiliation(s)
- Qi Nie
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| | - Wenqing Chen
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| | - Tianmei Zhang
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Shangrong Ye
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Zhongyu Ren
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Peng Zhang
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Jian Wen
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| |
Collapse
|