1
|
Wang M, Bergès R, Malfanti A, Préat V, Bastiancich C. Local delivery of doxorubicin prodrug via lipid nanocapsule-based hydrogel for the treatment of glioblastoma. Drug Deliv Transl Res 2024; 14:3322-3338. [PMID: 37889402 PMCID: PMC11499358 DOI: 10.1007/s13346-023-01456-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
Glioblastoma (GBM) recurrences appear in most cases around the resection cavity borders and arise from residual GBM cells that cannot be removed by surgery. Here, we propose a novel treatment that combines the advantages of nanomedicine and local drug delivery to target these infiltrating GBM cells. We developed an injectable lipid nanocapsule (LNC)-based formulation loaded with lauroyl-doxorubicin prodrug (DOXC12). Firstly, we demonstrated the efficacy of intratumoral administration of DOXC12 in GL261 GBM-bearing mice, which extended mouse survival. Then, we formulated an injectable hydrogel by mixing the appropriate amount of prodrug with the lipophilic components of LNC. We optimized the hydrogel by incorporating cytidine-C16 (CytC16) to achieve a mechanical stiffness adapted for an application in the brain post-surgery (DOXC12-LNCCL). DOXC12-LNCCL exhibited high DOXC12 encapsulation efficiency (95%) and a size of approximately 60 nm with sustained drug release for over 1 month in vitro. DOXC12-LNCCL exhibited enhanced cytotoxicity compared to free DOXC12 (IC50 of 349 and 86 nM, respectively) on GL261 GBM cells and prevented the growth of GL261 spheroids cultured on organotypic brain slices. In vivo, post-surgical treatment with DOXC12-LNCCL significantly improved the survival of GL261-bearing mice. The combination of this local treatment with the systemic administration of anti-inflammatory drug ibuprofen further delayed the onset of recurrences. In conclusion, our study presents a promising therapeutic approach for the treatment of GBM. By targeting residual GBM cells and reducing the inflammation post-surgery, we present a new strategy to delay the onset of recurrences in the gap period between surgery and standard of care therapy.
Collapse
Affiliation(s)
- Mingchao Wang
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium
| | - Raphaël Bergès
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 27 Boulevard Jean Moulin, Marseille, 13005, France
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium.
| | - Chiara Bastiancich
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium.
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 27 Boulevard Jean Moulin, Marseille, 13005, France.
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, Turin, 10125, Italy.
| |
Collapse
|
2
|
Amoah AS, Pestov NB, Korneenko TV, Prokhorenko IA, Kurakin GF, Barlev NA. Lipoxygenases at the Intersection of Infection and Carcinogenesis. Int J Mol Sci 2024; 25:3961. [PMID: 38612771 PMCID: PMC11011848 DOI: 10.3390/ijms25073961] [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: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The persisting presence of opportunistic pathogens like Pseudomonas aeruginosa poses a significant threat to many immunocompromised cancer patients with pulmonary infections. This review highlights the complexity of interactions in the host's defensive eicosanoid signaling network and its hijacking by pathogenic bacteria to their own advantage. Human lipoxygenases (ALOXs) and their mouse counterparts are integral elements of the innate immune system, mostly operating in the pro-inflammatory mode. Taking into account the indispensable role of inflammation in carcinogenesis, lipoxygenases have counteracting roles in this process. In addition to describing the structure-function of lipoxygenases in this review, we discuss their roles in such critical processes as cancer cell signaling, metastases, death of cancer and immune cells through ferroptosis, as well as the roles of ALOXs in carcinogenesis promoted by pathogenic infections. Finally, we discuss perspectives of novel oncotherapeutic approaches to harness lipoxygenase signaling in tumors.
Collapse
Affiliation(s)
- Abdul-Saleem Amoah
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Molecular Oncology, Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Nikolay B. Pestov
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
- Vavilov Institute of General Genetics, Moscow 119991, Russia
| | - Tatyana V. Korneenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Igor A. Prokhorenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Georgy F. Kurakin
- Department of Biochemistry, Pirogov Russian National Research Medical University, Moscow 117513, Russia;
| | - Nickolai A. Barlev
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
| |
Collapse
|
3
|
Gu X, He X, Wang H, Li J, Chen R, Liu H. Dynamic Susceptibility Contrast-Enhanced Perfusion-Weighted Imaging in Differentiation Between Recurrence and Pseudoprogression in High-Grade Glioma: A Meta-analysis. J Comput Assist Tomogr 2024; 48:303-310. [PMID: 37654056 DOI: 10.1097/rct.0000000000001543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
INTRODUCTION In glioma patients that have undergone surgical tumor resection, the ability to reliably distinguish between pseudoprogression (PsP) and a recurrent tumor (RT) is of key clinical importance. Accordingly, this meta-analysis evaluated the utility of dynamic susceptibility contrast-enhanced perfusion-weighted imaging as a means of distinguishing between PsP and RT when analyzing patients with high-grade glioma. MATERIALS AND METHODS The PubMed, Web of Science, and Wanfang databases were searched for relevant studies. Pooled analyses of sensitivity, specificity, positive likelihood ratio (PLR), and negative likelihood ratio (NLR) values were conducted, after which the area under the curve (AUC) for summary receiver operating characteristic curves was computed. RESULTS This meta-analysis ultimately included 21 studies enrolling 879 patients with 888 lesions. Cerebral blood volume-associated diagnostic results were reported in 20 of the analyzed studies, and the respective pooled sensitivity, specificity, PLR, and NLR values were 86% (95% confidence interval [CI], 0.81-0.89), 83% (95% CI, 0.77-0.87), 4.94 (95% CI, 3.61-6.75), and 0.18 (95% CI, 0.13-0.23) for these 20 studies. The corresponding AUC value was 0.91 (95% CI, 0.88-0.93), and the publication bias risk was low ( P = 0.976). Cerebral blood flow-related diagnostic results were additionally reported in 6 of the analyzed studies, with respective pooled sensitivity, specificity, PLR, and NLR values of 85% (95% CI, 0.78-0.90), 85% (95% CI, 0.76-0.91), 5.54 (95% CI, 3.40-9.01), and 0.18 (95% CI, 0.12-0.26). The corresponding AUC value was 0.92 (95% CI, 0.89-0.94), and the publication bias risk was low ( P = 0.373). CONCLUSIONS The present meta-analysis results suggest that dynamic susceptibility contrast-enhanced perfusion-weighted imaging represents an effective diagnostic approach to distinguishing between PsP and RT in high-grade glioma patients.
Collapse
Affiliation(s)
| | - Xining He
- From the Departments of Neurosurgery
| | - Hualong Wang
- Radiology, Binzhou People's Hospital, Binzhou, China
| | | | | | | |
Collapse
|
4
|
Xu M, Cheng Y, Meng R, Yang P, Chen J, Qiao Z, Wu J, Qian K, Li Y, Wang P, Zhou L, Wang T, Sheng D, Zhang Q. Enhancement of Microglia Functions by Developed Nano-Immuno-Synergist to Ameliorate Immunodeficiency for Malignant Glioma Treatment. Adv Healthc Mater 2023; 12:e2301861. [PMID: 37573475 DOI: 10.1002/adhm.202301861] [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: 06/12/2023] [Revised: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Resident microglia are key factors in mediating immunity against brain tumors, but the microglia in malignant glioma are functionally impaired. Little immunotherapy is explored to restore microglial function against glioma. Herein, oleanolic acid (OA) (microglia "restorer") and D PPA-1 peptide (immune checkpoint blockade) are integrated on a nano-immuno-synergist (D PAM@OA) to work coordinately. The self-assembled OA core is coated with macrophage membrane for efficient blood-brain barrier penetration and microglia targeting, on which D PPA-1 peptide is attached via acid-sensitive bonds for specific release in tumor microenvironment. With the enhanced accumulation of the dual drugs in their respective action sites, D PAM@OA effectively promotes the recruitment and activation of effector T cells by inhibiting aberrant activation of Signal transducer and activator of transcription (STAT-3) pathway in microglia, and assists activated effector T cells in killing tumor cells by blocking elevated immune checkpoint proteins in malignant glioma. Eventually, as adjuvant therapy, the rationally designed nano-immuno-synergist hinders malignant glioma progression and recurrence with or without temozolomide. The work demonstrates the feasibility of a nano-formulation for microglia-based immunotherapy, which may provide a new direction for the treatment of brain tumors.
Collapse
Affiliation(s)
- Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jian Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhen Qiao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
5
|
Babazadeh SM, Zolfaghari MR, Zargar M, Baesi K, Hosseini SY, Ghaemi A. Interleukin-24-mediated antitumor effects against human glioblastoma via upregulation of P38 MAPK and endogenous TRAIL-induced apoptosis and LC3-II activation-dependent autophagy. BMC Cancer 2023; 23:519. [PMID: 37280571 DOI: 10.1186/s12885-023-11021-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/29/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Melanoma differentiation-associated gene 7 (Mda-7) encodes IL-24, which can induce apoptosis in cancer cells. A novel gene therapy approach to treat deadly brain tumors, recombinant mda-7 adenovirus (Ad/mda-7) efficiently kills glioma cells. In this study, we investigated the factors affecting cell survival and apoptosis and autophagy mechanisms that destroy glioma cells by Ad/IL-24. METHODS Human glioblastoma U87 cell line was exposed to a multiplicity of infections of Ad/IL-24. Antitumor activities of Ad/IL-24 were assessed by cell proliferation (MTT) and lactate dehydrogenase (LDH) release analysis. Using flow cytometry, cell cycle arrest and apoptosis were investigated. Using the ELISA method, the tumor necrosis factor (TNF-α) level was determined as an apoptosis-promoting factor and Survivin level as an anti-apoptotic factor. The expression levels of TNF-related apoptosis inducing ligand(TRAIL) and P38 MAPK genes were assessed by the Reverse transcription-quantitative polymerase chain reaction(RT‑qPCR) method. The expression levels of caspase-3 and protein light chain 3-II (LC3-II) proteins were analyzed by flow cytometry as intervening factors in the processes of apoptosis and autophagy in the cell death signaling pathway, respectively. RESULTS The present findings demonstrated that transduction of IL-24 inhibited cell proliferation and induced cell cycle arrest and cell apoptosis in glioblastoma. Compared with cells of the control groups, Ad/IL24-infected U87 cells exhibited significantly increased elevated caspase-3, and TNF-α levels, while the survivin expression was decreased. TRAIL was shown to be upregulated in tumor cells after Ad/IL-24 infection and studies of the apoptotic cascade regulators indicate that Ad/IL-24 could further enhance the activation of apoptosis through the TNF family of death receptors. In the current study, we demonstrate that P38 MAPK is significantly activated by IL-24 expression. In addition, the overexpression of mda-7/IL-24 in GBM cells induced autophagy, which was triggered by the upregulation of LC3-II. CONCLUSIONS Our study demonstrates the antitumor effect of IL-24 on glioblastoma and may be a promising therapeutic approach for GBM cancer gene therapy.
Collapse
Affiliation(s)
- Seyedeh Maliheh Babazadeh
- Department of Microbiology, Faculty of Basic Science, Qom Branch, Islamic Azad University, Qom, Iran
| | - Mohammad Reza Zolfaghari
- Department of Microbiology, Faculty of Basic Science, Qom Branch, Islamic Azad University, Qom, Iran.
| | - Mohsen Zargar
- Department of Microbiology, Faculty of Basic Science, Qom Branch, Islamic Azad University, Qom, Iran
| | - Kazem Baesi
- Hepatitis and AIDS Department, Pasteur Institute of Iran, Tehran, Iran
| | - Sayed Younes Hosseini
- Bacteriology and Virology Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Ghaemi
- Department of Influenza and other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
6
|
Wang Y, Bastiancich C, Newland B. Injectable local drug delivery systems for glioblastoma: a systematic review and meta-analysis of progress to date. Biomater Sci 2023; 11:1553-1566. [PMID: 36655634 DOI: 10.1039/d2bm01534j] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Glioblastoma (GBM) is an aggressive malignant cancer associated with bleak prognosis and high mortality. The current standard of care for GBM is maximum surgical resection plus radiotherapy and temozolomide (TMZ) chemotherapy. The blood brain barrier (BBB) remains the main obstacle for chemotherapy and severely limits the choice of therapeutic agents. Local treatment allows drugs to circumvent the BBB and reduces systemic side effects. Despite much research effort, to date, no drug delivery system (DDS) designed to be directly injected into brain tumors has been clinically approved, and a systematic overview of the progress in this field, or lack thereof, is missing. In this review, a systematic search of pre-clinical literature was conducted which resulted in 36 original articles on injectable DDS for local treatment of GBM which met the inclusion criteria. A wide range of injectable DDS have been developed and tested pre-clinically which include nanoparticles, liposomes, microspheres, hydrogels and others. meta-Analyses of the included studies showed that, overall, local administration of injectable DDS was beneficial to increase the animal's survival time. Finally, this review summarized the therapeutic effect after local treatment and discussed the shortcomings of the experimental setting in in vivo studies.
Collapse
Affiliation(s)
- Yu Wang
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| | - Chiara Bastiancich
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13344 Marseille, France.,Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| |
Collapse
|
7
|
Wolin IAV, Nascimento APM, Seeger R, Poluceno GG, Zanotto-Filho A, Nedel CB, Tasca CI, Correia SEG, Oliveira MV, Pinto-Junior VR, Osterne VJS, Nascimento KS, Cavada BS, Leal RB. The lectin DrfL inhibits cell migration, adhesion and triggers autophagy-dependent cell death in glioma cells. Glycoconj J 2023; 40:47-67. [PMID: 36522582 DOI: 10.1007/s10719-022-10095-3] [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: 03/11/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of glioma, displaying atypical glycosylation pattern that may modulate signaling pathways involved in tumorigenesis. Lectins are glycan binding proteins with antitumor properties. The present study was designed to evaluate the antitumor capacity of the Dioclea reflexa lectin (DrfL) on glioma cell cultures. Our results demonstrated that DrfL induced morphological changes and cytotoxic effects in glioma cell cultures of C6, U-87MG and GBM1 cell lines. The action of DrfL was dependent upon interaction with glycans, and required a carbohydrate recognition domain (CRD), and the cytotoxic effect was apparently selective for tumor cells, not altering viability and morphology of primary astrocytes. DrfL inhibited tumor cell migration, adhesion, proliferation and survival, and these effects were accompanied by activation of p38MAPK and JNK (p46/54), along with inhibition of Akt and ERK1/2. DrfL also upregulated pro-apoptotic (BNIP3 and PUMA) and autophagic proteins (Atg5 and LC3 cleavage) in GBM cells. Noteworthy, inhibition of autophagy and caspase-8 were both able to attenuate cell death in GBM cells treated with DrfL. Our results indicate that DrfL cytotoxicity against GBM involves modulation of cell pathways, including MAPKs and Akt, which are associated with autophagy and caspase-8 dependent cell death.
Collapse
Affiliation(s)
- Ingrid A V Wolin
- Departamento de Bioquímica e Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Ana Paula M Nascimento
- Departamento de Bioquímica e Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Rodrigo Seeger
- Departamento de Bioquímica e Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Gabriela G Poluceno
- Departamento de Bioquímica e Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Alfeu Zanotto-Filho
- Departamento de Farmacologia e Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Claudia B Nedel
- Departamento de Biologia Celular, Embriologia e Genética, Programa Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Carla I Tasca
- Departamento de Bioquímica e Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Sarah Elizabeth Gomes Correia
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, CEP, 60020-181, BioMolLab, Fortaleza, Ceará, Brazil
| | - Messias Vital Oliveira
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, CEP, 60020-181, BioMolLab, Fortaleza, Ceará, Brazil
| | - Vanir Reis Pinto-Junior
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, CEP, 60020-181, BioMolLab, Fortaleza, Ceará, Brazil
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, Ceará, CEP, 60020-181, Brazil
| | - Vinicius Jose Silva Osterne
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, CEP, 60020-181, BioMolLab, Fortaleza, Ceará, Brazil
| | - Kyria Santiago Nascimento
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, CEP, 60020-181, BioMolLab, Fortaleza, Ceará, Brazil
| | - Benildo Sousa Cavada
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, CEP, 60020-181, BioMolLab, Fortaleza, Ceará, Brazil
| | - Rodrigo Bainy Leal
- Departamento de Bioquímica e Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Florianópolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
8
|
Muresan P, McCrorie P, Smith F, Vasey C, Taresco V, Scurr DJ, Kern S, Smith S, Gershkovich P, Rahman R, Marlow M. Development of nanoparticle loaded microneedles for drug delivery to a brain tumour resection site. Eur J Pharm Biopharm 2023; 182:53-61. [PMID: 36435313 DOI: 10.1016/j.ejpb.2022.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022]
Abstract
Systemic drug delivery to the central nervous system (CNS) has been historically impeded by the presence of the blood brain barrier rendering many therapies inefficacious to any cancer cells residing within the brain. Therefore, local drug delivery systems are being developed to overcome this shortfall. Here we have manufactured polymeric microneedle (MN) patches, which can be anchored within a resection cavity site following surgical removal of a tumour such as isocitrate dehydrogenase wild type glioblastoma (GBM). These MN patches have been loaded with polymer coated nanoparticles (NPs) containing cannabidiol (CBD) or olaparib (OLA) and applied to an in vitro brain simulant and ex vivo rat brain tissue to assess drug release and distance of penetration. MN patches loaded with methylene blue dye were placed into a cavity of 0.6 % agarose to simulate brain tissue. The results showed that clear channels were generated by the MNs and the dye spread laterally throughout the agarose. When loaded with CBD-NPs, the agarose showed a CBD concentration of 12.5 µg/g at 0.5 cm from the MN insertion site. Furthermore, high performance liquid chromatography of ex vivo brain tissue following CBD-NP/MN patch insertion showed successful delivery of 59.6 µg/g into the brain tissue. Similarly, OLA-NP loaded MN patches showed delivery of 5.2 µg/g OLA into agarose gel at 0.5 cm distance from the insertion site. Orbitrap secondary ion mass spectrometry (OrbiSIMS) analysis confirmed the presence of OLA and the MN patch at up to 6 mm away from the insertion site following its application to a rat brain hemisphere. This data has provided insight into the capabilities and versatility of MN patches for use in local brain drug delivery, giving promise for future research.
Collapse
Affiliation(s)
- Paula Muresan
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Phoebe McCrorie
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, NG7 2RD, UK
| | - Fiona Smith
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Catherine Vasey
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Vincenzo Taresco
- School of Chemistry, University of Nottingham, Nottingham NG7 2RD, UK
| | - David J Scurr
- Nanoscale and Microscale Research Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Stefanie Kern
- Nanoscale and Microscale Research Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Stuart Smith
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, NG7 2RD, UK
| | - Pavel Gershkovich
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Ruman Rahman
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, NG7 2RD, UK
| | - Maria Marlow
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
9
|
Wang M, Malfanti A, Bastiancich C, Préat V. Synergistic effect of doxorubicin lauroyl hydrazone derivative delivered by α-tocopherol succinate micelles for the treatment of glioblastoma. Int J Pharm X 2022; 5:100147. [PMID: 36620521 PMCID: PMC9813532 DOI: 10.1016/j.ijpx.2022.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
We hypothesized that tocopherol succinate (TOS) and D-α-tocopherol polyethylene2000 succinate (TPGS2000) micelles could work as a drug delivery system while enhancing the anti-cancer efficacy of doxorubicin lauryl hydrazone derivative (DOXC12) for the treatment of glioblastoma. The DOXC12-TOS-TPGS2000 micelles were formulated with synthesized DOXC12 and TPGS2000. They showed a high drug loading of hydrophobic DOXC12 (29%), a size of <100 nm and a pH sensitive drug release behaviour. In vitro, fast uptake of DOXC12-TOS-TPGS2000 micelles by GL261 cells was observed. For cytotoxicity, DOXC12-TOS-TPGS2000 micelles were evaluated on two glioblastoma cell lines and showed synergism between DOXC12 and TOS-TPGS2000. The higher cytotoxicity of DOXC12-TOS-TPGS2000 micelles was mainly caused by necrosis. The DOXC12-TOS-TPGS2000 micelles seem to be a promising delivery system for enhancing the anticancer efficacy of doxorubicin in glioblastoma (GBM).
Collapse
Affiliation(s)
- Mingchao Wang
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Alessio Malfanti
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Chiara Bastiancich
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France,Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Véronique Préat
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium,Corresponding author.
| |
Collapse
|
10
|
Tirgar F, Azizi Z, Hosseindoost S, Hadjighassem M. Preclinical gene therapy in glioblastoma multiforme: Using olfactory ensheathing cells containing a suicide gene. Life Sci 2022; 311:121132. [DOI: 10.1016/j.lfs.2022.121132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
|
11
|
Siddique R, Abideen SA, Nabi G, Awan FM, Noor Khan S, Ullah F, Khan S, Xue M. Fibroblast growth factor 2 is a druggable target against glioblastoma: A computational investigation. Front Chem 2022; 10:1071929. [PMID: 36505741 PMCID: PMC9732544 DOI: 10.3389/fchem.2022.1071929] [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: 10/20/2022] [Accepted: 11/08/2022] [Indexed: 11/26/2022] Open
Abstract
Fibroblast growth factor 2 (FGF2) is a key player in cancer and tissue homeostasis and regulates renewal of several stem cell types. The FGF2 role in malignant glioma is proven and tagged FGF2, a novel druggable target, is used for developing potent drugs against glioblastoma. In this study, Asinex 51412372, Asinex 51217461, and Asinex 51216586 were filtered to show the best binding affinity for FGF2 with binding energy scores of -8.3 kcal/mol, -8.2 kcal/mol, and -7.8 kcal/mol, respectively. The compounds showed chemical interactions with several vital residues of FGF2 along the compound length. The noticeable residues that interacted with the compounds were Arg15, Asp23, Arg63, and Gln105. In dynamic investigation in solution, the FGF2 reported unstable dynamics in the first 100 ns and gained structural equilibrium in the second phase of 100 ns. The maximum root mean square deviation (RMSD) value touched by the systems is 3 Å. Similarly, the residue flexibility of FGF2 in the presence of compounds was within a stable range and is compact along the simulation time length. The compounds showed robust atomic-level stable energies with FGF2, which are dominated by both van der Waals and electrostatic interactions. The net binding energy of systems varies between -40 kcal/mol and -86 kcal/mol, suggesting the formation of strong intermolecular docked complexes. The drug-likeness and pharmacokinetic properties also pointed toward good structures that are not toxic, have high gastric absorption, showed good distribution, and readily excreted from the body. In summary, the predicted compounds in this study might be ideal hits that might be further optimized for structure and activity during experimental studies.
Collapse
Affiliation(s)
- Rabeea Siddique
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou Uiversity, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Syed Ainul Abideen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ghulam Nabi
- Institute of Nature Conservation, Polish Academy of Sciences, KraKow, Poland
| | - Faryal Mehwish Awan
- Department of Medial Lab Technology, The University of Haripur, Haripur, Pakistan
| | - Sadiq Noor Khan
- Department of Medial Lab Technology, The University of Haripur, Haripur, Pakistan
| | - Fawad Ullah
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Centre of Biotechnology and Microbiology, University of Peshawar, Haripur, Pakistan
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou Uiversity, Zhengzhou, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Department of Medial Lab Technology, The University of Haripur, Haripur, Pakistan
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou Uiversity, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| |
Collapse
|
12
|
Ruiz-Molina D, Mao X, Alfonso-Triguero P, Lorenzo J, Bruna J, Yuste VJ, Candiota AP, Novio F. Advances in Preclinical/Clinical Glioblastoma Treatment: Can Nanoparticles Be of Help? Cancers (Basel) 2022; 14:4960. [PMID: 36230883 PMCID: PMC9563739 DOI: 10.3390/cancers14194960] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GB) is the most aggressive and frequent primary malignant tumor in the central nervous system (CNS), with unsatisfactory and challenging treatment nowadays. Current standard of care includes surgical resection followed by chemotherapy and radiotherapy. However, these treatments do not much improve the overall survival of GB patients, which is still below two years (the 5-year survival rate is below 7%). Despite various approaches having been followed to increase the release of anticancer drugs into the brain, few of them demonstrated a significant success, as the blood brain barrier (BBB) still restricts its uptake, thus limiting the therapeutic options. Therefore, enormous efforts are being devoted to the development of novel nanomedicines with the ability to cross the BBB and specifically target the cancer cells. In this context, the use of nanoparticles represents a promising non-invasive route, allowing to evade BBB and reducing systemic concentration of drugs and, hence, side effects. In this review, we revise with a critical view the different families of nanoparticles and approaches followed so far with this aim.
Collapse
Affiliation(s)
- Daniel Ruiz-Molina
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Xiaoman Mao
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Paula Alfonso-Triguero
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Julia Lorenzo
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Jordi Bruna
- Neuro-Oncology Unit, Bellvitge University Hospital-ICO (IDIBELL), Avinguda de la Gran Via de l’Hospitalet, 199-203, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Victor J. Yuste
- Instituto de Neurociencias. Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Fernando Novio
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Departament de Química, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
13
|
Long non-coding RNA LINC01018 inhibits human glioma cell proliferation and metastasis by directly targeting miRNA-182-5p. J Neurooncol 2022; 160:67-78. [PMID: 36094613 DOI: 10.1007/s11060-022-04113-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/04/2022] [Indexed: 10/14/2022]
Abstract
AIM Accumulating evidence suggests that lncRNAs are potential biomarkers and key regulators of tumor development and progression. However, the precise function of most lncRNAs in glioma remains unknown. In this study, we explored the role of long intergenic non-protein coding RNA 1018 (LINC01018) in human glioma. METHODS Expression levels of LINC01018 and miR-182-5p in clinical glioma tissues and cell lines were detected by quantitative real-time PCR (qRT-PCR). Cell proliferation, migration, and invasion were determined by Cell Counting Kit-8 (CCK-8) assay and Transwell assay. Epithelial-mesenchymal transition (EMT) related proteins were measured by Western blotting. Direct relationship between LINC01018 and miR-182-5p was tested by dual-luciferase reporter assay, RNA immunoprecipitation assay (RIP), and rescue assays. Lastly, bioinformatics analyses were conducted to predict the downstream factors of LINC01018/miR-182-5p axis in glioma. RESULTS LINC01018 was significantly down-regulated in glioma tissues and cell lines. Overexpression of LINC01018 dramatically inhibited cell proliferation, migration, and invasion and reverse EMT process in glioma. LINC01018 directly target to miR-182-5p. Forced up-regulation of miR-182-5p reversed the inhibitory effects on proliferative and metastatic abilities of glioma cells with LINC01018 overexpression. Lastly, the bioinformatics analyses revealed that LINC01018/miR-182-5p axis mediated a cluster of downstream genes (ADRA2C, RAB6B, RAB27B, RAPGEF5, STEAP2, TAGLN3, and UNC13C), which were potential key factors in the development of glioma. CONCLUSION LINC01018 inhibits cell proliferation and metastasis in human glioma by targeting miR-182-5p, and should be considered as a potential therapeutic target in this cancer.
Collapse
|
14
|
Elmtalab S, Abedi I, Alirezaei Z, Choopan Dastjerdi MH, Geraily G, Karimi AH. Semi-experimental assessment of neutron equivalent dose and secondary cancer risk for off-field organs in glioma patients undergoing 18-MV radiotherapy. PLoS One 2022; 17:e0271028. [PMID: 35905102 PMCID: PMC9337694 DOI: 10.1371/journal.pone.0271028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Neutron contamination as a source of out-of-field dose in radiotherapy is still of concern. High-energy treatment photons have the potential to overcome the binding energy of neutrons inside the nuclei. Fast neutrons emitting from the accelerator head can directly reach the patient’s bed. Considering that modern radiotherapy techniques can increase patient survival, concerns about unwanted doses and the lifetime risk of fatal cancer remain strong or even more prominent, especially in young adult patients. The current study addressed these concerns by quantifying the dose and risk of fatal cancer due to photo-neutrons for glioma patients undergoing 18-MV radiotherapy. In this study, an NRD model rem-meter detector was used to measure neutron ambient dose equivalent, H*(10), at the patient table. Then, the neutron equivalent dose received by each organ was estimated concerning the depth of each organ and by applying depth dose corrections to the measured H*(10). Finally, the effective dose and risk of secondary cancer were determined using NCRP 116 coefficients. Evidence revealed that among all organs, the breast (0.62 mSv/Gy) and gonads (0.58 mSv/Gy) are at risk of photoneutrons more than the other organs in such treatments. The neutron effective dose in the 18-MV conventional radiotherapy of the brain was 13.36 mSv. Among all organs, gonads (6.96 mSv), thyroid (1.86 mSv), and breasts (1.86 mSv) had more contribution to the effective dose, respectively. The total secondary cancer risk was estimated as 281.4 cases (per 1 million persons). The highest risk was related to the breast and gonads with 74.4 and, 34.8 cases per 1 million persons, respectively. Therefore, it is recommended that to prevent late complications (secondary cancer and genetic effects), these organs should be shielded from photoneutrons. This procedure not only improves the quality of the patient’s personal life but also the healthy childbearing in the community.
Collapse
Affiliation(s)
- Soheil Elmtalab
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Iraj Abedi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Alirezaei
- School of Paramedicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad Hossein Choopan Dastjerdi
- Reactor and Nuclear Safety Research School, Nuclear Science and Technology Research Institute, Isfahan, Iran
- * E-mail: (MHCD); (GG); (AHK)
| | - Ghazale Geraily
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- * E-mail: (MHCD); (GG); (AHK)
| | - Amir Hossein Karimi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- * E-mail: (MHCD); (GG); (AHK)
| |
Collapse
|
15
|
Cao X, Li S, Chen W, Lu H, Ye L, Min Z, Sun S, Teng C, Yin H, Zhang Q, He W, Wang X, Lv W, Lv L, Xin H. Multifunctional Hybrid Hydrogel System Enhanced the Therapeutic Efficacy of Treatments for Postoperative Glioma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:27623-27633. [PMID: 35673881 DOI: 10.1021/acsami.2c05147] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Glioma is the most lethal brain tumor with a poor prognosis, and a combination of multiple therapeutic strategies is critical for postoperative glioma treatment. Herein, a multifunctional hybrid hydrogel system (designated as CP&CL@RNPPTX-Gel) was developed for local treatment of postoperative glioma. The system was composed of self-illuminating chlorin e6 (Ce6) conjugated with luminol molecule (CL)-loaded glioma-targeting paclitaxel prodrug nanoparticles and copper peroxide nanodots (CP NDs) coembedded into a three-dimensional thermosensitive hydroxypropyl chitin hydrogel frame. After injection of CP&CL@RNPPTX-Gel into the cavity of postoperative glioma, the solution could be cross-linked into the gel as a drug reservoir under body temperature stimulation. Then, the sustained-released CP NDs decomposed into Cu2+ and H2O2 in the acidic microenvironment of the glioma cells to exert chemodynamic therapy (CDT). Meanwhile, Cu2+ could catalyze the self-luminescence of CL to induce photodynamic therapy (PDT) without external excitation light. Moreover, paclitaxel prodrug nanoparticles degraded into paclitaxel to restrain residual glioma cells in response to intracellular reduced glutathione (GSH). The in vitro and in vivo results showed that CP&CL@RNPPTX-Gel had great potential as a multifunctional hybrid hydrogel system with remarkable therapeutic effects for postoperative glioma treatment via a combination of chemotherapy, CDT, and PDT.
Collapse
Affiliation(s)
- Xiang Cao
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Shengnan Li
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wenli Chen
- Department of Rehabilitation Medicine, ZhongDa Hospital of Southeast University, Nanjing 210009, China
| | - Hongdan Lu
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lu Ye
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zhiyi Min
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Shanbo Sun
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chuanhui Teng
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Haoyuan Yin
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qi Zhang
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Weichong He
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiuzhen Wang
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wei Lv
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Wuxi 214400, China
| | - Lingyan Lv
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Wuxi 214400, China
| | - Hongliang Xin
- Department of Pharmaceutics and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
16
|
Application of Dual-Enhanced Surface-Enhanced Raman Scattering Probe Technology in the Diagnosis of Tumor Cells in Vitro. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113582. [PMID: 35684522 PMCID: PMC9182129 DOI: 10.3390/molecules27113582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022]
Abstract
With the development of precision medicine, antigen/antibody-targeted therapy has brought great hope to tumor patients; however, the migration of tumor cells, especially a small number of cells flowing into blood or other tissues, remains a clinical challenge. In particular, it is difficult to use functional gold nanomaterials for targeted clinical tumor diagnosis while simultaneously obtaining stable and highly sensitive Raman signals. Therefore, we developed a detection method for functional Au Nanostars (AuNSs) with dual signal enhancement that can specifically track location and obtain high-intensity surface-enhanced Raman scattering (SERS) signals. First, AuNSs with specific optical properties were synthesized and functionalized. The Raman dye 4-mercapto-hydroxybenzoic acid and polyethylene glycol were coupled with the tumor marker, epidermal growth factor receptor, to obtain the targeted SERS probes. In addition, a detection chip was prepared for Raman detection with physical enhancement, exhibiting a 40-times higher signal intensity than that of quartz glass. This study combines physical enhancement and SERS enhancement technologies to achieve dual enhancement, enabling the detection of a highly sensitive and stable Raman signal; this has potential clinical value for antigen/antibody-targeted tumor diagnosis and treatment.
Collapse
|
17
|
Lambride C, Vavourakis V, Stylianopoulos T. Convection-Enhanced Delivery In Silico Study for Brain Cancer Treatment. Front Bioeng Biotechnol 2022; 10:867552. [PMID: 35694227 PMCID: PMC9177080 DOI: 10.3389/fbioe.2022.867552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
Brain cancer therapy remains a formidable challenge in oncology. Convection-enhanced delivery (CED) is an innovative and promising local drug delivery method for the treatment of brain cancer, overcoming the challenges of the systemic delivery of drugs to the brain. To improve our understanding about CED efficacy and drug transport, we present an in silico methodology for brain cancer CED treatment simulation. To achieve this, a three-dimensional finite element formulation is utilized which employs a brain model representation from clinical imaging data and is used to predict the drug deposition in CED regimes. The model encompasses biofluid dynamics and the transport of drugs in the brain parenchyma. Drug distribution is studied under various patho-physiological conditions of the tumor, in terms of tumor vessel wall pore size and tumor tissue hydraulic conductivity as well as for drugs of various sizes, spanning from small molecules to nanoparticles. Through a parametric study, our contribution reports the impact of the size of the vascular wall pores and that of the therapeutic agent on drug distribution during and after CED. The in silico findings provide useful insights of the spatio-temporal distribution and average drug concentration in the tumor towards an effective treatment of brain cancer.
Collapse
Affiliation(s)
- Chryso Lambride
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Vasileios Vavourakis
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- *Correspondence: Vasileios Vavourakis, ; Triantafyllos Stylianopoulos,
| | - Triantafyllos Stylianopoulos
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
- *Correspondence: Vasileios Vavourakis, ; Triantafyllos Stylianopoulos,
| |
Collapse
|
18
|
Elmtalab S, Shanei A, Choopan Dastjerdi MH, Brkić H, Abedi I, Amouheidari A. DETERMINATION OF THE NEUTRON CONTAMINATION DURING BRAIN RADIOTHERAPY USING A MODERATED-BORON TRIFLUORIDE DETECTOR AND THE MCNP MONTE CARLO CODE. RADIATION PROTECTION DOSIMETRY 2022; 198:129-138. [PMID: 35137234 DOI: 10.1093/rpd/ncac001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 12/27/2021] [Accepted: 01/03/2022] [Indexed: 11/12/2022]
Abstract
This study aimed to determine the neutron dose equivalent to the thyroid gland and eye lens in brain tumor radiation therapy with 15- and 18-MV three-dimensional conformal methods (3D-CRT). A Monte Carlo simulation was performed using the Monte Carlo N-particle transport code to calculate neutron fluence and ambient dose equivalent (H*(10)). Afterward, these parameters were measured using a model NRD roentgen equivalent in man (REM) neutron detector (Thermo Electron Corporation, USA) equipped with Eberline's ASP-2e rate meter. Finally, the organ neutron dose equivalent was obtained by applying depth corrections to the measured ambient dose equivalent at the distance of the organ center from the central beam axis. The ratio of the out-of-field photon dose equivalent, measured previously, to the neutron dose equivalent in the eye lens was high due to its proximity to the radiation field. In contrast, this ratio remained unexpectedly high in the thyroid gland that is far from the central beam axis (about 15 cm). The calculated neutron parameters agreed with the measurements. The present study findings indicate that external field photon dose is the main source of thyroid gland biological effects in radiotherapy of brain tumors. In addition, it is appropriate to apply the model NRD REM neutron detector for measuring neutron contamination from high-energy linear accelerators inside and outside the treatment field.
Collapse
Affiliation(s)
- Soheil Elmtalab
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Shanei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hrvoje Brkić
- Department of biophysics and radiology, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
- Department of biophysics, biology and chemistry, Faculty of dental medicine and health, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Iraj Abedi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Amouheidari
- Department of Radiation Oncology, Isfahan Milad General Hospital, Isfahan, Iran
| |
Collapse
|
19
|
Soleimanbeigi M, Dousti F, Hassanzadeh F, Mirian M, Varshosaz J, Kasesaz Y, Rostami M. Boron Phenyl Alanine Targeted Chitosan-PNIPAAm Core-Shell Thermo-Responsive Nanoparticles; Boosting Drug Delivery to Glioblastoma in BNCT. Drug Dev Ind Pharm 2022; 47:1607-1623. [DOI: 10.1080/03639045.2022.2032132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Monireh Soleimanbeigi
- Master Student of Medicinal Chemistry, Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Dousti
- Master Student of Medicinal Chemistry, Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Science, Isfahan, Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Centre and Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Science, Isfahan, Iran
| | - Yaser Kasesaz
- Reactor and Nuclear Safety Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, Iran
| | - Mahboubeh Rostami
- Novel Drug Delivery Systems Research Centre and Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
20
|
Rationally designed drug delivery systems for the local treatment of resected glioblastoma. Adv Drug Deliv Rev 2021; 177:113951. [PMID: 34461201 DOI: 10.1016/j.addr.2021.113951] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023]
Abstract
Glioblastoma (GBM) is a particularly aggressive brain cancer associated with high recurrence and poor prognosis. The standard of care, surgical resection followed by concomitant radio- and chemotherapy, leads to low survival rates. The local delivery of active agents within the tumor resection cavity has emerged as an attractive means to initiate oncological treatment immediately post-surgery. This complementary approach bypasses the blood-brain barrier, increases the local concentration at the tumor site while reducing or avoiding systemic side effects. This review will provide a global overview on the local treatment for GBM with an emphasis on the lessons learned from past clinical trials. The main parameters to be considered to rationally design fit-of-purpose biomaterials and develop drug delivery systems for local administration in the GBM resection cavity to prevent the tumor recurrence will be described. The intracavitary local treatment of GBM should i) use materials that facilitate translation to the clinic; ii) be characterized by easy GMP effective scaling up and easy-handling application by the neurosurgeons; iii) be adaptable to fill the tumor-resected niche, mold to the resection cavity or adhere to the exposed brain parenchyma; iv) be biocompatible and possess mechanical properties compatible with the brain; v) deliver a therapeutic dose of rationally-designed or repurposed drug compound(s) into the GBM infiltrative margin. Proof of concept with high translational potential will be provided. Finally, future perspectives to facilitate the clinical translation of the local perisurgical treatment of GBM will be discussed.
Collapse
|
21
|
Bastiancich C, Bozzato E, Henley I, Newland B. Does local drug delivery still hold therapeutic promise for brain cancer? A systematic review. J Control Release 2021; 337:296-305. [PMID: 34298055 DOI: 10.1016/j.jconrel.2021.07.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Despite the gold standard treatment combining surgical resection, radiation and adjuvant plus concomitant chemotherapy with the alkylating agent temozolomide (TMZ), the prognosis remains poor (5-year survival rate < 10%). Over the last three decades, a vast array of drug delivery systems (DDS) have been developed for the local treatment of GBM, with the majority of the characterization being undertaken in pre-clinical models. We aimed to gain an overview of the potential efficacy of such local delivery systems in comparison to the systemic drug administration. METHODS In this paper, a systematic search of Pubmed, Web of Science, and Scopus was performed using pre-determined search terms. Studies were assessed for eligibility based on specific inclusion and exclusion criteria. A total of fifteen publications were included for analysis of local vs systemic group median survival, tumor volume and adverse events, with five brought forward for a meta-analysis. RESULTS The majority of studies showed local delivery to be more efficacious than systemic administration, regardless of the drug, animal model, type of DDS used, or duration of the study. The meta-analysis also showed that the mean difference between median survival ratios was statistically significantly in favor of local delivery. CONCLUSION Preclinical evidence shows that there is a firm rationale for further developing DDS for local therapeutic delivery to GBM and other brain cancers.
Collapse
Affiliation(s)
- C Bastiancich
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
| | - E Bozzato
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - I Henley
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - B Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
22
|
Li A, Zhang T, Huang T, Lin R, Mu J, Su Y, Sun H, Jiang X, Wu H, Xu D, Cao H, Sun X, Ling D, Gao J. Iron Oxide Nanoparticles Promote Cx43-Overexpression of Mesenchymal Stem Cells for Efficient Suicide Gene Therapy during Glioma Treatment. Theranostics 2021; 11:8254-8269. [PMID: 34373740 PMCID: PMC8344020 DOI: 10.7150/thno.60160] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) have been applied as a promising vehicle for tumour-targeted delivery of suicide genes in the herpes simplex virus thymidine kinase (HSV-tk)/ganciclovir (GCV) suicide gene therapy against malignant gliomas. The efficiency of this strategy is largely dependent on the bystander effect, which relies on high suicide gene expression levels and efficient transportation of activated GCV towards glioma cells. However, up to now, the methods to enhance the bystander effect of this strategy in an efficient and safe way are still lacking and new approaches to improve this therapeutic strategy are required. Methods: In this study, MSCs were gene transfected using magnetosome-like ferrimagnetic iron oxide nanochains (MFIONs) to highly express HSV-tk. Both the suicide and bystander effects of HSV-tk expressed MSCs (MSCs-tk) were quantitatively evaluated. Connexin 43 (Cx43) expression by MSCs and glioma cells was measured under different treatments. Intercellular communication between MSCs and C6 glioma cells was examined using a dye transfer assay. Glioma tropism and the bio-distribution of MSCs-tk were observed. Anti-tumour activity was investigated in the orthotopic glioma of rats after intravenous administration of MSCs-tk followed by intraperitoneal injection of GCV. Results: Gene transfection using MFIONs achieved sufficient expression of HSV-tk and triggered Cx43 overexpression in MSCs. These Cx43 overexpressing MSCs promoted gap junction intercellular communication (GJIC) between MSCs and glioma cells, resulting in significantly inhibited growth of glioma through an improved bystander effect. Outstanding tumour targeting and significantly prolonged survival with decreased tumour size were observed after the treatment using MFION-transfected MSCs in glioma model rats. Conclusion: Our results show that iron oxide nanoparticles have the potential to improve the suicide gene expression levels of transfected MSCs, while promoting the GJIC formation between MSCs and tumour cells, which enhances the sensitivity of glioma cells to HSV-tk/GCV suicide gene therapy.
Collapse
Affiliation(s)
- Ai Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Ting Huang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ruyi Lin
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiafu Mu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanqin Su
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hao Sun
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinchi Jiang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Honghui Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Donghang Xu
- Department of Pharmacy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaoyi Sun
- Department of Pharmacy, Zhejiang University City College, Hangzhou 310015, China
| | - Daishun Ling
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
23
|
Retinal toxicities of systemic anticancer drugs. Surv Ophthalmol 2021; 67:97-148. [PMID: 34048859 DOI: 10.1016/j.survophthal.2021.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 01/07/2023]
Abstract
Newer anticancer drugs have revolutionized cancer treatment in the last decade, but conventional chemotherapy still occupies a central position in many cancers, with combination therapy and newer methods of delivery increasing their efficacy while minimizing toxicities. We discuss the retinal toxicities of anticancer drugs with an emphasis on the mechanism of toxicity. Uveitis is seen with the use of v-raf murine sarcoma viral oncogene homolog B editing anticancer inhibitors as well as immunotherapy. Most of the cases are mild with only anterior uveitis, but severe cases of posterior uveitis, panuveitis, and Vogt-Koyanagi-Harada-like disease may also occur. In the retina, a transient neurosensory detachment is observed in almost all patients on mitogen-activated protein kinase kinase (MEK) inhibitors. Microvasculopathy is often seen with interferon α, but vascular occlusion is a more serious toxicity caused by interferon α and MEK inhibitors. Crystalline retinopathy with or without macular edema may occur with tamoxifen; however, even asymptomatic patients may develop cavitatory spaces seen on optical coherence tomography. A unique macular edema with angiographic silence is characteristic of taxanes. Delayed dark adaptation has been observed with fenretinide. Interestingly, this drug is finding potential application in Stargardt disease and age-related macular degeneration.
Collapse
|
24
|
Maleki H, Hosseini Najafabadi MR, Webster TJ, Hadjighassem MR, Sadroddiny E, Ghanbari H, Khosravani M, Adabi M. Effect of Paclitaxel/etoposide co-loaded polymeric nanoparticles on tumor size and survival rate in a rat model of glioblastoma. Int J Pharm 2021; 604:120722. [PMID: 34022255 DOI: 10.1016/j.ijpharm.2021.120722] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/18/2022]
Abstract
The aim of this work is to co-load paclitaxel (PTX) and etoposide (ETP) in methoxy poly(ethylene glycol)-poly(lactic-co-glycolic acid) nanoparticles (mPEG-PLGA NPs) to overcome pharmacokinetics and physiological limitations and enhance therapeutic efficacy for treating intracranial glioblastoma. Both drugs were loaded into mPEG-PLGA NPs by a nano-precipitation method. The resultant NPs demonstrated an enhanced cytotoxic effect indicated by lower IC50 values and augmented cell apoptosis to U87 and C6 glioma cell lines compared to both free drugs. Additionally, blood compatibility assays showed that the PTX/ETP co-loaded mPEG-PLGA NPs did not induce blood hemolysis, blood clotting, or platelet aggregation. In vivo anti-glioma efficacy evaluation in rats bearingintracranialC6glioma revealed a superior anti-glioma activity for the treatment with PTX/ETP co-loaded mPEG-PLGA NPs compared to other formulations, particularly a significantly longer median survival, 76 days compared to 36 days for free PTX and 37 days for free ETP treatment, respectively, and higher tumor regression, proved by magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Hassan Maleki
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Hosseini Najafabadi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Thomas J Webster
- Chemical Engineering Department, Northeastern University, Boston, MA 02115, USA
| | - Mahmoud Reza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Curry RN, Glasgow SM. The Role of Neurodevelopmental Pathways in Brain Tumors. Front Cell Dev Biol 2021; 9:659055. [PMID: 34012965 PMCID: PMC8127784 DOI: 10.3389/fcell.2021.659055] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Disruptions to developmental cell signaling pathways and transcriptional cascades have been implicated in tumor initiation, maintenance and progression. Resurgence of aberrant neurodevelopmental programs in the context of brain tumors highlights the numerous parallels that exist between developmental and oncologic mechanisms. A deeper understanding of how dysregulated developmental factors contribute to brain tumor oncogenesis and disease progression will help to identify potential therapeutic targets for these malignancies. In this review, we summarize the current literature concerning developmental signaling cascades and neurodevelopmentally-regulated transcriptional programs. We also examine their respective contributions towards tumor initiation, maintenance, and progression in both pediatric and adult brain tumors and highlight relevant differentiation therapies and putative candidates for prospective treatments.
Collapse
Affiliation(s)
- Rachel N. Curry
- Department of Neuroscience, Baylor College of Medicine, Center for Cell and Gene Therapy, Houston, TX, United States
- Integrative Molecular and Biomedical Sciences, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX, United States
| | - Stacey M. Glasgow
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
- Neurosciences Graduate Program, University of California, San Diego, San Diego, CA, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
26
|
McCrorie P, Vasey CE, Smith SJ, Marlow M, Alexander C, Rahman R. Biomedical engineering approaches to enhance therapeutic delivery for malignant glioma. J Control Release 2020; 328:917-931. [DOI: 10.1016/j.jconrel.2020.11.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/23/2022]
|
27
|
Unbehau R, Luthringer-Feyerabend BJC, Willumeit-Römer R. The impact of brain cell metabolism and extracellular matrix on magnesium degradation. Acta Biomater 2020; 116:426-437. [PMID: 32890748 DOI: 10.1016/j.actbio.2020.08.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/13/2020] [Accepted: 08/28/2020] [Indexed: 01/09/2023]
Abstract
Due to its degradability, magnesium holds potential for the application as a base material for local treatment systems. Particularly for the therapy of severe brain-related diseases, local approaches are advantageous. To confirm the suitability of magnesium as a material for neural implants, information on the interaction of brain cells with magnesium is essential. Initial steps of such an evaluation need to include not only cytocompatibility tests but also the analysis of the in vitro material degradation to predict in vivo material performance. Considering the sensitivity and functional importance of neural tissue, an in-depth understanding of the processes involved is of particular relevance. Here, we investigate the influence of four different brain cell types and fibroblasts on magnesium degradation in direct material contact. Our findings indicate cell type as well as cell density-dependent degradation behavior. Metabolic activity (lactate content) appears to be crucial for degradation promotion. Extracellular matrix composition, distribution, and matrix/cell ratios are analyzed to elucidate the cell-material interactions further. Statement of Significance Thanks to their degradability, magnesium (Mg)-based materials could be promising biomaterials for local ion or even drug delivery strategies for the treatment of severe brain-related diseases. To confirm the suitability of Mg as a neural implant material, information on the interaction of brain cells with Mg is essential. Initial steps of such an evaluation need to include cytocompatibility tests and the analysis of the in vitro material degradation to predict in vivo material performance. The present study provides data on the influence of different brain cell types on Mg degradation in direct material contact. Our findings indicate cell type and cell density-dependent degradation behavior, and elucidate the role of cell metabolites and extracellular matrix molecules in the underlying degradation mechanisms.
Collapse
Affiliation(s)
- Reneé Unbehau
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Max-Planck-Str. 1, 21502 Geesthacht, Germany
| | - Bérengère J C Luthringer-Feyerabend
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Max-Planck-Str. 1, 21502 Geesthacht, Germany.
| | - Regine Willumeit-Römer
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Max-Planck-Str. 1, 21502 Geesthacht, Germany
| |
Collapse
|
28
|
Liang P, Wang G, Liu X, Wang Z, Wang J, Gao W. Spatiotemporal combination of thermosensitive polypeptide fused interferon and temozolomide for post-surgical glioblastoma immunochemotherapy. Biomaterials 2020; 264:120447. [PMID: 33069137 DOI: 10.1016/j.biomaterials.2020.120447] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
Cancer recurrence post surgical resection is of considerable challenge especially in glioblastoma (GBM) therapy. Herein, we demonstrate that interferon-alpha (IFN) fused to a body temperature-sensitive elastin-like polypeptide (IFN-ELP(V)) formed a depot in situ when injected into GBM resection cavity in a mouse brain orthotopic model of GBM. Notably, IFN-ELP(V) in the depot showed a zero-order release kinetics, resulting in dramatically improved pharmacokinetics and biodistribution, and thus inhibited GBM recurrence by stimulating antitumor immunoresponse as compared to IFN. More importantly, when combined with subsequent intraperitoneal injection of temozolomide (TMZ), IFN-ELP(V) could much more effectively suppress post-surgical GBM recurrence than IFN, leading to a remarkably enhanced GBM-free survival rate (60%) over IFN (12.5%). Our findings implicate that the spatiotemporally-programmed combination of IFN-ELP(V) and TMZ leads to the synergy of post-surgical GBM immunochemotherapy, thereby providing a new and effective strategy for cancer therapy.
Collapse
Affiliation(s)
- Ping Liang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Guihuai Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Xinyu Liu
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China
| | - Zhuoran Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Jing Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Weiping Gao
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
29
|
Cha GD, Kang T, Baik S, Kim D, Choi SH, Hyeon T, Kim DH. Advances in drug delivery technology for the treatment of glioblastoma multiforme. J Control Release 2020; 328:350-367. [PMID: 32896613 DOI: 10.1016/j.jconrel.2020.09.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is a particularly aggressive and malignant type of brain tumor, notorious for its high recurrence rate and low survival rate. The treatment of GBM is challenging mainly because several issues associated with the GBM microenvironment have not yet been resolved. These obstacles originate from a variety of factors such as genetics, anatomy, and cytology, all of which collectively hinder the treatment of GBM. Recent advances in materials and device engineering have presented new perspectives with regard to unconventional drug administration methods for GBM treatment. Such novel drug delivery approaches, based on the clear understanding of the intrinsic properties of GBM, have shown promise in overcoming some of the obstacles. In this review, we first recapitulate the first-line therapy and clinical challenges in the current treatment of GBM. Afterwards, we introduce the latest technological advances in drug delivery strategies to improve the efficiency for GBM treatment, mainly focusing on materials and devices. We describe such efforts by classifying them into two categories, systemic and local drug delivery. Finally, we discuss unmet challenges and prospects for the clinical translation of these drug delivery technologies.
Collapse
Affiliation(s)
- Gi Doo Cha
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Taegyu Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Seungmin Baik
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dokyoon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan 15588, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
30
|
Zhang X, Zhang W, Mao XG, Cao WD, Zhen HN, Hu SJ. Malignant Intracranial High Grade Glioma and Current Treatment Strategy. Curr Cancer Drug Targets 2020; 19:101-108. [PMID: 29848277 DOI: 10.2174/1568009618666180530090922] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/06/2017] [Accepted: 12/19/2017] [Indexed: 12/17/2022]
Abstract
Malignant high-grade glioma (HGG) is the most common and extremely fatal type of primary intracranial tumor. These tumors recurred within 2 to 3 cm of the primary region of tumor resection in the majority of cases. Furthermore, the blood-brain barrier significantly limited the access of many systemically administered chemotherapeutics to the tumor, pointing towards a stringent need for new therapeutic patterns. Therefore, targeting therapy using local drug delivery for HGG becomes a priority for the development of novel therapeutic strategies. The main objectives to the effective use of chemotherapy for HGG include the drug delivery to the tumor region and the infusion of chemotherapeutic agents into the vascular supply of a tumor directly, which could improve the pharmacokinetic profile by enhancing drug delivery to the neoplasm tissue. Herein, we reviewed clinical and molecular features, different methods of chemotherapy application in HGGs, especially the existing and promising targeting therapies using local drug delivery for HGG which could effectively inhibit tumor invasion, proliferation and recurrence of HGG to combat the deadly disease. Undoubtedly, novel chemical medicines targeting these HGG may represent one of the most important directions in the Neuro-oncology.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing-Gang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei-Dong Cao
- Department of Neurosurgery, Navy General Hospital, PLA, Beijing, 100048, China
| | - Hai-Ning Zhen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Shi-Jie Hu
- Department of Neuro-oncology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
31
|
Rasch F, Schmitt C, Saure LM, Meyer R, Adamski V, Dengiz D, Scherließ R, Lucius R, Synowitz M, Mishra YK, Hattermann K, Adelung R, Held-Feindt J, Schütt F. Macroscopic Silicone Microchannel Matrix for Tailored Drug Release and Localized Glioblastoma Therapy. ACS Biomater Sci Eng 2020; 6:3388-3397. [DOI: 10.1021/acsbiomaterials.0c00094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Florian Rasch
- Chair for Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiser Str. 2, 24143 Kiel, Germany
| | - Christina Schmitt
- Department of Anatomy, Kiel University, Otto-Hahn-Platz 8, 24118 Kiel, Germany
| | - Lena M. Saure
- Chair for Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiser Str. 2, 24143 Kiel, Germany
| | - Rieke Meyer
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, Arnold-Heller-Str. 3, House D, 24105 Kiel, Germany
| | - Vivian Adamski
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, Arnold-Heller-Str. 3, House D, 24105 Kiel, Germany
| | - Duygu Dengiz
- Chair for Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiser Str. 2, 24143 Kiel, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany
| | - Ralph Lucius
- Department of Anatomy, Kiel University, Otto-Hahn-Platz 8, 24118 Kiel, Germany
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, Arnold-Heller-Str. 3, House D, 24105 Kiel, Germany
| | - Yogendra K. Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, 6400 Sønderborg, Denmark
| | - Kirsten Hattermann
- Department of Anatomy, Kiel University, Otto-Hahn-Platz 8, 24118 Kiel, Germany
| | - Rainer Adelung
- Chair for Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiser Str. 2, 24143 Kiel, Germany
| | - Janka Held-Feindt
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, Arnold-Heller-Str. 3, House D, 24105 Kiel, Germany
| | - Fabian Schütt
- Chair for Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiser Str. 2, 24143 Kiel, Germany
| |
Collapse
|
32
|
Song Q, Zhang C, Chen X, Cheng Y. Comparing amide proton transfer imaging with dynamic susceptibility contrast-enhanced perfusion in predicting histological grades of gliomas: a meta-analysis. Acta Radiol 2020; 61:549-557. [PMID: 31495179 DOI: 10.1177/0284185119871667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background As a subtype of chemical exchange saturation transfer imaging without contrast agent administration, amide proton transfer (APT) imaging has demonstrated the potential for differentiating the histologic grades of gliomas. Dynamic susceptibility contrast-enhanced perfusion, a perfusion-weighted imaging technique, is a well-established technique in grading gliomas. Purpose To compare the ability of amide proton transfer and dynamic susceptibility contrast-enhanced imaging for predicting the grades of gliomas. Material and Methods A comprehensive literature search was performed independently by two observers to identify articles about the diagnostic performance of amide proton transfer and dynamic susceptibility contrast-enhanced perfusion in predicting the grade of gliomas. Summary estimates of diagnostic accuracy were obtained by using a random-effects model. Results Of 179 studies identified, 23 studies were included the analysis. Eight studies evaluated amide proton transfer and 16 studies evaluated dynamic susceptibility contrast-enhanced perfusion with the parameter rCBV. The pooled sensitivities and specificities of each study’s best performing parameter were 88% (95% confidence interval [CI] 74–95) and 89% (95% CI 78–95) for amide proton transfer, and 95% (95% CI 87–98), 88% (95% CI 81–93) for perfusion-weighted imaging–dynamic susceptibility contrast-enhanced perfusion, respectively. The pooled sensitivities and specificities for grading gliomas using the two most commonly evaluated parameters, were 92% (95% CI 80–97) and 90% (95% CI 75–96) for APTmax, and 97% (95% CI 91–99) and 87% (95% CI 80–92) for rCBVmax, respectively. Conclusion Considering the similar performance of APT and dynamic susceptibility contrast-enhanced (DSC) in predicting glioma grade, the former method appears preferable since it needs no contrast agent.
Collapse
Affiliation(s)
- Qingxu Song
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Chencheng Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, PR China
| | - Xin Chen
- Department of MR, Shandong Medical Imaging Research Institute, Shandong University, Jinan, PR China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, PR China
| |
Collapse
|
33
|
Turan O, Bielecki P, Perera V, Lorkowski M, Covarrubias G, Tong K, Yun A, Loutrianakis G, Raghunathan S, Park Y, Moon T, Cooley S, Dixit D, Griswold M, Ghaghada K, Peiris P, Rich J, Karathanasis E. Treatment of glioblastoma using multicomponent silica nanoparticles. ADVANCED THERAPEUTICS 2019; 2:1900118. [PMID: 32953978 PMCID: PMC7500584 DOI: 10.1002/adtp.201900118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Indexed: 01/12/2023]
Abstract
Glioblastomas (GBMs) remain highly lethal. This partially stems from the presence of brain tumor initiating cells (BTICs), a highly plastic cellular subpopulation that is resistant to current therapies. In addition to resistance, the blood-brain barrier limits the penetration of most drugs into GBMs. To effectively deliver a BTIC-specific inhibitor to brain tumors, we developed a multicomponent nanoparticle, termed Fe@MSN, which contains a mesoporous silica shell and an iron oxide core. Fibronectin-targeting ligands directed the nanoparticle to the near-perivascular areas of GBM. After Fe@MSN particles deposited in the tumor, an external low-power radiofrequency (RF) field triggered rapid drug release due to mechanical tumbling of the particle resulting in penetration of high amounts of drug across the blood-brain tumor interface and widespread drug delivery into the GBM. We loaded the nanoparticle with the drug 1400W, which is a potent inhibitor of the inducible nitric oxide synthase (iNOS). It has been shown that iNOS is preferentially expressed in BTICs and is required for their maintenance. Using the 1400W-loaded Fe@MSN and RF-triggered release, in vivo studies indicated that the treatment disrupted the BTIC population in hypoxic niches, suppressed tumor growth and significantly increased survival in BTIC-derived GBM xenografts.
Collapse
Affiliation(s)
- O. Turan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - P.A. Bielecki
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - V. Perera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - M. Lorkowski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - G. Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - K. Tong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - A. Yun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Georgia Loutrianakis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - S. Raghunathan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Y. Park
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - T. Moon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - S. Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - D. Dixit
- Department of Neurosciences, University of California, San Diego, California
| | - M.A. Griswold
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - K.B. Ghaghada
- Edward B. Singleton Department of Pediatric Radiology, Texas Children’s Hospital, Houston, Texas
| | - P.M. Peiris
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - J.N. Rich
- Department of Neurosciences, University of California, San Diego, California
| | - E. Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
34
|
Novel Semi-Replicative Retroviral Vector Mediated Double Suicide Gene Transfer Enhances Antitumor Effects in Patient-Derived Glioblastoma Models. Cancers (Basel) 2019; 11:cancers11081090. [PMID: 31370279 PMCID: PMC6721803 DOI: 10.3390/cancers11081090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/12/2019] [Accepted: 07/30/2019] [Indexed: 01/10/2023] Open
Abstract
As glioblastomas are mostly localized infiltrative lesions, gene therapy based on the retroviral replicating vector (RRV) system is considered an attractive strategy. Combinations of multiple suicide genes can circumvent the limitations associated with each gene, achieving direct and synergistic cytotoxic effects, along with bystander cell killing. In this study, we constructed a semi-and pseudotyped-RRV (sp-RRV) system harboring two suicide genes—herpes simplex virus type 1 thymidine kinase (TK) and yeast cytosine deaminase (CD)—to verify the dissemination and antitumor efficacy of our sp-RRV system (spRRVe-sEF1α-TK/sRRVgp-sEF1α-CD) in seven patient-derived glioblastoma stem-like cells (GSCs). Flow cytometry and high-content analysis revealed a wide range of transduction efficiency and good correlation between the delivery of therapeutic genes and susceptibility to the prodrugs ganciclovir and 5-fluorocytosine in patient-derived GSCs in vitro. Intra-tumoral delivery of spRRVe-sEF1α-TK/sRRVgp-sEF1α-CD, combined with prodrug treatment, synergistically inhibited cell proliferation and angiogenesis while increasing apoptosis and the depletion of tumor-associated macrophages in orthotopic glioblastoma xenografts. Genomic profiling of patient-derived GSCs revealed that the key genes preventing sp-RRV infection and transmission were associated with cell adhesion, migration, development, differentiation, and proliferation. This is the first report demonstrating that a novel sp-RRV-mediated TK/CD double suicide gene transfer system has high oncolytic power against extremely heterogeneous and treatment-refractory glioblastomas.
Collapse
|
35
|
Darge HF, Andrgie AT, Tsai HC, Lai JY. Polysaccharide and polypeptide based injectable thermo-sensitive hydrogels for local biomedical applications. Int J Biol Macromol 2019; 133:545-563. [DOI: 10.1016/j.ijbiomac.2019.04.131] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 01/19/2023]
|
36
|
Turan O, Bielecki P, Perera V, Lorkowski M, Covarrubias G, Tong K, Yun A, Rahmy A, Ouyang T, Raghunathan S, Gopalakrishnan R, Griswold MA, Ghaghada KB, Peiris PM, Karathanasis E. Delivery of drugs into brain tumors using multicomponent silica nanoparticles. NANOSCALE 2019; 11:11910-11921. [PMID: 31187845 PMCID: PMC7776621 DOI: 10.1039/c9nr02876e] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Glioblastomas are highly lethal cancers defined by resistance to conventional therapies and rapid recurrence. While new brain tumor cell-specific drugs are continuously becoming available, efficient drug delivery to brain tumors remains a limiting factor. We developed a multicomponent nanoparticle, consisting of an iron oxide core and a mesoporous silica shell that can effectively deliver drugs across the blood-brain barrier into glioma cells. When exposed to alternating low-power radiofrequency (RF) fields, the nanoparticle's mechanical tumbling releases the entrapped drug molecules from the pores of the silica shell. After directing the nanoparticle to target the near-perivascular regions and altered endothelium of the brain tumor via fibronectin-targeting ligands, rapid drug release from the nanoparticles is triggered by RF facilitating wide distribution of drug delivery across the blood-brain tumor interface.
Collapse
Affiliation(s)
- O Turan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zhang H, Wang T, Liu H, Ren F, Qiu W, Sun Q, Yan F, Zheng H, Li Z, Gao M. Second near-infrared photodynamic therapy and chemotherapy of orthotopic malignant glioblastoma with ultra-small Cu 2-xSe nanoparticles. NANOSCALE 2019; 11:7600-7608. [PMID: 30968107 DOI: 10.1039/c9nr01789e] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The treatment of malignant glioblastoma is a huge challenge due to the existence of the blood-brain barrier. Herein, we report the treatment of orthotopic malignant glioblastoma with imaging guided second near-infrared (NIR-II) photodynamic therapy and chemotherapy by using drug-loaded ultra-small Cu2-xSe theranostic nanoparticles (NPs). Ultra-small Cu2-xSe NPs possess a strong absorbance in the NIR-II window, and their absorption at 1064 nm is around 2 times that at 808 nm. Their strong NIR-II absorbance and the deeper-tissue penetration of NIR-II light ensure excellent photodynamic therapy performance under irradiation with a 1064 nm laser. We also demonstrate that ultra-small Cu2-xSe NPs can produce vast amounts of reactive oxygen species via electron transfer (for ˙OH generation) and energy transfer (for 1O2 generation) mechanisms under irradiation. In addition, these NPs can be effectively and locally transported into orthotopic malignant glioblastoma with the assistance of focused ultrasound. The deposited Cu2-xSe NPs can be used for photoacoustic imaging to guide the combined NIR-II photodynamic therapy and chemotherapy. The results show that the tumor growth can be significantly suppressed. This work demonstrates the great potential of drug-loaded ultra-small Cu2-xSe NPs as a promising therapeutic agent for the treatment of orthotopic malignant glioblastoma.
Collapse
Affiliation(s)
- Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mahzouni P, Shavakhi M. Prostate-Specific Membrane Antigen Expression in Neovasculature of Glioblastoma Multiforme. Adv Biomed Res 2019; 8:18. [PMID: 30993088 PMCID: PMC6425742 DOI: 10.4103/abr.abr_209_18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Glioblastoma (GBM) is the most malignant brain tumor with a poor prognosis that can be very difficult to cure, and the current treatment options have no optimal outcomes. Hence, it is essential to find new treatment modalities. Histologically, this tumor has high microvascular density that makes it desirable for vascular target agent drugs. Prostate-specific membrane antigen (PSMA) is a novel antigen with unique features that expresses in the vascular endothelium of some malignant tumors. Materials and Methods: Formalin-fixed paraffin-embedded tissues from sixty patients who underwent GBM tumor resection from 2012 to 2016 were evaluated for the expression of PSMA by immunohistochemistry. Sections were also assessed for the extent and intensity of endothelial staining in tumor microvessels and for clinicopathologic factor correlation. Results: A considerable PSMA expression level was detected in 66% of the cases, and the intensity was strong and moderate in 63%. There was no significant correlation neither between PSMA expression with tumor site, presence of necrosis, and endothelial proliferation nor with age and sex. Conclusion: The expression of PSMA in GBM, as observed in the current study, may suggest a new role of PSMA-targeted therapy and indicate more investigations focused on complementary treatment strategies that specifically target tumor vasculature.
Collapse
Affiliation(s)
- Parvin Mahzouni
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mitra Shavakhi
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
39
|
Bae Y, Thuy LT, Lee YH, Ko KS, Han J, Choi JS. Polyplexes of Functional PAMAM Dendrimer/Apoptin Gene Induce Apoptosis of Human Primary Glioma Cells In Vitro. Polymers (Basel) 2019; 11:E296. [PMID: 30960280 PMCID: PMC6419211 DOI: 10.3390/polym11020296] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/01/2019] [Accepted: 02/07/2019] [Indexed: 01/21/2023] Open
Abstract
Highly efficient and safe gene delivery has become an important aspect of neuronal gene therapy. We evaluated the ability of polyamidoamine (PAMAM) dendrimer grafted with phenylalanine, histidine, and arginine (PAMAM-FHR), a nonviral gene delivery vector, to deliver a therapeutic, tumor cell-specific killer gene, apoptin, into the human primary glioma cell line GBL-14 and human dermal fibroblasts. We performed a transfection assay using plasmids of luciferase and enhanced green fluorescent protein (EGFP) and assessed cell viability. Both cell lines were treated with complexes of PAMAM-FHR and apoptin after which their intracellular uptake and localization were examined by fluorescence-activated cell sorting (FACS)analysis and confocal laser scanning microscopy. Confocal microscopy showed that the PAMAM-FHR escaped from the endo-lysosome into the cytosol. Cell cycle phase distribution analysis, annexin V staining, and a tetramethylrhodamine ethyl ester (TMRE) assay established that apoptin triggered apoptosis in the GBL-14 cell line but not in normal fibroblasts. These results indicated that the PAMAM-FHR/apoptin complex is an effective gene vehicle for cancer therapy in vitro.
Collapse
Affiliation(s)
- Yoonhee Bae
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.
| | - Le Thi Thuy
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea.
| | - Young Hwa Lee
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea.
| | - Kyung Soo Ko
- Department of Internal Medicine, Sanggye Paik Hospital, Cardiovascular and Metabolic Disease Center, Inje University, Seoul 100-032, Korea.
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.
| | - Joon Sig Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea.
| |
Collapse
|
40
|
Pang L, Zhu Y, Qin J, Zhao W, Wang J. Primary M1 macrophages as multifunctional carrier combined with PLGA nanoparticle delivering anticancer drug for efficient glioma therapy. Drug Deliv 2018; 25:1922-1931. [PMID: 30465444 PMCID: PMC6263108 DOI: 10.1080/10717544.2018.1502839] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022] Open
Abstract
Glioma remains difficult to treat because of the infiltrative growth of tumor cells and their resistance to standard therapy. Despite rapid development of targeted drug delivery system, the current therapeutic efficacy is still challenging. Based on our previous studies, macrophages have been proved to be promising drug carrier for active glioma delivery. To make full use of macrophage carrier, primary M1 macrophages were proposed to replace regular macrophage to deliver nanodrugs into glioma, because M1 macrophages not only have the natural ability to home into tumor tissues, but they also have stronger phagocytic capability than other types of macrophage, which can enable them to uptake enough drug-loaded nanoparticles for therapy. In addition, M1 macrophages are not easily affected by harsh tumor microenvironment and inhibit tumor growth themselves. In this study, M1 macrophage-loaded nanoparticles (M1-NPs) were prepared by incubating poly(lactide-co-glycolide) (PLGA) nanoparticles with primary M1 macrophages. In vitro cell assays demonstrated M1 macrophage still maintained good tumor tropism capability after particle loading, and could efficiently carry particles across endothelial barrier into tumor tissues. In vivo imaging verified that M1-NPs exhibited higher brain tumor distribution than free nanoparticles. DOX@M1-NPs (doxorubicin-loaded M1-NPs) presented significantly enhanced anti-glioma effect with prolonged survival median and increased cell apoptosis. In conclusion, the results provided a new strategy exploiting M1 macrophage as carrier for drug delivery, which improved targeting efficiency and therapeutic efficacy of chemodrugs for glioma therapy.
Collapse
Affiliation(s)
- Liang Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, Schoolof Pharmacy, Fudan University, Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, Shanghai, China
| | - Ying Zhu
- Insitituteof Clinical Pharmacology, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jing Qin
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, Schoolof Pharmacy, Fudan University, Shanghai, China
| | - Wenjie Zhao
- Shanghai Institute of Pharmaceutical Industry, Shanghai, China
| | - Jianxin Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, Schoolof Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Kim Y, Kang H, Powathil G, Kim H, Trucu D, Lee W, Lawler S, Chaplain M. Role of extracellular matrix and microenvironment in regulation of tumor growth and LAR-mediated invasion in glioblastoma. PLoS One 2018; 13:e0204865. [PMID: 30286133 PMCID: PMC6171904 DOI: 10.1371/journal.pone.0204865] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
The cellular dispersion and therapeutic control of glioblastoma, the most aggressive type of primary brain cancer, depends critically on the migration patterns after surgery and intracellular responses of the individual cancer cells in response to external biochemical cues in the microenvironment. Recent studies have shown that miR-451 regulates downstream molecules including AMPK/CAB39/MARK and mTOR to determine the balance between rapid proliferation and invasion in response to metabolic stress in the harsh tumor microenvironment. Surgical removal of the main tumor is inevitably followed by recurrence of the tumor due to inaccessibility of dispersed tumor cells in normal brain tissue. In order to address this complex process of cell proliferation and invasion and its response to conventional treatment, we propose a mathematical model that analyzes the intracellular dynamics of the miR-451-AMPK- mTOR-cell cycle signaling pathway within a cell. The model identifies a key mechanism underlying the molecular switches between proliferative phase and migratory phase in response to metabolic stress in response to fluctuating glucose levels. We show how up- or down-regulation of components in these pathways affects the key cellular decision to infiltrate or proliferate in a complex microenvironment in the absence and presence of time delays and stochastic noise. Glycosylated chondroitin sulfate proteoglycans (CSPGs), a major component of the extracellular matrix (ECM) in the brain, contribute to the physical structure of the local brain microenvironment but also induce or inhibit glioma invasion by regulating the dynamics of the CSPG receptor LAR as well as the spatiotemporal activation status of resident astrocytes and tumor-associated microglia. Using a multi-scale mathematical model, we investigate a CSPG-induced switch between invasive and non-invasive tumors through the coordination of ECM-cell adhesion and dynamic changes in stromal cells. We show that the CSPG-rich microenvironment is associated with non-invasive tumor lesions through LAR-CSGAG binding while the absence of glycosylated CSPGs induce the critical glioma invasion. We illustrate how high molecular weight CSPGs can regulate the exodus of local reactive astrocytes from the main tumor lesion, leading to encapsulation of non-invasive tumor and inhibition of tumor invasion. These different CSPG conditions also change the spatial profiles of ramified and activated microglia. The complex distribution of CSPGs in the tumor microenvironment can determine the nonlinear invasion behaviors of glioma cells, which suggests the need for careful therapeutic strategies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
| | - Hyunji Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Gibin Powathil
- Department of Mathematics, Swansea University, Swansea, United Kingdom
| | - Hyeongi Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Dumitru Trucu
- Division of Mathematics, University of Dundee, Dundee, United Kingdom
| | - Wanho Lee
- National Institute for Mathematical Sciences, Daejeon, Republic of Korea
| | - Sean Lawler
- Department of neurosurgery, Brigham and Women’s Hospital & Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mark Chaplain
- School of Mathematics and Statistics, Mathematical Institute, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
42
|
Liu H, Zhou M, Sheng Z, Chen Y, Yeh CK, Chen W, Liu J, Liu X, Yan F, Zheng H. Theranostic nanosensitizers for highly efficient MR/fluorescence imaging-guided sonodynamic therapy of gliomas. J Cell Mol Med 2018; 22:5394-5405. [PMID: 30156368 PMCID: PMC6201228 DOI: 10.1111/jcmm.13811] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/25/2018] [Accepted: 06/29/2018] [Indexed: 12/22/2022] Open
Abstract
Glioma is the most frequent primary brain tumour of the central nervous system. Its high aggressiveness and deep‐seated brain lesion make it a great challenge to develop a non‐invasive, precise and effective treatment approach. Here, we report a multifunctional theranostic agent that can integrate imaging and therapy into a single nano‐platform for imaging‐guided sonodynamic therapy (SDT). The SDT agents were fabricated by encapsulation of sinoporphyrin sodium (DVDMS) chelating with manganese ions into nanoliposomes (DVDMS‐Mn‐LPs). DVDMS‐Mn‐LPs are physiologically stable and biologically compatible, and they can produce singlet oxygen upon ultrasound irradiation to kill cancer cells. Both cell and animal studies demonstrated that SDT with DVDMS‐Mn‐LPs can significantly improve the antitumour growth efficiency even in the presence of skull. In addition, DVDMS‐Mn‐LPs are good for MR and fluorescence imaging. Thus, DVDMS‐Mn‐LPs reported here may provide a promising strategy for imaging‐guided modality for glioma treatment.
Collapse
Affiliation(s)
- Hongmei Liu
- Department of Ultrasonography, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Meijun Zhou
- Department of Ultrasonography, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramic and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Taiwan
| | - Wenting Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Jia Liu
- Department of Ultrasonography, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fei Yan
- Department of Ultrasonography, Guangdong Second Provincial General Hospital, Guangzhou, China.,Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Department of Ultrasonography, Guangdong Second Provincial General Hospital, Guangzhou, China.,Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
43
|
Duan S. Silencing the autophagy-specific gene Beclin-1 contributes to attenuated hypoxia-induced vasculogenic mimicry formation in glioma. Cancer Biomark 2018; 21:565-574. [PMID: 29278874 DOI: 10.3233/cbm-170444] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To explore the influence of Beclin-1 on vasculogenic mimicry (VM) induced by hypoxia in glioma. METHODS CD34-PAS staining was carried out to observe VM formation, and immunohistochemistry was used to determine the expression levels of Beclin-1, HIF-1α, VEGF and MMP2 in 105 patients with primary glioma. Human glioma U87MG cells were divided into Normoxia, Hypoxia, Hypoxia + NC siRNA and Hypoxia + Beclin-1 siRNA groups. Cobalt chloride (CoCl2) was used to stimulate hypoxic conditions, and a VM tube formation assay was used to detect VM formation. Wound healing and Transwell invasion assays were used to detect the invasive and migratory abilities of U87MG cells, respectively. Fluorescent LC3 puncta analysis was performed to examine the status of autophagic flux. Expression levels of Beclin-1 and VM-related molecules were determined using real-time quantitative-polymerase chain reaction (RT-qPCR) and western blotting. RESULTS There were 34 VM-positive cases and 71 VM-negative cases among 105 glioma patients, and VM formation was correlated with pathological grade and the expression of Beclin-1, HIF-1α, VEGF and MMP2. Positive relations were found between Beclin-1 and the expression of HIF-1α, VEGF and MMP2. Under hypoxic conditions, significant increases in the total length of tubes, migration rate, invasion cell number and expression of VM-related molecules were found in U87MG cells. Silencing Beclin-1 markedly decreased hypoxia-induced VM formation and the invasive and migratory abilities, together with the expression of VM-related molecules, in U87MG cells and significantly inhibited the autophagic flux. CONCLUSION Silencing Beclin-1 can attenuate hypoxia-induced VM formation and the metastatic ability of U87MG cells and is a potential target for VM inhibition in glioma.
Collapse
|
44
|
Basso J, Miranda A, Nunes S, Cova T, Sousa J, Vitorino C, Pais A. Hydrogel-Based Drug Delivery Nanosystems for the Treatment of Brain Tumors. Gels 2018; 4:E62. [PMID: 30674838 PMCID: PMC6209281 DOI: 10.3390/gels4030062] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 07/09/2018] [Accepted: 07/18/2018] [Indexed: 11/16/2022] Open
Abstract
Chemotherapy is commonly associated with limited effectiveness and unwanted side effects in normal cells and tissues, due to the lack of specificity of therapeutic agents to cancer cells when systemically administered. In brain tumors, the existence of both physiological barriers that protect tumor cells and complex resistance mechanisms to anticancer drugs are additional obstacles that hamper a successful course of chemotherapy, thus resulting in high treatment failure rates. Several potential surrogate therapies have been developed so far. In this context, hydrogel-based systems incorporating nanostructured drug delivery systems (DDS) and hydrogel nanoparticles, also denoted nanogels, have arisen as a more effective and safer strategy than conventional chemotherapeutic regimens. The former, as a local delivery approach, have the ability to confine the release of anticancer drugs near tumor cells over a long period of time, without compromising healthy cells and tissues. Yet, the latter may be systemically administered and provide both loading and targeting properties in their own framework, thus identifying and efficiently killing tumor cells. Overall, this review focuses on the application of hydrogel matrices containing nanostructured DDS and hydrogel nanoparticles as potential and promising strategies for the treatment and diagnosis of glioblastoma and other types of brain cancer. Some aspects pertaining to computational studies are finally addressed.
Collapse
Affiliation(s)
- João Basso
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-354, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal.
| | - Ana Miranda
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-354, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal.
| | - Sandra Nunes
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, Coimbra 3004-535, Portugal.
| | - Tânia Cova
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, Coimbra 3004-535, Portugal.
| | - João Sousa
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-354, Portugal.
- LAQV REQUIMTE, Group of Pharmaceutical Technology, Porto 4051-401, Portugal.
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-354, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal.
- LAQV REQUIMTE, Group of Pharmaceutical Technology, Porto 4051-401, Portugal.
| | - Alberto Pais
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, Coimbra 3004-535, Portugal.
| |
Collapse
|
45
|
Peng Y, Huang J, Xiao H, Wu T, Shuai X. Codelivery of temozolomide and siRNA with polymeric nanocarrier for effective glioma treatment. Int J Nanomedicine 2018; 13:3467-3480. [PMID: 29942129 PMCID: PMC6007389 DOI: 10.2147/ijn.s164611] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background The treatment of glioma remains a challenge because conventional chemotherapy is often ineffective by drug resistance. Combinative therapy using chemotherapeutic agents and siRNA has demonstrated potential to improve anticancer outcome through a synergistic effect in various cancers. The current study aims to achieve better glioma treatment through a combinative therapy based on a folate-targeted nanocarrier carrying both temozolomide (TMZ) and anti-BCL-2 siRNA. Methods A polymeric micelle (TMZ-FaPEC@siRNA) incorporating TMZ and anti-BCL-2 siRNA was prepared based on folate-conjugated triblock copolymer (Fa-PEG-PEI-PCL, Fa-PEC) of poly(ε-caprolactone) (PCL), poly(ethylenimine) (PEI) and poly(ethylene glycol) (PEG). The physicochemical properties and drug release profile of TMZ-FaPEC@siRNA were tested. The Fa-targeted drug delivery and joint effect of siRNA and TMZ to induce glioma apoptosis and tumor growth inhibition were evaluated both in vitro and in vivo. Results In vitro cell study demonstrated that the nanocarrier effectively facilitates codelivery of siRNA and TMZ into C6 cells, resulting in a strong apoptotic response of cancer cells by silencing the antiapoptosis BCL-2 gene and activating the proapoptotic Bax gene simultaneously. In vivo study in rat bearing orthotropic glioma showed that tumor growth was inhibited and meanwhile animal survival was prolonged remarkably through intracranial injection of TMZ-FaPEC@siRNA. Conclusion Our results evidence the strong efficacy of the folate-targeted nanomedicine carrying TMZ and BCL-2 siRNA in treating glioma.
Collapse
Affiliation(s)
- Yuan Peng
- Center of Biomedical Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.,Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, Department of Polymer Science, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jinsheng Huang
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, Department of Polymer Science, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Hong Xiao
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, Department of Polymer Science, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Teng Wu
- Center of Biomedical Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xintao Shuai
- Center of Biomedical Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.,Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, Department of Polymer Science, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
46
|
Nam L, Coll C, Erthal LCS, de la Torre C, Serrano D, Martínez-Máñez R, Santos-Martínez MJ, Ruiz-Hernández E. Drug Delivery Nanosystems for the Localized Treatment of Glioblastoma Multiforme. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E779. [PMID: 29751640 PMCID: PMC5978156 DOI: 10.3390/ma11050779] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/30/2018] [Accepted: 05/01/2018] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme is one of the most prevalent and malignant forms of central nervous system tumors. The treatment of glioblastoma remains a great challenge due to its location in the intracranial space and the presence of the blood⁻brain tumor barrier. There is an urgent need to develop novel therapy approaches for this tumor, to improve the clinical outcomes, and to reduce the rate of recurrence and adverse effects associated with present options. The formulation of therapeutic agents in nanostructures is one of the most promising approaches to treat glioblastoma due to the increased availability at the target site, and the possibility to co-deliver a range of drugs and diagnostic agents. Moreover, the local administration of nanostructures presents significant additional advantages, since it overcomes blood⁻brain barrier penetration issues to reach higher concentrations of therapeutic agents in the tumor area with minimal side effects. In this paper, we aim to review the attempts to develop nanostructures as local drug delivery systems able to deliver multiple agents for both therapeutic and diagnostic functions for the management of glioblastoma.
Collapse
Affiliation(s)
- L Nam
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin (TCD), Dublin 2, Ireland.
- Trinity Biomedical Sciences Institute, TCD, Dublin 2, Ireland.
| | - C Coll
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin (TCD), Dublin 2, Ireland.
- Trinity Biomedical Sciences Institute, TCD, Dublin 2, Ireland.
| | - L C S Erthal
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin (TCD), Dublin 2, Ireland.
- Trinity Biomedical Sciences Institute, TCD, Dublin 2, Ireland.
| | - C de la Torre
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46010 València, Spain.
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| | - D Serrano
- Departamento de Farmacia Galenica y Tecnologia Alimentaria, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - R Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46010 València, Spain.
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| | - M J Santos-Martínez
- Trinity Biomedical Sciences Institute, TCD, Dublin 2, Ireland.
- School of Medicine, Trinity College Dublin (TCD), Dublin 2, Ireland.
| | - E Ruiz-Hernández
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin (TCD), Dublin 2, Ireland.
- Trinity Biomedical Sciences Institute, TCD, Dublin 2, Ireland.
| |
Collapse
|
47
|
Lee CW, Hsu LF, Lee MH, Lee IT, Liu JF, Chiang YC, Tsai MH. Extracts of Artocarpus communis Induce Mitochondria-Associated Apoptosis via Pro-oxidative Activity in Human Glioblastoma Cells. Front Pharmacol 2018; 9:411. [PMID: 29770114 PMCID: PMC5941989 DOI: 10.3389/fphar.2018.00411] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 04/09/2018] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an extremely aggressive and devastating malignant tumor in the central nervous system. Its incidence is increasing and the prognosis is poor. Artocarpin is a natural prenylated flavonoid with various anti-inflammatory and anti-tumor properties. Studies have shown that artocarpin is associated with cell death of primary glioblastoma cells. However, the in vivo effects and the cellular and molecular mechanisms modulating the anticancer activities of artocarpin remain unknown. In this study, we demonstrated that treating the glioblastoma cell lines U87 and U118 cells with artocarpin induced apoptosis. Artocarpin-induced apoptosis is associated with caspase activation and poly (ADP-ribose) polymerase (PARP) cleavage and is mediated by the mitochondrial pathway. This is associated with mitochondrial depolarization, mitochondrial-derived reactive oxidative species (ROS) production, cytochrome c release, Bad and Bax upregulations, and Bcl-2 downregulation. Artocarpin induced NADPH oxidase/ROS generation plays an important role in the mitochondrial pathway activation. Furthermore, we found artocarpin-induced ROS production in mitochondria is associated with Akt- and ERK1/2 activation. After treatment with artocarpin, ROS causes PI3K/Akt/ERK1/2-induced cell death of these tumor cells. These observations were further verified by the results from the implantation of both U87 and U118 cells into in vivo mouse. In conclusion, our findings suggest that artocarpin induces mitochondria-associated apoptosis of glioma cells, suggesting that artocarpine can be a potential chemotherapeutic agent for future GBM treatment.
Collapse
Affiliation(s)
- Chiang-Wen Lee
- Division of Basic Medical Sciences, Department of Nursing, Chang Gung University of Science and Technology, Chiayi, Taiwan.,Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi, Taiwan.,Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Lee-Fen Hsu
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Ming-Hsueh Lee
- Division of Neurosurgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - I-Ta Lee
- The Center of Translational Medicine, Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ju-Fang Liu
- Central Laboratory, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yao-Chang Chiang
- Division of Basic Medical Sciences, Department of Nursing, Chang Gung University of Science and Technology, Chiayi, Taiwan.,Center for Drug Abuse and Addiction, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ming-Horng Tsai
- Department of Pediatrics, Division of Neonatology and Pediatric Hematology/Oncology, Yunlin Chang Gung Memorial Hospital, Yunlin County, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
48
|
El-Boubbou K. Magnetic iron oxide nanoparticles as drug carriers: preparation, conjugation and delivery. Nanomedicine (Lond) 2018; 13:929-952. [PMID: 29546817 DOI: 10.2217/nnm-2017-0320] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Magnetic nanoparticles (MNPs), particularly made of iron oxides, have been extensively studied as diagnostic imaging agents and therapeutic delivery vehicles. In this review, special emphasis is set on the 'recent advancements of drug-conjugated MNPs used for therapeutic applications'. The most prevalent preparation methods and chemical functionalization strategies required for translational biomedical nanoformulations are outlined. Particular attention is, then, devoted to the tailored conjugation of drugs to the MNP carrier according to either noncovalent or covalent attachments, with advantages and drawbacks of both pathways conferred. Notable examples are presented to demonstrate the advantages of MNPs in respective drug-delivery applications. Understanding of the preparation, conjugation and delivery processes will definitely bring, in the next decades, a novel magneto-nanovehicle for effective theranostics.
Collapse
Affiliation(s)
- Kheireddine El-Boubbou
- Department of Basic Sciences, College of Science & Health Professions, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11481, Saudi Arabia.,King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Hospital, Riyadh 11426, Saudi Arabia
| |
Collapse
|
49
|
Keu KV, Witney TH, Yaghoubi S, Rosenberg J, Kurien A, Magnusson R, Williams J, Habte F, Wagner JR, Forman S, Brown C, Allen-Auerbach M, Czernin J, Tang W, Jensen MC, Badie B, Gambhir SS. Reporter gene imaging of targeted T cell immunotherapy in recurrent glioma. Sci Transl Med 2018; 9. [PMID: 28100832 DOI: 10.1126/scitranslmed.aag2196] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/14/2016] [Indexed: 12/12/2022]
Abstract
High-grade gliomas are aggressive cancers that often become rapidly fatal. Immunotherapy using CD8+ cytotoxic T lymphocytes (CTLs), engineered to express both herpes simplex virus type 1 thymidine kinase (HSV1-TK) and interleukin-13 (IL-13) zetakine chimeric antigen receptor (CAR), is a treatment strategy with considerable potential. To optimize this and related immunotherapies, it would be helpful to monitor CTL viability and trafficking to glioma cells. We show that noninvasive positron emission tomography (PET) imaging with 9-[4-[18F]fluoro-3-(hydroxymethyl)butyl]guanine ([18F]FHBG) can track HSV1-tk reporter gene expression present in CAR-engineered CTLs. [18F]FHBG imaging was safe and enabled the longitudinal imaging of T cells stably transfected with a PET reporter gene in patients. Further optimization of this imaging approach for monitoring in vivo cell trafficking should greatly benefit various cell-based therapies for cancer.
Collapse
Affiliation(s)
- Khun Visith Keu
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States.,Division of Nuclear Medicine, Hôpital de la Cité-de-la-Santé de Laval, QC, H7M 3L9, Canada
| | - Timothy H Witney
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States.,Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Shahriar Yaghoubi
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States
| | - Jarrett Rosenberg
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States
| | - Anita Kurien
- Neurosurgery, City of Hope, Duarte, CA, 91010, United States
| | | | - John Williams
- Molecular & Medical Pharmacology, UCLA, Los Angeles, CA, 90095, United States
| | - Frezghi Habte
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States
| | - Jamie R Wagner
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, United States
| | - Stephen Forman
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, United States
| | - Christine Brown
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, United States
| | | | - Johannes Czernin
- Molecular & Medical Pharmacology, UCLA, Los Angeles, CA, 90095, United States
| | - Winson Tang
- Sangamo BioSciences Inc, Richmond, CA 94804, United States
| | - Michael C Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, 98145, United States
| | - Behnam Badie
- Neurosurgery, City of Hope, Duarte, CA, 91010, United States
| | - Sanjiv S Gambhir
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States.,Department of Bioengineering, Department of Materials Science & Engineering, Bio-X, Stanford University, Palo Alto, CA, 94305, United States
| |
Collapse
|
50
|
Wu D, Wang L, Yang Y, Huang J, Hu Y, Shu Y, Zhang J, Zheng J. MAD2-p31 comet axis deficiency reduces cell proliferation, migration and sensitivity of microtubule-interfering agents in glioma. Biochem Biophys Res Commun 2018; 498:157-163. [PMID: 29408509 DOI: 10.1016/j.bbrc.2018.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/02/2018] [Indexed: 11/16/2022]
Abstract
Mitotic arrest deficient-like-1 (MAD2, also known as MAD2L1) is thought to be an important spindle assembly checkpoint protein, which ensures accurate chromosome segregation and is closely associated with poor prognosis in many cancer. As a MAD2 binding protein, p31comet counteracts the function of MAD2 and leads to mitotic checkpoint silence. In this study, we explore the function of MAD2-p31comet axis in malignant glioma cells. Our results showed that disruption of MAD2-p31comet axis by MAD2 knockdown or p31comet overexpression suppressed cell proliferation, survival and migration of glioma, indicating that MAD2-p31comet axis is required for maintaining glioma cells malignancy. It is noted that MAD2 depletion or p31comet overexpression reduced the sensitivity of glioma cells to microtubule-interfering agents paclitaxel and vinblastine, providing clinical guidance for application of such drugs. Taken together, our findings suggest that MAD2-p31comet axis may serve as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Dang Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lepeng Wang
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Yanhong Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yuhua Hu
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Yongwei Shu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jingyu Zhang
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jing Zheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|