1
|
Lubitz LJ, Haffner MP, Rieger H, Leneweit G. Increased Cellular Uptake of ApoE3- or c(RGD)-Modified Liposomes for Glioblastoma Therapy Depending on the Target Cells. Pharmaceutics 2024; 16:1112. [PMID: 39339149 PMCID: PMC11434700 DOI: 10.3390/pharmaceutics16091112] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
As effective treatment of glioblastoma is still an unmet need, targeted delivery systems for efficient treatment are of utmost interest. Therefore, in this paper, surface modifications with a small peptide c(RGD) or physiological protein (ApoE3) were investigated. Cellular uptake in murine endothelial cells (bEnd.3) and different glioma cells (human U-87 MG, rat F98) was tested to elucidate possible differences and to correlate the uptake to the receptor expression. Different liposomal formulations were measured at 1 and 3 h for three lipid incubation concentrations. We calculated the liposomal uptake saturation S and the saturation half-time t1/2. An up to 9.6-fold increased uptake for ApoE3-modified liposomes, primarily in tumor cells, was found. Contrarily, c(RGD) liposomes showed a stronger increase in uptake in endothelial cells (up to 40.5-fold). The uptake of modified liposomes revealed enormous differences in S and t1/2 when comparing different tumor cell lines. However, for ApoE3-modified liposomes, we proved comparable saturation values (~25,000) for F98 cells and U-87 MG cells despite a 6-fold lower expression of LRP1 in F98 cells and a 5-fold slower uptake rate. Our findings suggest that cellular uptake of surface-modified liposomes depends more on the target structure than the ligand type, with significant differences between cell types of different origins.
Collapse
Affiliation(s)
- Larissa J. Lubitz
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute, 75223 Niefern-Öschelbronn, Germany
- Department of Chemical and Process Engineering, Institute of Mechanical Process Engineering and Mechanics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | | | - Harden Rieger
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute, 75223 Niefern-Öschelbronn, Germany
| | - Gero Leneweit
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute, 75223 Niefern-Öschelbronn, Germany
- Department of Chemical and Process Engineering, Institute of Mechanical Process Engineering and Mechanics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| |
Collapse
|
2
|
Malone K, Dugas M, Earl N, Alain T, LaCasse EC, Beug ST. Astrocytes and the tumor microenvironment inflammatory state dictate the killing of glioblastoma cells by Smac mimetic compounds. Cell Death Dis 2024; 15:592. [PMID: 39147758 PMCID: PMC11327263 DOI: 10.1038/s41419-024-06971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Smac mimetic compounds (SMCs) are small molecule drugs that sensitize cancer cells to TNF-α-induced cell death and have multiple immunostimulatory effects through alterations in NF-κB signaling. The combination of SMCs with immunotherapies has been reported to result in durable cures of up to 40% in syngeneic, orthotopic murine glioblastoma (GBM) models. Herein, we find that SMC resistance is not due to a cell-intrinsic mechanism of resistance. We thus evaluated the contribution of GBM and brain stromal components to identify parameters leading to SMC efficacy and resistance. The common physiological features of GBM tumors, such as hypoxia, hyaluronic acid, and glucose deprivation were found not to play a significant role in SMC efficacy. SMCs induced the death of microglia and macrophages, which are the major immune infiltrates in the tumor microenvironment. This death of microglia and macrophages then enhances the ability of SMCs to induce GBM cell death. Conversely, astrocytes promoted GBM cell growth and abrogated the ability of SMCs to induce death of GBM cells. The astrocyte-mediated resistance can be overcome in the presence of exogenous TNF-α. Overall, our results highlight that SMCs can induce death of microglia and macrophages, which then provides a source of death ligands for GBM cells, and that the targeting of astrocytes is a potential mechanism for overcoming SMC resistance for the treatment of GBM.
Collapse
Affiliation(s)
- Kyle Malone
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Melanie Dugas
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Nathalie Earl
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Tommy Alain
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Eric C LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Shawn T Beug
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
3
|
Liu LH, Liu YF, Zhang HB, Liu XL, Zhang HW, Huang B, Lin F, Li WH. A Novel ANG-BSA/BCNU/ICG MNPs Integrated for Targeting Therapy of Glioblastoma. Technol Cancer Res Treat 2024; 23:15330338241281321. [PMID: 39444362 PMCID: PMC11526396 DOI: 10.1177/15330338241281321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
PURPOSE Develop an albumin nanoparticle-based nanoprobe for targeted glioblastoma (GBM) diagnosis and treatment, utilizing Angopep-2 for low-density lipoprotein receptor-related protein (LRP) targeting. METHODS Combined albumin-coated superparamagnetic iron oxide (SPIO), Carmustine (BCNU), and indocyanine green (ICG). Assessed morphology, size, Zeta potential, fluorescence, and drug encapsulation. Conducted in vitro fluorescence/MRI imaging and cell viability assays, and in vivo nanoprobe accumulation evaluation in brain tumors. RESULTS ANG-BSA/BCNU/ICG MNPs exhibited superior targeting and cytotoxicity against GBM cells in vitro. In vivo, enhanced brain tumor accumulation during imaging was observed. CONCLUSION This targeted imaging and drug delivery system holds promise for efficient GBM therapy and intraoperative localization, addressing Blood-brain barrier (BBB) limitations with precise drug delivery and imaging capabilities.
Collapse
Affiliation(s)
- Li-Hong Liu
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Yu-Feng Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Hong-Bo Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Xiao-Lei Liu
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Han-Wen Zhang
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Biao Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Fan Lin
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Wei-Hua Li
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| |
Collapse
|
4
|
Angom RS, Nakka NMR, Bhattacharya S. Advances in Glioblastoma Therapy: An Update on Current Approaches. Brain Sci 2023; 13:1536. [PMID: 38002496 PMCID: PMC10669378 DOI: 10.3390/brainsci13111536] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary malignant brain tumor characterized by a high grade of malignancy and an extremely unfavorable prognosis. The current efficacy of established treatments for GBM is insufficient, necessitating the prompt development of novel therapeutic approaches. The progress made in the fundamental scientific understanding of GBM is swiftly translated into more advanced stages of therapeutic studies. Despite extensive efforts to identify new therapeutic approaches, GBM exhibits a high mortality rate. The current efficacy of treatments for GBM patients is insufficient due to factors such as tumor heterogeneity, the blood-brain barrier, glioma stem cells, drug efflux pumps, and DNA damage repair mechanisms. Considering this, pharmacological cocktail therapy has demonstrated a growing efficacy in addressing these challenges. Towards this, various forms of immunotherapy, including the immune checkpoint blockade, chimeric antigen receptor T (CAR T) cell therapy, oncolytic virotherapy, and vaccine therapy have emerged as potential strategies for enhancing the prognosis of GBM. Current investigations are focused on exploring combination therapies to mitigate undesirable side effects and enhance immune responses against tumors. Furthermore, clinical trials are underway to evaluate the efficacy of several strategies to circumvent the blood-brain barrier (BBB) to achieve targeted delivery in patients suffering from recurrent GBM. In this review, we have described the biological and molecular targets for GBM therapy, pharmacologic therapy status, prominent resistance mechanisms, and new treatment approaches. We also discuss these promising therapeutic approaches to assess prospective innovative therapeutic agents and evaluated the present state of preclinical and clinical studies in GBM treatment. Overall, this review attempts to provide comprehensive information on the current status of GBM therapy.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA; (R.S.A.); (N.M.R.N.)
| | - Naga Malleswara Rao Nakka
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA; (R.S.A.); (N.M.R.N.)
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA; (R.S.A.); (N.M.R.N.)
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| |
Collapse
|
5
|
Kubelt C, Hellmold D, Peschke E, Hauck M, Will O, Schütt F, Lucius R, Adelung R, Scherließ R, Hövener JB, Jansen O, Synowitz M, Held-Feindt J. Establishment of a Rodent Glioblastoma Partial Resection Model for Chemotherapy by Local Drug Carriers-Sharing Experience. Biomedicines 2023; 11:1518. [PMID: 37371613 DOI: 10.3390/biomedicines11061518] [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: 04/28/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Local drug delivery systems (LDDS) represent a promising therapy strategy concerning the most common and malignant primary brain tumor glioblastoma (GBM). Nevertheless, to date, only a few systems have been clinically applied, and their success is very limited. Still, numerous new LDDS approaches are currently being developed. Here, (partial resection) GBM animal models play a key role, as such models are needed to evaluate the therapy prior to any human application. However, such models are complex to establish, and only a few reports detail the process. Here, we report our results of establishing a partial resection glioma model in rats suitable for evaluating LDDS. C6-bearing Wistar rats and U87MG-spheroids- and patient-derived glioma stem-like cells-bearing athymic rats underwent tumor resection followed by the implantation of an exemplary LDDS. Inoculation, tumor growth, residual tumor tissue, and GBM recurrence were reliably imaged using high-resolution Magnetic Resonance Imaging. The release from an exemplary LDDS was verified in vitro and in vivo using Fluorescence Molecular Tomography. The presented GBM partial resection model appears to be well suited to determine the efficiency of LDDS. By sharing our expertise, we intend to provide a powerful tool for the future testing of these very promising systems, paving their way into clinical application.
Collapse
Affiliation(s)
- Carolin Kubelt
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, 24105 Kiel, Germany
| | - Dana Hellmold
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, 24105 Kiel, Germany
| | - Eva Peschke
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, Kiel University, 24118 Kiel, Germany
| | - Margarethe Hauck
- Functional Nanomaterials, Department of Materials Science, Faculty of Engineering, Kiel University, 24143 Kiel, Germany
| | - Olga Will
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, Kiel University, 24118 Kiel, Germany
| | - Fabian Schütt
- Functional Nanomaterials, Department of Materials Science, Faculty of Engineering, Kiel University, 24143 Kiel, Germany
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, 24118 Kiel, Germany
| | - Ralph Lucius
- Institute of Anatomy, Kiel University, 24118 Kiel, Germany
| | - Rainer Adelung
- Functional Nanomaterials, Department of Materials Science, Faculty of Engineering, Kiel University, 24143 Kiel, Germany
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, 24118 Kiel, Germany
| | - Regina Scherließ
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, 24118 Kiel, Germany
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, 24118 Kiel, Germany
| | - Jan-Bernd Hövener
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, Kiel University, 24118 Kiel, Germany
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, 24118 Kiel, Germany
| | - Olav Jansen
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, 24118 Kiel, Germany
- Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, 24105 Kiel, Germany
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, 24105 Kiel, Germany
| | - Janka Held-Feindt
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, UKSH Campus Kiel, 24105 Kiel, Germany
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, 24118 Kiel, Germany
| |
Collapse
|
6
|
Duerinck J, Tuyaerts S, Movahedi K, Neyns B. Overcoming the immune suppressive nature of glioblastoma by leveraging the surgical intervention - current status and future perspectives. Front Immunol 2023; 14:1183641. [PMID: 37275902 PMCID: PMC10237336 DOI: 10.3389/fimmu.2023.1183641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Despite relentless efforts to improve outcome, the prognosis of glioblastoma (GBM) remains poor. Standard therapy at first diagnosis consists of maximal safe surgical resection followed by radiochemotherapy, but treatment options at recurrence are scarce and have limited efficacy. Immunotherapy is a broad term that covers several treatment strategies, including immune checkpoint inhibition (ICI). The successes of systemically administered therapeutic monoclonal antibodies that block the Programmed death receptor or ligand (PD-(L)1) and Cytotoxic T-Lymphocyte associated protein (CTLA)-4 immune checkpoints in other cancer types could not be reproduced in glioblastoma. This is considered to be related to the intrinsic low immunogenicity and strong immunosuppressive tumor microenvironment of glioblastoma, in addition to the presence of a blood-glioma and blood-brain barrier that limits many systemically administered therapeutic agents from reaching their target. In this mini-review, we address the specific aspects of immune suppression in glioblastoma and discuss potential strategies that could help to overcome it. The potential advantages of incorporating surgical resection in clinical trials of immunotherapy for glioblastoma, including window-of-opportunity studies, are highlighted. Combination strategies that include surgical resection, as well as local administration of therapeutic agents in the brain are discussed as a potential strategy to achieve an effective immunological response against glioblastoma.
Collapse
Affiliation(s)
- Johnny Duerinck
- Department of Neurosurgery, Universitair Ziekenhuis Brussel (UZ Brussels), Brussels, Belgium
- C4N - Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sandra Tuyaerts
- Laboratory for Medical & Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussels), Brussels, Belgium
| | - Kiavash Movahedi
- Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Bart Neyns
- Laboratory for Medical & Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussels), Brussels, Belgium
| |
Collapse
|
7
|
Lu ZG, Shen J, Yang J, Wang JW, Zhao RC, Zhang TL, Guo J, Zhang X. Nucleic acid drug vectors for diagnosis and treatment of brain diseases. Signal Transduct Target Ther 2023; 8:39. [PMID: 36650130 PMCID: PMC9844208 DOI: 10.1038/s41392-022-01298-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/08/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
Nucleic acid drugs have the advantages of rich target selection, simple in design, good and enduring effect. They have been demonstrated to have irreplaceable superiority in brain disease treatment, while vectors are a decisive factor in therapeutic efficacy. Strict physiological barriers, such as degradation and clearance in circulation, blood-brain barrier, cellular uptake, endosome/lysosome barriers, release, obstruct the delivery of nucleic acid drugs to the brain by the vectors. Nucleic acid drugs against a single target are inefficient in treating brain diseases of complex pathogenesis. Differences between individual patients lead to severe uncertainties in brain disease treatment with nucleic acid drugs. In this Review, we briefly summarize the classification of nucleic acid drugs. Next, we discuss physiological barriers during drug delivery and universal coping strategies and introduce the application methods of these universal strategies to nucleic acid drug vectors. Subsequently, we explore nucleic acid drug-based multidrug regimens for the combination treatment of brain diseases and the construction of the corresponding vectors. In the following, we address the feasibility of patient stratification and personalized therapy through diagnostic information from medical imaging and the manner of introducing contrast agents into vectors. Finally, we take a perspective on the future feasibility and remaining challenges of vector-based integrated diagnosis and gene therapy for brain diseases.
Collapse
Affiliation(s)
- Zhi-Guo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| | - Jie Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jing-Wen Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Rui-Chen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Tian-Lu Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| |
Collapse
|
8
|
Fahmy MA, Farghaly AA, Hassan EE, Hassan ZM, Abd-Alla HI. Protective role of Codiaeum variegatum against genotoxicity induced by carmustine in somatic and germ cells of male mice. Mol Biol Rep 2022; 49:9543-9553. [PMID: 36053281 PMCID: PMC9515021 DOI: 10.1007/s11033-022-07845-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Carmustine (Cr) is an important chemotherapeutic drug, widely used in the treatment of brain tumors. Herein, the protective role of Codiaeum variegatum leaves ethyl acetate fraction was determined against genotoxicity of Cr. The technique HPLC-qTOF-MS/MS was used to identify the constituents in C. variegatum. MATERIALS 90 male mice were used to evaluate micronuclei (MPCEs) in bone marrow, chromosomal aberration (CAs) in bone marrow and mouse spermatocytes, sperm abnormalities, and gene expression (qRT-PCR). The following groups were included, I: Negative control (ethanol 30%), II: Positive control (i.p injected once with 30 mg/kg Cr), III: Control orally treated with C. variegatum at 500 mg/kg, four days. IV-VI: treated with 100, 300, and 500 mg/kg of the plant (4 days) plus a single dose of Cr. RESULTS In bone marrow, Cr induced significant increase in MPCEs and CAs by 3 and 7-folds respectively over the control. Cr also induced a significant percentage of CAs in spermatocytes in meiosis in the form of univalent (X-Y and autosomal univalent) and also a significant percentage of morphological sperm abnormalities was recorded. A large number of coiled tail abnormalities were detected indicating the effect of Cr in sperm motility. Cr induced an overexpression of p53 gene. C. variegatum mitigated all deleterious genotoxic effects of Cr. Chemical analysis showed that flavones (35.21%) and phenolic acids (17.62%) constitute the main components. CONCLUSIONS The results indicated that Cr is genotoxic in both somatic and germ cells. The active components in C. variegatum together participate in the obtained protective role.
Collapse
Affiliation(s)
- Maha A Fahmy
- Department of Genetics and Cytology, National Research Centre, Giza, 12622, Egypt
| | - Ayman A Farghaly
- Department of Genetics and Cytology, National Research Centre, Giza, 12622, Egypt
| | - Entesar E Hassan
- Department of Genetics and Cytology, National Research Centre, Giza, 12622, Egypt
| | - Zeinab M Hassan
- Chemistry of Natural Compounds Department, National Research Centre, Giza, 12622, Egypt
| | - Howaida I Abd-Alla
- Chemistry of Natural Compounds Department, National Research Centre, Giza, 12622, Egypt.
| |
Collapse
|
9
|
Sun S, Kiang KMY, Leung GKK. Chaperone protein P4HB predicts temozolomide response and prognosis in malignant glioma. Oncol Lett 2022; 24:264. [PMID: 35765277 DOI: 10.3892/ol.2022.13385] [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: 03/03/2022] [Accepted: 05/12/2022] [Indexed: 11/06/2022] Open
Abstract
Prolyl 4-hydroxylase beta polypeptide (P4HB) is a chaperone protein associated with temozolomide (TMZ) resistance through the unfolded protein response. Cancer cells with constitutive activation of endoplasmic reticulum stress and upregulation of P4HB have been observed to show resistance against chemotherapies. The present study focused on the evaluation of the prognostic value of P4HB in subtypes of glioma with or without O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. P4HB expression was assessed by immunohistochemical staining in 73 grade I-IV gliomas and its association with the clinicopathological data was determined. It was indicated that P4HB expression was significantly associated with several parameters, including age, tumour grade and the number of TMZ treatment cycles received. In the Kaplan-Meier analysis, P4HB expression was positively associated with risk of mortality and disease progression. In patients treated with TMZ, high P4HB expression was significantly associated with poor overall survival (OS) and progression-free survival (PFS). The association between MGMT promoter methylation and P4HB expression was also assessed. Patients with MGMTMethP4HBLow tumours had the most favourable PFS (48 months) among cases with various combinations of MGMT methylation status and P4HB expression. Multivariate analysis revealed that P4HB may be used as an independent prognostic indicator of OS, particularly in high-grade gliomas. The present study uncovered the potential role of P4HB in a nuanced pathological stratification during clinical decision-making with respect to MGMT promoter methylation status and TMZ treatment.
Collapse
Affiliation(s)
- Stella Sun
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR 999077, P.R. China
| | - Karrie Mei-Yee Kiang
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR 999077, P.R. China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR 999077, P.R. China
| |
Collapse
|
10
|
El Kheir W, Marcos B, Virgilio N, Paquette B, Faucheux N, Lauzon MA. Drug Delivery Systems in the Development of Novel Strategies for Glioblastoma Treatment. Pharmaceutics 2022; 14:1189. [PMID: 35745762 PMCID: PMC9227363 DOI: 10.3390/pharmaceutics14061189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a grade IV glioma considered the most fatal cancer of the central nervous system (CNS), with less than a 5% survival rate after five years. The tumor heterogeneity, the high infiltrative behavior of its cells, and the blood-brain barrier (BBB) that limits the access of therapeutic drugs to the brain are the main reasons hampering the current standard treatment efficiency. Following the tumor resection, the infiltrative remaining GBM cells, which are resistant to chemotherapy and radiotherapy, can further invade the surrounding brain parenchyma. Consequently, the development of new strategies to treat parenchyma-infiltrating GBM cells, such as vaccines, nanotherapies, and tumor cells traps including drug delivery systems, is required. For example, the chemoattractant CXCL12, by binding to its CXCR4 receptor, activates signaling pathways that play a critical role in tumor progression and invasion, making it an interesting therapeutic target to properly control the direction of GBM cell migration for treatment proposes. Moreover, the interstitial fluid flow (IFF) is also implicated in increasing the GBM cell migration through the activation of the CXCL12-CXCR4 signaling pathway. However, due to its complex and variable nature, the influence of the IFF on the efficiency of drug delivery systems is not well understood yet. Therefore, this review discusses novel drug delivery strategies to overcome the GBM treatment limitations, focusing on chemokines such as CXCL12 as an innovative approach to reverse the migration of infiltrated GBM. Furthermore, recent developments regarding in vitro 3D culture systems aiming to mimic the dynamic peritumoral environment for the optimization of new drug delivery technologies are highlighted.
Collapse
Affiliation(s)
- Wiam El Kheir
- Advanced Dynamic Cell Culture Systems Laboratory, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
| | - Bernard Marcos
- Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
| | - Nick Virgilio
- Department of Chemical Engineering, Polytechnique Montréal, 2500 Chemin de Polytechnique, Montréal, QC H3T 1J4, Canada;
| | - Benoit Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada;
- Clinical Research Center of the Centre Hospitalier Universitaire de l’Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Nathalie Faucheux
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Clinical Research Center of the Centre Hospitalier Universitaire de l’Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Marc-Antoine Lauzon
- Advanced Dynamic Cell Culture Systems Laboratory, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Research Center on Aging, 1036 Rue Belvédère Sud, Sherbrooke, QC J1H 4C4, Canada
| |
Collapse
|
11
|
Li B, Chen X, Qiu W, Zhao R, Duan J, Zhang S, Pan Z, Zhao S, Guo Q, Qi Y, Wang W, Deng L, Ni S, Sang Y, Xue H, Liu H, Li G. Synchronous Disintegration of Ferroptosis Defense Axis via Engineered Exosome-Conjugated Magnetic Nanoparticles for Glioblastoma Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105451. [PMID: 35508804 PMCID: PMC9189685 DOI: 10.1002/advs.202105451] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/09/2022] [Indexed: 05/14/2023]
Abstract
Glioblastoma (GBM) is one of the most fatal central nervous system tumors and lacks effective or sufficient therapies. Ferroptosis is a newly discovered method of programmed cell death and opens a new direction for GBM treatment. However, poor blood-brain barrier (BBB) penetration, reduced tumor targeting ability, and potential compensatory mechanisms hinder the effectiveness of ferroptosis agents during GBM treatment. Here, a novel composite therapeutic platform combining the magnetic targeting features and drug delivery properties of magnetic nanoparticles with the BBB penetration abilities and siRNA encapsulation properties of engineered exosomes for GBM therapy is presented. This platform can be enriched in the brain under local magnetic localization and angiopep-2 peptide-modified engineered exosomes can trigger transcytosis, allowing the particles to cross the BBB and target GBM cells by recognizing the LRP-1 receptor. Synergistic ferroptosis therapy of GBM is achieved by the combined triple actions of the disintegration of dihydroorotate dehydrogenase and the glutathione peroxidase 4 ferroptosis defense axis with Fe3 O4 nanoparticle-mediated Fe2+ release. Thus, the present findings show that this system can serve as a promising platform for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Boyan Li
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Xin Chen
- State Key Laboratory of Crystal MaterialsShandong UniversityJinan250100P. R. China
| | - Wei Qiu
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Rongrong Zhao
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Jiazhi Duan
- State Key Laboratory of Crystal MaterialsShandong UniversityJinan250100P. R. China
| | - Shouji Zhang
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Ziwen Pan
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Shulin Zhao
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Qindong Guo
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Yanhua Qi
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Wenhan Wang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinan250100P. R. China
| | - Lin Deng
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Shilei Ni
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Yuanhua Sang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinan250100P. R. China
| | - Hao Xue
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| | - Hong Liu
- State Key Laboratory of Crystal MaterialsShandong UniversityJinan250100P. R. China
- Institute for Advanced Interdisciplinary Research (IAIR)University of JinanJinan250022P. R. China
| | - Gang Li
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinan250012P. R. China
| |
Collapse
|
12
|
Lastakchi S, Olaloko MK, McConville C. A Potential New Treatment for High-Grade Glioma: A Study Assessing Repurposed Drug Combinations against Patient-Derived High-Grade Glioma Cells. Cancers (Basel) 2022; 14:2602. [PMID: 35681582 PMCID: PMC9179370 DOI: 10.3390/cancers14112602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 02/05/2023] Open
Abstract
Repurposed drugs have demonstrated in vitro success against high-grade gliomas; however, their clinical success has been limited due to the in vitro model not truly representing the clinical scenario. In this study, we used two distinct patient-derived tumour fragments (tumour core (TC) and tumour margin (TM)) to generate a heterogeneous, clinically relevant in vitro model to assess if a combination of repurposed drugs (irinotecan, pitavastatin, disulfiram, copper gluconate, captopril, celecoxib, itraconazole and ticlopidine), each targeting a different growth promoting pathway, could successfully treat high-grade gliomas. To ensure the clinical relevance of our data, TC and TM samples from 11 different patients were utilized. Our data demonstrate that, at a concentration of 100µm or lower, all drug combinations achieved lower LogIC50 values than temozolomide, with one of the combinations almost eradicating the cancer by achieving cell viabilities below 4% in five of the TM samples 6 days after treatment. Temozolomide was unable to stop tumour growth over the 14-day assay, while combination 1 stopped tumour growth, with combinations 2, 3 and 4 slowing down tumour growth at higher doses. To validate the cytotoxicity data, we used two distinct assays, end point MTT and real-time IncuCyte life analysis, to evaluate the cytotoxicity of the combinations on the TC fragment from patient 3, with the cell viabilities comparable across both assays. The local administration of combinations of repurposed drugs that target different growth promoting pathways of high-grade gliomas have the potential to be translated into the clinic as a novel treatment strategy for high-grade gliomas.
Collapse
Affiliation(s)
| | | | - Christopher McConville
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (S.L.); (M.K.O.)
| |
Collapse
|
13
|
Hauck M, Hellmold D, Kubelt C, Synowitz M, Adelung R, Schütt F, Held‐Feindt J. Localized Drug Delivery Systems in High‐Grade Glioma Therapy – From Construction to Application. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Margarethe Hauck
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Dana Hellmold
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Carolin Kubelt
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Michael Synowitz
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Rainer Adelung
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Fabian Schütt
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Janka Held‐Feindt
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| |
Collapse
|
14
|
Fabrication and Modelling of a Reservoir-Based Drug Delivery System for Customizable Release. Pharmaceutics 2022; 14:pharmaceutics14040777. [PMID: 35456611 PMCID: PMC9025308 DOI: 10.3390/pharmaceutics14040777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Localized therapy approaches have emerged as an alternative drug administration route to overcome the limitations of systemic therapies, such as the crossing of the blood–brain barrier in the case of brain tumor treatment. For this, implantable drug delivery systems (DDS) have been developed and extensively researched. However, to achieve an effective localized treatment, the release kinetics of DDS needs to be controlled in a defined manner, so that the concentration at the tumor site is within the therapeutic window. Thus, a DDS, with patient-specific release kinetics, is crucial for the improvement of therapy. Here, we present a computationally supported reservoir-based DDS (rDDS) development towards patient-specific release kinetics. The rDDS consists of a reservoir surrounded by a polydimethylsiloxane (PDMS) microchannel membrane. By tailoring the rDDS, in terms of membrane porosity, geometry, and drug concentration, the release profiles can be precisely adapted, with respect to the maximum concentration, release rate, and release time. The release is investigated using a model dye for varying parameters, leading to different distinct release profiles, with a maximum release of up to 60 days. Finally, a computational simulation, considering exemplary in vivo conditions (e.g., exchange of cerebrospinal fluid), is used to study the resulting drug release profiles, demonstrating the customizability of the system. The establishment of a computationally supported workflow, for development towards a patient-specific rDDS, in combination with the transfer to suitable drugs, could significantly improve the efficacy of localized therapy approaches.
Collapse
|
15
|
Nance E, Pun SH, Saigal R, Sellers DL. Drug delivery to the central nervous system. NATURE REVIEWS. MATERIALS 2022; 7:314-331. [PMID: 38464996 PMCID: PMC10923597 DOI: 10.1038/s41578-021-00394-w] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 03/12/2024]
Abstract
Despite the rising global incidence of central nervous system (CNS) disorders, CNS drug development remains challenging, with high costs, long pathways to clinical use and high failure rates. The CNS is highly protected by physiological barriers, in particular, the blood-brain barrier and the blood-cerebrospinal fluid barrier, which limit access of most drugs. Biomaterials can be designed to bypass or traverse these barriers, enabling the controlled delivery of drugs into the CNS. In this Review, we first examine the effects of normal and diseased CNS physiology on drug delivery to the brain and spinal cord. We then discuss CNS drug delivery designs and materials that are administered systemically, directly to the CNS, intranasally or peripherally through intramuscular injections. Finally, we highlight important challenges and opportunities for materials design for drug delivery to the CNS and the anticipated clinical impact of CNS drug delivery.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Rajiv Saigal
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| |
Collapse
|
16
|
Singh M, Jindal D, Agarwal V, Pathak D, Sharma M, Pancham P, Mani S, Rachana. New phase therapeutic pursuits for targeted drug delivery in glioblastoma multiforme. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:866-888. [PMID: 36654821 PMCID: PMC9834280 DOI: 10.37349/etat.2022.00118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/19/2022] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is known as the most aggressive and prevalent brain tumor with a high mortality rate. It is reported in people who are as young as 10 years old to as old as over 70 years old, exhibiting inter and intra tumor heterogeneity. There are several genomic and proteomic investigations that have been performed to find the unexplored potential targets of the drug against GBM. Therefore, certain effective targets have been taken to further validate the studies embarking on the robustness in the field of medicinal chemistry followed by testing in clinical trials. Also, The Cancer Genome Atlas (TCGA) project has identified certain overexpressed targets involved in the pathogenesis of GBM in three major pathways, i.e., tumor protein 53 (p53), retinoblastoma (RB), and receptor tyrosine kinase (RTK)/rat sarcoma virus (Ras)/phosphoinositide 3-kinase (PI3K) pathways. This review focuses on the compilation of recent developments in the fight against GBM thus, directing future research into the elucidation of pathogenesis and potential cure for GBM. Also, it highlights the potential biomarkers that have undergone extensive research and have promising prognostic and predictive values. Additionally, this manuscript analyses the advent of gene therapy and immunotherapy, unlocking the way to consider treatment approaches other than, or in addition to, conventional chemo-radiation therapies. This review study encompasses all the relevant research studies associated with the pathophysiology, occurrence, diagnostic tools, and therapeutic intervention for GBM. It highlights the evolution of various therapeutic perspectives against GBM from the most conventional form of radiotherapy to the recent advancement of gene/cell/immune therapy. Further, the review focuses on various targeted therapies for GBM including chemotherapy sensitization, radiotherapy, nanoparticles based, immunotherapy, cell therapy, and gene therapy which would offer a comprehensive account for exploring several facets related to GBM prognostics.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India,Correspondence: Manisha Singh, Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India.
| | - Divya Jindal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Vinayak Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Deepanshi Pathak
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Mansi Sharma
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Pranav Pancham
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Rachana
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| |
Collapse
|
17
|
Rechberger JS, Thiele F, Daniels DJ. Status Quo and Trends of Intra-Arterial Therapy for Brain Tumors: A Bibliometric and Clinical Trials Analysis. Pharmaceutics 2021; 13:pharmaceutics13111885. [PMID: 34834300 PMCID: PMC8625566 DOI: 10.3390/pharmaceutics13111885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Intra-arterial drug delivery circumvents the first-pass effect and is believed to increase both efficacy and tolerability of primary and metastatic brain tumor therapy. The aim of this update is to report on pertinent articles and clinical trials to better understand the research landscape to date and future directions. Elsevier's Scopus and ClinicalTrials.gov databases were reviewed in August 2021 for all possible articles and clinical trials of intra-arterial drug injection as a treatment strategy for brain tumors. Entries were screened against predefined selection criteria and various parameters were summarized. Twenty clinical trials and 271 articles satisfied all inclusion criteria. In terms of articles, 201 (74%) were primarily clinical and 70 (26%) were basic science, published in a total of 120 different journals. Median values were: publication year, 1986 (range, 1962-2021); citation count, 15 (range, 0-607); number of authors, 5 (range, 1-18). Pertaining to clinical trials, 9 (45%) were phase 1 trials, with median expected start and completion years in 2011 (range, 1998-2019) and 2022 (range, 2008-2025), respectively. Only one (5%) trial has reported results to date. Glioma was the most common tumor indication reported in both articles (68%) and trials (75%). There were 215 (79%) articles investigating chemotherapy, while 13 (65%) trials evaluated targeted therapy. Transient blood-brain barrier disruption was the commonest strategy for articles (27%) and trials (60%) to optimize intra-arterial therapy. Articles and trials predominately originated in the United States (50% and 90%, respectively). In this bibliometric and clinical trials analysis, we discuss the current state and trends of intra-arterial therapy for brain tumors. Most articles were clinical, and traditional anti-cancer agents and drug delivery strategies were commonly studied. This was reflected in clinical trials, of which only a single study had reported outcomes. We anticipate future efforts to involve novel therapeutic and procedural strategies based on recent advances in the field.
Collapse
Affiliation(s)
- Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA;
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence:
| | - Frederic Thiele
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA;
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
18
|
Alghamdi M, Gumbleton M, Newland B. Local delivery to malignant brain tumors: potential biomaterial-based therapeutic/adjuvant strategies. Biomater Sci 2021; 9:6037-6051. [PMID: 34357362 DOI: 10.1039/d1bm00896j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most aggressive malignant brain tumor and is associated with a very poor prognosis. The standard treatment for newly diagnosed patients involves total tumor surgical resection (if possible), plus irradiation and adjuvant chemotherapy. Despite treatment, the prognosis is still poor, and the tumor often recurs within two centimeters of the original tumor. A promising approach to improving the efficacy of GBM therapeutics is to utilize biomaterials to deliver them locally at the tumor site. Local delivery to GBM offers several advantages over systemic administration, such as bypassing the blood-brain barrier and increasing the bioavailability of the therapeutic at the tumor site without causing systemic toxicity. Local delivery may also combat tumor recurrence by maintaining sufficient drug concentrations at and surrounding the original tumor area. Herein, we critically appraised the literature on local delivery systems based within the following categories: polymer-based implantable devices, polymeric injectable systems, and hydrogel drug delivery systems. We also discussed the negative effect of hypoxia on treatment strategies and how one might utilize local implantation of oxygen-generating biomaterials as an adjuvant to enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Faculty of Pharmacy, King Abdulaziz University, Jeddah, 22522, Kingdom of Saudi Arabia
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| |
Collapse
|
19
|
Watts C, Ashkan K, Jenkinson MD, Price SJ, Santarius T, Matys T, Zhang TT, Finch A, Collins P, Allinson K, Jefferies SJ, Scoffings DJ, Zisakis A, Phillips M, Wanek K, Smith P, Clifton-Hadley L, Counsell N. An Evaluation of the Tolerability and Feasibility of Combining 5-Amino-Levulinic Acid (5-ALA) with BCNU Wafers in the Surgical Management of Primary Glioblastoma. Cancers (Basel) 2021; 13:cancers13133241. [PMID: 34209555 PMCID: PMC8267684 DOI: 10.3390/cancers13133241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 02/02/2023] Open
Abstract
Simple Summary This reseach explored the safety and feasibility of combining local chemotherapy with fluorescence-guided resection in patients with a brain cancer, glioblastoma. The aim was to determine if the combination of fluorescence-guided surgery using 5-aminolevulinic acid and BCNU wafers left in the tumour cavity at the end of the operation was safe and did not prevent patients getting subsequent chemo-radiotherapy. The results showed that combining local chemotherapy with fluorescence-guided resection was tolerable in terms of surgical morbidity and overall toxicity. However, any potential therapeutic benefit requires further investigation, preferably with improved local delivery technologies. Abstract Background Glioblastoma (GBM) is the commonest primary malignant brain tumour in adults and effective treatment options are limited. Combining local chemotherapy with enhanced surgical resection using 5-aminolevulinic acid (5-ALA) could improve outcomes. Here we assess the safety and feasibility of combining BCNU wafers with 5-ALA-guided surgery. Methods We conducted a multicentre feasibility study of 5-ALA with BCNU wafers followed by standard-of-care chemoradiotherapy (chemoRT) in patients with suspected GBM. Patients judged suitable for radical resection were administered 5-ALA pre-operatively and BCNU wafers at the end resection. Post-operative treatment continued as per routine clinical practice. The primary objective was to establish if combining 5-ALA and BCNU wafers is safe without compromising patients from receiving standard chemoRT. Results Seventy-two patients were recruited, sixty-four (88.9%) received BCNU wafer implants, and fifty-nine (81.9%) patients remained eligible following formal histological diagnosis. Seven (11.9%) eligible patients suffered surgical complications but only two (3.4%) were not able to begin chemoRT, four (6.8%) additional patients did not begin chemoRT within 6 weeks of surgery due to surgical complications. Eleven (18.6%) patients did not begin chemoRT for other reasons (other toxicity (n = 3), death (n = 3), lost to follow-up/withdrew (n = 3), clinical decision (n = 1), poor performance status (n = 1)). Median progression-free survival was 8.7 months (95% CI: 6.4–9.8) and median overall survival was 14.7 months (95% CI: 11.7–16.8). Conclusions Combining BCNU wafers with 5-ALA-guided surgery in newly diagnosed GBM patients is both feasible and tolerable in terms of surgical morbidity and overall toxicity. Any potential therapeutic benefit for the sequential use of 5-ALA and BCNU with chemoRT requires further investigation with improved local delivery technologies.
Collapse
Affiliation(s)
- Colin Watts
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK;
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham B15 2WB, UK;
- Correspondence:
| | - Keyoumars Ashkan
- Department of Neurosurgery, King’s College Hospital, London SE5 9RS, UK;
| | - Michael D. Jenkinson
- Department of Neurosurgery, The Walton Centre, Liverpool L9 7LJ, UK;
- Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Stephen J. Price
- Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Thomas Santarius
- Academic Neurosurgery Department, University of Cambridge, Cambridge CB2 0QQ, UK;
| | - Tomasz Matys
- Department of Clinical Neurosciences, Cambridge University Hospitals Foundation Trust, Cambridge CB2 0QQ, UK; (T.S.); (P.C.)
| | - Ting Ting Zhang
- Department of Clinical Neurosciences, Cambridge University Hospitals Foundation Trust, Cambridge CB2 0QQ, UK; (T.S.); (P.C.)
| | - Alina Finch
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Peter Collins
- Academic Neurosurgery Department, University of Cambridge, Cambridge CB2 0QQ, UK;
| | - Kieren Allinson
- Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals Foundation Trust, Cambridge CB2 0QQ, UK; (T.M.); (T.T.Z.); (D.J.S.)
| | - Sarah J. Jefferies
- Department of Histopathology, Cambridge University Hospitals Foundation Trust, Cambridge CB2 0QQ, UK;
| | - Daniel J. Scoffings
- Department of Clinical Neurosciences, Cambridge University Hospitals Foundation Trust, Cambridge CB2 0QQ, UK; (T.S.); (P.C.)
| | - Athanasios Zisakis
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham B15 2WB, UK;
| | - Mark Phillips
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK;
| | - Katharina Wanek
- Cancer Institute, University College London, London WC1E 6DD, UK;
| | - Paul Smith
- Cancer Institute, University College London, London WC1E 6DD, UK;
| | | | | |
Collapse
|
20
|
Yang TC, Liu SJ, Lo WL, Chen SM, Tang YL, Tseng YY. Enhanced Anti-Tumor Activity in Mice with Temozolomide-Resistant Human Glioblastoma Cell Line-Derived Xenograft Using SN-38-Incorporated Polymeric Microparticle. Int J Mol Sci 2021; 22:ijms22115557. [PMID: 34074038 PMCID: PMC8197307 DOI: 10.3390/ijms22115557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) has remained one of the most lethal and challenging cancers to treat. Previous studies have shown encouraging results when irinotecan was used in combination with temozolomide (TMZ) for treating GBM. However, irinotecan has a narrow therapeutic index: a slight dose increase in irinotecan can induce toxicities that outweigh its therapeutic benefits. SN-38 is the active metabolite of irinotecan that accounts for both its anti-tumor efficacy and toxicity. In our previous paper, we showed that SN-38 embedded into 50:50 biodegradable poly[(d,l)-lactide-co-glycolide] (PLGA) microparticles (SMPs) provides an efficient delivery and sustained release of SN-38 from SMPs in the brain tissues of rats. These properties of SMPs give them potential for therapeutic application due to their high efficacy and low toxicity. In this study, we tested the anti-tumor activity of SMP-based interstitial chemotherapy combined with TMZ using TMZ-resistant human glioblastoma cell line-derived xenograft models. Our data suggest that treatment in which SMPs are combined with TMZ reduces tumor growth and extends survival in mice bearing xenograft tumors derived from both TMZ-resistant and TMZ-sensitive human glioblastoma cell lines. Our findings demonstrate that combining SMPs with TMZ may have potential as a promising strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Tao-Chieh Yang
- Department of Neurosurgery, School of Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (S.-J.L.); (Y.-L.T.)
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33302, Taiwan
| | - Wei-Lun Lo
- Division of Neurosurgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan;
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
| | - Shu-Mei Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110301, Taiwan
| | - Ya-Ling Tang
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (S.-J.L.); (Y.-L.T.)
| | - Yuan-Yun Tseng
- Division of Neurosurgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan;
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
- Correspondence: ; Tel.: +886-2-22490088 (ext. 8120); Fax: +886-2-22480900
| |
Collapse
|
21
|
Mansouri A, Beyn ME, Pancholi A, Chow CT, Wang R, Boutet A, Elias GJB, Germann J, Loh A, Voisin MR, Lozano AM, Chiocca EA, Vogelbaum MA, Zadeh G. Evolution of the Neurosurgeon's Role in Clinical Trials for Glioblastoma: A Systematic Overview of the Clinicaltrials.Gov Database. Neurosurgery 2021; 89:196-203. [PMID: 33989408 DOI: 10.1093/neuros/nyab169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 03/12/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The therapeutic challenge of glioblastoma (GBM) has catalyzed the development of clinical trials to evaluate novel interventions. With increased understanding of GBM biology and technological advances, the neurosurgeon's role in neuro-oncology has evolved. OBJECTIVE To evaluate the current landscape of procedure-based clinical trials for GBM to characterize this evolution, gain insight into past failures, and accordingly outline implications for future research and practice that may inform future studies. METHODS The ClinicalTrials.gov database was searched for surgical/procedural trials in individuals with GBM. Demographics, specific intervention, trial phase, and main outcome measures were abstracted. RESULTS A total of 224 of 2311 GBM trials (9.7%) were identified as procedural, with the majority being based in the United States (155/224, 69.2%), single-center (155/224, 69.2%), and not randomized (176/224, 78.6%). Primary and recurrent GBMs were evenly addressed. The leading interventions were local delivery of therapeutics (50.0%), surgical techniques (33.9%), such as image-guided surgery, and novel device applications (14.3%). Phase I designs predominated (82/224, 36.6%). The top primary outcome was safety/tolerability/feasibility (88/224, 39.3%), followed by survival (46/224, 20.5%). Approximately 17% of studies were terminated, withdrawn, or suspended. Fifty-two linked publications were identified, among which 42 were classified as having a positive result. CONCLUSION Procedural interventions comprised ∼10% of all registered GBM trials. Local delivery of therapeutics, use of surgical imaging techniques and novel device applications, predominantly through phase I designs, represent the evolved role of the neurosurgeon in neuro-oncology. Improved reporting of trial designs, outcomes, and results are needed to better inform the field and increase efficiency.
Collapse
Affiliation(s)
- Alireza Mansouri
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, Pennsylvania, USA.,Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | | | | | | | - Ryan Wang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alexandre Boutet
- University Health Network, Toronto, Ontario, Canada.,Joint Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Aaron Loh
- University Health Network, Toronto, Ontario, Canada
| | - Mathew R Voisin
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Andres M Lozano
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.,Krembil Research Institute, Toronto, Ontario, Canada
| | - E Antonio Chiocca
- Harvey W. Cushing Neuro-Oncology Laboratories (HCNL), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Gelareh Zadeh
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.,MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Recent Advances in the Use of Lipid-Based Nanoparticles Against Glioblastoma Multiforme. Arch Immunol Ther Exp (Warsz) 2021; 69:8. [PMID: 33772646 DOI: 10.1007/s00005-021-00609-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant brain tumor in adults. Although the overall incidence is less than 10 per 100,000 individuals, its poor prognosis and low survival rate make GBM a crucial public health issue. The main challenges for GBM treatment are related to tumor location and its complex and heterogeneous biology. In this sense, a broad range of nanoparticles with different sizes, architectures, and surface properties, have been engineered as brain drug delivery systems. Among them, lipid-based nanoparticles, such as liposomes, have been pointed out as promising materials to deliver antitumoral drugs to the central nervous system and thus, to improve brain drug targeting and therapeutic efficiency. Here, we describe the synthesis and general characteristics of lipid-based nanoparticles, as well as evidence in the past 5 years regarding their potential use to treat GBM.
Collapse
|
23
|
Poonaki E, Ariakia F, Jalili-Nik M, Shafiee Ardestani M, Tondro G, Samini F, Ghasemi S, Sahab-Negah S, Gorji A. Targeting BMI-1 with PLGA–PEG nanoparticle-containing PTC209 modulates the behavior of human glioblastoma stem cells and cancer cells. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00078-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AbstractDespite advances in glioblastoma (GBM) treatments, current approaches have failed to improve the overall survival of patients. The oncogene BMI-1, a core member of the polycomb group proteins, is a potential novel therapeutic target for GBM. To enhance the efficacy and reduce the toxicity, PTC209, a BMI-1 inhibitor, was loaded into a PLGA–PEG nanoparticle conjugated with CD133 antibody (Nano-PTC209) and its effect on the behavior of human GBM stem-like cells (GSCs) and the human glioblastoma cell line (U87MG) was assessed. Nano-PTC209 has a diameter of ~ 75 nm with efficient drug loading and controlled release. The IC50 values of Nano-PTC209 for GSCs and U87MG cells were considerably lower than PTC209. Nano-PTC209 significantly decreased the viability of both GSCs and U87MG cells in a dose-dependent manner and caused a significant enhancement of apoptosis and p53 levels as well as inhibition of AKT and JNK signaling pathways. Furthermore, Nano-PTC209 significantly inhibited the migration ability, decreased the activity of metalloproteinase-2 and -9, and increased the generation of reactive oxygen species in both GSCs and U87MG cells. Our data indicate that PLGA–PEG nanoparticle conjugated with CD133 antibody could be an ideal nanocarrier to deliver PTC209 and effectively target BMI-1 for potential approaches in the treatment of GBM.
Collapse
|
24
|
Zhou X, Smith QR, Liu X. Brain penetrating peptides and peptide-drug conjugates to overcome the blood-brain barrier and target CNS diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1695. [PMID: 33470550 DOI: 10.1002/wnan.1695] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Nearly one in six people worldwide suffer from disorders of the central nervous system (CNS). There is an urgent need for effective strategies to improve the success rates in CNS drug discovery and development. The lack of effective technologies for delivering drugs and genes to the brain due to the blood-brain barrier (BBB), a structural barrier that effectively blocks most neurotherapeutic agents from reaching the brain, has posed a formidable hurdle for CNS drug development. Brain-homing and brain-penetrating molecular transport vectors, such as brain permeable peptides or BBB shuttle peptides, have shown promise in overcoming the BBB and ferrying the drug molecules to the brain. The BBB shuttle peptides are discovered by phage display technology or derived from natural neurotropic proteins or certain viruses and harness the receptor-mediated transcytosis molecular machinery for crossing the BBB. Brain permeable peptide-drug conjugates (PDCs), composed of BBB shuttle peptides, linkers, and drug molecules, have emerged as a promising CNS drug delivery system by taking advantage of the endogenous transcytosis mechanism and tricking the brain into allowing these bioactive molecules to pass the BBB. Here, we examine the latest development of brain-penetrating peptide shuttles and brain-permeable PDCs as molecular vectors to deliver small molecule drug payloads across the BBB to reach brain parenchyma. Emerging knowledge of the contribution of the peptides and their specific receptors expressed on the brain endothelial cells, choice of drug payloads, the design of PDCs, brain entry mechanisms, and delivery efficiency to the brain are highlighted. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Quentin R Smith
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| |
Collapse
|
25
|
Grego EA, Siddoway AC, Uz M, Liu L, Christiansen JC, Ross KA, Kelly SM, Mallapragada SK, Wannemuehler MJ, Narasimhan B. Polymeric Nanoparticle-Based Vaccine Adjuvants and Delivery Vehicles. Curr Top Microbiol Immunol 2021; 433:29-76. [PMID: 33165869 PMCID: PMC8107186 DOI: 10.1007/82_2020_226] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As vaccine formulations have progressed from including live or attenuated strains of pathogenic components for enhanced safety, developing new adjuvants to more effectively generate adaptive immune responses has become necessary. In this context, polymeric nanoparticles have emerged as a promising platform with multiple advantages, including the dual capability of adjuvant and delivery vehicle, administration via multiple routes, induction of rapid and long-lived immunity, greater shelf-life at elevated temperatures, and enhanced patient compliance. This comprehensive review describes advances in nanoparticle-based vaccines (i.e., nanovaccines) with a particular focus on polymeric particles as adjuvants and delivery vehicles. Examples of the nanovaccine approach in respiratory infections, biodefense, and cancer are discussed.
Collapse
Affiliation(s)
- Elizabeth A Grego
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Alaric C Siddoway
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Metin Uz
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Luman Liu
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - John C Christiansen
- Departments of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, 50011, USA
| | - Kathleen A Ross
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Sean M Kelly
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Surya K Mallapragada
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Michael J Wannemuehler
- Departments of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, 50011, USA
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Balaji Narasimhan
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA.
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
26
|
Griffith JI, Rathi S, Zhang W, Zhang W, Drewes LR, Sarkaria JN, Elmquist WF. Addressing BBB Heterogeneity: A New Paradigm for Drug Delivery to Brain Tumors. Pharmaceutics 2020; 12:E1205. [PMID: 33322488 PMCID: PMC7763839 DOI: 10.3390/pharmaceutics12121205] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Effective treatments for brain tumors remain one of the most urgent and unmet needs in modern oncology. This is due not only to the presence of the neurovascular unit/blood-brain barrier (NVU/BBB) but also to the heterogeneity of barrier alteration in the case of brain tumors, which results in what is referred to as the blood-tumor barrier (BTB). Herein, we discuss this heterogeneity, how it contributes to the failure of novel pharmaceutical treatment strategies, and why a "whole brain" approach to the treatment of brain tumors might be beneficial. We discuss various methods by which these obstacles might be overcome and assess how these strategies are progressing in the clinic. We believe that by approaching brain tumor treatment from this perspective, a new paradigm for drug delivery to brain tumors might be established.
Collapse
Affiliation(s)
- Jessica I. Griffith
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Sneha Rathi
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Wenqiu Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Wenjuan Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School—Duluth, Duluth, MN 55812, USA;
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902, USA;
| | - William F. Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| |
Collapse
|
27
|
Lv QL, Wang LC, Li DC, Lin QX, Shen XL, Liu HY, Li M, Ji YL, Qin CZ, Chen SH. Knockdown lncRNA DLEU1 Inhibits Gliomas Progression and Promotes Temozolomide Chemosensitivity by Regulating Autophagy. Front Pharmacol 2020; 11:560543. [PMID: 33362537 PMCID: PMC7756250 DOI: 10.3389/fphar.2020.560543] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Gliomas are the most fatal malignant cerebral tumors. Temozolomide (TMZ), as the primary chemotherapy drug, has been widely used in clinics. However, resistance of TMZ still remains to poor defined. LncRNAs have been reported to play crucial roles in progression of various cancers and resistance of multiple drugs. However, the biological function and underlying mechanisms of most lncRNAs in glioma still remains unclear. Based on the TCGA database, a total of 94 differentially expressed lncRNAs, including 16 up-regulated genes and 78 downregulated genes were identified between gliomas and normal brain tissues. Subsequently, lncRNA DLEU1, HOTAIR, and LOC00132111 were tested to be significantly related to overall survival (OS) between high- and low-expression groups. Additionally, we verified that lncRNA DLEU1 was high expressed in 108 gliomas, compared with 19 normal brain tissues. And high expression of lncRNA DLEU1 predicted a poor prognosis (HR = 1.703, 95%CI: 1.133–2.917, p-value = 0.0159). Moreover, functional assays revealed that knockdown of lncRNA DLEU1 could suppress the proliferation by inducing cell cycle arrest at G1 phase and reducing the S phase by down-regulating the CyclinD1 and p-AKT, as the well as migration and invasion by inhibiting the epithelial–mesenchymal transition (EMT) markers, such as ZEB1, N-cadherin, β-catenin and snail in glioma cells. Furthermore, silencing lncRNA DLEU1 suppressed TMZ-activated autophagy via regulating the expression of P62 and LC3, and promoted sensitivity of glioma cells to TMZ by triggering apoptosis. Conclusively, our study indicated that lncRNA DLEU1 might perform as a prognostic potential target and underlying therapeutic target for sensitivity of glioma to TMZ.
Collapse
Affiliation(s)
- Qiao-Li Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Department of Head and Neck Surgery, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Li-Chong Wang
- Jiangxi Key Laboratory of Translational Cancer Research, Department of Head and Neck Surgery, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China.,Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dang-Chi Li
- Jiangxi University of Technology High School, Nanchang, China
| | - Qian-Xia Lin
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Xiao-Li Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hai-Yun Liu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Min Li
- Department of Head and Neck Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Yu-Long Ji
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Chong-Zhen Qin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shu-Hui Chen
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
28
|
Abstract
Although surgical resection of the solid tumor component of glioblastoma has been shown to provide a survival advantage, it will never be a curative procedure. Yet, systemically applied adjuvants (radiation therapy and chemotherapy) also are not curative and their options are limited by the inability of most agents to cross the blood-brain barrier. Direct delivery of adjuvant therapies during a surgical procedure potentially provides an approach to bypass the blood-brain barrier and effectively treat residual tumor cells. This article summarizes the approaches and therapeutics that have been evaluated to date, and challenges that remain to be overcome.
Collapse
|
29
|
Alghamdi M, Chierchini F, Eigel D, Taplan C, Miles T, Pette D, Welzel PB, Werner C, Wang W, Neto C, Gumbleton M, Newland B. Poly(ethylene glycol) based nanotubes for tuneable drug delivery to glioblastoma multiforme. NANOSCALE ADVANCES 2020; 2:4498-4509. [PMID: 36132909 PMCID: PMC9418774 DOI: 10.1039/d0na00471e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/20/2020] [Indexed: 06/16/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of malignant brain tumour, which is associated with a poor two-year survival rate and a high rate of fatal recurrence near the original tumour. Focal/local drug delivery devices hold promise for improving therapeutic outcomes for GBM by increasing drug concentrations locally at the tumour site, or by facilitating the use of potent anti-cancer drugs that are poorly permeable across the blood brain barrier (BBB). For inoperable tumours, stereotactic delivery to the tumour necessitates the development of nanoscale/microscale injectable drug delivery devices. Herein we assess the ability of a novel class of polymer nanotube (based on poly(ethylene glycol) (PEG)) to load doxorubicin (a mainstay breast cancer therapeutic with poor BBB permeability) and release it slowly. The drug loading properties of the PEG nanotubes could be tuned by varying the degree of carboxylic acid functionalisation and hence the capacity of the nanotubes to electrostatically bind and load doxorubicin. 70% of the drug was released over the first seven days followed by sustained drug release for the remaining two weeks tested. Unloaded PEG nanotubes showed no toxicity to any of the cell types analysed, whereas doxorubicin loaded nanotubes decreased GBM cell viability (C6, U-87 and U-251) in a dose dependent manner in 2D in vitro culture. Finally, doxorubicin loaded PEG nanotubes significantly reduced the viability of in vitro 3D GBM models whilst unloaded nanotubes showed no cytotoxicity. Taken together, these findings show that polymer nanotubes could be used to deliver alternative anti-cancer drugs for local therapeutic strategies against brain cancers.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
- School of Pharmacy, King Abdulaziz University Jeddah 21589 Saudi Arabia
| | - Filippo Chierchini
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Christian Taplan
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Thomas Miles
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Dagmar Pette
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Petra B Welzel
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin Ireland
| | - Catia Neto
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| |
Collapse
|
30
|
Gomez-Zepeda D, Taghi M, Scherrmann JM, Decleves X, Menet MC. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019; 12:pharmaceutics12010020. [PMID: 31878061 PMCID: PMC7022905 DOI: 10.3390/pharmaceutics12010020] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Drug delivery into the brain is regulated by the blood-brain interfaces. The blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the blood-arachnoid barrier (BAB) regulate the exchange of substances between the blood and brain parenchyma. These selective barriers present a high impermeability to most substances, with the selective transport of nutrients and transporters preventing the entry and accumulation of possibly toxic molecules, comprising many therapeutic drugs. Transporters of the ATP-binding cassette (ABC) superfamily have an important role in drug delivery, because they extrude a broad molecular diversity of xenobiotics, including several anticancer drugs, preventing their entry into the brain. Gliomas are the most common primary tumors diagnosed in adults, which are often characterized by a poor prognosis, notably in the case of high-grade gliomas. Therapeutic treatments frequently fail due to the difficulty of delivering drugs through the brain barriers, adding to diverse mechanisms developed by the cancer, including the overexpression or expression de novo of ABC transporters in tumoral cells and/or in the endothelial cells forming the blood-brain tumor barrier (BBTB). Many models have been developed to study the phenotype, molecular characteristics, and function of the blood-brain interfaces as well as to evaluate drug permeability into the brain. These include in vitro, in vivo, and in silico models, which together can help us to better understand their implication in drug resistance and to develop new therapeutics or delivery strategies to improve the treatment of pathologies of the central nervous system (CNS). In this review, we present the principal characteristics of the blood-brain interfaces; then, we focus on the ABC transporters present on them and their implication in drug delivery; next, we present some of the most important models used for the study of drug transport; finally, we summarize the implication of ABC transporters in glioma and the BBTB in drug resistance and the strategies to improve the delivery of CNS anticancer drugs.
Collapse
Affiliation(s)
- David Gomez-Zepeda
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| | - Méryam Taghi
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Xavier Decleves
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Biologie du médicament et toxicologie, Hôpital Cochin, AP HP, 75006 Paris, France
| | - Marie-Claude Menet
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Hormonologie adulte, Hôpital Cochin, AP HP, 75006 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| |
Collapse
|
31
|
Banstola A, Pham TT, Jeong JH, Yook S. Polydopamine-tailored paclitaxel-loaded polymeric microspheres with adhered NIR-controllable gold nanoparticles for chemo-phototherapy of pancreatic cancer. Drug Deliv 2019; 26:629-640. [PMID: 31237149 PMCID: PMC6598510 DOI: 10.1080/10717544.2019.1628118] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022] Open
Abstract
Chemotherapeutic drugs often used as a first-line treatment of pancreatic cancer (PC) exhibit challenges due to resistance development, lack of selectivity, and tumor heterogeneity. Currently, combination chemo-photothermal therapy is known to enhance the therapeutic efficacy of chemotherapeutic drugs in PC. In this study, we develop adherent gold nanoparticles (GNPs) and paclitaxel (PTX)-loaded PLGA microspheres for the treatment of PC. Polydopamine (pD) was used as a linker to adhere GNPs to the surface of PLGA-Ms and characterized using TEM. Short-term cytotoxicity of GNPs-pD-PTX-PLGA-Ms with or without NIR treatment was evaluated using CCK-8 assays. ROS and western blot assay were performed to determine the intensity of ROS following the treatment of GNPs-pD-PTX-PLGA-Ms with or without NIR in Panc-1 cell line. Successful adhesion of GNPs on the microspheres was confirmed by TEM. CCK-8 assay revealed that GNPs-pD-PTX-PLGA-Ms with NIR showed three-fold higher cytotoxicity, compared to the group without NIR. Furthermore, ROS and western blot assay suggest that GNPs-pD-PTX-PLGA-Ms with NIR showed more ROS generation, followed by downregulation of the expression levels of antioxidant enzyme (SOD2 and CATALASE). These results suggest that the GNPs-pD-PTX-PLGA-Ms in combination with NIR irradiation can provide a synergistic chemo-photothermal therapy for the treatment of PC.
Collapse
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Tung Thanh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu, South Korea
| |
Collapse
|
32
|
Duwa R, Emami F, Lee S, Jeong JH, Yook S. Polymeric and lipid-based drug delivery systems for treatment of glioblastoma multiforme. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2019.06.050] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Wen ZP, Zeng WJ, Chen YH, Li H, Wang JY, Cheng Q, Yu J, Zhou HH, Liu ZZ, Xiao J, Chen XP. Knockdown ATG4C inhibits gliomas progression and promotes temozolomide chemosensitivity by suppressing autophagic flux. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:298. [PMID: 31291988 PMCID: PMC6617611 DOI: 10.1186/s13046-019-1287-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/24/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Gliomas are the most common primary tumors in central nervous system. Despite advances in diagnosis and therapy, the prognosis of glioma remains gloomy. Autophagy is a cellular catabolic process that degrades proteins and damaged organelles, which is implicated in tumorigenesis and tumor progression. Autophagy related 4C cysteine peptidase (ATG4C) is an autophagy regulator responsible for cleaving of pro-LC3 and delipidation of LC3 II. This study was designed to investigate the role of ATG4C in glioma progression and temozolomide (TMZ) chemosensitivity. METHODS The association between ATG4C mRNA expression and prognosis of gliomas patients was analyzed using the TCGA datasets. The role of ATG4C in proliferation, apoptosis, autophagy, and TMZ chemosensitivity were investigated by silencing ATG4C in vivo. Ectopic xenograft nude mice model was established to investigate the effects of ATG4C on glioma growth in vivo. RESULTS The median overall survival (OS) time of patients with higher ATG4C expression was significantly reduced (HR: 1.48, p = 9.91 × 10- 7). ATG4C mRNA expression was evidently increased with the rising of glioma grade (p = 2.97 × 10- 8). Knockdown ATG4C suppressed glioma cells proliferation by inducing cell cycle arrest at G1 phase. ATG4C depletion suppressed autophagy and triggered apoptosis through ROS accumulation. Depletion of ATG4C suppressed TMZ-activated autophagy and promoted sensitivity of glioma cells to TMZ. Additionally, ATG4C knockdown suppressed the growth of glioma remarkably in nude mice. CONCLUSION ATG4C is a potential prognostic predictor for glioma patient. Targeting ATG4C may provide promising therapy strategies for gliomas treatment.
Collapse
Affiliation(s)
- Zhi-Peng Wen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Wen-Jing Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Yan-Hong Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Medical Genetic Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, Hunan, People's Republic of China
| | - He Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Jie-Ya Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Jing Yu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Zheng-Zheng Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan province, China.
| | - Jian Xiao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China. .,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
34
|
Clavreul A, Pourbaghi-Masouleh M, Roger E, Menei P. Nanocarriers and nonviral methods for delivering antiangiogenic factors for glioblastoma therapy: the story so far. Int J Nanomedicine 2019; 14:2497-2513. [PMID: 31040671 PMCID: PMC6461002 DOI: 10.2147/ijn.s194858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is an essential component of glioblastoma (GB) progression. The development of angiogenesis inhibitor therapy, including treatments targeting vascular endothelial growth factor (VEGF) in particular, raised new hopes for the treatment of GB, but no Phase III clinical trial to date has reported survival benefits relative to standard treatment. There are several possible reasons for this limited efficacy, including VEGF-independent angiogenesis, induction of tumor invasion, and inefficient antiangiogenic factor delivery to the tumor. Efforts have been made to overcome these limitations by identifying new angiogenesis inhibitors that target angiogenesis through different mechanisms of action without inducing tumor invasion, and through the development of viral and nonviral delivery methods to improve antiangiogenic activity. Herein, we describe the nonviral methods, including convection-enhanced delivery devices, implantable polymer devices, nanocarriers, and cellular vehicles, to deliver antiangiogenic factors. We focus on those evaluated in intracranial (orthotopic) animal models of GB, the most relevant models of this disease, as they reproduce the clinical scenario of tumor progression and therapy response encountered in GB patients.
Collapse
Affiliation(s)
- Anne Clavreul
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| | - Milad Pourbaghi-Masouleh
- CRCINA, INSERM, University of Nantes, University of Angers, Angers, France, .,Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emilie Roger
- MINT, INSERM 1066, CNRS 6021, University of Angers, Angers, France
| | - Philippe Menei
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| |
Collapse
|
35
|
Su BC, Pan CY, Chen JY. Antimicrobial Peptide TP4 Induces ROS-Mediated Necrosis by Triggering Mitochondrial Dysfunction in Wild-Type and Mutant p53 Glioblastoma Cells. Cancers (Basel) 2019; 11:cancers11020171. [PMID: 30717309 PMCID: PMC6406555 DOI: 10.3390/cancers11020171] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
Antimicrobial peptide tilapia piscidin 4 (TP4) from Oreochromis niloticus exhibits potent bactericidal and anti-tumorigenic effects. In a variety of cancers, the mutation status of p53 is a decisive factor for therapeutic sensitivity. Therefore, we investigated the impact of p53 status on TP4-induced cytotoxicity in glioblastoma cell lines and the molecular mechanisms that govern cytotoxic effects. Both U87MG (wild-type/WT p53) and U251 (mutant p53) glioblastoma cell lines were sensitive to TP4-induced cytotoxicity. The necrosis inhibitors Necrostatin-1 and GSK’872 attenuated TP4-induced cytotoxicity, and TP4 treatment induced the release of cyclophilin A, a biomarker of necrosis. Moreover, TP4 induced mitochondrial hyperpolarization and dysfunction, which preceded the elevation of intracellular reactive oxygen species, DNA damage, and necrotic cell death in both U87MG and U251 glioblastoma cells. p38 was also activated by TP4, but did not contribute to cytotoxicity. SB202190, a specific p38 inhibitor, enhanced TP4-induced oxidative stress, mitochondrial dysfunction, and cytotoxicity, suggesting a protective role of p38. Furthermore, TP4-induced cytotoxicity, oxidative stress, phosphorylation of p38, and DNA damage were all attenuated by the mitochondrial-targeted reactive oxygen species (ROS) scavenger MitoTEMPO, or the reactive oxygen species scavenger N-acetyl-L-cysteine. Based on these data, we conclude that TP4 induces necrosis in both WT and mutant p53 glioblastoma cells through a mitochondrial ROS-dependent pathway.
Collapse
Affiliation(s)
- Bor-Chyuan Su
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan.
| | - Chieh-Yu Pan
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan.
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan.
| |
Collapse
|
36
|
Lundy DJ, Lee KJ, Peng IC, Hsu CH, Lin JH, Chen KH, Tien YW, Hsieh PCH. Inducing a Transient Increase in Blood-Brain Barrier Permeability for Improved Liposomal Drug Therapy of Glioblastoma Multiforme. ACS NANO 2019; 13:97-113. [PMID: 30532951 DOI: 10.1021/acsnano.8b03785] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The blood-brain barrier (BBB) selectively controls the passage of endogenous and exogenous molecules between systemic circulation and the brain parenchyma. Nanocarrier-based drugs such as liposomes and nanoparticles are an attractive prospect for cancer therapy since they can carry a drug payload and be modified to improve targeting and retention at the desired site. However, the BBB prevents most therapeutic drugs from entering the brain, including physically restricting the passage of liposomes and nanoparticles. In this paper, we show that a low dose of systemically injected recombinant human vascular endothelial growth factor induces a short period of increased BBB permeability. We have shown increased delivery of a range of nanomedicines to the brain including contrast agents for imaging, varying sizes of nanoparticles, small molecule chemotherapeutics, tracer dyes, and liposomal chemotherapeutics. However, this effect was not uniform across all brain regions, and permeability varied depending on the drug or molecule measured. We have found that this window of BBB permeability effect is transient, with normal BBB integrity restored within 4 h. This strategy, combined with liposomal doxorubicin, was able to significantly extend survival in a mouse model of human glioblastoma. We have found no evidence of systemic toxicity, and the technique was replicated in pigs, demonstrating that this technique could be scaled up and potentially be translated to the clinic, thus allowing the use of nanocarrier-based therapies for brain disorders.
Collapse
Affiliation(s)
- David J Lundy
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
- Graduate Institute of Biomedical Materials and Tissue Engineering , Taipei Medical University , Taipei 110 , Taiwan
| | - Keng-Jung Lee
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - I-Chia Peng
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Chia-Hsin Hsu
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Jen-Hao Lin
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Kun-Hung Chen
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Yu-Wen Tien
- Department of Surgery , National Taiwan University and Hospital , Taipei 100 , Taiwan
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
- Department of Surgery , National Taiwan University and Hospital , Taipei 100 , Taiwan
- Institute of Medical Genomics and Proteomics , National Taiwan University , Taipei 100 , Taiwan
- Institute of Clinical Medicine , National Taiwan University , Taipei 100 , Taiwan
| |
Collapse
|
37
|
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
|
38
|
Sippl C, Ketter R, Bohr L, Kim YJ, List M, Oertel J, Urbschat S. MiRNA-181d Expression Significantly Affects Treatment Responses to Carmustine Wafer Implantation. Neurosurgery 2018; 85:147-155. [DOI: 10.1093/neuros/nyy214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/07/2018] [Indexed: 01/17/2023] Open
Affiliation(s)
- Christoph Sippl
- Department of Neurosurgery, Faculty of Medicine, University of Saarland, Kirrbergerstraße, Homburg/ Saar, Germany
| | - Ralf Ketter
- Department of Neurosurgery, Faculty of Medicine, University of Saarland, Kirrbergerstraße, Homburg/ Saar, Germany
| | - Lisa Bohr
- Department of Neurosurgery, Faculty of Medicine, University of Saarland, Kirrbergerstraße, Homburg/ Saar, Germany
| | - Yoo Jin Kim
- Institute of Pathology, Faculty of Medicine, University of Saarland, Kaiserslautern, Germany
| | - Markus List
- Max-Planck-Institute of Informatics, Computational Biology and Applied Algorithmics, Saarbrücken, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine, University of Saarland, Kirrbergerstraße, Homburg/ Saar, Germany
| | - Steffi Urbschat
- Department of Neurosurgery, Faculty of Medicine, University of Saarland, Kirrbergerstraße, Homburg/ Saar, Germany
| |
Collapse
|
39
|
Cornelison RC, Munson JM. Perspective on Translating Biomaterials Into Glioma Therapy: Lessons From in vitro Models. FRONTIERS IN MATERIALS 2018; 5:27. [PMID: 30911536 PMCID: PMC6430582 DOI: 10.3389/fmats.2018.00027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Glioblastoma (GBM) is the most common and malignant form of brain cancer. Even with aggressive standard of care, GBM almost always recurs because its diffuse, infiltrative nature makes these tumors difficult to treat. The use of biomaterials is one strategy that has been, and is being, employed to study and overcome recurrence. Biomaterials have been used in GBM in two ways: in vitro as mediums in which to model the tumor microenvironment, and in vivo to sustain release of cytotoxic therapeutics. In vitro systems are a useful platform for studying the effects of drugs and tissue-level effectors on tumor cells in a physiologically relevant context. These systems have aided examination of how glioma cells respond to a variety of natural, synthetic, and semi-synthetic biomaterials with varying substrate properties, biochemical factor presentations, and non-malignant parenchymal cell compositions in both 2D and 3D environments. The current in vivo paradigm is completely different, however. Polymeric implants are simply used to line the post-surgical resection cavities and deliver secondary therapies, offering moderate impacts on survival. Instead, perhaps we can use the data generated from in vitro systems to design novel biomaterial-based treatments for GBM akin to a tissue engineering approach. Here we offer our perspective on the topic, summarizing how biomaterials have been used to identify facets of glioma biology in vitro and discussing the elements that show promise for translating these systems in vivo as new therapies for GBM.
Collapse
Affiliation(s)
- R. Chase Cornelison
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jennifer M. Munson
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
40
|
Pawlowska E, Szczepanska J, Szatkowska M, Blasiak J. An Interplay between Senescence, Apoptosis and Autophagy in Glioblastoma Multiforme-Role in Pathogenesis and Therapeutic Perspective. Int J Mol Sci 2018; 19:ijms19030889. [PMID: 29562589 PMCID: PMC5877750 DOI: 10.3390/ijms19030889] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy, cellular senescence, programmed cell death and necrosis are key responses of a cell facing a stress. These effects are partly interconnected, but regulation of their mutual interactions is not completely clear. That regulation seems to be especially important in cancer cells, which have their own program of development and demand more nutrition and energy than normal cells. Glioblastoma multiforme (GBM) belongs to the most aggressive and most difficult to cure cancers, so studies on its pathogenesis and new therapeutic strategies are justified. Using an animal model, it was shown that autophagy is required for GBM development. Temozolomide (TMZ) is the key drug in GBM chemotherapy and it was reported to induce senescence, autophagy and apoptosis in GBM. In some GBM cells, TMZ induces small toxicity despite its significant concentration and GBM cells can be intrinsically resistant to apoptosis. Resveratrol, a natural compound, was shown to potentiate anticancer effect of TMZ in GBM cells through the abrogation G2-arrest and mitotic catastrophe resulting in senescence of GBM cells. Autophagy is the key player in TMZ resistance in GBM. TMZ can induce apoptosis due to selective inhibition of autophagy, in which autophagic vehicles accumulate as their fusion with lysosomes is blocked. Modulation of autophagic action of TMZ with autophagy inhibitors can result in opposite outcomes, depending on the step targeted in autophagic flux. Studies on relationships between senescence, autophagy and apoptosis can open new therapeutic perspectives in GBM.
Collapse
Affiliation(s)
- Elzbieta Pawlowska
- Department of Orthodontics, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Magdalena Szatkowska
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| | - Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| |
Collapse
|
41
|
Lillehei KO, Kalkanis SN, Liau LM, Mydland DE, Olson J, Paleologos NA, Ryken T, Johnson T, Scullin E. Rationale and design of the 500-patient, 3-year, and prospective Vigilant ObservatIon of GlIadeL WAfer ImplaNT registry. CNS Oncol 2017; 7:CNS08. [PMID: 29206049 PMCID: PMC5977275 DOI: 10.2217/cns-2017-0036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Implantation of biodegradable wafers impregnated with carmustine (BCNU) is one of the few chemotherapeutic modalities that have been evaluated in Phase III trials and approved by the US FDA for treatment of newly diagnosed high-grade glioma and recurrent glioblastoma. Enrolling up to 500 patients for 3-year follow-up at over 30 sites, the prospective Vigilant ObservatIon of GlIadeL WAfer ImplaNT (VIGILANT) registry (NCT02684838) will evaluate BCNU wafers for treatment of CNS malignancies in contemporary practice and in the new era of molecular tumor analysis. Subgroup analyses will include tumor type, molecular marker status, and treatment combinations. Interim analyses from the VIGILANT registry will be reported until complete results are available in 2024.
Collapse
Affiliation(s)
- Kevin O Lillehei
- Department of Neurosurgery, University of Colorado School of Medicine, Denver, CO, 80045, USA
| | - Steven N Kalkanis
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, 90095, CA, USA
| | | | - Jeffrey Olson
- Department of Neurosurgery, Emory University, Atlanta, GA, 30322, USA
| | - Nina A Paleologos
- Department of Neurology, Advocate Healthcare, Chicago, IL, 60515, USA
| | - Timothy Ryken
- Department of Neurosurgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | | | - Evan Scullin
- Arbor Pharmaceuticals, LLC, Atlanta, GA, 30328, USA
| |
Collapse
|
42
|
Leelakanok N, Geary S, Salem A. Fabrication and Use of Poly(d,l-lactide-co-glycolide)-Based Formulations Designed for Modified Release of 5-Fluorouracil. J Pharm Sci 2017; 107:513-528. [PMID: 29045885 DOI: 10.1016/j.xphs.2017.10.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/03/2017] [Accepted: 10/06/2017] [Indexed: 12/14/2022]
Abstract
5-fluorouracil (5-FU) is a chemotherapeutic agent that has been used for the treatment of a variety of malignancies since its initial introduction to the clinic in 1957. Owing to its short biological half-life, multiple dosings are generally required to maintain effective 5-FU plasma concentrations throughout the therapeutic period. Clinical studies have shown that continuous 5-FU administration is generally superior to bolus injection as exhibited by lower toxicities and increased therapeutic efficacy. Optimal therapeutic efficacy, however, is often compromised by the limiting therapeutic index. Whilst oral formulations are also used, these suffer from the drawbacks of variable bioavailability and first-pass metabolism. As a result, sustained release formulations of 5-FU have been investigated in an effort to mimic the kinetics of continuous infusion particularly for situations where local delivery is considered appropriate. The biocompatible, biodegradable, and highly tunable synthetic polymer, poly(d,l-lactide-co-glycolide) (PLGA), is widely used as a vector for sustained drug delivery, however, issues such as insufficient loading and inappropriate burst release kinetics have dogged progress into the clinic for small hydrophilic drugs such as 5-FU. This review provides introductory information about the mechanism of action, pharmacokinetic and physicochemical properties, and clinical use of 5-FU that have contributed to the development of PLGA-based 5-FU release platforms. In addition, this review provides information on fabrication methods used for a range of 5-FU-loaded PLGA formulations and discusses factors affecting the release kinetics of 5-FU as well as the in vitro and in vivo antitumor or antiproliferative efficacy of these platforms.
Collapse
Affiliation(s)
- Nattawut Leelakanok
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, Iowa City, Iowa 52242
| | - Sean Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, Iowa City, Iowa 52242
| | - Aliasger Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, Iowa City, Iowa 52242.
| |
Collapse
|
43
|
Wang L, Su J, Zhao Z, Hou Y, Yin X, Zheng N, Zhou X, Yan J, Xia J, Wang Z. MiR-26b reverses temozolomide resistance via targeting Wee1 in glioma cells. Cell Cycle 2017; 16:1954-1964. [PMID: 28898169 DOI: 10.1080/15384101.2017.1367071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence has demonstrated that microRNAs (miRNA) play a critical role in chemotherapy-induced epithelial-mesenchymal transition (EMT) in glioma. However, the underlying mechanism of chemotherapy-triggered EMT has not been fully understood. In the current study, we determined the role of miR-26b in regulation of EMT in stable temozolomide (TMZ)-resistant (TR) glioma cells, which have displayed mesenchymal features. Our results illustrated that miR-26b was significantly downregulated in TR cells. Moreover, ectopic expression of miR-26b by its mimics reversed the phenotype of EMT in TR cells. Furthermore, we found that miR-26b governed TR-mediate EMT partly due to governing its target Wee1. Notably, overexpression of miR-26b sensitized TR cells to TMZ. These findings suggest that upregulation of miR-26b or targeting Wee1 could serve as novel approaches to reverse chemotherapy resistance in glioma.
Collapse
Affiliation(s)
- Lixia Wang
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Jingna Su
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Zhe Zhao
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Yingying Hou
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Xuyuan Yin
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Nana Zheng
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Xiuxia Zhou
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Jingzhe Yan
- b Department of Abdominal Oncosurgery , Jilin Province Cancer Hospital , Changchun , Jilin , China
| | - Jun Xia
- c Department of Biochemistry and Molecular Biology , Bengbu Medical College , Anhui , China
| | - Zhiwei Wang
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China.,c Department of Biochemistry and Molecular Biology , Bengbu Medical College , Anhui , China.,d Department of Pathology , Beth Israel Deaconess Medical Center, Harvard Medical School , MA , USA
| |
Collapse
|
44
|
Lucca LE, Hafler DA. Co-inhibitory blockade while preserving tolerance: checkpoint inhibitors for glioblastoma. Immunol Rev 2017; 276:9-25. [PMID: 28258696 DOI: 10.1111/imr.12529] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The introduction of immunotherapy with checkpoint receptor blockade has changed the treatment of advanced cancers, at times inducing prolonged remission. Nevertheless, the success rate of the approach is variable across patients and different tumor types, and treatment is often accompanied by severe immune-related side effects, suggesting the importance of co-inhibitory pathway for both prevention of autoimmunity and failure of tumor rejection. A better understanding of how to uncouple anti-tumor activity from loss of self-tolerance is necessary to increase the therapeutic efficacy of checkpoint immunotherapy. In this review, we describe basic concepts of T-cell exhaustion that occur in cancer, highlighting the role of co-inhibitory receptors in contributing to this process while preventing immunopathology. By providing an overview of the current therapeutic success and immune-related burden of secondary effects of checkpoint immunotherapy, we illustrate the "double-edged sword" related to interference with immune-regulatory pathways. Finally, since achieving tumor rejection while preserving self-tolerance is particularly important for the central nervous system, we analyze the case for checkpoint immunotherapy in glioblastoma, the most common adult brain tumor.
Collapse
Affiliation(s)
- Liliana E Lucca
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
45
|
Shi L, Li H, Zhan Y. All-trans retinoic acid enhances temozolomide-induced autophagy in human glioma cells U251 via targeting Keap1/Nrf2/ARE signaling pathway. Oncol Lett 2017; 14:2709-2714. [PMID: 28927033 PMCID: PMC5588105 DOI: 10.3892/ol.2017.6482] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/17/2017] [Indexed: 01/03/2023] Open
Abstract
The present study evaluated the retinoic acid (RA) enhancement of temozolomide (TMZ) effects on the human glioma cells U251 and explored its underlying molecular mechanism. The cell growth was detected using the MTT assay and the cell cycle was assessed by flow cytometry. Cell apoptosis was analyzed by Annexin V/propidium iodide staining, and the cell morphology was evaluated using transmission electron microscopy (TEM). Additionally, reverse transcription-PCR and western blot analysis were applied to detect the mRNA and protein levels. The RA treatment in combination with TMZ in the human U251 cells further inhibited cells growth, arresting cell cycle progression at the G0/G1 phase, and significantly induced apoptosis of U251 cells. After the RA+TMZ treatment of U251 cells, autophagy associated proteins Beclin 1 and LC3B were significantly increased, and the TEM analysis were consistent with autophagy protein levels. Moreover, Keap1/Nrf2/ARE expression was downregulated significantly, indicating the involvement in the mechanisms that RA treatment could enhance the TMZ effects on U251 cells. RA treatment in combination with TMZ may provide some experimental evidence for the possible effect of RA+TMZ against the growth and the proliferation of glioma cells. Therefore, the RA+TMZ administration may have potential utility for glioblastoma treatment.
Collapse
Affiliation(s)
- Lin Shi
- Department of Neurosurgery, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Hongyuan Li
- Department of Neurosurgery, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Yang Zhan
- Department of Pathology, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| |
Collapse
|
46
|
Urbschat S, Sippl C, Engelhardt J, Kammers K, Oertel J, Ketter R. Importance of biomarkers in glioblastomas patients receiving local BCNU wafer chemotherapy. Mol Cytogenet 2017; 10:16. [PMID: 28484518 PMCID: PMC5418867 DOI: 10.1186/s13039-017-0317-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 04/24/2017] [Indexed: 12/18/2022] Open
Abstract
Background To assess the influence of molecular markers with potential prognostic value to groups of patients with newly diagnosed glioblastoma patients were examined: group A with 36 patients (surgical resection plus standard combined chemoradiotherapy) and group B with 36 patients (surgical resection, standard combined chemoradiotherapy plus carmustine wafer implantation). Our aim was to determine chromosomal alterations, methylation status of MGMT, p15, and p16 (CDKN2A) in order to analyse the influence on patient survival time as well as radio- and chemotherapy responses. Promoter hypermethylation of MGMT, p16, and p15 genes were determined by MS-PCR. Comparative genomic hybridisation (CGH) analyses were performed with isolated, labelled DNA of each tumor to detect genetic alterations. Results Age of onset of the disease showed a significant effect on overall survival (OS) (p < 0.0001). Additional treatment with carmustine wafer (group B) compared to the control group (group A) did not result in improved OS (p = 0.562). Patients with a methylated MGMT promotor showed a significant longer OS compared to those patients with unmethylated MGMT promotor (p = 0.041). Subgroup analyses revealed that patients with methylated p15 showed a significant shorter OS when administered to group B rather than in group A (p = 0.0332). In patients additionally treated with carmustine wafer an amplification of 4q12 showed a significant impact on a reduced OS (p = 0.00835). In group B, a loss of 13q was significantly associated with a longer OS (p = 0.0364). If a loss of chromosome 10 occurred, patients in group B showed a significantly longer OS (p = 0.0123). Conclusion A clinical benefit for the widespread use of additional carmustine wafer implantation could not be found. However, carmustine wafer implantation shows a significantly improved overall survival if parts of chromosome 10 or chromosome 13 are deleted. In cases of 4q12 amplification and in cases of a methylated p15 promotor, the use of carmustine wafers is especially not recommended. The MGMT promoter methylation is a strong prognostic Biomarker for benefit from temozolomide and BCNU chemotherapy.
Collapse
Affiliation(s)
- Steffi Urbschat
- Department of Neurosurgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Christoph Sippl
- Department of Neurosurgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Jana Engelhardt
- Department of Neurosurgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Kai Kammers
- Division of Biostatistics and Bioinformatics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Ralf Ketter
- Department of Neurosurgery, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|
47
|
Cote DJ, Balak N, Brennum J, Holsgrove DT, Kitchen N, Kolenda H, Moojen WA, Schaller K, Robe PA, Mathiesen T, Broekman ML. Ethical difficulties in the innovative surgical treatment of patients with recurrent glioblastoma multiforme. J Neurosurg 2017; 126:2045-2050. [PMID: 28430037 DOI: 10.3171/2016.11.jns162488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- David J Cote
- Cushing Neurosurgical Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Naci Balak
- Department of Neurosurgery, Göztepe Education and Research Hospital, Istanbul, Turkey
| | - Jannick Brennum
- Department of Neurosurgery, Rigshopsitalet, University of Copenhagen, Denmark
| | - Daniel T Holsgrove
- Department of Neurosurgery, Salford Royal Hospital, Salford, United Kingdom
| | - Neil Kitchen
- The National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Herbert Kolenda
- Department of Neurosurgery, Agaplesion Diakonieklinikum, Rotenburg, Germany
| | - Wouter A Moojen
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands.,Department of Neurosurgery, Medical Center Haaglanden, The Hague, The Netherlands
| | | | - Pierre A Robe
- Department of Neurosurgery, University Medical Center, Utrecht, The Netherlands
| | - Tiit Mathiesen
- Department of Neurosurgery, Karolinska Institute, Stockholm, Sweden ; and
| | - Marike L Broekman
- Department of Neurosurgery, University Medical Center, Utrecht, The Netherlands.,Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
48
|
Li K, Tu Y, Liu Q, Ouyang Y, He M, Luo M, Chen J, Pi R, Liu A. PT93, a novel caffeic acid amide derivative, suppresses glioblastoma cells migration, proliferation and MMP-2/-9 expression. Oncol Lett 2017; 13:1990-1996. [PMID: 28454354 DOI: 10.3892/ol.2017.5663] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/02/2016] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant type of primary brain tumor in adults and can diffusely infiltrate adjacent normal tissue. GBM is therefore rarely cured by surgery or radiation therapy. Matrix metalloproteinases (MMPs) are involved in tissue remodeling and numerous other physiological progresses. The MMPs MMP-2 and MMP-9 are associated with the invasion ability of GBM. PT93 is a novel caffeic acid amide derivative that was first synthesized in 2013. In the present study, the human GBM T98G, U87 and U251 cell lines and the normal mouse neuron HT22 cell line were used to investigate the anticancer and cytotoxic effects of PT93 in vitro. The cytotoxicity of PT93 was measured using MTT and lactate dehydrogenase assays. The anti-proliferation effect was tested using a cell colony formation assay. Gelatin zymography analysis and a scratch test were used to investigate the anti-migration mechanism of PT93. Western blot analysis was used to measure the expression of MMP-2/-9. The experimental results showed that PT93 suppressed the proliferation of T98G cells, and showed cytotoxicity effects at high concentration in T98G, U87, U251 and HT22 cell lines. Furthermore, PT93 limited the migration ability of the cells and inhibited the extracellular MMP-2 and MMP-9 activity of T98G and U251 cells. Finally, the present study confirmed that PT93 affects the level of MMP-2/-9 expression in T98G cells in a concentration-dependent manner. The present study indicates that PT93, as a novel caffeic acid amide derivative, may be used in the treatment of GBM.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yalin Tu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China.,International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, Guangdong 510006, P.R. China
| | - Qingyu Liu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Ying Ouyang
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Mingliang He
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Ming Luo
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jingkao Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China.,International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, Guangdong 510006, P.R. China
| | - Rongbiao Pi
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China.,International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, Guangdong 510006, P.R. China
| | - Anmin Liu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
49
|
Luo B, Wang S, Rao R, Liu X, Xu H, Wu Y, Yang X, Liu W. Conjugation Magnetic PAEEP-PLLA Nanoparticles with Lactoferrin as a Specific Targeting MRI Contrast Agent for Detection of Brain Glioma in Rats. NANOSCALE RESEARCH LETTERS 2016; 11:227. [PMID: 27119155 PMCID: PMC4848283 DOI: 10.1186/s11671-016-1421-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/12/2016] [Indexed: 05/09/2023]
Abstract
The diagnosis of malignant brain gliomas is largely based on magnetic resonance imaging (MRI) with contrast agents. In recent years, nano-sized contrast agents have been developed for improved MRI diagnosis. In this study, oleylamine-coated Fe3O4 magnetic nanoparticles (OAM-MNPs) were synthesized with thermal decomposition method and encapsulated in novel amphiphilic poly(aminoethyl ethylene phosphate)/poly(L-lactide) (PAEEP-PLLA) copolymer nanoparticles. The OAM-MNP-loaded PAEEP-PLLA nanoparticles (M-PAEEP-PLLA-NPs) were further conjugated with lactoferrin (Lf) for glioma tumor targeting. The Lf-conjugated M-PAEEP-PLLA-NPs (Lf-M-PAEEP-PLLA-NPs) were characterized by photon correlation spectroscopy (PCS), transmission electron microscopy (TEM), Fourier transform infrared (FTIR), thermo-gravimetric analysis (TGA), X-ray diffraction (XRD), and vibrating sample magnetometer (VSM). The average size of OAM-MNPs, M-PAEEP-PLLA-NPs, and Lf-M-PAEEP-PLLA-NPs were 8.6 ± 0.3, 165.7 ± 0.6, and 218.2 ± 0.4 nm, with polydispersity index (PDI) of 0.185 ± 0.023, 0.192 ± 0.021, and 0.224 ± 0.036, respectively. TEM imaging showed that OAM-MNPs were monodisperse and encapsulated in Lf-M-PAEEP-PLLA-NPs. TGA analysis showed that the content of iron oxide nanoparticles was 92.8 % in OAM-MNPs and 45.2 % in Lf-M-PAEEP-PLLA-NPs. VSM results indicated that both OAM-MNPs and Lf-M-PAEEP-PLLA-NPs were superparamagnetic, and the saturated magnetic intensity were 77.1 and 74.8 emu/g Fe. Lf-M-PAEEP-PLLA-NPs exhibited good biocompatibility in cytotoxicity assay. The high cellular uptake of Lf-M-PAEEP-PLLA-NPs in C6 cells indicated that Lf provided effective targeting for the brain tumor cells. The T 2 relaxation rate (r 2) of M-PAEEP-PLLA-NPs and Lf-M-PAEEP-PLLA-NPs were calculated to be 167.2 and 151.3 mM(-1) s(-1). In MRI on Wistar rat-bearing glioma tumor, significant contrast enhancement could clearly appear at 4 h after injection and last 48 h. Prussian blue staining of the section clearly showed the retention of Lf-M-PAEEP-PLLA-NPs in tumor tissues. The results from the in vitro and in vivo MRI indicated that Lf-M-PAEEP-PLLA-NPs possessed strong, long-lasting, tumor targeting, and enhanced tumor MRI contrast ability. Lf-M-PAEEP-PLLA-NPs represent a promising nano-sized MRI contrast agent for brain glioma targeting MRI.
Collapse
Affiliation(s)
- Binhua Luo
- College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China
- College of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Siqi Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, People's Republic of China
| | - Rong Rao
- College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, China
| | - Xuhan Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, China
| | - Haibo Xu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, People's Republic of China.
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Yun Wu
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, USA
| | - Xiangliang Yang
- College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, China.
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
50
|
Tao J, Zhang J, Hu Y, Yang Y, Gou Z, Du T, Mao J, Gou M. A conformal hydrogel nanocomposite for local delivery of paclitaxel. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2016; 28:107-118. [PMID: 27765001 DOI: 10.1080/09205063.2016.1250344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Jie Tao
- School of Materials Science and Engineering, Sichuan University, Chengdu, China
| | - Jiumeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yu Hu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiyuan Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ting Du
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jian Mao
- School of Materials Science and Engineering, Sichuan University, Chengdu, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|