51
|
Kang JH, Cho J, Ko YT. Investigation on the effect of nanoparticle size on the blood-brain tumour barrier permeability by in situ perfusion via internal carotid artery in mice. J Drug Target 2018; 27:103-110. [PMID: 29972326 DOI: 10.1080/1061186x.2018.1497037] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a limiting factor in nanoparticle drug delivery to the brain, and various attempts have been made to overcome it for efficient drug delivery. Nowadays, it was considered as further issue for brain-drug delivery that the nanoparticle delivered to brain through the BBB reach cancer cells in tumour tissue. In this study, we investigated the effect of nanoparticle size on blood-brain tumour barrier (BBTB) permeation of fluorescence-labelled gold nanoparticles (AuNPs) in a mouse model of orthotopic glioblastoma multiforme (GBM), established by intracranial implantation of luciferase-expressing human glioblastoma U87MG cells. AuNPs sized 10, 50, and 100 nm were perfused into the GBM mice via internal carotid artery (ICA) for 5 min. Immediately after perfusion, the brains were fixed and prepared for LSCM observation. The AuNPs distribution in the normal and tumorous brain tissues was analysed qualitatively and quantitatively. Higher distribution of AuNPs was observed in the tumorous tissue than in the normal tissue. Furthermore, the smallest nanoparticle, 10 nm AuNPs, was widely distributed in the brain tumour tissue, whereas the 50 and 100 nm AuNPs were located near the blood vessels. Therefore, nanoparticle size affected the permeation of nanoparticles from the blood into brain tumour tissue.
Collapse
Affiliation(s)
- Ji Hee Kang
- a College of Pharmacy, Gachon Institute of Pharmaceutical Sciences , Gachon University , Incheon , South Korea
| | - Jinsung Cho
- a College of Pharmacy, Gachon Institute of Pharmaceutical Sciences , Gachon University , Incheon , South Korea
| | - Young Tag Ko
- a College of Pharmacy, Gachon Institute of Pharmaceutical Sciences , Gachon University , Incheon , South Korea
| |
Collapse
|
52
|
Li M, Shi K, Tang X, Wei J, Cun X, Chen X, Yu Q, Zhang Z, He Q. pH-sensitive folic acid and dNP2 peptide dual-modified liposome for enhanced targeted chemotherapy of glioma. Eur J Pharm Sci 2018; 124:240-248. [PMID: 30071282 DOI: 10.1016/j.ejps.2018.07.055] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/27/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023]
Abstract
Effective chemotherapy for clinical glioma treatment is still lacking due to the poor penetration of blood-brain barrier (BBB) and the poor internalization into tumor cells. To facilitate the transmigration across the BBB as well as the glioma targeting of chemotherapeutics, we constructed cell penetrating peptide dNP2 and tumor microenvironment-cleavable folic acid (FA) dual modified, paclitaxel (PTX) loaded liposome for the targeted delivery of glioma. The modification of dNP2 significantly enhanced the transmigration across the BBB in an in vitro BBB model. The acid-cleavable cFd-Lip/PTX exhibited sensitive cleavage of FA at pH 6.8, which led to enhanced cellular uptake mediated by both cell penetrating peptide dNP2 and the interaction between FA and folate receptor (FR) on the glioma cells. After intravenous injection, compared with non-cleavable Fd-Lip and single modified liposomes, cFd-Lip enhanced the accumulation in orthotropic glioma and improved the anti-tumor effect of glioma-bearing mice. The dual modified liposomes also facilitated deep penetration into tumor cells and consequently enhanced the cytotoxicity of PTX-loaded liposomes. The acid-cleavable dual modified strategy retained the BBB penetrating and tumor targeting ability, meanwhile, the cleavage of FA further maximized the cell permeability of dNP2, exhibiting enhanced tumor targeting effect. The multi-targeting strategy provides a promising approach towards targeted chemotherapy for glioma.
Collapse
Affiliation(s)
- Man Li
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Kairong Shi
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xian Tang
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jiaojie Wei
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xingli Cun
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaoxiao Chen
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qianwen Yu
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qin He
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
53
|
Barriers to Effective Drug Treatment for Brain Metastases: A Multifactorial Problem in the Delivery of Precision Medicine. Pharm Res 2018; 35:177. [PMID: 30003344 DOI: 10.1007/s11095-018-2455-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022]
Abstract
The treatment of metastatic lesions in the brain represents a serious unmet medical need in the field of neuro-oncology. Even though many effective compounds have demonstrated success in treating peripheral (non-CNS) tumors with targeted agents, one aspect of this lack of success in the brain may be related to poor delivery of otherwise effective compounds. Many factors can influence the brain delivery of these agents, but one key barrier is a heterogeneously "leaky" BBB that expresses efflux transporters that limit the BBB permeability for many targeted agents. Future success in therapeutics for brain metastases must take into account the adequate delivery of "active, free drug" to the target, and may include combinations of targeted drugs that are appropriate to address each individual patient's tumor type. This review discusses some issues that are pertinent to precision medicine for brain metastases, using specific examples of tumor types that have a high incidence of brain metastases.
Collapse
|
54
|
Alfonso JCL, Talkenberger K, Seifert M, Klink B, Hawkins-Daarud A, Swanson KR, Hatzikirou H, Deutsch A. The biology and mathematical modelling of glioma invasion: a review. J R Soc Interface 2018; 14:rsif.2017.0490. [PMID: 29118112 DOI: 10.1098/rsif.2017.0490] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022] Open
Abstract
Adult gliomas are aggressive brain tumours associated with low patient survival rates and limited life expectancy. The most important hallmark of this type of tumour is its invasive behaviour, characterized by a markedly phenotypic plasticity, infiltrative tumour morphologies and the ability of malignant progression from low- to high-grade tumour types. Indeed, the widespread infiltration of healthy brain tissue by glioma cells is largely responsible for poor prognosis and the difficulty of finding curative therapies. Meanwhile, mathematical models have been established to analyse potential mechanisms of glioma invasion. In this review, we start with a brief introduction to current biological knowledge about glioma invasion, and then critically review and highlight future challenges for mathematical models of glioma invasion.
Collapse
Affiliation(s)
- J C L Alfonso
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| | - K Talkenberger
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| | - M Seifert
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - B Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany.,German Cancer Consortium (DKTK), partner site, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Hawkins-Daarud
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - K R Swanson
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - H Hatzikirou
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| | - A Deutsch
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| |
Collapse
|
55
|
Therapeutic Potential and Utility of Elacridar with Respect to P-glycoprotein Inhibition: An Insight from the Published In Vitro, Preclinical and Clinical Studies. Eur J Drug Metab Pharmacokinet 2018; 42:915-933. [PMID: 28374336 DOI: 10.1007/s13318-017-0411-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The occurrence of efflux mechanisms via Permeability-glycoprotein (P-gp) recognized as an important physiological process impedes drug entry or transport across membranes into tissues. In some instances, either low oral bioavailability or lack of brain penetration has been attributed to P-gp mediated efflux activity. Therefore, the objective of development of P-gp inhibitors was to facilitate the attainment of higher drug exposures in tissues. Many third-generation P-gp inhibitors such as elacridar, tariquidar, zosuquidar, etc. have entered clinical development to fulfil the promise. The body of evidence from in vitro and in vivo preclinical and clinical data reviewed in this paper provides the basis for an effective blockade of P-gp efflux mechanism by elacridar. However, clinical translation of the promise has been elusive not just for elacridar but also for other P-gp inhibitors in this class. The review provides introspection and perspectives on the lack of clinical translation of this class of drugs and a broad framework of strategies and considerations in the potential application of elacridar and other P-gp inhibitors in oncology therapeutics.
Collapse
|
56
|
Vogelbaum MA. Targeted Therapies for Brain Tumors: Will They Ever Deliver? Clin Cancer Res 2018; 24:3790-3791. [PMID: 29798907 DOI: 10.1158/1078-0432.ccr-18-0855] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/03/2018] [Accepted: 04/17/2018] [Indexed: 11/16/2022]
Abstract
The strategy of using biologically targeted therapeutics for cancer has yet to translate into effective treatment of gliomas. The neuro-oncology community is beginning to recognize that phase 0 studies should be performed to account for the impact of the blood-brain barrier on the ability of a therapeutic to reach its target(s). Clin Cancer Res; 24(16); 3790-1. ©2018 AACRSee related article by Sanai et al., p. 3820.
Collapse
|
57
|
Sattiraju A, Sai KKS, Mintz A. Glioblastoma Stem Cells and Their Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:119-140. [PMID: 29204831 DOI: 10.1007/978-3-319-69194-7_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant astrocytoma associated with a poor patient survival. Apart from arising de novo, GBMs also occur due to progression of slower growing grade III astrocytomas. GBM is characterized by extensive hypoxia, angiogenesis, proliferation and invasion. Standard treatment options such as surgical resection, radiation therapy and chemotherapy have increased median patient survival to 14.6 months in adults but recurrent disease arising from treatment resistant cancer cells often results in patient mortality. These treatment resistant cancer cells have been found to exhibit stem cell like properties. Strategies to identify or target these Glioblastoma Stem Cells (GSC) have proven to be unsuccessful so far. Studies on cancer stem cells (CSC) within GBM and other cancers have highlighted the importance of paracrine signaling networks within their microenvironment on the growth and maintenance of CSCs. The study of GSCs and their communication with various cell populations within their microenvironment is therefore not only important to understand the biology of GBMs but also to predict response to therapies and to identify novel targets which could stymy support to treatment resistant cancer cells and prevent disease recurrence. The purpose of this chapter is to introduce the concept of GSCs and to detail the latest findings indicating the role of various cellular subtypes within their microenvironment on their survival, proliferation and differentiation.
Collapse
Affiliation(s)
- Anirudh Sattiraju
- Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | | - Akiva Mintz
- Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
58
|
Yu KKH, Taylor JT, Pathmanaban ON, Youshani AS, Beyit D, Dutko-Gwozdz J, Benson R, Griffiths G, Peers I, Cueppens P, Telfer BA, Williams KJ, McBain C, Kamaly-Asl ID, Bigger BW. High content screening of patient-derived cell lines highlights the potential of non-standard chemotherapeutic agents for the treatment of glioblastoma. PLoS One 2018; 13:e0193694. [PMID: 29499065 PMCID: PMC5834163 DOI: 10.1371/journal.pone.0193694] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/19/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common primary brain malignancy in adults, yet survival outcomes remain poor. First line treatment is well established, however disease invariably recurs and improving prognosis is challenging. With the aim of personalizing therapy at recurrence, we have established a high content screening (HCS) platform to analyze the sensitivity profile of seven patient-derived cancer stem cell lines to 83 FDA-approved chemotherapy drugs, with and without irradiation. METHODS Seven cancer stem cell lines were derived from patients with GBM and, along with the established cell line U87-MG, each patient-derived line was cultured in tandem in serum-free conditions as adherent monolayers and three-dimensional neurospheres. Chemotherapeutics were screened at multiple concentrations and cells double-stained to observe their effect on both cell death and proliferation. Sensitivity was classified using high-throughput algorithmic image analysis. RESULTS Cell line specific drug responses were observed across the seven patient-derived cell lines. Few agents were seen to have radio-sensitizing effects, yet some drug classes showed a marked difference in efficacy between monolayers and neurospheres. In vivo validation of six drugs suggested that cell death readout in a three-dimensional culture scenario is a more physiologically relevant screening model and could be used effectively to assess the chemosensitivity of patient-derived GBM lines. CONCLUSION The study puts forward a number of non-standard chemotherapeutics that could be useful in the treatment of recurrent GBM, namely mitoxantrone, bortezomib and actinomycin D, whilst demonstrating the potential of HCS to be used for personalized treatment based on the chemosensitivity profile of patient tumor cells.
Collapse
Affiliation(s)
- Kenny Kwok-Hei Yu
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - Jessica T. Taylor
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - Omar N. Pathmanaban
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Manchester Academic Health Sciences Centre, Salford, United Kingdom
| | - Amir Saam Youshani
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - Deniz Beyit
- Imagen Therapeutics, Manchester, United Kingdom
| | | | | | | | - Ian Peers
- Inferstats Consulting, Alderley Park, Biohub, Cheshire, United Kingdom
| | - Peter Cueppens
- Inferstats Consulting, Alderley Park, Biohub, Cheshire, United Kingdom
| | - Brian A. Telfer
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kaye J. Williams
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Catherine McBain
- Department of Clinical Oncology, The Christie NHS FT, Manchester, United Kingdom
| | - Ian D. Kamaly-Asl
- Children’s Brain Tumour Research Network (CBTRN), Royal Manchester Children’s Hospital, Manchester, United Kingdom
- Department of Neurosurgery, Royal Manchester Children’s Hospital, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Brian W. Bigger
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
59
|
Fisusi FA, Schätzlein AG, Uchegbu IF. Nanomedicines in the treatment of brain tumors. Nanomedicine (Lond) 2018; 13:579-583. [PMID: 29376468 DOI: 10.2217/nnm-2017-0378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Funmilola A Fisusi
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.,Drug Research & Production Unit, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Andreas G Schätzlein
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.,Nanomerics Ltd., New Bridge Street House, 30-34 New Bridge Street, London, EC4V 6BJ, UK
| | - Ijeoma F Uchegbu
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.,Nanomerics Ltd., New Bridge Street House, 30-34 New Bridge Street, London, EC4V 6BJ, UK
| |
Collapse
|
60
|
Mazur J, Roy K, Kanwar JR. Recent advances in nanomedicine and survivin targeting in brain cancers. Nanomedicine (Lond) 2017; 13:105-137. [PMID: 29161215 DOI: 10.2217/nnm-2017-0286] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Brain cancer is a highly lethal disease, especially devastating toward both the elderly and children. This cancer has no therapeutics available to combat it, predominately due to the blood-brain barrier (BBB) preventing treatments from maintaining therapeutic levels within the brain. Recently, nanoparticle technology has entered the forefront of cancer therapy due to its ability to deliver therapeutic effects while potentially passing physiological barriers. Key nanoparticles for brain cancer treatment include glutathione targeted PEGylated liposomes, gold nanoparticles, superparamagnetic iron oxide nanoparticles and nanoparticle-albumin bound drugs, with these being discussed throughout this review. Recently, the survivin protein has gained attention as it is over-expressed in a majority of tumors. This review will briefly discuss the properties of survivin, while focusing on how both nanoparticles and survivin-targeting treatments hold potential as brain cancer therapies. This review may provide useful insight into new brain cancer treatment options, particularly survivin inhibition and nanomedicine.
Collapse
Affiliation(s)
- Jake Mazur
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Kislay Roy
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Jagat R Kanwar
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| |
Collapse
|
61
|
Yasunaga M, Manabe S, Tsuji A, Furuta M, Ogata K, Koga Y, Saga T, Matsumura Y. Development of Antibody-Drug Conjugates Using DDS and Molecular Imaging. Bioengineering (Basel) 2017; 4:bioengineering4030078. [PMID: 28952557 PMCID: PMC5615324 DOI: 10.3390/bioengineering4030078] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 12/04/2022] Open
Abstract
Antibody-drug conjugate (ADC), as a next generation of antibody therapeutics, is a combination of an antibody and a drug connected via a specialized linker. ADC has four action steps: systemic circulation, the enhanced permeability and retention (EPR) effect, penetration within the tumor tissue, and action on cells, such as through drug delivery system (DDS) drugs. An antibody with a size of about 10 nm has the same capacity for passive targeting as some DDS carriers, depending on the EPR effect. In addition, some antibodies are capable of active targeting. A linker is stable in the bloodstream but should release drugs efficiently in the tumor cells or their microenvironment. Thus, the linker technology is actually a typical controlled release technology in DDS. Here, we focused on molecular imaging. Fluorescent and positron emission tomography (PET) imaging is useful for the visualization and evaluation of antibody delivery in terms of passive and active targeting in the systemic circulation and in tumors. To evaluate the controlled release of the ADC in the targeted area, a mass spectrometry imaging (MSI) with a mass microscope, to visualize the drug released from ADC, was used. As a result, we succeeded in confirming the significant anti-tumor activity of anti-fibrin, or anti-tissue factor-ADC, in preclinical settings by using DDS and molecular imaging.
Collapse
Affiliation(s)
- Masahiro Yasunaga
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa 277-8577, Japan.
| | - Shino Manabe
- Synthetic Cellular Chemistry Laboratory, RIKEN, Wako 351-0198, Japan.
| | - Atsushi Tsuji
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, QST, Chiba 263-8555, Japan; .
| | | | | | - Yoshikatsu Koga
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa 277-8577, Japan.
| | - Tsuneo Saga
- Department of Diagnostic Radiology, Kyoto University Hospital; Kyoto 606-8501, Japan.
| | - Yasuhiro Matsumura
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa 277-8577, Japan.
| |
Collapse
|
62
|
Shi J, Hou S, Huang J, Wang S, Huan W, Huang C, Liu X, Jiang R, Qian W, Lu J, Wang X, Shi W, Huang R, Chen J. An MSN-PEG-IP drug delivery system and IL13Rα2 as targeted therapy for glioma. NANOSCALE 2017; 9:8970-8981. [PMID: 28443896 DOI: 10.1039/c6nr08786h] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A combination of gene therapy and chemotherapy has recently received interest as a targeted therapy for glioma. A mesoporous silica nanoparticle (MSN)-based vehicle coated with IL13Rα2-targeted peptide (IP) using polyethylene glycol (PEG), MSN-PEG-IP (MPI), was constructed and confirmed as a potential glioma-targeted drug delivery system in vitro. In this work, tissue microarray (TMA) results revealed that IL13Rα2 was over-expressed in human glioma tissues and that high expression of IL13Rα2 in patients was associated with poor survival. Doxorubicin (DOX)-loaded MPI (MPI/D) crossed the blood-brain barrier, specifically targeting glioma cells and significantly enhancing the cellular uptake of DOX in glioma cells compared with MSN/DOX (M/D) and MSN-PEG/DOX (MP/D), whereas the normal brain was not affected. Magnetic Resonance Imaging (MRI) examinations showed that the tumour size of glioma-bearing rats in the MPI/D-treated group was much smaller than those in the M/D and MP/D treated groups. Immunofluorescence results demonstrated that MPI/D treatment induced more apoptosis and much less proliferation than the other two treatments. However, the therapeutic effect was weak when IL13Rα2 was knocked down. Furthermore, U87 cells treated with IL-13 and MPI together could increase both STAT6 and P63 expression, which attenuated glioma cell proliferation, invasion and migration compared with cells treated with IL-13 alone. The results of the subcutaneous tumour model also revealed that IL13Rα2 knockdown could hinder cell proliferation and induce more apoptosis. The promising results suggested that MPI can not only deliver DOX to glioma in a targeted manner but also occupy IL13Rα2, which can promote IL-13 binding to IL13Rα1 and activation of the JAK-STAT pathway to induce an anti-glioma effect.
Collapse
Affiliation(s)
- Jinlong Shi
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong 226001, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Sattiraju A, Xiong X, Pandya DN, Wadas TJ, Xuan A, Sun Y, Jung Y, Sai KKS, Dorsey JF, Li KC, Mintz A. Alpha Particle Enhanced Blood Brain/Tumor Barrier Permeabilization in Glioblastomas Using Integrin Alpha-v Beta-3-Targeted Liposomes. Mol Cancer Ther 2017; 16:2191-2200. [PMID: 28619756 DOI: 10.1158/1535-7163.mct-16-0907] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/26/2017] [Accepted: 06/08/2017] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant astrocytoma characterized by extensive invasion, angiogenesis, hypoxia, and micrometastasis. Despite the relatively leaky nature of GBM blood vessels, effective delivery of antitumor therapeutics has been a major challenge due to the complications caused by the blood-brain barrier (BBB) and the highly torturous nature of newly formed tumor vasculature (blood tumor barrier-BTB). External beam radiotherapy was previously shown to be an effective means of permeabilizing central nervous system (CNS) barriers. By using targeted short-ranged radionuclides, we show for the first time that our targeted actinium-225-labeled αvβ3-specific liposomes (225Ac-IA-TLs) caused catastrophic double stranded DNA breaks and significantly enhanced the permeability of BBB and BTB in mice bearing orthotopic GBMs. Histologic studies revealed characteristic α-particle induced double strand breaks within tumors but was not significantly present in normal brain regions away from the tumor where BBB permeability was observed. These findings indicate that the enhanced vascular permeability in these distal regions did not result from direct α-particle-induced DNA damage. On the basis of these results, in addition to their direct antitumor effects, 225Ac-IA-TLs can potentially be used to enhance the permeability of BBB and BTB for effective delivery of systemically administered antitumor therapeutics. Mol Cancer Ther; 16(10); 2191-200. ©2017 AACR.
Collapse
Affiliation(s)
- Anirudh Sattiraju
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Xiaobing Xiong
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Darpan N Pandya
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Thaddeus J Wadas
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ang Xuan
- Department of Nuclear Medicine and Radiology, the People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yao Sun
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Youngkyoo Jung
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - Jay F Dorsey
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - King C Li
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina. .,Columbia University, New York, New York
| |
Collapse
|
64
|
Monaco I, Camorani S, Colecchia D, Locatelli E, Calandro P, Oudin A, Niclou S, Arra C, Chiariello M, Cerchia L, Comes Franchini M. Aptamer Functionalization of Nanosystems for Glioblastoma Targeting through the Blood-Brain Barrier. J Med Chem 2017; 60:4510-4516. [PMID: 28471660 DOI: 10.1021/acs.jmedchem.7b00527] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polymeric nanoparticles (PNPs) may efficiently deliver in vivo therapeutics to tumors when conjugated to specific targeting agents. Gint4.T aptamer specifically recognizes platelet-derived growth factor receptor β and can cross the blood-brain barrier (BBB). We synthesized Gint4.T-conjugated PNPs able of high uptake into U87MG glioblastoma (GBM) cells and with astonishing EC50 value (38 pM) when loaded with a PI3K-mTOR inhibitor. We also demonstrated in vivo BBB passage and tumor accumulation in a GBM orthotopic model.
Collapse
Affiliation(s)
- Ilaria Monaco
- Department of Industrial Chemistry "Toso Montanari", University of Bologna , Viale Risorgimento 4, 40136 Bologna, Italy
| | - Simona Camorani
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore" (IEOS), Consiglio Nazionale delle Ricerche (CNR) , Via S. Pansini 5, 80131 Naples, Italy
| | - David Colecchia
- Istituto Toscano Tumori (ITT), Core Research Laboratory and Consiglio Nazionale delle Ricerche (CNR), Istituto di Fisiologia Clinica , Via Fiorentina 1, 53100, Siena, Italy
| | - Erica Locatelli
- Department of Industrial Chemistry "Toso Montanari", University of Bologna , Viale Risorgimento 4, 40136 Bologna, Italy
| | - Pierpaolo Calandro
- Istituto Toscano Tumori (ITT), Core Research Laboratory and Consiglio Nazionale delle Ricerche (CNR), Istituto di Fisiologia Clinica , Via Fiorentina 1, 53100, Siena, Italy
| | - Anais Oudin
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health , 84 Val Fleuri, Luxembourg L-1586, Luxembourg
| | - Simone Niclou
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health , 84 Val Fleuri, Luxembourg L-1586, Luxembourg
| | - Claudio Arra
- Animal Facility Unit, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", IRCCS , via M. Semmola, 80131 Naples, Italy
| | - Mario Chiariello
- Istituto Toscano Tumori (ITT), Core Research Laboratory and Consiglio Nazionale delle Ricerche (CNR), Istituto di Fisiologia Clinica , Via Fiorentina 1, 53100, Siena, Italy
| | - Laura Cerchia
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore" (IEOS), Consiglio Nazionale delle Ricerche (CNR) , Via S. Pansini 5, 80131 Naples, Italy
| | - Mauro Comes Franchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna , Viale Risorgimento 4, 40136 Bologna, Italy
| |
Collapse
|
65
|
Mangani D, Weller M, Roth P. The network of immunosuppressive pathways in glioblastoma. Biochem Pharmacol 2017; 130:1-9. [DOI: 10.1016/j.bcp.2016.12.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/13/2016] [Indexed: 12/21/2022]
|
66
|
Liu C, Liu XN, Wang GL, Hei Y, Meng S, Yang LF, Yuan L, Xie Y. A dual-mediated liposomal drug delivery system targeting the brain: rational construction, integrity evaluation across the blood-brain barrier, and the transporting mechanism to glioma cells. Int J Nanomedicine 2017; 12:2407-2425. [PMID: 28405164 PMCID: PMC5378461 DOI: 10.2147/ijn.s131367] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
As the global population ages, cancer rates increase worldwide, and degenerative diseases of the central nervous system (CNS), brain tumors, and inflammation threaten human health more frequently. We designed a dual-mediated (receptor-mediated and adsorption-mediated) liposome, named transferrin–cell penetrating peptide–sterically stabilized liposome (TF-CPP-SSL), to improve therapy for gliomas through combining molecular recognition of transferrin receptors (TF-Rs) on the blood–brain barrier (BBB) and glioma cells with the internalization and lysosomal escaping ability of CPP. Based on the systematic investigation of structure–activity relations on the cellular level, we constructed TF-CPP-SSL rationally by conjugating TF and CPP moieties to the liposomes via PEG3.4K and PEG2.0K, respectively, and found the optimum densities of TF and CPP were 1.8% and 4%, respectively. These liposomes had the highest targeting efficacy for brain microvascular endothelial cell and C6 cell uptake but avoided capture by normal cells. Fluorescence resonance energy transfer technology and coculture models of BBB and glioma C6 cells indicated that TF-CPP-SSL was transported across the BBB without drug leakage, liposome breakup, or cleavage of ligand. TF-CPP-SSL offered advantages for crossing the BBB and entering into glioma C6 cells. Real-time confocal viewing revealed that TF-CPP-SSL was entrapped in endosomes of glioma C6 cells and then escaped from lysosomes successfully to release the liposomal contents into the cytosol. Entrapped contents, such as doxorubicin, could then enter the nucleus to exert pharmacological effects.
Collapse
Affiliation(s)
- Chang Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Pharmaceutics, School of Pharmaceutical Sciences
| | - Xiao-Na Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Pharmaceutics, School of Pharmaceutical Sciences
| | - Gui-Ling Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Pharmaceutics, School of Pharmaceutical Sciences
| | - Yu Hei
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Pharmaceutics, School of Pharmaceutical Sciences
| | - Shuai Meng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Pharmaceutics, School of Pharmaceutical Sciences
| | - Ling-Fei Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Pharmaceutics, School of Pharmaceutical Sciences
| | - Lan Yuan
- Medical and Healthy Analysis Center, Peking University, Beijing, People's Republic of China
| | - Ying Xie
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Pharmaceutics, School of Pharmaceutical Sciences
| |
Collapse
|
67
|
Jensen SS, Petterson SA, Halle B, Aaberg-Jessen C, Kristensen BW. Effects of the lysosomal destabilizing drug siramesine on glioblastoma in vitro and in vivo. BMC Cancer 2017; 17:178. [PMID: 28270132 PMCID: PMC5341392 DOI: 10.1186/s12885-017-3162-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/01/2017] [Indexed: 11/10/2022] Open
Abstract
Background Glioblastoma is the most frequent and most malignant brain tumor with the patients having a median survival of only 14.6 months. Although glioblastoma patients are treated with surgery, radiation and chemotherapy recurrence is inevitable. A stem-like population of radio- and chemoresistant brain tumor-initiating cells combined with the invasive properties of the tumors is believed to be critical for treatment resistance. In the present study, the aim was to investigate the effect of a novel therapeutic strategy using the lysosomotropic detergent siramesine on glioblastomas. Methods Standard glioma cell lines and patient-derived spheroids cultures with tumor-initiating stem-like cells were used to investigate effects of siramesine on proliferation and cell death. Responsible mechanisms were investigated by inhibitors of caspases and cathepsins. Effects of siramesine on migrating tumor cells were investigated by a flat surface migration assay and by implanting spheroids into organotypic rat brain slice cultures followed by confocal time-lapse imaging. Finally the effect of siramesine was investigated in an orthotopic mouse glioblastoma model. Results obtained in vitro and in vivo were confirmed by immunohistochemical staining of histological sections of spheroids, spheroids in brain slice cultures and tumors in mice brains. Results The results showed that siramesine killed standard glioma cell lines in vitro, and loss of acridine orange staining suggested a compromised lysosomal membrane. Co-treatment of the cell lines with inhibitors of caspases and cathepsins suggested differential involvement in cell death. Siramesine caused tumor cell death and reduced secondary spheroid formation of patient-derived spheroid cultures. In the flat surface migration model siramesine caused tumor cell death and inhibited tumor cell migration. This could not be reproduced in the organotypic three dimensional spheroid-brain slice culture model or in the mice xenograft model. Conclusions In conclusion the in vitro results obtained with tumor cells and spheroids suggest a potential of lysosomal destabilizing drugs in killing glioblastoma cells, but siramesine was without effect in the organotypic spheroid-brain slice culture model and the in vivo xenograft model. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3162-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stine S Jensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3. floor, 5000, Odense C, Denmark.,Institute of Clinical Research, University of Southern Denmark, Winsløwparken 19.3, 5000, Odense C, Denmark
| | - Stine A Petterson
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3. floor, 5000, Odense C, Denmark.,Institute of Clinical Research, University of Southern Denmark, Winsløwparken 19.3, 5000, Odense C, Denmark
| | - Bo Halle
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3. floor, 5000, Odense C, Denmark.,Institute of Clinical Research, University of Southern Denmark, Winsløwparken 19.3, 5000, Odense C, Denmark.,Department of Neurosurgery, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Charlotte Aaberg-Jessen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3. floor, 5000, Odense C, Denmark.,Institute of Clinical Research, University of Southern Denmark, Winsløwparken 19.3, 5000, Odense C, Denmark
| | - Bjarne W Kristensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3. floor, 5000, Odense C, Denmark. .,Institute of Clinical Research, University of Southern Denmark, Winsløwparken 19.3, 5000, Odense C, Denmark.
| |
Collapse
|
68
|
Near infrared fluorescent imaging of brain tumor with IR780 dye incorporated phospholipid nanoparticles. J Transl Med 2017; 15:18. [PMID: 28114956 PMCID: PMC5260002 DOI: 10.1186/s12967-016-1115-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 12/27/2016] [Indexed: 12/11/2022] Open
Abstract
Background Near-IR fluorescence (NIRF) imaging is becoming a promising approach in preclinical tumor detection and clinical image-guided oncological surgery. While heptamethine cyanine dye IR780 has excellent tumor targeting and imaging potential, its hydrophobic property limits its clinical use. In this study, we developed nanoparticle formulations to facilitate the use of IR780 for fluorescent imaging of malignant brain tumor. Methods Self-assembled IR780-liposomes and IR780-phospholipid micelles were prepared and their NIRF properties were characterized. The intracellular accumulation of IR780-nanoparticles in glioma cells were determined using confocal microscopy. The in vivo brain tumor targeting and NIRF imaging capacity of IR780-nanoparticles were evaluated using U87MG glioma ectopic and orthotopic xenograft models and a spontaneous glioma mouse model driven by RAS/RTK activation. Results The loading of IR780 into liposomes or phospholipid micelles was efficient. The particle diameter of IR780-liposomes and IR780-phospholipid micelles were 95 and 26 nm, respectively. While stock solutions of each preparation were maintained at ready-to-use condition, the IR780-phospholipid micelles were more stable. In tissue culture cells, IR780-nanoparticles prepared by either method accumulated in mitochondria, however, in animals the IR780-phospholipid micelles showed enhanced intra-tumoral accumulation in U87MG ectopic tumors. Moreover, IR780-phospholipid micelles also showed preferred intracranial tumor accumulation and potent NIRF signal intensity in glioma orthotopic models at a real-time, non-invasive manner. Conclusion The IR780-phospholipid micelles demonstrated tumor-specific NIRF imaging capacity in glioma preclinical mouse models, providing great potential for clinical imaging and image-guided surgery of brain tumors. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1115-2) contains supplementary material, which is available to authorized users.
Collapse
|
69
|
Abstract
Over the last decade mass spectrometry imaging (MSI) has been integrated in to many areas of drug discovery and development. It can have significant impact in oncology drug discovery as it allows efficacy and safety of compounds to be assessed against the backdrop of the complex tumour microenvironment. We will discuss the roles of MSI in investigating compound and metabolite biodistribution and defining pharmacokinetic -pharmacodynamic relationships, analysis that is applicable to all drug discovery projects. We will then look more specifically at how MSI can be used to understand tumour metabolism and other applications specific to oncology research. This will all be described alongside the challenges of applying MSI to industry research with increased use of metrology for MSI.
Collapse
|
70
|
Mechanisms of radiotherapy-associated cognitive disability in patients with brain tumours. Nat Rev Neurol 2016; 13:52-64. [PMID: 27982041 DOI: 10.1038/nrneurol.2016.185] [Citation(s) in RCA: 351] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Standard treatment of primary and metastatic brain tumours includes high-dose megavoltage-range radiation to the cranial vault. About half of patients survive >6 months, and many attain long-term control or cure. However, 50-90% of survivors exhibit disabling cognitive dysfunction. The radiation-associated cognitive syndrome is poorly understood and has no effective prevention or long-term treatment. Attention has primarily focused on mechanisms of disability that appear at 6 months to 1 year after radiotherapy. However, recent studies show that CNS alterations and dysfunction develop much earlier following radiation exposure. This finding has prompted the hypothesis that subtle early forms of radiation-induced CNS damage could drive chronic pathophysiological processes that lead to permanent cognitive decline. This Review presents evidence of acute radiation-triggered CNS inflammation, injury to neuronal lineages, accessory cells and their progenitors, and loss of supporting structure integrity. Moreover, injury-related processes initiated soon after irradiation could synergistically alter the signalling microenvironment in progenitor cell niches in the brain and the hippocampus, which is a structure critical to memory and cognition. Progenitor cell niche degradation could cause progressive neuronal loss and cognitive disability. The concluding discussion addresses future directions and potential early treatments that might reverse degenerative processes before they can cause permanent cognitive disability.
Collapse
|
71
|
Mehta N, Lyon JG, Patil K, Mokarram N, Kim C, Bellamkonda RV. Bacterial Carriers for Glioblastoma Therapy. MOLECULAR THERAPY-ONCOLYTICS 2016; 4:1-17. [PMID: 28345020 PMCID: PMC5363759 DOI: 10.1016/j.omto.2016.12.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/04/2016] [Indexed: 01/22/2023]
Abstract
Treatment of aggressive glioblastoma brain tumors is challenging, largely due to diffusion barriers preventing efficient drug dosing to tumors. To overcome these barriers, bacterial carriers that are actively motile and programmed to migrate and localize to tumor zones were designed. These carriers can induce apoptosis via hypoxia-controlled expression of a tumor suppressor protein p53 and a pro-apoptotic drug, Azurin. In a xenograft model of human glioblastoma in rats, bacterial carrier therapy conferred a significant survival benefit with 19% overall long-term survival of >100 days in treated animals relative to a median survival of 26 days in control untreated animals. Histological and proteomic analyses were performed to elucidate the safety and efficacy of these carriers, showing an absence of systemic toxicity and a restored neural environment in treated responders. In the treated non-responders, proteomic analysis revealed competing mechanisms of pro-apoptotic and drug-resistant activity. This bacterial carrier opens a versatile avenue to overcome diffusion barriers in glioblastoma by virtue of its active motility in extracellular space and can lead to tailored therapies via tumor-specific expression of tumoricidal proteins.
Collapse
Affiliation(s)
- Nalini Mehta
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Johnathan G Lyon
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Drive, Durham, NC 27708-0271, USA
| | - Ketki Patil
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Nassir Mokarram
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Drive, Durham, NC 27708-0271, USA
| | - Christine Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Drive, Durham, NC 27708-0271, USA
| |
Collapse
|
72
|
Salphati L, Alicke B, Heffron TP, Shahidi-Latham S, Nishimura M, Cao T, Carano RA, Cheong J, Greve J, Koeppen H, Lau S, Lee LB, Nannini-Pepe M, Pang J, Plise EG, Quiason C, Rangell L, Zhang X, Gould SE, Phillips HS, Olivero AG. Brain Distribution and Efficacy of the Brain Penetrant PI3K Inhibitor GDC-0084 in Orthotopic Mouse Models of Human Glioblastoma. Drug Metab Dispos 2016; 44:1881-1889. [PMID: 27638506 DOI: 10.1124/dmd.116.071423] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/09/2016] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. Limited treatment options have only marginally impacted patient survival over the past decades. The phophatidylinositol 3-kinase (PI3K) pathway, frequently altered in GBM, represents a potential target for the treatment of this glioma. 5-(6,6-Dimethyl-4-morpholino-8,9-dihydro-6H-[1,4]oxazino[4,3-e]purin-2-yl)pyrimidin-2-amine (GDC-0084) is a PI3K inhibitor that was specifically optimized to cross the blood-brain barrier. The goals of our studies were to characterize the brain distribution, pharmacodynamic (PD) effect, and efficacy of GDC-0084 in orthotopic xenograft models of GBM. GDC-0084 was tested in vitro to assess its sensitivity to the efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) and in vivo in mice to evaluate its effects on the PI3K pathway in intact brain. Mice bearing U87 or GS2 intracranial tumors were treated with GDC-0084 to assess its brain distribution by matrix-assisted laser desorption ionization (MALDI) imaging and measure its PD effects and efficacy in GBM orthotopic models. Studies in transfected cells indicated that GDC-0084 was not a substrate of P-gp or BCRP. GDC-0084 markedly inhibited the PI3K pathway in mouse brain, causing up to 90% suppression of the pAkt signal. MALDI imaging showed GDC-0084 distributed evenly in brain and intracranial U87 and GS2 tumors. GDC-0084 achieved significant tumor growth inhibition of 70% and 40% against the U87 and GS2 orthotopic models, respectively. GDC-0084 distribution throughout the brain and intracranial tumors led to potent inhibition of the PI3K pathway. Its efficacy in orthotopic models of GBM suggests that it could be effective in the treatment of GBM. GDC-0084 is currently in phase I clinical trials.
Collapse
Affiliation(s)
- Laurent Salphati
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Bruno Alicke
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Timothy P Heffron
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Sheerin Shahidi-Latham
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Merry Nishimura
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Tim Cao
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Richard A Carano
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Jonathan Cheong
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Joan Greve
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Hartmut Koeppen
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Shari Lau
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Leslie B Lee
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Michelle Nannini-Pepe
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Jodie Pang
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Emile G Plise
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Cristine Quiason
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Linda Rangell
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Xiaolin Zhang
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Stephen E Gould
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Heidi S Phillips
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| | - Alan G Olivero
- Departments of Drug Metabolism and Pharmacokinetics (L.S., S.S.-L., J.C., J.P., E.G.P., C.Q., X.Z.), Discovery Chemistry (T.P.H., A.G.O.), Cancer Signaling and Translational Oncology (B.A., M.N., M.N.-P., L.B.L., S.E.G., H.S.P.), Biomedical Imaging (T.C., R.A.C., J.G.), and Pathology (H.K., S.L., L.R.), Genentech Inc., South San Francisco, California
| |
Collapse
|
73
|
Salomon RG. Carboxyethylpyrroles: From Hypothesis to the Discovery of Biologically Active Natural Products. Chem Res Toxicol 2016; 30:105-113. [PMID: 27750413 DOI: 10.1021/acs.chemrestox.6b00304] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Our research on the roles of lipid oxidation in human disease is guided by chemical intuition. For example, we postulated that 2-(ω-carboxyethyl)pyrrole (CEP) derivatives of primary amines would be produced through covalent adduction of a γ-hydroxyalkenal generated, in turn, through oxidative fragmentation of docosahexaenoates. Our studies confirmed the natural occurrence of this chemistry, and the biological activities of these natural products and their extensive involvements in human physiology (wound healing) and pathology (age-related macular degeneration, autism, atherosclerosis, sickle cell disease, and tumor growth) continue to emerge. This perspective recounts these discoveries and proposes new frontiers where further developments are likely. Perhaps more significantly, it depicts an effective chemistry-based approach to the discovery of novel biochemistry.
Collapse
Affiliation(s)
- Robert G Salomon
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| |
Collapse
|
74
|
Hicks MJ, Chiuchiolo MJ, Ballon D, Dyke JP, Aronowitz E, Funato K, Tabar V, Havlicek D, Fan F, Sondhi D, Kaminsky SM, Crystal RG. Anti-Epidermal Growth Factor Receptor Gene Therapy for Glioblastoma. PLoS One 2016; 11:e0162978. [PMID: 27711187 PMCID: PMC5053413 DOI: 10.1371/journal.pone.0162978] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/31/2016] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary intracranial brain tumor in adults with a mean survival of 14 to 15 months. Aberrant activation of the epidermal growth factor receptor (EGFR) plays a significant role in GBM progression, with amplification or overexpression of EGFR in 60% of GBM tumors. To target EGFR expressed by GBM, we have developed a strategy to deliver the coding sequence for cetuximab, an anti-EGFR antibody, directly to the CNS using an adeno-associated virus serotype rh.10 gene transfer vector. The data demonstrates that single, local delivery of an anti-EGFR antibody by an AAVrh.10 vector coding for cetuximab (AAVrh.10Cetmab) reduces GBM tumor growth and increases survival in xenograft mouse models of a human GBM EGFR-expressing cell line and patient-derived GBM. AAVrh10.CetMab-treated mice displayed a reduction in cachexia, a significant decrease in tumor volume and a prolonged survival following therapy. Adeno-associated-directed delivery of a gene encoding a therapeutic anti-EGFR monoclonal antibody may be an effective strategy to treat GBM.
Collapse
Affiliation(s)
- Martin J Hicks
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Maria J Chiuchiolo
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Douglas Ballon
- Department of Radiology, Weill Cornell Medical College, New York, New York, United States of America
| | - Jonathan P Dyke
- Department of Radiology, Weill Cornell Medical College, New York, New York, United States of America
| | - Eric Aronowitz
- Department of Radiology, Weill Cornell Medical College, New York, New York, United States of America
| | - Kosuke Funato
- Department of Neurosurgery, Memorial Sloan-Kettering Cancer Institute, New York, NY, United States of America
| | - Viviane Tabar
- Department of Neurosurgery, Memorial Sloan-Kettering Cancer Institute, New York, NY, United States of America
| | - David Havlicek
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Fan Fan
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
75
|
Kamran N, Calinescu A, Candolfi M, Chandran M, Mineharu Y, Asad AS, Koschmann C, Nunez FJ, Lowenstein PR, Castro MG. Recent advances and future of immunotherapy for glioblastoma. Expert Opin Biol Ther 2016; 16:1245-64. [PMID: 27411023 PMCID: PMC5014608 DOI: 10.1080/14712598.2016.1212012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/08/2016] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Outcome for glioma (GBM) remains dismal despite advances in therapeutic interventions including chemotherapy, radiotherapy and surgical resection. The overall survival benefit observed with immunotherapies in cancers such as melanoma and prostate cancer has fuelled research into evaluating immunotherapies for GBM. AREAS COVERED Preclinical studies have brought a wealth of information for improving the prognosis of GBM and multiple clinical studies are evaluating a wide array of immunotherapies for GBM patients. This review highlights advances in the development of immunotherapeutic approaches. We discuss the strategies and outcomes of active and passive immunotherapies for GBM including vaccination strategies, gene therapy, check point blockade and adoptive T cell therapies. We also focus on immunoediting and tumor neoantigens that can impact the efficacy of immunotherapies. EXPERT OPINION Encouraging results have been observed with immunotherapeutic strategies; some clinical trials are reaching phase III. Significant progress has been made in unraveling the molecular and genetic heterogeneity of GBM and its implications to disease prognosis. There is now consensus related to the critical need to incorporate tumor heterogeneity into the design of therapeutic approaches. Recent data also indicates that an efficacious treatment strategy will need to be combinatorial and personalized to the tumor genetic signature.
Collapse
Affiliation(s)
- Neha Kamran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Alexandra Calinescu
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Marianela Candolfi
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Mayuri Chandran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Yohei Mineharu
- d Department of Neurosurgery , Kyoto University Graduate School of Medicine , Kyoto , Japan
| | - Antonela S Asad
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Carl Koschmann
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Felipe J Nunez
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Pedro R Lowenstein
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Maria G Castro
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| |
Collapse
|
76
|
Ferluga S, Tomé CML, Herpai DM, D'Agostino R, Debinski W. Simultaneous targeting of Eph receptors in glioblastoma. Oncotarget 2016; 7:59860-59876. [PMID: 27494882 PMCID: PMC5312354 DOI: 10.18632/oncotarget.10978] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 07/16/2016] [Indexed: 12/20/2022] Open
Abstract
Eph tyrosine kinase receptors are frequently overexpressed and functional in many cancers, and they are attractive candidates for targeted therapy. Here, we analyzed the expression of Eph receptor A3, one of the most up-regulated factors in glioblastoma cells cultured under tumorsphere-forming conditions, together with EphA2 and EphB2 receptors. EphA3 was overexpressed in up to 60% of glioblastoma tumors tested, but not in normal brain. EphA3 was localized in scattered areas of the tumor, the invasive ring, and niches near tumor vessels. EphA3 co-localized with macrophage/leukocyte markers, suggesting EphA3 expression on tumor-infiltrating cells of bone marrow origin. We took advantage of the fact that ephrinA5 (eA5) is a ligand that binds EphA3, EphA2 and EphB2 receptors, and used it to construct a novel targeted anti-glioblastoma cytotoxin. The eA5-based cytotoxin potently and specifically killed glioblastoma cells with an IC50 of at least 10-11 M. This and similar cytotoxins will simultaneously target different compartments of glioblastoma tumors while mitigating tumor heterogeneity.
Collapse
Affiliation(s)
- Sara Ferluga
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Carla Maria Lema Tomé
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Denise Mazess Herpai
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Ralph D'Agostino
- Department of Biostatistical Sciences, Section on Biostatistics, Wake Forest University Health Sciences, Winston-Salem, NC, 27157, USA
| | - Waldemar Debinski
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|
77
|
Tournier N, Goutal S, Auvity S, Traxl A, Mairinger S, Wanek T, Helal OB, Buvat I, Soussan M, Caillé F, Langer O. Strategies to Inhibit ABCB1- and ABCG2-Mediated Efflux Transport of Erlotinib at the Blood-Brain Barrier: A PET Study on Nonhuman Primates. J Nucl Med 2016; 58:117-122. [PMID: 27493269 DOI: 10.2967/jnumed.116.178665] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/12/2016] [Indexed: 12/12/2022] Open
Abstract
The tyrosine kinase inhibitor erlotinib poorly penetrates the blood-brain barrier (BBB) because of efflux transport by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2), thereby limiting its utility in the treatment of non-small cell lung cancer metastases in the brain. Pharmacologic strategies to inhibit ABCB1/ABCG2-mediated efflux transport at the BBB have been successfully developed in rodents, but it remains unclear whether these can be translated to humans given the pronounced species differences in ABCG2/ABCB1 expression ratios at the BBB. We assessed the efficacy of two different ABCB1/ABCG2 inhibitors to enhance brain distribution of 11C-erlotinib in nonhuman primates as a model of the human BBB. METHODS Papio anubis baboons underwent PET scans of the brain after intravenous injection of 11C-erlotinib under baseline conditions (n = 4) and during intravenous infusion of high-dose erlotinib (10 mg/kg/h, n = 4) or elacridar (12 mg/kg/h, n = 3). RESULTS Under baseline conditions, 11C-erlotinib distribution to the brain (total volume of distribution [VT], 0.22 ± 0.015 mL/cm3) was markedly lower than its distribution to muscle tissue surrounding the skull (VT, 0.86 ± 0.10 mL/cm3). Elacridar infusion resulted in a 3.5 ± 0.9-fold increase in 11C-erlotinib distribution to the brain (VT, 0.81 ± 0.21 mL/cm3, P < 0.01), reaching levels comparable to those in muscle tissue, without changing 11C-erlotinib plasma pharmacokinetics. During high-dose erlotinib infusion, 11C-erlotinib brain distribution was also significantly (1.7 ± 0.2-fold) increased (VT, 0.38 ± 0.033 mL/cm3, P < 0.05), with a concomitant increase in 11C-erlotinib plasma exposure. CONCLUSION We successfully implemented ABCB1/ABCG2 inhibition protocols in nonhuman primates resulting in pronounced increases in brain distribution of 11C-erlotinib. For patients with brain tumors, such inhibition protocols may ultimately be applied to create more effective treatments using drugs that undergo efflux transport at the BBB.
Collapse
Affiliation(s)
- Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Sebastien Goutal
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Sylvain Auvity
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Alexander Traxl
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Severin Mairinger
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Ourkia-Badia Helal
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Irène Buvat
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Michael Soussan
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Oliver Langer
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; and.,Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
78
|
Affiliation(s)
- Timothy P. Heffron
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
79
|
Gao H. Progress and perspectives on targeting nanoparticles for brain drug delivery. Acta Pharm Sin B 2016; 6:268-86. [PMID: 27471668 PMCID: PMC4951594 DOI: 10.1016/j.apsb.2016.05.013] [Citation(s) in RCA: 317] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 02/06/2023] Open
Abstract
Due to the ability of the blood-brain barrier (BBB) to prevent the entry of drugs into the brain, it is a challenge to treat central nervous system disorders pharmacologically. The development of nanotechnology provides potential to overcome this problem. In this review, the barriers to brain-targeted drug delivery are reviewed, including the BBB, blood-brain tumor barrier (BBTB), and nose-to-brain barrier. Delivery strategies are focused on overcoming the BBB, directly targeting diseased cells in the brain, and dual-targeted delivery. The major concerns and perspectives on constructing brain-targeted delivery systems are discussed.
Collapse
|
80
|
Gao H. Perspectives on Dual Targeting Delivery Systems for Brain Tumors. J Neuroimmune Pharmacol 2016; 12:6-16. [PMID: 27270720 DOI: 10.1007/s11481-016-9687-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/31/2016] [Indexed: 12/30/2022]
Abstract
Brain tumor remains one of the most serious threats to human beings. Different from peripheral tumors, drug delivery to brain tumor is largely restricted by the blood brain barrier (BBB). To fully conquer this barrier and specifically deliver drugs to brain tumor, dual targeting delivery systems were explored, which are functionalized with two active targeting ligands: one to the BBB and the other to the brain tumor. The development of dual targeting delivery system is still in its early stage, and attentions need to be paid to issues and concerns that remain unresolved in future studies.
Collapse
Affiliation(s)
- Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China.
| |
Collapse
|
81
|
Chimeric adeno-associated virus and bacteriophage: a potential targeted gene therapy vector for malignant glioma. Ther Deliv 2016; 5:975-90. [PMID: 25375341 DOI: 10.4155/tde.14.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The incipient development of gene therapy for cancer has fuelled its progression from bench to bedside in mere decades. Of all malignancies that exist, gliomas are the largest class of brain tumors, and are renowned for their aggressiveness and resistance to therapy. In order for gene therapy to achieve clinical success, a multitude of barriers ranging from glioma tumor physiology to vector biology must be overcome. Many viral gene delivery systems have been subjected to clinical investigation; however, with highly limited success. In this review, the current progress and challenges of gene therapy for malignant glioma are discussed. Moreover, we highlight the hybrid adeno-associated virus and bacteriophage vector as a potential candidate for targeted gene delivery to brain tumors.
Collapse
|
82
|
Fisusi FA, Siew A, Chooi KW, Okubanjo O, Garrett N, Lalatsa K, Serrano D, Summers I, Moger J, Stapleton P, Satchi-Fainaro R, Schätzlein AG, Uchegbu IF. Lomustine Nanoparticles Enable Both Bone Marrow Sparing and High Brain Drug Levels - A Strategy for Brain Cancer Treatments. Pharm Res 2016; 33:1289-303. [PMID: 26903051 PMCID: PMC4820487 DOI: 10.1007/s11095-016-1872-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 02/02/2016] [Indexed: 12/11/2022]
Abstract
PURPOSE The blood brain barrier compromises glioblastoma chemotherapy. However high blood concentrations of lipophilic, alkylating drugs result in brain uptake, but cause myelosuppression. We hypothesised that nanoparticles could achieve therapeutic brain concentrations without dose-limiting myelosuppression. METHODS Mice were dosed with either intravenous lomustine Molecular Envelope Technology (MET) nanoparticles (13 mg kg(-1)) or ethanolic lomustine (6.5 mg kg(-1)) and tissues analysed. Efficacy was assessed in an orthotopic U-87 MG glioblastoma model, following intravenous MET lomustine (daily 13 mg kg(-1)) or ethanolic lomustine (daily 1.2 mg kg(-1) - the highest repeated dose possible). Myelosuppression and MET particle macrophage uptake were also investigated. RESULTS The MET formulation resulted in modest brain targeting (brain/ bone AUC0-4h ratios for MET and ethanolic lomustine = 0.90 and 0.53 respectively and brain/ liver AUC0-4h ratios for MET and ethanolic lomustine = 0.24 and 0.15 respectively). The MET formulation significantly increased mice (U-87 MG tumours) survival times; with MET lomustine, ethanolic lomustine and untreated mean survival times of 33.2, 22.5 and 21.3 days respectively and there were no material treatment-related differences in blood and femoral cell counts. Macrophage uptake is slower for MET nanoparticles than for liposomes. CONCLUSIONS Particulate drug formulations improved brain tumour therapy without major bone marrow toxicity.
Collapse
Affiliation(s)
- Funmilola A Fisusi
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Adeline Siew
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Kar Wai Chooi
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Omotunde Okubanjo
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Natalie Garrett
- School of Physics, University of Exeter, Exeter, EX4 4QL, UK
| | - Katerina Lalatsa
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Dolores Serrano
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Ian Summers
- School of Physics, University of Exeter, Exeter, EX4 4QL, UK
| | - Julian Moger
- School of Physics, University of Exeter, Exeter, EX4 4QL, UK
| | - Paul Stapleton
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Andreas G Schätzlein
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
- Nanomerics Ltd. Euro House, 1394 High Road, London, N20 9YZ, UK
| | - Ijeoma F Uchegbu
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
- Nanomerics Ltd. Euro House, 1394 High Road, London, N20 9YZ, UK.
| |
Collapse
|
83
|
Shi C, Wu JB, Pan D. Review on near-infrared heptamethine cyanine dyes as theranostic agents for tumor imaging, targeting, and photodynamic therapy. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:50901. [PMID: 27165449 DOI: 10.1117/1.jbo.21.5.050901] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/20/2016] [Indexed: 05/21/2023]
Abstract
A class of near-infrared fluorescence (NIRF) heptamethine cyanine dyes that are taken up and accumulated specifically in cancer cells without chemical conjugation have recently emerged as promising tools for tumor imaging and targeting. In addition to their fluorescence and nuclear imaging-based tumor-imaging properties, these dyes can be developed as drug carriers to safely deliver chemotherapy drugs to tumors. They can also be used as effective agents for photodynamic therapy with remarkable tumoricidal activity via photodependent cytotoxic activity. The preferential uptake of dyes into cancer but not normal cells is co-operatively mediated by the prevailing activation of a group of organic anion-transporting polypeptides on cancer cell membranes, as well as tumor hypoxia and increased mitochondrial membrane potential in cancer cells. Such mechanistic explorations have greatly advanced the current application and future development of NIRF dyes and their derivatives as anticancer theranostic agents. This review summarizes current knowledge and emerging advances in NIRF dyes, including molecular characterization, photophysical properties, multimodal development and uptake mechanisms, and their growing potential for preclinical and clinical use.
Collapse
Affiliation(s)
- Changhong Shi
- Fourth Military Medical University, Laboratory Animal Center, 169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Jason Boyang Wu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Uro-Oncology Research Program, Department of Medicine, Los Angeles, California 90048, United States
| | - Dongfeng Pan
- University of Virginia, Department of Radiology, Charlottesville, Virginia 22908, United States
| |
Collapse
|
84
|
Gupta SK, Kizilbash SH, Carlson BL, Mladek AC, Boakye-Agyeman F, Bakken KK, Pokorny JL, Schroeder MA, Decker PA, Cen L, Eckel-Passow JE, Sarkar G, Ballman KV, Reid JM, Jenkins RB, Verhaak RG, Sulman EP, Kitange GJ, Sarkaria JN. Delineation of MGMT Hypermethylation as a Biomarker for Veliparib-Mediated Temozolomide-Sensitizing Therapy of Glioblastoma. J Natl Cancer Inst 2016; 108:djv369. [PMID: 26615020 PMCID: PMC4862419 DOI: 10.1093/jnci/djv369] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/23/2015] [Accepted: 10/29/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Sensitizing effects of poly-ADP-ribose polymerase inhibitors have been studied in several preclinical models, but a clear understanding of predictive biomarkers is lacking. In this study, in vivo efficacy of veliparib combined with temozolomide (TMZ) was evaluated in a large panel of glioblastoma multiforme (GBM) patient-derived xenografts (PDX) and potential biomarkers were analyzed. METHODS The efficacy of TMZ alone vs TMZ/veliparib was compared in a panel of 28 GBM PDX lines grown as orthotopic xenografts (8-10 mice per group); all tests of statistical significance were two-sided. DNA damage was analyzed by γH2AX immunostaining and promoter methylation of DNA repair gene O6-methylguanine-DNA-methyltransferase (MGMT) by Clinical Laboratory Improvement Amendments-approved methylation-specific polymerase chain reaction. RESULTS The combination of TMZ/veliparib statistically significantly extended survival of GBM models (P < .05 by log-rank) compared with TMZ alone in five of 20 MGMT-hypermethylated lines (average extension in median survival = 87 days, range = 20-150 days), while the combination was ineffective in six MGMT-unmethylated lines. In the MGMT promoter-hypermethylated GBM12 line (median survival with TMZ+veliparib = 189 days, 95% confidence interval [CI] = 59 to 289 days, vs TMZ alone = 98 days, 95% CI = 49 to 210 days, P = .04), the profound TMZ-sensitizing effect of veliparib was lost when MGMT was overexpressed (median survival with TMZ+veliparib = 36 days, 95% CI = 28 to 38 days, vs TMZ alone = 35 days, 95% CI = 32 to 37 days, P = .87), and a similar association was observed in two nearly isogenic GBM28 sublines with an intact vs deleted MGMT locus. In comparing DNA damage signaling after dosing with veliparib/TMZ or TMZ alone, increased phosphorylation of damage-responsive proteins (KAP1, Chk1, Chk2, and H2AX) was observed only in MGMT promoter-hypermethylated lines. CONCLUSION Veliparib statistically significantly enhances (P < .001) the efficacy of TMZ in tumors with MGMT promoter hypermethylation. Based on these data, MGMT promoter hypermethylation is being used as an eligibility criterion for A071102 (NCT02152982), the phase II/III clinical trial evaluating TMZ/veliparib combination in patients with GBM.
Collapse
Affiliation(s)
- Shiv K Gupta
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Sani H Kizilbash
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Brett L Carlson
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Ann C Mladek
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Felix Boakye-Agyeman
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Katrina K Bakken
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Jenny L Pokorny
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Mark A Schroeder
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Paul A Decker
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Ling Cen
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Jeanette E Eckel-Passow
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Gobinda Sarkar
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Karla V Ballman
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Joel M Reid
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Robert B Jenkins
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Roeland G Verhaak
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Erik P Sulman
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Gaspar J Kitange
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS)
| | - Jann N Sarkaria
- Department of Radiation Oncology (SKG, BLC, ACM, KKB, JLP, MAS, LC, GJK, JNS), Division of Medical Oncology (SHK), Molecular Pharmacology and Experimental Therapeutics (FBA, JMR), Division of Biomedical Statistics and Informatics (PAD, JEEP, KVB), and Laboratory Medicine and Pathology (GS, RBJ), Mayo Clinic, Rochester MN; The University of Texas, MD Anderson Cancer Center, Houston, TX (RGV, EPS).
| |
Collapse
|
85
|
Tamborini M, Locatelli E, Rasile M, Monaco I, Rodighiero S, Corradini I, Franchini MC, Passoni L, Matteoli M. A Combined Approach Employing Chlorotoxin-Nanovectors and Low Dose Radiation To Reach Infiltrating Tumor Niches in Glioblastoma. ACS NANO 2016; 10:2509-2520. [PMID: 26745323 DOI: 10.1021/acsnano.5b07375] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of glioma, with life expectancy of around 2 years after diagnosis, due to recidivism and to the blood-brain barrier (BBB) limiting the amount of drugs which reach the residual malignant cells, thus contributing to the failure of chemotherapies. To bypass the obstacles imposed by the BBB, we investigated the use of nanotechnologies combined with radiotherapy, as a potential therapeutic strategy for GBM. We used poly(lactic-co-glycolic acid) (PLGA) nanoparticles (PNP) conjugated to chlorotoxin (CTX), a peptide reported to bind selectively to glioma cells. Silver nanoparticles were entrapped inside the functionalized nanoparticles (Ag-PNP-CTX), to allow detection and quantification of the cellular uptake by confocal microscopy, both in vitro and in vivo. In vitro experiments performed with different human glioblastoma cell lines showed higher cytoplasmic uptake of Ag-PNP-CTX, with respect to nonfunctionalized nanoparticles. In vivo experiments showed that Ag-NP-CTX efficiently targets the tumor, but are scarcely effective in crossing the blood brain barrier in the healthy brain, where dispersed metastatic cells are present. We show here that single whole brain X-ray irradiation, performed 20 h before nanoparticle injection, enhances the expression of the CTX targets, MMP-2 and ClC-3, and, through BBB permeabilization, potently increases the amount of internalized Ag-PNP-CTX even in dispersed cells, and generated an efficient antitumor synergistic effect able to inhibit in vivo tumor growth. Notably, the application of Ag-PNP-CTX to irradiated tumor cells decreases the extracellular activity of MMP-2. By targeting dispersed GBM cells and reducing MMP-2 activity, the combined use of CTX-nanovectors with radiotherapy may represent a promising therapeutic approach toward GBM.
Collapse
Affiliation(s)
- Matteo Tamborini
- Department of Medical Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
- CNR Institute of Neuroscience, Via Vanvitelli 32, 20129 Milano, Italy
| | - Erica Locatelli
- Department of Industrial Chemistry "Toso Montanari", University of Bologna , Viale Risorgimento 4, 40136 Bologna, Italy
| | - Marco Rasile
- Department of Medical Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
- Laboratory of Pharmacology and Brain Pathology, Humanitas Research Hospital , Via Manzoni 56, Rozzano, 20089 Milano, Italy
| | - Ilaria Monaco
- Department of Industrial Chemistry "Toso Montanari", University of Bologna , Viale Risorgimento 4, 40136 Bologna, Italy
| | | | - Irene Corradini
- Department of Medical Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
- CNR Institute of Neuroscience, Via Vanvitelli 32, 20129 Milano, Italy
| | - Mauro Comes Franchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna , Viale Risorgimento 4, 40136 Bologna, Italy
| | - Lorena Passoni
- Department of Medical Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
- Fondazione Filarete, Viale Ortles 22/4, 20139 Milano, Italy
| | - Michela Matteoli
- CNR Institute of Neuroscience, Via Vanvitelli 32, 20129 Milano, Italy
- Laboratory of Pharmacology and Brain Pathology, Humanitas Research Hospital , Via Manzoni 56, Rozzano, 20089 Milano, Italy
| |
Collapse
|
86
|
Parrish KE, Cen L, Murray J, Calligaris D, Kizilbash S, Mittapalli RK, Carlson BL, Schroeder MA, Sludden J, Boddy AV, Agar NYR, Curtin NJ, Elmquist WF, Sarkaria JN. Efficacy of PARP Inhibitor Rucaparib in Orthotopic Glioblastoma Xenografts Is Limited by Ineffective Drug Penetration into the Central Nervous System. Mol Cancer Ther 2015; 14:2735-43. [PMID: 26438157 DOI: 10.1158/1535-7163.mct-15-0553] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/14/2015] [Indexed: 12/22/2022]
Abstract
PARP inhibition can enhance the efficacy of temozolomide and prolong survival in orthotopic glioblastoma (GBM) xenografts. The aim of this study was to evaluate the combination of the PARP inhibitor rucaparib with temozolomide and to correlate pharmacokinetic and pharmacodynamic studies with efficacy in patient-derived GBM xenograft models. The combination of rucaparib with temozolomide was highly effective in vitro in short-term explant cultures derived from GBM12, and, similarly, the combination of rucaparib and temozolomide (dosed for 5 days every 28 days for 3 cycles) significantly prolonged the time to tumor regrowth by 40% in heterotopic xenografts. In contrast, the addition of rucaparib had no impact on the efficacy of temozolomide in GBM12 or GBM39 orthotopic models. Using Madin-Darby canine kidney (MDCK) II cells stably expressing murine BCRP1 or human MDR1, cell accumulation studies demonstrated that rucaparib is transported by both transporters. Consistent with the influence of these efflux pumps on central nervous system drug distribution, Mdr1a/b(-/-)Bcrp1(-/-) knockout mice had a significantly higher brain to plasma ratio for rucaparib (1.61 ± 0.25) than wild-type mice (0.11 ± 0.08). A pharmacokinetic and pharmacodynamic evaluation after a single dose confirmed limited accumulation of rucaparib in the brain is associated with substantial residual PARP enzymatic activity. Similarly, matrix-assisted laser desorption/ionization mass spectrometric imaging demonstrated significantly enhanced accumulation of drug in flank tumor compared with normal brain or orthotopic tumors. Collectively, these results suggest that limited drug delivery into brain tumors may significantly limit the efficacy of rucaparib combined with temozolomide in GBM.
Collapse
Affiliation(s)
- Karen E Parrish
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota
| | - Ling Cen
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - James Murray
- Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Calligaris
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sani Kizilbash
- Department of Medical Oncology Mayo Clinic, Rochester, Minnesota
| | | | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Mark A Schroeder
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Alan V Boddy
- Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - William F Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota
| | - Jann N Sarkaria
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
87
|
Traxl A, Wanek T, Mairinger S, Stanek J, Filip T, Sauberer M, Müller M, Kuntner C, Langer O. Breast Cancer Resistance Protein and P-Glycoprotein Influence In Vivo Disposition of 11C-Erlotinib. J Nucl Med 2015; 56:1930-6. [PMID: 26359257 DOI: 10.2967/jnumed.115.161273] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/31/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED (11)C-erlotinib is a PET tracer to distinguish responders from nonresponders to epidermal growth factor receptor-targeted tyrosine kinase inhibitors and may also be of interest to predict distribution of erlotinib to tissues targeted for treatment. The aim of this study was to investigate if the known interaction of erlotinib with the multidrug efflux transporters breast cancer resistance protein (humans, ABCG2; rodents, Abcg2) and P-glycoprotein (humans, ABCB1; rodents, Abcb1a/b) affects tissue distribution and excretion of (11)C-erlotinib and has an influence on the ability of (11)C-erlotinib PET to predict erlotinib tissue distribution at therapeutic doses. METHODS Wild-type and Abcb1a/b or Abcg2 knockout mice underwent (11)C-erlotinib PET/MR scans, with or without the coinjection of a pharmacologic dose of erlotinib (10 mg/kg) or after pretreatment with the ABCB1/ABCG2 inhibitor elacridar (10 mg/kg). Integration plot analysis was used to determine organ uptake (CLuptake) and biliary excretion (CLbile) clearances of radioactivity. RESULTS (11)C-erlotinib distribution to the brain was restricted by Abcb1a/b and Abcg2, and CLuptake into the brain was only significantly increased when both Abcb1a/b and Abcg2 were absent (wild-type mice, 0.017 ± 0.004 mL/min/g of tissue; Abcb1a/b((-/-))Abcg2((-/-)) mice, 0.079 ± 0.013 mL/min/g of tissue; P < 0.001). The pretreatment of wild-type mice with elacridar increased CLuptake into the brain to levels comparable to Abcb1a/b((-/-))Abcg2((-/-)) mice (0.090 ± 0.007 mL/min/g of tissue, P < 0.001). The absence of Abcb1a/b and Abcg2 led to a 2.6-fold decrease in CLbile (wild-type mice, 0.025 ± 0.005 mL/min/g of tissue; Abcb1a/b((-/-))Abcg2((-/-)) mice, 0.0095 ± 0.001 mL/min/g of tissue; P < 0.001). There were pronounced differences in distribution of (11)C-erlotinib to the brain, liver, kidney, and lung and hepatobiliary excretion into intestine between animals injected with a microdose and pharmacologic dose of erlotinib. CONCLUSION ABCG2, ABCB1, and possibly other transporters influence in vivo disposition of (11)C-erlotinib and thereby affect its distribution to normal and potentially also tumor tissue. Saturable transport of erlotinib leads to nonlinear pharmacokinetics, possibly compromising the prediction of erlotinib tissue distribution at therapeutic doses from PET with a microdose of (11)C-erlotinib. The inhibition of ABCB1 and ABCG2 is a promising approach to enhance brain distribution of erlotinib to increase its efficacy in the treatment of brain tumors.
Collapse
Affiliation(s)
- Alexander Traxl
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and
| | - Thomas Wanek
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and
| | - Severin Mairinger
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and
| | - Johann Stanek
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Filip
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and
| | - Michael Sauberer
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and
| | - Markus Müller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Claudia Kuntner
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and
| | - Oliver Langer
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; and Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
88
|
Oberoi RK, Parrish KE, Sio TT, Mittapalli RK, Elmquist WF, Sarkaria JN. Strategies to improve delivery of anticancer drugs across the blood-brain barrier to treat glioblastoma. Neuro Oncol 2015; 18:27-36. [PMID: 26359209 DOI: 10.1093/neuonc/nov164] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 07/15/2015] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma (GBM) is a lethal and aggressive brain tumor that is resistant to conventional radiation and cytotoxic chemotherapies. Molecularly targeted agents hold great promise in treating these genetically heterogeneous tumors, yet have produced disappointing results. One reason for the clinical failure of these novel therapies can be the inability of the drugs to achieve effective concentrations in the invasive regions beyond the bulk tumor. In this review, we describe the influence of the blood-brain barrier on the distribution of anticancer drugs to both the tumor core and infiltrative regions of GBM. We further describe potential strategies to overcome these drug delivery limitations. Understanding the key factors that limit drug delivery into brain tumors will guide future development of approaches for enhanced delivery of effective drugs to GBM.
Collapse
Affiliation(s)
- Rajneet K Oberoi
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (R.K.O., K.E.P., R.K.M., W.F.E.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (T.T.S., J.N.S.)
| | - Karen E Parrish
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (R.K.O., K.E.P., R.K.M., W.F.E.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (T.T.S., J.N.S.)
| | - Terence T Sio
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (R.K.O., K.E.P., R.K.M., W.F.E.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (T.T.S., J.N.S.)
| | - Rajendar K Mittapalli
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (R.K.O., K.E.P., R.K.M., W.F.E.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (T.T.S., J.N.S.)
| | - William F Elmquist
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (R.K.O., K.E.P., R.K.M., W.F.E.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (T.T.S., J.N.S.)
| | - Jann N Sarkaria
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (R.K.O., K.E.P., R.K.M., W.F.E.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (T.T.S., J.N.S.)
| |
Collapse
|
89
|
The TWEAK receptor Fn14 is a potential cell surface portal for targeted delivery of glioblastoma therapeutics. Oncogene 2015; 35:2145-55. [PMID: 26300004 DOI: 10.1038/onc.2015.310] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/14/2015] [Accepted: 07/14/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Fibroblast growth factor-inducible 14 (Fn14; TNFRSF12A) is the cell surface receptor for the tumor necrosis factor (TNF) family member TNF-like weak inducer of apoptosis (TWEAK). The Fn14 gene is normally expressed at low levels in healthy tissues but expression is significantly increased after tissue injury and in many solid tumor types, including glioblastoma (GB; formerly referred to as 'GB multiforme'). GB is the most common and aggressive primary malignant brain tumor and the current standard-of-care therapeutic regimen has a relatively small impact on patient survival, primarily because glioma cells have an inherent propensity to invade into normal brain parenchyma, which invariably leads to tumor recurrence and patient death. Despite major, concerted efforts to find new treatments, a new GB therapeutic that improves survival has not been introduced since 2005. In this review article, we summarize studies indicating that (i) Fn14 gene expression is low in normal brain tissue but is upregulated in advanced brain cancers and, in particular, in GB tumors exhibiting the mesenchymal molecular subtype; (ii) Fn14 expression can be detected in glioma cells residing in both the tumor core and invasive rim regions, with the maximal levels found in the invading glioma cells located within normal brain tissue; and (iii) TWEAK Fn14 engagement as well as Fn14 overexpression can stimulate glioma cell migration, invasion and resistance to chemotherapeutic agents in vitro. We also discuss two new therapeutic platforms that are currently in development that leverage Fn14 overexpression in GB tumors as a way to deliver cytotoxic agents to the glioma cells remaining after surgical resection while sparing normal healthy brain cells.
Collapse
|
90
|
Wu JB, Shi C, Chu GCY, Xu Q, Zhang Y, Li Q, Yu JS, Zhau HE, Chung LWK. Near-infrared fluorescence heptamethine carbocyanine dyes mediate imaging and targeted drug delivery for human brain tumor. Biomaterials 2015. [PMID: 26197410 DOI: 10.1016/j.biomaterials.2015.07.028] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain tumors and brain metastases are among the deadliest malignancies of all human cancers, largely due to the cellular blood-brain and blood-tumor barriers that limit the delivery of imaging and therapeutic agents from the systemic circulation to tumors. Thus, improved strategies for brain tumor visualization and targeted treatment are critically needed. Here we identified and synthesized a group of near-infrared fluorescence (NIRF) heptamethine carbocyanine dyes and derivative NIRF dye-drug conjugates for effective imaging and therapeutic targeting of brain tumors of either primary or metastatic origin in mice, which is mechanistically mediated by tumor hypoxia and organic anion-transporting polypeptide genes. We also demonstrate that these dyes, when conjugated to chemotherapeutic agents such as gemcitabine, significantly restricted the growth of both intracranial glioma xenografts and prostate tumor brain metastases and prolonged survival in mice. These results show promise in the application of NIRF dyes as novel theranostic agents for the detection and treatment of brain tumors.
Collapse
Affiliation(s)
- Jason Boyang Wu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Changhong Shi
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Gina Chia-Yi Chu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Qijin Xu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yi Zhang
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Qinlong Li
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - John S Yu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
91
|
Raub TJ, Wishart GN, Kulanthaivel P, Staton BA, Ajamie RT, Sawada GA, Gelbert LM, Shannon HE, Sanchez-Martinez C, De Dios A. Brain Exposure of Two Selective Dual CDK4 and CDK6 Inhibitors and the Antitumor Activity of CDK4 and CDK6 Inhibition in Combination with Temozolomide in an Intracranial Glioblastoma Xenograft. Drug Metab Dispos 2015; 43:1360-71. [PMID: 26149830 DOI: 10.1124/dmd.114.062745] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/02/2015] [Indexed: 11/22/2022] Open
Abstract
Effective treatments for primary brain tumors and brain metastases represent a major unmet medical need. Targeting the CDK4/CDK6-cyclin D1-Rb-p16/ink4a pathway using a potent CDK4 and CDK6 kinase inhibitor has potential for treating primary central nervous system tumors such as glioblastoma and some peripheral tumors with high incidence of brain metastases. We compared central nervous system exposures of two orally bioavailable CDK4 and CDK6 inhibitors: abemaciclib, which is currently in advanced clinical development, and palbociclib (IBRANCE; Pfizer), which was recently approved by the U.S. Food and Drug Administration. Abemaciclib antitumor activity was assessed in subcutaneous and orthotopic glioma models alone and in combination with standard of care temozolomide (TMZ). Both inhibitors were substrates for xenobiotic efflux transporters P-glycoprotein and breast cancer resistant protein expressed at the blood-brain barrier. Brain Kp,uu values were less than 0.2 after an equimolar intravenous dose indicative of active efflux but were approximately 10-fold greater for abemaciclib than palbociclib. Kp,uu increased 2.8- and 21-fold, respectively, when similarly dosed in P-gp-deficient mice. Abemaciclib had brain area under the curve (0-24 hours) Kp,uu values of 0.03 in mice and 0.11 in rats after a 30 mg/kg p.o. dose. Orally dosed abemaciclib significantly increased survival in a rat orthotopic U87MG xenograft model compared with vehicle-treated animals, and efficacy coincided with a dose-dependent increase in unbound plasma and brain exposures in excess of the CDK4 and CDK6 Ki values. Abemaciclib increased survival time of intracranial U87MG tumor-bearing rats similar to TMZ, and the combination of abemaciclib and TMZ was additive or greater than additive. These data show that abemaciclib crosses the blood-brain barrier and confirm that both CDK4 and CDK6 inhibitors reach unbound brain levels in rodents that are expected to produce enzyme inhibition; however, abemaciclib brain levels are reached more efficiently at presumably lower doses than palbociclib and are potentially on target for a longer period of time.
Collapse
Affiliation(s)
- Thomas J Raub
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Graham N Wishart
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Palaniappan Kulanthaivel
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Brian A Staton
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Rose T Ajamie
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Geri A Sawada
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Lawrence M Gelbert
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Harlan E Shannon
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Concepcion Sanchez-Martinez
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Alfonso De Dios
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| |
Collapse
|
92
|
Mendes B, Marques C, Carvalho I, Costa P, Martins S, Ferreira D, Sarmento B. Influence of glioma cells on a new co-culture in vitro blood–brain barrier model for characterization and validation of permeability. Int J Pharm 2015; 490:94-101. [PMID: 25981617 DOI: 10.1016/j.ijpharm.2015.05.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 05/07/2015] [Accepted: 05/09/2015] [Indexed: 01/11/2023]
|
93
|
Mason WP. Blood-brain barrier-associated efflux transporters: a significant but underappreciated obstacle to drug development in glioblastoma. Neuro Oncol 2015; 17:1181-2. [PMID: 26138634 DOI: 10.1093/neuonc/nov122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 01/28/2023] Open
Affiliation(s)
- Warren P Mason
- Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada (W.P.M.)
| |
Collapse
|
94
|
Barboza T, Gomes T, Mizurini DM, Monteiro RQ, König S, Francischetti IMB, Signoretti PVP, Ramos IP, Gutfilen B, Souza SAL. (99m)Tc-ixolaris targets glioblastoma-associated tissue factor: in vitro and pre-clinical applications. Thromb Res 2015; 136:432-9. [PMID: 26070446 DOI: 10.1016/j.thromres.2015.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 04/18/2015] [Accepted: 05/30/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND The clotting initiator protein tissue factor (TF) has recently been described as a potential target that can be exploited to image aggressive tumors. Ixolaris is a specific TF inhibitor that blocks tumor cell procoagulant activity and tumor growth. OBJECTIVE Herein we evaluated the ability of (99m)Tc-ixolaris to target tumor-derived TF using an orthotopic glioblastoma (GBM) model in mice. METHODS The right forebrains of Swiss mice were stereotactically inoculated with U87-MG human GBM cells. Histological and immunohistochemical analyses were performed on the resulting tumors after 35-45 days. The biodistribution of (99m)Tc-ixolaris was evaluated by semi-quantitative whole-body scintigraphy and a quantitative analysis of radioactivity in isolated organs. RESULTS No (99m)Tc-ixolaris uptake was observed in brain of tumor-free mice, independently of the integrity of brain-blood barrier. In contrast, the presence of TF-expressing brain tumor masses determined a significant (99m)Tc-ixolaris uptake. CONCLUSION (99m)Tc-ixolaris recognized TF-expressing GBM cells in vivo. Given the proposed role of TF in tumor progression, (99m)Tc-ixolaris is a promising radiopharmaceutical agent for quantifying cancer-associated TF in aggressive tumors, including GBM.
Collapse
Affiliation(s)
- Thiago Barboza
- Laboratório de Marcação de Células e Moléculas, Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Tainá Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil; Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Daniella M Mizurini
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Robson Q Monteiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Sandra König
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Ivo M B Francischetti
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Paula V P Signoretti
- Laboratório de Físico-Química Biológica Aída Hassón Voloch e Laboratório Intermediário de Biomembranas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Isalira P Ramos
- Laboratório de Cardiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Bianca Gutfilen
- Laboratório de Marcação de Células e Moléculas, Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Sergio A L Souza
- Laboratório de Marcação de Células e Moléculas, Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
95
|
Fakhoury M. Drug delivery approaches for the treatment of glioblastoma multiforme. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1365-73. [PMID: 26046399 DOI: 10.3109/21691401.2015.1052467] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
CONTEXT Glioblastoma multiforme (GBM) is by far the most common and aggressive form of glial tumor. It is characterized by a highly proliferative population of cells that invade surrounding tissue and that frequently recur after surgical resection and chemotherapy. Over the last decades, a number of promising novel pharmacological approaches have been investigated, but most of them have failed clinical trials due to some side-effects such as toxicity and poor drug delivery to the brain. The major obstacle in the treatment of GBM is the presence of the blood-brain barrier (BBB). Due to their relatively high molecular weight, most therapeutic drugs fail to cross the BBB from the blood circulation. OBJECTIVE This paper sheds light on the characteristics of GBM and the challenges of current pharmacological treatments. A closer look is given to the role of nanotechnology in the field of drug delivery, and its application in the treatment of brain tumors such as GBM. METHOD For this purpose, effort was made to select the most recent studies using predefined search criteria that included at least one of the following keywords in the PubMed and Medline databases: glioblastoma, drug delivery, blood-brain barrier, nanotechnology, and nanoparticle. CONCLUSION Breakthrough in nanotechnology offers promising applications in cancer therapy and targeted drug delivery. However, more efforts need to be devoted to the development of novel therapeutic strategies that enable the delivery of drugs to desired areas of the brain with limited side-effects and higher therapeutic efficiency.
Collapse
Affiliation(s)
- Marc Fakhoury
- a Department of Neurosciences , University of Montreal , Montreal , QC , Canada
| |
Collapse
|
96
|
Applied Pharmaceutical Analysis India 2014 conference report. Bioanalysis 2015; 6:1891-6. [PMID: 25158960 DOI: 10.4155/bio.14.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Applied Pharmaceutical Analysis (APA) India 23-26 February 2014, Ahmedabad, India The fifth Applied Pharmaceutical Analysis (APA) India meeting was held in February 2014 at Hyatt Ahmedabad, India. With the theme of 'The Science of Measurement: Current status and Future trends in Bioanalysis, Biotransformation and Drug Discovery Platforms', the conference was attended by over 160 delegates. The agenda comprised advanced and relevant research topics in the key areas of bioanalysis and drug metabolism. APA India 2014 provided a unique platform for networking and professional linking to participants, innovators and policy-makers. As part of the global research community, APA India continues to grow and receive considerable attention from the drug discovery and development community of India.
Collapse
|
97
|
Zhang F, Xu CL, Liu CM. Drug delivery strategies to enhance the permeability of the blood-brain barrier for treatment of glioma. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2089-100. [PMID: 25926719 PMCID: PMC4403597 DOI: 10.2147/dddt.s79592] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gliomas are amongst the most insidious and destructive types of brain cancer and are associated with a poor prognosis, frequent recurrences, and extremely high lethality despite combination treatment of surgery, radiotherapy, and chemotherapy. The existence of the blood–brain barrier (BBB) restricts the delivery of therapeutic molecules into the brain and offers the clinical efficacy of many pharmaceuticals that have been demonstrated to be effective for other kinds of tumors. This challenge emphasizes the need to be able to deliver drugs effectively across the BBB to reach the brain parenchyma. Enhancement of the permeability of the BBB and being able to transport drugs across it has been shown to be a promising strategy to improve drug absorption and treatment efficacy. This review highlights the innovative technologies that have been introduced to enhance the permeability of the BBB and to obtain an optimal distribution and concentration of drugs in the brain to treat gliomas, such as nanotechniques, hyperthermia techniques, receptor-mediated transport, cell-penetrating peptides, and cell-mediated delivery.
Collapse
Affiliation(s)
- Fang Zhang
- School of Pharmacy, National First-Class Key Discipline for Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Chun-Lei Xu
- School of Pharmacy, National First-Class Key Discipline for Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Chun-Mei Liu
- School of Pharmacy, National First-Class Key Discipline for Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
98
|
Stylli SS, Luwor RB, Ware TM, Tan F, Kaye AH. Mouse models of glioma. J Clin Neurosci 2015; 22:619-26. [DOI: 10.1016/j.jocn.2014.10.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/15/2014] [Indexed: 10/24/2022]
|
99
|
Azuaje F, Tiemann K, Niclou SP. Therapeutic control and resistance of the EGFR-driven signaling network in glioblastoma. Cell Commun Signal 2015; 13:23. [PMID: 25885672 PMCID: PMC4391485 DOI: 10.1186/s12964-015-0098-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/10/2015] [Indexed: 12/31/2022] Open
Abstract
The alteration of the epidermal growth factor receptor (EGFR)-driven signaling network is a characteristic feature of glioblastomas (GBM), and its inhibition represents a treatment strategy. However, EGFR-targeted interventions have been largely ineffective. Complex perturbations in this system are likely to be central to tumor cells with high adaptive capacity and resistance to therapies. We review key concepts and mechanisms relevant to EGFR-targeted treatment resistance at a systems level. Our understanding of treatment resistance as a systems-level phenomenon is necessary to develop effective therapeutic options for GBM patients. This is allowing us to go beyond the notion of therapeutic targets as single molecular components, into strategies that can weaken cancer signaling robustness and boost inherent network-level vulnerabilities.
Collapse
Affiliation(s)
- Francisco Azuaje
- Department of Oncology, NorLux Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg.
| | - Katja Tiemann
- Department of Oncology, NorLux Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg.
| | - Simone P Niclou
- Department of Oncology, NorLux Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg.
| |
Collapse
|
100
|
Mechanisms regulating glioma invasion. Cancer Lett 2015; 362:1-7. [PMID: 25796440 DOI: 10.1016/j.canlet.2015.03.015] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/10/2015] [Accepted: 03/11/2015] [Indexed: 01/10/2023]
Abstract
Glioblastoma (GBM) is the most aggressive, deadliest, and most common brain malignancy in adults. Despite the advances made in surgical techniques, radiotherapy and chemotherapy, the median survival for GBM patients has remained at a mere 14 months. GBM poses several unique challenges to currently available treatments for the disease. For example, GBM cells have the propensity to aggressively infiltrate/invade into the normal brain tissues and along the vascular tracks, which prevents complete resection of all malignant cells and limits the effect of localized radiotherapy while sparing normal tissue. Although anti-angiogenic treatment exerts anti-edematic effect in GBM, unfortunately, tumors progress with acquired increased invasiveness. Therefore, it is an important task to gain a deeper understanding of the intrinsic and post-treatment invasive phenotypes of GBM in hopes that the gained knowledge would lead to novel GBM treatments that are more effective and less toxic. This review will give an overview of some of the signaling pathways that have been shown to positively and negatively regulate GBM invasion, including, the PI3K/Akt, Wnt, sonic hedgehog-GLI1, and microRNAs. The review will also discuss several approaches to cancer therapies potentially altering GBM invasiveness.
Collapse
|