601
|
Li Z, Ma J, Liu L, Liu X, Wang P, Liu Y, Li Z, Zheng J, Chen J, Tao W, Xue Y. Endothelial-Monocyte Activating Polypeptide II Suppresses the In Vitro Glioblastoma-Induced Angiogenesis by Inducing Autophagy. Front Mol Neurosci 2017; 10:208. [PMID: 28701921 PMCID: PMC5488748 DOI: 10.3389/fnmol.2017.00208] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 06/14/2017] [Indexed: 12/18/2022] Open
Abstract
The obstacle in delivering therapeutics to glioblastoma (GBM) is tumor-induced angiogenesis which leads to the formation of abnormal vessels and a dysfunctional blood-tumor barrier. Here, we elucidated the effect of endothelial-monocyte activating polypeptide II (EMAP II) on the GBM-induced angiogenesis as well as its potential mechanisms. Our results proved that EMAP II inhibited the viability, mitochondrial membrane potential, migration and tube formation of GBM-induced endothelial cells (GECs) by inducing cell autophagy, demonstrated by cell viability assay, JC-1 staining assay, transwell assay and tube formation assay, respectively. Cell autophagy was induced by EMAP II through the observation of autophagic vacuoles formation and the up-regulation of microtubule-associated protein-1 light chain-3 (LC3)-II and p62/SQSTM1 expression, demonstrated by transmission electron microscopy analysis, immunofluorescence assay and Western blot assay. The activity of PI3K/AKT/mTOR signal pathway could be inhibited by the EMAP II treatment. Furthermore, unfolded protein response (UPR)-related proteins (GRP78, eIF2α, and CHOP) were up-regulated by EMAP II, which suggest that GECs exposed to EMAP II experienced endoplasmic reticulum stress. Further, mechanistic investigations found that EMAP II reduced the miR-96 expression which could directly target the 3'-UTR of these UPR-related proteins, and over-expression of miR-96 inhibited LC3 and p62/SQSTM1 expression by down-regulating these UPR-related proteins in GECs. Moreover, the combination of EMAP II with miR-96 inhibitor showed the inhibitory effect on the viability, migration, and in vitro tube formation of GECs, which are critical for angiogenesis. Taken together, we have demonstrated the fact that EMAP II resulted in the decreased GBM-induced angiogenesis by inducing autophagy, which might contribute to establishing potential strategies for human GBM treatment.
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical UniversityShenyang, China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical UniversityShenyang, China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical UniversityShenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China
- Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical UniversityShenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China
- Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China
- Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China
- Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Jiajia Chen
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical UniversityShenyang, China
| | - Wei Tao
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical UniversityShenyang, China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical UniversityShenyang, China
| |
Collapse
|
602
|
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
|
603
|
Zhao HF, Wang J, Shao W, Wu CP, Chen ZP, To SST, Li WP. Recent advances in the use of PI3K inhibitors for glioblastoma multiforme: current preclinical and clinical development. Mol Cancer 2017; 16:100. [PMID: 28592260 PMCID: PMC5463420 DOI: 10.1186/s12943-017-0670-3] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/26/2017] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive malignant primary tumor in the central nervous system. One of the most widely used chemotherapeutic drugs for GBM is temozolomide, which is a DNA-alkylating agent and its efficacy is dependent on MGMT methylation status. Little progress in improving the prognosis of GBM patients has been made in the past ten years, urging the development of more effective molecular targeted therapies. Hyper-activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway is frequently found in a variety of cancers including GBM, and it plays a central role in the regulation of tumor cell survival, growth, motility, angiogenesis and metabolism. Numerous PI3K inhibitors including pan-PI3K, isoform-selective and dual PI3K/mammalian target of rapamycin (mTOR) inhibitors have exhibited favorable preclinical results and entered clinical trials in a range of hematologic malignancies and solid tumors. Furthermore, combination of inhibitors targeting PI3K and other related pathways may exert synergism on suppressing tumor growth and improving patients' prognosis. Currently, only a handful of PI3K inhibitors are in phase I/II clinical trials for GBM treatment. In this review, we focus on the importance of PI3K/Akt pathway in GBM, and summarize the current development of PI3K inhibitors alone or in combination with other inhibitors for GBM treatment from preclinical to clinical studies.
Collapse
Affiliation(s)
- Hua-fu Zhao
- Department of Neurosurgery & Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035 China
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 China
| | - Jing Wang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 China
| | - Wei Shao
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Chang-peng Wu
- Department of Neurosurgery & Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035 China
- College of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
| | - Zhong-ping Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 China
| | - Shing-shun Tony To
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Wei-ping Li
- Department of Neurosurgery & Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035 China
| |
Collapse
|
604
|
Ziegler J, Bastian A, Lerner M, Bailey-Downs L, Saunders D, Smith N, Sutton J, Battiste JD, Ihnat MA, Gangjee A, Towner RA. AG488 as a therapy against gliomas. Oncotarget 2017; 8:71833-71844. [PMID: 29069750 PMCID: PMC5641093 DOI: 10.18632/oncotarget.18284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/05/2017] [Indexed: 11/25/2022] Open
Abstract
High-grade gliomas such as glioblastomas (GBM) present a deadly prognosis following diagnosis and very few effective treatment options. Here, we investigate if the small molecule AG488 can be an effective therapy against GBM with both anti-angiogenic as well as an anti-microtubule inhibiting modalities, using a human G55 glioma xenograft model in nude mice. From in vitro studies, we report that AG488 incubation reduced cell viability in G55 and HMEC-1 cells more so than TMZ treatment, and AG488 treatment also decreased cell viability in normal astrocytes, but not as much as for G55 cells (p<0.0001). In vivo investigations indicated that AG488 therapy helped reduce tumor volumes (p<0.0001), prolong survival (p<0.01), increase tumor perfusion (p<0.01), and decrease microvessel density (MVD) (p<0.05), compared to untreated mice or mice treated with non-specific IgG, in the G55 xenograft model. Additionally, AG488 did not induce apoptosis in normal mouse brain tissue. Animal survival and tumor volume changes for AG488 were comparable to TMZ or anti-VEGF therapies, however AG488 was found to be more effective in decreasing tumor-related vascularity (perfusion and MVD). AG488 is a potential novel therapy against high-grade gliomas.
Collapse
Affiliation(s)
- Jadith Ziegler
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anja Bastian
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Megan Lerner
- Department of Surgery Research Laboratory, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Lora Bailey-Downs
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jake Sutton
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - James D Battiste
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael A Ihnat
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Aleem Gangjee
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
605
|
Abstract
Malignant brain tumors represent one of the most devastating forms of cancer with abject survival rates that have not changed in the past 60years. This is partly because the brain is a critical organ, and poses unique anatomical, physiological, and immunological barriers. The unique interplay of these barriers also provides an opportunity for creative engineering solutions. Cancer immunotherapy, a means of harnessing the host immune system for anti-tumor efficacy, is becoming a standard approach for treating many cancers. However, its use in brain tumors is not widespread. This review discusses the current approaches, and hurdles to these approaches in treating brain tumors, with a focus on immunotherapies. We identify critical barriers to immunoengineering brain tumor therapies and discuss possible solutions to these challenges.
Collapse
|
606
|
Sewing ACP, Lagerweij T, van Vuurden DG, Meel MH, Veringa SJE, Carcaboso AM, Gaillard PJ, Peter Vandertop W, Wesseling P, Noske D, Kaspers GJL, Hulleman E. Preclinical evaluation of convection-enhanced delivery of liposomal doxorubicin to treat pediatric diffuse intrinsic pontine glioma and thalamic high-grade glioma. J Neurosurg Pediatr 2017; 19:518-530. [PMID: 28291423 DOI: 10.3171/2016.9.peds16152] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Pediatric high-grade gliomas (pHGGs) including diffuse intrinsic pontine gliomas (DIPGs) are primary brain tumors with high mortality and morbidity. Because of their poor brain penetrance, systemic chemotherapy regimens have failed to deliver satisfactory results; however, convection-enhanced delivery (CED) may be an alternative mode of drug delivery. Anthracyclines are potent chemotherapeutics that have been successfully delivered via CED in preclinical supratentorial glioma models. This study aims to assess the potency of anthracyclines against DIPG and pHGG cell lines in vitro and to evaluate the efficacy of CED with anthracyclines in orthotopic pontine and thalamic tumor models. METHODS The sensitivity of primary pHGG cell lines to a range of anthracyclines was tested in vitro. Preclinical CED of free doxorubicin and pegylated liposomal doxorubicin (PLD) to the brainstem and thalamus of naïve nude mice was performed. The maximum tolerated dose (MTD) was determined based on the observation of clinical symptoms, and brains were analyzed after H & E staining. Efficacy of the MTD was tested in adult glioma E98-FM-DIPG and E98-FM-thalamus models and in the HSJD-DIPG-007-Fluc primary DIPG model. RESULTS Both pHGG and DIPG cells were sensitive to anthracyclines in vitro. Doxorubicin was selected for further preclinical evaluation. Convection-enhanced delivery of the MTD of free doxorubicin and PLD in the pons was 0.02 mg/ml, and the dose tolerated in the thalamus was 10 times higher (0.2 mg/ml). Free doxorubicin or PLD via CED was ineffective against E98-FM-DIPG or HSJD-DIPG-007-Fluc in the brainstem; however, when applied in the thalamus, 0.2 mg/ml of PLD slowed down tumor growth and increased survival in a subset of animals with small tumors. CONCLUSIONS Local delivery of doxorubicin to the brainstem causes severe toxicity, even at doxorubicin concentrations that are safe in the thalamus. As a consequence, the authors could not establish a therapeutic window for treating orthotopic brainstem tumors in mice. For tumors in the thalamus, therapeutic concentrations to slow down tumor growth could be reached. These data suggest that anatomical location determines the severity of toxicity after local delivery of therapeutic agents and that caution should be used when translating data from supratentorial CED studies to treat infratentorial tumors.
Collapse
Affiliation(s)
- A Charlotte P Sewing
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Tonny Lagerweij
- Neurosurgery, and.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Dannis G van Vuurden
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Michaël H Meel
- Departments of 1 Pediatric Oncology.,Neurosurgery, and.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Susanna J E Veringa
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Angel M Carcaboso
- Preclinical Therapeutics and Drug Delivery Research Program, Department of Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | | | - W Peter Vandertop
- Neurosurgery, and.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Pieter Wesseling
- Pathology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam.,2-BBB Medicines, Leiden.,Department of Pathology, RadboudUMC, Nijmegen
| | - David Noske
- Neurosurgery, and.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Gertjan J L Kaspers
- Neuro-Oncology Research Group.,Academy of Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands ; and
| | - Esther Hulleman
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| |
Collapse
|
607
|
Huang WC, Lu IL, Chiang WH, Lin YW, Tsai YC, Chen HH, Chang CW, Chiang CS, Chiu HC. Tumortropic adipose-derived stem cells carrying smart nanotherapeutics for targeted delivery and dual-modality therapy of orthotopic glioblastoma. J Control Release 2017; 254:119-130. [DOI: 10.1016/j.jconrel.2017.03.035] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/30/2016] [Accepted: 03/19/2017] [Indexed: 01/07/2023]
|
608
|
Characterization and carboplatin loaded chitosan nanoparticles for the chemotherapy against breast cancer in vitro studies. Int J Biol Macromol 2017; 97:115-122. [DOI: 10.1016/j.ijbiomac.2016.12.090] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/02/2016] [Accepted: 12/30/2016] [Indexed: 11/17/2022]
|
609
|
Peng C, Gao X, Xu J, Du B, Ning X, Tang S, Bachoo RM, Yu M, Ge WP, Zheng J. Targeting orthotopic gliomas with renal-clearable luminescent gold nanoparticles. NANO RESEARCH 2017; 10:1366-1376. [PMID: 29034063 PMCID: PMC5639726 DOI: 10.1007/s12274-017-1472-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
A major clinical translational challenge in nanomedicine is the potential of toxicity associated with the uptake and long-term retention of non-degradable nanoparticles (NPs) in major organs. The development of inorganic NPs that undergo renal clearance could potentially resolve this significant biosafety concern. However, it remains unclear whether inorganic NPs that can be excreted by the kidneys remain capable of targeting tumors with poor permeability. Glioblastoma multiforme, the most malignant orthotopic brain tumor, presents a unique challenge for NP delivery because of the blood-brain barrier and robust blood-tumor barrier of reactive microglia and macroglia in the tumor microenvironment. Herein, we used an orthotopic murine glioma model to investigate the passive targeting of glutathione-coated gold nanoparticles (AuNPs) of 3 nm in diameter that undergo renal clearance and 18-nm AuNPs that fail to undergo renal clearance. Remarkably, we report that 3-nm AuNPs were able to target intracranial tumor tissues with higher efficiency (2.3× relative to surrounding non-tumor normal brain tissues) and greater specificity (3.0×) than did the larger AuNPs. Pharmacokinetics studies suggested that the higher glioma targeting ability of the 3-nm AuNPs may be attributed to the longer retention time in circulation. The total accumulation of the 3-nm AuNPs in major organs was significantly less (8.4×) than that of the 18-nm AuNPs. Microscopic imaging of blood vessels and renal-clearable AuNPs showed extravasation of NPs from the leaky blood-tumor barrier into the tumor interstitium. Taken together, our results suggest that the 3-nm AuNPs, characterized by enhanced permeability and retention, are able to target brain tumors and undergo renal clearance.
Collapse
Affiliation(s)
- Chuanqi Peng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Xiaofei Gao
- Children's Research Institute, Department of Pediatrics, Department of Neuroscience, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jing Xu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Bujie Du
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Xuhui Ning
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shaoheng Tang
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Robert M Bachoo
- Simmons Cancer Center, Annette G. Strauss Center for Neuro-Oncology, Department of Internal Medicine, Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mengxiao Yu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Woo-Ping Ge
- Children's Research Institute, Department of Pediatrics, Department of Neuroscience, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jie Zheng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
610
|
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]
|
611
|
Hutter S, Bolin S, Weishaupt H, Swartling FJ. Modeling and Targeting MYC Genes in Childhood Brain Tumors. Genes (Basel) 2017; 8:genes8040107. [PMID: 28333115 PMCID: PMC5406854 DOI: 10.3390/genes8040107] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 11/16/2022] Open
Abstract
Brain tumors are the second most common group of childhood cancers, accounting for about 20%–25% of all pediatric tumors. Deregulated expression of the MYC family of transcription factors, particularly c-MYC and MYCN genes, has been found in many of these neoplasms, and their expression levels are often correlated with poor prognosis. Elevated c-MYC/MYCN initiates and drives tumorigenesis in many in vivo model systems of pediatric brain tumors. Therefore, inhibition of their oncogenic function is an attractive therapeutic target. In this review, we explore the roles of MYC oncoproteins and their molecular targets during the formation, maintenance, and recurrence of childhood brain tumors. We also briefly summarize recent progress in the development of therapeutic approaches for pharmacological inhibition of MYC activity in these tumors.
Collapse
Affiliation(s)
- Sonja Hutter
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| | - Sara Bolin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| | - Holger Weishaupt
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
612
|
Gao Z, Chen Y, Cai X, Xu R. Predict drug permeability to blood-brain-barrier from clinical phenotypes: drug side effects and drug indications. Bioinformatics 2017; 33:901-908. [PMID: 27993785 PMCID: PMC5860495 DOI: 10.1093/bioinformatics/btw713] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/16/2016] [Accepted: 11/19/2016] [Indexed: 12/25/2022] Open
Abstract
Motivation Blood-Brain-Barrier (BBB) is a rigorous permeability barrier for maintaining homeostasis of Central Nervous System (CNS). Determination of compound's permeability to BBB is prerequisite in CNS drug discovery. Existing computational methods usually predict drug BBB permeability from chemical structure and they generally apply to small compounds passing BBB through passive diffusion. As abundant information on drug side effects and indications has been recorded over time through extensive clinical usage, we aim to explore BBB permeability prediction from a new angle and introduce a novel approach to predict BBB permeability from drug clinical phenotypes (drug side effects and drug indications). This method can apply to both small compounds and macro-molecules penetrating BBB through various mechanisms besides passive diffusion. Results We composed a training dataset of 213 drugs with known brain and blood steady-state concentrations ratio and extracted their side effects and indications as features. Next, we trained SVM models with polynomial kernel and obtained accuracy of 76.0%, AUC 0.739, and F 1 score (macro weighted) 0.760 with Monte Carlo cross validation. The independent test accuracy was 68.3%, AUC 0.692, F 1 score 0.676. When both chemical features and clinical phenotypes were available, combining the two types of features achieved significantly better performance than chemical feature based approach (accuracy 85.5% versus 72.9%, AUC 0.854 versus 0.733, F 1 score 0.854 versus 0.725; P < e -90 ). We also conducted de novo prediction and identified 110 drugs in SIDER database having the potential to penetrate BBB, which could serve as start point for CNS drug repositioning research. Availability and Implementation https://github.com/bioinformatics-gao/CASE-BBB-prediction-Data. Contact rxx@case.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zhen Gao
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Yang Chen
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Xiaoshu Cai
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, OH, USA
| | - Rong Xu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
613
|
Chen C, Duan Z, Yuan Y, Li R, Pang L, Liang J, Xu X, Wang J. Peptide-22 and Cyclic RGD Functionalized Liposomes for Glioma Targeting Drug Delivery Overcoming BBB and BBTB. ACS APPLIED MATERIALS & INTERFACES 2017; 9:5864-5873. [PMID: 28128553 DOI: 10.1021/acsami.6b15831] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Chemotherapy outcomes for the treatment of glioma remain unsatisfied due to the inefficient drug transport across BBB/BBTB and poor drug accumulation in the tumor site. Nanocarriers functionalized with different targeting ligands are considered as one of the most promising alternatives. However, few studies were reported to compare the targeting efficiency of the ligands and develop nanoparticles to realize BBB/BBTB crossing and brain tumor targeting simultaneously. In this study, six peptide-based ligands (Angiopep-2, T7, Peptide-22, c(RGDfK), D-SP5 and Pep-1), widely used for brain delivery, were selected to decorate liposomes, respectively, so as to compare their targeting ability to BBB or BBTB. Based on the in vitro cellular uptake results on BCECs and HUVECs, Peptide-22 and c(RGDfK) were picked to construct a BBB/BBTB dual-crossing, glioma-targeting liposomal drug delivery system c(RGDfK)/Pep-22-DOX-LP. In vitro cellular uptake demonstrated that the synergetic effect of c(RGDfK) and Peptide-22 could significantly increase the internalization of liposomes on U87 cells. In vivo imaging further verified that c(RGDfK)/Pep-22-LP exhibited higher brain tumor distribution than single ligand modified liposomes. The median survival time of glioma-bearing mice treated with c(RGDfK)/Pep-22-DOX-LP (39.5 days) was significantly prolonged than those treated with free doxorubicin or other controls. In conclusion, the c(RGDfK) and Peptide-22 dual-modified liposome was constructed based on the targeting ability screening of various ligands. The system could effectively overcome BBB/BBTB barriers, target to tumor cells and inhibit the growth of glioma, which proved its potential for improving the efficacy of chemotherapeutics for glioma therapy.
Collapse
Affiliation(s)
- Cuitian Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Ziqing Duan
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Yan Yuan
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Ruixiang Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Liang Pang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Jianming Liang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Xinchun Xu
- Shanghai Xuhui Central Hospital , Shanghai 200031, People's Republic of China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| |
Collapse
|
614
|
Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W. Glioma: experimental models and reality. Acta Neuropathol 2017; 133:263-282. [PMID: 28074274 PMCID: PMC5250671 DOI: 10.1007/s00401-017-1671-4] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
Abstract
In theory, in vitro and in vivo models for human gliomas have great potential to not only enhance our understanding of glioma biology, but also to facilitate the development of novel treatment strategies for these tumors. For reliable prediction and validation of the effects of different therapeutic modalities, however, glioma models need to comply with specific and more strict demands than other models of cancer, and these demands are directly related to the combination of genetic aberrations and the specific brain micro-environment gliomas grow in. This review starts with a brief introduction on the pathological and molecular characteristics of gliomas, followed by an overview of the models that have been used in the last decades in glioma research. Next, we will discuss how these models may play a role in better understanding glioma development and especially in how they can aid in the design and optimization of novel therapies. The strengths and weaknesses of the different models will be discussed in light of genotypic, phenotypic and metabolic characteristics of human gliomas. The last part of this review provides some examples of how therapy experiments using glioma models can lead to deceptive results when such characteristics are not properly taken into account.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Roel Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
615
|
Marianecci C, Rinaldi F, Hanieh PN, Di Marzio L, Paolino D, Carafa M. Drug delivery in overcoming the blood-brain barrier: role of nasal mucosal grafting. Drug Des Devel Ther 2017; 11:325-335. [PMID: 28184152 PMCID: PMC5291459 DOI: 10.2147/dddt.s100075] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The blood-brain barrier (BBB) plays a fundamental role in protecting and maintaining the homeostasis of the brain. For this reason, drug delivery to the brain is much more difficult than that to other compartments of the body. In order to bypass or cross the BBB, many strategies have been developed: invasive techniques, such as temporary disruption of the BBB or direct intraventricular and intracerebral administration of the drug, as well as noninvasive techniques. Preliminary results, reported in the large number of studies on the potential strategies for brain delivery, are encouraging, but it is far too early to draw any conclusion about the actual use of these therapeutic approaches. Among the most recent, but still pioneering, approaches related to the nasal mucosa properties, the permeabilization of the BBB via nasal mucosal engrafting can offer new potential opportunities. It should be emphasized that this surgical procedure is quite invasive, but the implication for patient outcome needs to be compared to the gold standard of direct intracranial injection, and evaluated whilst keeping in mind that central nervous system diseases and lysosomal storage diseases are chronic and severely debilitating and that up to now no therapy seems to be completely successful.
Collapse
Affiliation(s)
- Carlotta Marianecci
- Department of Drug Chemistry and Technology, University of Rome “Sapienza”, Rome, Italy
| | - Federica Rinaldi
- Center for Life Nano Science@ Sapienza, Fondazione Istituto Italiano di Tecnologia, Rome, Italy
| | - Patrizia Nadia Hanieh
- Department of Drug Chemistry and Technology, University of Rome “Sapienza”, Rome, Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University “G. d’Annunzio”, Chieti, Italy
| | - Donatella Paolino
- IRC FSH-Interregional Research Center for Food Safety & Health, Campus Universitario “S. Venuta”, University of Catanzaro “Magna Græcia”, Catanzaro, Italy
- Department of Health Sciences, Campus Universitario “S. Venuta”, University of Catanzaro “Magna Græcia”, Catanzaro, Italy
| | - Maria Carafa
- Department of Drug Chemistry and Technology, University of Rome “Sapienza”, Rome, Italy
| |
Collapse
|
616
|
Jiang Y, Wang X, Liu X, Lv W, Zhang H, Zhang M, Li X, Xin H, Xu Q. Enhanced Antiglioma Efficacy of Ultrahigh Loading Capacity Paclitaxel Prodrug Conjugate Self-Assembled Targeted Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2017; 9:211-217. [PMID: 27976583 DOI: 10.1021/acsami.6b13805] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Glioblastoma multiforme (GBM) presents one of the most lethal brain tumor with a dismal prognosis. And nanodrug delivery system (nano-DDS) have raised a lot of concern, while the conventional nanoformulations addressed many limitations, especially the low drug loading capacity and poor stability in vivo. Herein, we proposed PTX prodrug (PTX-SS-C18) conjugate self-assembled nanoparticles (PSNPs) functionalized with Pep-1, glioma homing peptide, to overcome the blood brain tumor barrier (BBTB) via interleukin 13 receptor α2 (IL-13Rα2)-mediated endocytosis for targeting GMB. This nanocarrier was with ultrahigh drug loading capacity (56.03%) and redox-sensitivity to the up-expression of glutathione in glioma tumors. And compared with PEG-PSNPs, Pep-PSNPs could significantly enhance cellular uptake in U87MG cells via IL-13Rα2-mediated endocytosis. Enhanced cytotoxicity of Pep-PSNPs against U87MG cells and BCEC cells pretreated with glutathione monoester (GSH-OEt) confirmed that this nanosystem was sensitive to reduction environment, and there was significant difference between targeting and nontargeting groups in MTT assay. Real-time fluorescence image of intracranialU87MG glioma-bearing mice revealed that Pep-PSNPs could more efficiently accumulate at tumor site and improve the penetration. Furthermore, the ex vivo fluorescence imaging and corresponding semiquantitative results displayed that the glioma fluorescence intensity of Pep-PSNPs group was 1.74-fold higher than that of nontargeting group. Pep-PSNPs exhibited remarkable antiglioblastoma efficacy with an extended median survival time. In conclusion, Pep-PSNPs had a promising perspective as a targeting drug delivery system of PTX for glioma treatment.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Xiuzhen Wang
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Xin Liu
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Wei Lv
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Hongjuan Zhang
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Mingwan Zhang
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Xinrui Li
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| | - Qunwei Xu
- Department of Pharmaceutics, School of Pharmacy, and §Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, China
| |
Collapse
|
617
|
Sawicki E, Schellens JHM, Beijnen JH, Nuijen B. Pharmaceutical development of an amorphous solid dispersion formulation of elacridar hydrochloride for proof-of-concept clinical studies. Drug Dev Ind Pharm 2017; 43:584-594. [PMID: 28010129 DOI: 10.1080/03639045.2016.1274901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE A novel tablet formulation containing an amorphous solid dispersion (ASD) of elacridar hydrochloride was developed with the purpose to resolve the drug's low solubility in water and to conduct proof-of-concept clinical studies. SIGNIFICANCE Elacridar is highly demanded for proof-of-concept clinical trials that study the drug's suitability to boost brain penetration and bioavailability of numerous anticancer agents. Previously, clinical trials with elacridar were performed with a tablet containing elacridar hydrochloride. However, this tablet formulation resulted in poor and unpredictable absorption which was caused by the low aqueous solubility of elacridar hydrochloride. METHODS Twenty four different ASDs were produced and dissolution was compared to crystalline elacridar hydrochloride and a crystalline physical mixture. The formulation with highest dissolution was characterized for amorphicity. Subsequently, a tablet was developed and monitored for chemical/physical stability for 12 months at +15-25 °C, +2-8 °C and -20 °C. RESULTS The ASD powder was composed of freeze dried elacridar hydrochloride-povidone K30-sodium dodecyl sulfate (1:6:1, w/w/w), appeared fully amorphous and resulted in complete dissolution whereas crystalline elacridar hydrochloride resulted in only 1% dissolution. The ASD tablets contained 25 mg elacridar hydrochloride and were stable for at least 12 months at -20 °C. CONCLUSIONS The ASD tablet was considered feasible for proof-of-concept clinical studies and is now used as such.
Collapse
Affiliation(s)
- E Sawicki
- a Department of Pharmacy and Pharmacology , Antoni van Leeuwenhoek Hospital/MC Slotervaart , Amsterdam , The Netherlands
| | - J H M Schellens
- b Department of Clinical Pharmacology , the Netherlands Cancer Institute , Amsterdam , the Netherlands.,c Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology , Utrecht University , Utrecht , the Netherlands
| | - J H Beijnen
- a Department of Pharmacy and Pharmacology , Antoni van Leeuwenhoek Hospital/MC Slotervaart , Amsterdam , The Netherlands.,b Department of Clinical Pharmacology , the Netherlands Cancer Institute , Amsterdam , the Netherlands.,c Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology , Utrecht University , Utrecht , the Netherlands
| | - B Nuijen
- a Department of Pharmacy and Pharmacology , Antoni van Leeuwenhoek Hospital/MC Slotervaart , Amsterdam , The Netherlands
| |
Collapse
|
618
|
d,l-Sulforaphane Induces ROS-Dependent Apoptosis in Human Gliomablastoma Cells by Inactivating STAT3 Signaling Pathway. Int J Mol Sci 2017; 18:ijms18010072. [PMID: 28054986 PMCID: PMC5297707 DOI: 10.3390/ijms18010072] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/11/2016] [Accepted: 12/26/2016] [Indexed: 12/25/2022] Open
Abstract
d,l-Sulforaphane (SFN), a synthetic analogue of broccoli-derived isomer l-SFN, exerts cytotoxic effects on multiple tumor cell types through different mechanisms and is more potent than the l-isomer at inhibiting cancer growth. However, the means by which SFN impairs glioblastoma (GBM) cells remains poorly understood. In this study, we investigated the anti-cancer effect of SFN in GBM cells and determined the underlying molecular mechanisms. Cell viability assays, flow cytometry, immunofluorescence, and Western blot results revealed that SFN could induced apoptosis of GBM cells in a dose- and time-dependent manner, via up-regulation of caspase-3 and Bax, and down-regulation of Bcl-2. Mechanistically, SFN treatment led to increase the intracellular reactive oxygen species (ROS) level in GBM cells. Meanwhile, SFN also suppressed both constitutive and IL-6-induced phosphorylation of STAT3, and the activation of upstream JAK2 and Src tyrosine kinases, dose- and time-dependently. Moreover, blockage of ROS production by using the ROS inhibitor N-acetyl-l-cysteine totally reversed SFN-mediated down-regulation of JAK2/Src-STAT3 signaling activation and the subsequent effects on apoptosis by blocking the induction of apoptosis-related genes in GBM cells. Taken together, our data suggests that SFN induces apoptosis in GBM cells via ROS-dependent inactivation of STAT3 phosphorylation. These findings motivate further evaluation of SFN as a cancer chemopreventive agent in GBM treatment.
Collapse
|
619
|
Peyrl A, Frischer J, Hainfellner JA, Preusser M, Dieckmann K, Marosi C. Brain tumors - other treatment modalities. HANDBOOK OF CLINICAL NEUROLOGY 2017; 145:547-560. [PMID: 28987193 DOI: 10.1016/b978-0-12-802395-2.00034-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Management of tumors of the central nervous system is challenging for clinicians for various reasons, including complex diagnostic procedures, limited penetration of drugs into brain tissue, and the prerequisite to preserve brain function in any case of therapeutic intervention. Therapeutic success is dependent on the efforts, skills, and cooperation of involved specialists and disciplines. Knowledge and ability to apply adequate therapeutic modalities in an interdisciplinary approach in due time are crucial, necessitating coordination of diagnostic procedures and therapeutic interventions by means of multidisciplinary brain tumor boards. In this chapter we present in brief the essential current standards and future perspectives for therapy modalities that complement surgery of brain tumors.
Collapse
Affiliation(s)
- Andreas Peyrl
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Josa Frischer
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center - Central Nervous System Tumors Unit (CCC-CNS), Medical University of Vienna, Vienna, Austria
| | - Johannes A Hainfellner
- Comprehensive Cancer Center - Central Nervous System Tumors Unit (CCC-CNS), Medical University of Vienna, Vienna, Austria; Institute of Neurology, Medical University of Vienna, Vienna, Austria.
| | - Matthias Preusser
- Comprehensive Cancer Center - Central Nervous System Tumors Unit (CCC-CNS), Medical University of Vienna, Vienna, Austria; Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Karin Dieckmann
- Comprehensive Cancer Center - Central Nervous System Tumors Unit (CCC-CNS), Medical University of Vienna, Vienna, Austria; Department of Radiotherapy, Medical University of Vienna, Vienna, Austria
| | - Christine Marosi
- Comprehensive Cancer Center - Central Nervous System Tumors Unit (CCC-CNS), Medical University of Vienna, Vienna, Austria; Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
620
|
Hron RJ, Jursic BS, Neumann DM. Synthesis of N-aryl and N-arylcarbamoylamino derivatives of 1,3-diazinane-5-carboxamide and their activity against glioblastoma LN-229 cell line. Bioorg Med Chem 2016; 24:6183-6193. [DOI: 10.1016/j.bmc.2016.09.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/28/2016] [Accepted: 09/30/2016] [Indexed: 02/02/2023]
|
621
|
Pinto MP, Arce M, Yameen B, Vilos C. Targeted brain delivery nanoparticles for malignant gliomas. Nanomedicine (Lond) 2016; 12:59-72. [PMID: 27876436 DOI: 10.2217/nnm-2016-0307] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Brain tumors display the highest mortality rates of all childhood cancers, and over the last decade its prevalence has steadily increased in elderly. To date, effective treatments for brain tumors and particularly for malignant gliomas remain a challenge mainly due to the low permeability and high selectivity of the blood-brain barrier (BBB) to conventional anticancer drugs. In recent years, the elucidation of the cellular mechanisms involved in the transport of substances into the brain has boosted the development of therapeutic-targeted nanoparticles (NPs) with the ability to cross the BBB. Here, we present a comprehensive overview of the available therapeutic strategies developed against malignant gliomas based on 'actively targeted' NPs, the challenges of crossing the BBB and blood-brain tumor barrier as well as its mechanisms and a critical assessment of clinical studies that have used targeted NPs for the treatment of malignant gliomas. Finally, we discuss the potential of actively targeted NP-based strategies in clinical settings, its possible side effects and future directions for therapeutic applications. First draft submitted: 4 October 2016; Accepted for publication: 14 October 2016; Published online: 23 November 2016.
Collapse
Affiliation(s)
- Mauricio P Pinto
- Laboratory of Immunology of Reproduction, Faculty of Chemistry & Biology, Universidad de Santiago de Chile, 9170022 Santiago, Chile
| | - Maximiliano Arce
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Basit Yameen
- Laboratory of Nanomedicine & Biomaterials, Department of Anesthesiology, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02115, USA.,Department of Chemistry, SBA School of Science & Engineering, Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Cristian Vilos
- Laboratory of Nanomedicine & Targeted Delivery, Center for Integrative Medicine & Innovative Science, Faculty of Medicine, Universidad Andres Bello, Santiago, 8370071 Santiago, Chile.,Center for Bioinformatics & Integrative Biology, Faculty of Biological Sciences, Universidad Andres Bello, Santiago, 8370071 Santiago, Chile.,Center for the Development of Nanoscience & Nanotechnology, CEDENNA, 9170124 Santiago, Chile
| |
Collapse
|
622
|
Jiang Y, Lv L, Shi H, Hua Y, Lv W, Wang X, Xin H, Xu Q. PEGylated Polyamidoamine dendrimer conjugated with tumor homing peptide as a potential targeted delivery system for glioma. Colloids Surf B Biointerfaces 2016; 147:242-249. [DOI: 10.1016/j.colsurfb.2016.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/30/2016] [Accepted: 08/02/2016] [Indexed: 10/21/2022]
|
623
|
Gao S, Tian H, Xing Z, Zhang D, Guo Y, Guo Z, Zhu X, Chen X. A non-viral suicide gene delivery system traversing the blood brain barrier for non-invasive glioma targeting treatment. J Control Release 2016; 243:357-369. [PMID: 27794494 DOI: 10.1016/j.jconrel.2016.10.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/06/2016] [Accepted: 10/25/2016] [Indexed: 12/27/2022]
Abstract
Herpes simplex virus type I thymidine kinase gene (HSV-TK) in viral vector is a promising strategy against glioblastoma multiforme (GBM). However, the biosafety risk restricts its application in clinic. In this work, poly (l-lysine)-grafted polyethylenimine (PEI-PLL), which combines the high transfection efficiency of polyethylenimine and the good biodegradability of poly (l-lysine), was adopted as the non-viral vector backbone. Angiopep-2, a blood brain barrier (BBB) crossing and glioma targeting bifunctional peptide was conjugated on PEI-PLL via polyethyleneglycol (PEG) and designated as PPA. The optimal transfection ratio of PPA/DNA complexes nanoparticles (PPA NPs) was firstly characterized. Next, the glioma targeting of the PPA NPs was confirmed through cellular uptake and transfection analysis. The in vivo imaging studies demonstrated that the PPA NPs could not only penetrate BBB but also accumulate in striatum and cortex via systemic administration. Moreover, the PPA/HSV-TK NPs showed remarkably anti-glioma effect and survival benefit in an invasive orthotopic human GBM mouse model through inhibiting proliferation and inducing apoptosis (p<0.05 vs control). This study firstly illustrated that the cationic polymer PPA could be exploited as an efficient gene vector to cross the BBB, and innovatively provided a potential non-viral nanomedicine for noninvasive suicide gene therapy in the glioma treatment.
Collapse
Affiliation(s)
- Shiqian Gao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Graduate School of Chinese Academy of Sciences, Beijing 100039, PR China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Zhenkai Xing
- School of Life Science, Northeast Normal University, Changchun 130024, PR China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Ye Guo
- School of Life Science, Northeast Normal University, Changchun 130024, PR China
| | - Zhaopei Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Xiaojuan Zhu
- School of Life Science, Northeast Normal University, Changchun 130024, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| |
Collapse
|
624
|
You Y, Yang L, He L, Chen T. Tailored mesoporous silica nanosystem with enhanced permeability of the blood-brain barrier to antagonize glioblastoma. J Mater Chem B 2016; 4:5980-5990. [PMID: 32263487 DOI: 10.1039/c6tb01329e] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cancer-targeted drug delivery systems with permeability of the blood-brain barrier (BBB) have become of great interest for the rational design of high-efficiency anticancer agents. Herein, a tailored mesoporous silica nanoparticles (MSNs) nanosystem modified by RGD (arginine-glycine-aspartate) peptide was designed and tested for use as a carrier of anticancer agents, by using a novel organic selenium compound BSeC as a model molecule. As expected, the nanosystem (BSeC@MSNs-RGD) could effectively enhance the BBB permeability and the cellular uptake of BSeC in tumor cells. The internalized BSeC@MSNs-RGD triggered mitochondrial dysfunction and intracellular ROS overproduction, which subsequently activated the p53 and MAPKs pathways. Moreover, the nanosystem could inhibit the U87 tumor spheroids growth, significantly prolong the blood circulation time of the loaded drug in vivo and effectively reduce its in vivo toxicity. Taken together, this study provides a strategy for the rational design of a tailored nanomedicine with enhanced BBB permeability to treat human brain glioma.
Collapse
Affiliation(s)
- Yuanyuan You
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | | | | | | |
Collapse
|
625
|
Lin F, de Gooijer MC, Hanekamp D, Chandrasekaran G, Buil LCM, Thota N, Sparidans RW, Beijnen JH, Würdinger T, van Tellingen O. PI3K-mTOR Pathway Inhibition Exhibits Efficacy Against High-grade Glioma in Clinically Relevant Mouse Models. Clin Cancer Res 2016; 23:1286-1298. [PMID: 27553832 DOI: 10.1158/1078-0432.ccr-16-1276] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/08/2016] [Accepted: 08/17/2016] [Indexed: 11/16/2022]
Abstract
Purpose: The PI3K-AKT-mTOR signaling pathway is frequently activated in glioblastoma and offers several druggable targets. However, clinical efficacy of PI3K/mTOR inhibitors in glioblastoma has not yet been demonstrated. Insufficient drug delivery may limit the efficacy of PI3K/mTOR inhibitors against glioblastoma. The presence of the efflux transporters ABCB1/Abcb1 (P-glycoprotein, MDR1) and ABCG2/Abcg2 (BCRP) at the blood-brain barrier (BBB) restricts the brain penetration of many drugs.Experimental Design: We used in vitro drug transport assays and performed pharmacokinetic/pharmacodynamic studies in wild-type and ABC-transporter knockout mice. The efficacy of PI3K-mTOR inhibition was established using orthotopic allograft and genetically engineered spontaneous glioblastoma mouse models.Results: The mTOR inhibitors rapamycin and AZD8055 are substrates of ABCB1, whereas the dual PI3K/mTOR inhibitor NVP-BEZ235 and the PI3K inhibitor ZSTK474 are not. Moreover, ABCG2 transports NVP-BEZ235 and AZD8055, but not ZSTK474 or rapamycin. Concordantly, Abcb1a/b-/-;Abcg2-/- mice revealed increased brain penetration of rapamycin (13-fold), AZD8055 (7.7-fold), and NVP-BEZ235 (4.5-fold), but not ZSTK474 relative to WT mice. Importantly, ABC transporters limited rapamycin brain penetration to subtherapeutic levels, while the reduction in NVP-BEZ235 brain penetration did not prevent target inhibition. NVP-BEZ235 and ZSTK474 demonstrated antitumor efficacy with improved survival against U87 orthotopic gliomas, although the effect of ZSTK474 was more pronounced. Finally, ZSTK474 prolonged overall survival in Cre-LoxP conditional transgenic Pten;p16Ink4a/p19Arf;K-Rasv12;LucR mice, mainly by delaying tumor onset.Conclusions: PI3K/mTOR inhibitors with weak affinities for ABC transporters can achieve target inhibition in brain (tumors), but have modest single-agent efficacy and combinations with (BBB penetrable) inhibitors of other activated pathways may be required. Clin Cancer Res; 23(5); 1286-98. ©2016 AACR.
Collapse
Affiliation(s)
- Fan Lin
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Amsterdam, the Netherlands
| | - Mark C de Gooijer
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Amsterdam, the Netherlands
| | - Diana Hanekamp
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Amsterdam, the Netherlands
| | - Gayathri Chandrasekaran
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Amsterdam, the Netherlands
| | - Levi C M Buil
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Amsterdam, the Netherlands
| | - Nishita Thota
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Amsterdam, the Netherlands
| | - Rolf W Sparidans
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam, the Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam, the Netherlands.,Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht University, Utrecht, the Netherlands
| | - Tom Würdinger
- Neuro-oncology Research Group, Departments of Neurosurgery and Pediatric Oncology/Hematology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.,Molecular Neurogenetics Unit, Departments of Neurology and Radiology, Massachusetts General Hospital, and Neuroscience Program, Harvard Medical School, Boston, Massachusetts
| | - Olaf van Tellingen
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Amsterdam, the Netherlands.
| |
Collapse
|
626
|
Roberts NB, Wadajkar AS, Winkles JA, Davila E, Kim AJ, Woodworth GF. Repurposing platinum-based chemotherapies for multi-modal treatment of glioblastoma. Oncoimmunology 2016; 5:e1208876. [PMID: 27757301 DOI: 10.1080/2162402x.2016.1208876] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 10/21/2022] Open
Abstract
Glioblastoma (GBM) is a fatal brain cancer for which new treatment options are sorely needed. Platinum-based drugs have been investigated extensively for GBM treatment but few have shown significant efficacy without major central nervous system (CNS) and systemic toxicities. The relative success of platinum drugs for treatment of non-CNS cancers indicates great therapeutic potential when effectively delivered to the tumor region(s). New insights into the broad anticancer effects of platinum drugs, particularly immunomodulatory effects, and innovative delivery strategies that can maximize these multi-modal effects and minimize toxicities may promote the re-purposing of this chemotherapeutic drug class for GBM treatment.
Collapse
Affiliation(s)
- Nathan B Roberts
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aniket S Wadajkar
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeffrey A Winkles
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eduardo Davila
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA; Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
627
|
Sales TT, Resende FFB, Chaves NL, Titze-De-Almeida SS, Báo SN, Brettas ML, Titze-De-Almeida R. Suppression of the Eag1 potassium channel sensitizes glioblastoma cells to injury caused by temozolomide. Oncol Lett 2016; 12:2581-2589. [PMID: 27698831 PMCID: PMC5038559 DOI: 10.3892/ol.2016.4992] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 06/16/2016] [Indexed: 01/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of human primary brain tumor. The standard treatment protocol includes radiotherapy in combination with temozolomide (TMZ). Despite advances in GBM treatment, the survival time of patients diagnosed with glioma is 14.5 months. Regarding tumor biology, various types of cancer cell overexpress the ether à go-go 1 (Eag1) potassium channel. Therefore, the present study examined the role of Eag1 in the cell damage caused by TMZ on the U87MG glioblastoma cell line. Eag1 was inhibited using a channel blocker (astemizole) or silenced by a short-hairpin RNA expression vector (pKv10.1-3). pKv10.1-3 (0.2 µg) improved the Eag1 silencing caused by 250 µM TMZ, as determined by reverse transcription-quantitative polymerase chain reaction and immunocytochemistry. Additionally, inhibiting Eag1 with the vector or astemizole (5 µM) reduced glioblastoma cell viability and sensitized cells to TMZ. Cell viability decreased by 63% for pKv10.1-3 + TMZ compared with 34% for TMZ alone, and by 77% for astemizole + TMZ compared with 46% for TMZ alone, as determined by MTT assay. In addition, both the vector and astemizole increased the apoptosis rate of glioblastoma cells triggered by TMZ, as determined by an Annexin V apoptosis assay. Collectively, the current data reveal that Eag1 has a role in the damage caused to glioblastoma by TMZ. Furthermore, suppression of this channel can improve the action of TMZ on U87MG glioblastoma cells. Thus, silencing Eag1 is a promising strategy to improve GBM treatment and merits additional studies in animal models of glioma.
Collapse
Affiliation(s)
- Thais Torquato Sales
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, Faculty of Agronomy and Veterinary Medicine, University of Brasília, Brasília DF 70910-900, Brazil
| | - Fernando Francisco Borges Resende
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, Faculty of Agronomy and Veterinary Medicine, University of Brasília, Brasília DF 70910-900, Brazil
| | - Natália Lemos Chaves
- Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília DF 70910-900, Brazil
| | - Simoneide Souza Titze-De-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, Faculty of Agronomy and Veterinary Medicine, University of Brasília, Brasília DF 70910-900, Brazil
| | - Sônia Nair Báo
- Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília DF 70910-900, Brazil
| | - Marcella Lemos Brettas
- Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília DF 70910-900, Brazil; Planaltina Campus, University of Brasília, Brasília DF 70910-900, Brazil
| | - Ricardo Titze-De-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, Faculty of Agronomy and Veterinary Medicine, University of Brasília, Brasília DF 70910-900, Brazil
| |
Collapse
|
628
|
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
|
629
|
Arvold ND, Lee EQ, Mehta MP, Margolin K, Alexander BM, Lin NU, Anders CK, Soffietti R, Camidge DR, Vogelbaum MA, Dunn IF, Wen PY. Updates in the management of brain metastases. Neuro Oncol 2016; 18:1043-65. [PMID: 27382120 PMCID: PMC4933491 DOI: 10.1093/neuonc/now127] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/09/2016] [Indexed: 12/16/2022] Open
Abstract
The clinical management/understanding of brain metastases (BM) has changed substantially in the last 5 years, with key advances and clinical trials highlighted in this review. Several of these changes stem from improvements in systemic therapy, which have led to better systemic control and longer overall patient survival, associated with increased time at risk for developing BM. Development of systemic therapies capable of preventing BM and controlling both intracranial and extracranial disease once BM are diagnosed is paramount. The increase in use of stereotactic radiosurgery alone for many patients with multiple BM is an outgrowth of the desire to employ treatments focused on local control while minimizing cognitive effects associated with whole brain radiotherapy. Complications from BM and their treatment must be considered in comprehensive patient management, especially with greater awareness that the majority of patients do not die from their BM. Being aware of significant heterogeneity in prognosis and therapeutic options for patients with BM is crucial for appropriate management, with greater attention to developing individual patient treatment plans based on predicted outcomes; in this context, recent prognostic models of survival have been extensively revised to incorporate molecular markers unique to different primary cancers.
Collapse
Affiliation(s)
| | | | | | - Kim Margolin
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - Brian M. Alexander
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - Nancy U. Lin
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - Carey K. Anders
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - Riccardo Soffietti
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - D. Ross Camidge
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - Michael A. Vogelbaum
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - Ian F. Dunn
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| | - Patrick Y. Wen
- St. Luke's Radiation Oncology Associates, St. Luke's Cancer Center, Whiteside Institute for Clinical Research and University of Minnesota Duluth, Duluth, Minnesota (N.D.A.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (E.Q.L., P.Y.W.); Harvard Medical School, Boston, Massachusetts (E.Q.L., B.M.A., N.U.L., I.F.D., P.Y.W.); Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland (M.P.M.); Department of Medical Oncology, City of Hope, Duarte, California (K.M.); Department of Radiation Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (B.M.A.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (N.U.L.); Department of Medicine, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, North Carolina (C.K.A.); Department of Neurology/Neuro-Oncology, University of Turin, Turin, Italy (R.S.); Division of Medical Oncology, University of Colorado Denver, Denver, Colorado (D.R.C.); Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio (M.A.V.); Department of Neurosurgery, Brigham & Women's Hospital, Boston, Massachusetts (I.F.D.)
| |
Collapse
|
630
|
Bauer M, Römermann K, Karch R, Wulkersdorfer B, Stanek J, Philippe C, Maier‐Salamon A, Haslacher H, Jungbauer C, Wadsak W, Jäger W, Löscher W, Hacker M, Zeitlinger M, Langer O. Pilot PET Study to Assess the Functional Interplay Between ABCB1 and ABCG2 at the Human Blood-Brain Barrier. Clin Pharmacol Ther 2016; 100:131-41. [PMID: 26940368 PMCID: PMC4979595 DOI: 10.1002/cpt.362] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/20/2016] [Accepted: 02/28/2016] [Indexed: 01/16/2023]
Abstract
ABCB1 and ABCG2 work together at the blood-brain barrier (BBB) to limit brain distribution of dual ABCB1/ABCG2 substrates. In this pilot study we used positron emission tomography (PET) to assess brain distribution of two model ABCB1/ABCG2 substrates ([(11) C]elacridar and [(11) C]tariquidar) in healthy subjects without (c.421CC) or with (c.421CA) the ABCG2 single-nucleotide polymorphism (SNP) c.421C>A. Subjects underwent PET scans under conditions when ABCB1 and ABCG2 were functional and during ABCB1 inhibition with high-dose tariquidar. In contrast to the ABCB1-selective substrate (R)-[(11) C]verapamil, [(11) C]elacridar and [(11) C]tariquidar showed only moderate increases in brain distribution during ABCB1 inhibition. This provides evidence for a functional interplay between ABCB1 and ABCG2 at the human BBB and suggests that both ABCB1 and ABCG2 need to be inhibited to achieve substantial increases in brain distribution of dual ABCB1/ABCG2 substrates. During ABCB1 inhibition c.421CA subjects had significantly higher increases in [(11) C]tariquidar brain distribution than c.421CC subjects, pointing to impaired cerebral ABCG2 function.
Collapse
Affiliation(s)
- M Bauer
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| | - K Römermann
- Department of Pharmacology, Toxicology & PharmacyUniversity of Veterinary MedicineHannoverGermany
| | - R Karch
- Center for Medical Statistics, Informatics and Intelligent SystemsMedical University of ViennaViennaAustria
| | - B Wulkersdorfer
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| | - J Stanek
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
- Health and Environment DepartmentAIT Austrian Institute of Technology GmbHSeibersdorfAustria
| | - C Philippe
- Department of Biomedical Imaging und Image‐guided Therapy, Division of Nuclear MedicineMedical University of ViennaViennaAustria
| | - A Maier‐Salamon
- Department of Clinical Pharmacy and DiagnosticsUniversity of ViennaViennaAustria
| | - H Haslacher
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - C Jungbauer
- Austrian Red Cross Blood Transfusion ServicesViennaAustria
| | - W Wadsak
- Department of Biomedical Imaging und Image‐guided Therapy, Division of Nuclear MedicineMedical University of ViennaViennaAustria
- Medical Imaging ClusterMedical University of ViennaViennaAustria
| | - W Jäger
- Department of Clinical Pharmacy and DiagnosticsUniversity of ViennaViennaAustria
| | - W Löscher
- Department of Pharmacology, Toxicology & PharmacyUniversity of Veterinary MedicineHannoverGermany
| | - M Hacker
- Department of Biomedical Imaging und Image‐guided Therapy, Division of Nuclear MedicineMedical University of ViennaViennaAustria
- Medical Imaging ClusterMedical University of ViennaViennaAustria
| | - M Zeitlinger
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| | - O Langer
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
- Health and Environment DepartmentAIT Austrian Institute of Technology GmbHSeibersdorfAustria
- Department of Biomedical Imaging und Image‐guided Therapy, Division of Nuclear MedicineMedical University of ViennaViennaAustria
- Medical Imaging ClusterMedical University of ViennaViennaAustria
| |
Collapse
|
631
|
Mallick S, Benson R, Hakim A, Rath GK. Management of glioblastoma after recurrence: A changing paradigm. J Egypt Natl Canc Inst 2016; 28:199-210. [PMID: 27476474 DOI: 10.1016/j.jnci.2016.07.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/30/2016] [Accepted: 07/03/2016] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma remains the most common primary brain tumor after the age of 40years. Maximal safe surgery followed by adjuvant chemoradiotherapy has remained the standard treatment for glioblastoma (GBM). But recurrence is an inevitable event in the natural history of GBM with most patients experiencing it after 6-9months of primary treatment. Recurrent GBM poses great challenge to manage with no well-defined management protocols. The challenge starts from differentiating radiation necrosis from true local progression. A fine balance needs to be maintained on improving survival and assuring a better quality of life. Treatment options are limited and ranges from re-excision, re-irradiation, systemic chemotherapy or a combination of these. Re-excision and re-irradiation must be attempted in selected patients and has been shown to improve survival outcomes. To facilitate the management of GBM recurrences, a treatment algorithm is proposed.
Collapse
Affiliation(s)
- Supriya Mallick
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India.
| | - Rony Benson
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Abdul Hakim
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Goura K Rath
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
632
|
Advances in Targeted Drug Delivery Approaches for the Central Nervous System Tumors: The Inspiration of Nanobiotechnology. J Neuroimmune Pharmacol 2016; 12:84-98. [DOI: 10.1007/s11481-016-9698-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/06/2016] [Indexed: 12/21/2022]
|
633
|
Carradori D, Gaudin A, Brambilla D, Andrieux K. Application of Nanomedicine to the CNS Diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 130:73-113. [PMID: 27678175 DOI: 10.1016/bs.irn.2016.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Drug delivery to the brain is a challenge because of the many mechanisms that protect the brain from the entry of foreign substances. Numerous molecules which could be active against brain disorders are not clinically useful due to the presence of the blood-brain barrier. Nanoparticles can be used to deliver these drugs to the brain. Encapsulation within colloidal systems can allow the passage of nontransportable drugs across this barrier by masking their physicochemical properties. It should be noted that the status of the blood-brain barrier is different depending on the brain disease. In fact, in some pathological situations such as tumors or inflammatory disorders, its permeability is increased allowing very easy translocation of carriers. This chapter gathers the promising results obtained by using nanoparticles as drug delivery systems with the aim to improve the therapy of some CNS diseases such as brain tumor, Alzheimer's disease, and stroke. The data show that several approaches can be investigated: (1) carrying drug through a permeabilized barrier, (2) crossing the barrier thanks to receptor-mediated transcytosis pathway in order to deliver drug into the brain parenchyma, and also (3) targeting and treating the endothelial cells themselves to preserve locally the brain tissue. The examples given in this chapter contribute to demonstrate that delivering drugs into the brain is one of the most promising applications of nanotechnology in clinical neuroscience.
Collapse
Affiliation(s)
- D Carradori
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Bruxelles, Belgium
| | - A Gaudin
- Yale University, New Haven, CT, United States
| | - D Brambilla
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - K Andrieux
- Université Paris Descartes, Université Paris-Sorbonne, UTCBS, UMR CNRS 8258, UE1022 INSERM, Paris, France.
| |
Collapse
|
634
|
Kopylov AT, Myasoedov NF, Dadayan AK, Zgoda VG, Medvedev AE, Zolotarev YA. Use of deuterium labeling by high-temperature solid-state hydrogen-exchange reaction for mass spectrometric analysis of bradykinin biotransformation. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2016; 30:1283-1294. [PMID: 27173110 DOI: 10.1002/rcm.7558] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/04/2016] [Accepted: 03/10/2016] [Indexed: 06/05/2023]
Abstract
RATIONALE Studies of molecular biodegradation by mass spectrometry often require synthetic compounds labeled with stable isotopes as internal standards. However, labeling is very expensive especially when a large number of compounds are needed for analysis of biotransformation. Here we describe an approach for qualitative and quantitative analysis using bradykinin (BK) and its in vitro degradation metabolites as an example. Its novelty lies in the use of deuterated peptides which are obtained by a high-temperature solid-state exchange (HSCIE) reaction. METHODS Deuterated and native BK were analyzed by positive electrospray ionization high-resolution mass spectrometry (ESI-HRMS) using an Orbitrap Fusion mass spectrometer. High-energy collision-induced dissociation (HCD) experiments were performed on [M+H](+) and [M+2H](2+) ions in targeted-MS(2) mode with adjusted normalized HCD value. RESULTS After the HSCIE reaction, each amino acid residue of the deuterated peptide contained deuterium atoms and the average degree of substitution was 5.5 atoms per the peptide molecule. The deuterated peptide demonstrated the same chromatographic mobility as the unlabeled counterpart, and lack of racemization during substitution with deuterium. Deuterium-labeled and unlabeled BKs were incubated with human plasma and their corresponding fragments BK(1-5) and BK(1-7), well known as the major metabolites, were detected. CONCLUSIONS Quantitative assays demonstrated applicability of the heavy peptide for both sequencing and quantification of generated fragments. Applicability of the HSCIE deuterated peptide for analysis of routes of its degradation has been shown in in vitro experiments. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Arthur T Kopylov
- Institute of Biomedical Chemistry, bld.8, 10 Pogodinskaya str., 119121, Moscow, Russian Federation
| | - Nikolay F Myasoedov
- Institute of Molecular Genetics, 2 Academic Kurchatov Sq, 123182, Moscow, Russian Federation
| | - Alexander K Dadayan
- Institute of Molecular Genetics, 2 Academic Kurchatov Sq, 123182, Moscow, Russian Federation
| | - Victor G Zgoda
- Institute of Biomedical Chemistry, bld.8, 10 Pogodinskaya str., 119121, Moscow, Russian Federation
| | - Alexei E Medvedev
- Institute of Biomedical Chemistry, bld.8, 10 Pogodinskaya str., 119121, Moscow, Russian Federation
| | - Yurii A Zolotarev
- Institute of Molecular Genetics, 2 Academic Kurchatov Sq, 123182, Moscow, Russian Federation
| |
Collapse
|
635
|
Touat M, Duran-Peña A, Alentorn A, Lacroix L, Massard C, Idbaih A. Emerging circulating biomarkers in glioblastoma: promises and challenges. Expert Rev Mol Diagn 2016; 15:1311-23. [PMID: 26394701 DOI: 10.1586/14737159.2015.1087315] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM) is the most common and devastating primary malignant brain tumor in adults. The past few years have seen major progress in our understanding of the molecular basis of GBM. These advances, which have contributed to the development of novel targeted therapies, will change the paradigms in GBM therapy from disease-based to individually tailored molecular target-based treatment. No validated circulating biomarkers have yet been integrated into clinical practice for GBM. There is thus a critical need to implement minimally invasive clinical tests enabling molecular stratification and prognosis assessment, as well as the prediction and monitoring of treatment response. After examination of data from recent studies exploring several categories of tumor-associated biomarkers (circulating tumor cells, extracellular vesicles, nucleic acids and oncometabolites) identified in the blood, cerebrospinal fluid and urine, this article discusses the challenges and prospects for the development of circulating biomarkers in GBM.
Collapse
Affiliation(s)
- Mehdi Touat
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,b 2 Département d'innovations thérapeutiques précoces, Gustave Roussy, F-94805 Villejuif, France
| | - Alberto Duran-Peña
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France
| | - Agusti Alentorn
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France.,d 4 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Ludovic Lacroix
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,e 5 Département de biologie médicale et de pathologie, Gustave Roussy, F-94805 Villejuif, France.,f 6 Laboratoire de recherche translationnelle et centre de ressources biologiques, Gustave Roussy, F-94805 Villejuif, France
| | - Christophe Massard
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,b 2 Département d'innovations thérapeutiques précoces, Gustave Roussy, F-94805 Villejuif, France
| | - Ahmed Idbaih
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France.,d 4 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| |
Collapse
|
636
|
Gmeiner WH, Debinski W, Milligan C, Caudell D, Pardee TS. The applications of the novel polymeric fluoropyrimidine F10 in cancer treatment: current evidence. Future Oncol 2016; 12:2009-20. [PMID: 27279153 DOI: 10.2217/fon-2016-0091] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
F10 is a novel polymeric fluoropyrimidine drug candidate with strong anticancer activity in multiple preclinical models. F10 has strong potential for impacting cancer treatment because it displays high cytotoxicity toward proliferating malignant cells with minimal systemic toxicities thus providing an improved therapeutic window relative to traditional fluoropyrimidine drugs, such as 5-fluorouracil. F10 has a unique mechanism that involves dual targeting of thymidylate synthase and Top1. In this review, the authors provide an overview of the studies that revealed the novel aspects of F10's cytotoxic mechanism and summarize results obtained in preclinical models of acute myeloid leukemia, acute lymphocytic leukemia, glioblastoma and prostate cancer that demonstrate the strong potential of F10 to improve treatment outcomes.
Collapse
Affiliation(s)
- William H Gmeiner
- Wake Forest Baptist Medical Center Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Waldemar Debinski
- Wake Forest Baptist Medical Center Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Carol Milligan
- Wake Forest Baptist Medical Center Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,Department of Neurobiology & Anatomy, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - David Caudell
- Wake Forest Baptist Medical Center Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Timothy S Pardee
- Wake Forest Baptist Medical Center Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,Department of Hematology/Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
637
|
Carboplatin loaded Surface modified PLGA nanoparticles: Optimization, characterization, and in vivo brain targeting studies. Colloids Surf B Biointerfaces 2016; 142:307-314. [DOI: 10.1016/j.colsurfb.2016.02.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 11/18/2022]
|
638
|
Kim DG, Kim KH, Seo YJ, Yang H, Marcusson EG, Son E, Lee K, Sa JK, Lee HW, Nam DH. Anti-miR delivery strategies to bypass the blood-brain barrier in glioblastoma therapy. Oncotarget 2016; 7:29400-11. [PMID: 27102443 PMCID: PMC5045404 DOI: 10.18632/oncotarget.8837] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/28/2016] [Indexed: 12/19/2022] Open
Abstract
Small non-coding RNAs called miRNAs are key regulators in various biological processes, including tumor initiation, propagation, and metastasis in glioblastoma as well as other cancers. Recent studies have shown the potential for oncogenic miRNAs as therapeutic targets in glioblastoma. However, the application of antisense oligomers, or anti-miRs, to the brain is limited due to the blood-brain barrier (BBB), when administered in the traditional systemic manner. To induce a therapeutic effect in glioblastoma, anti-miR therapy requires a robust and effective delivery system to overcome this obstacle. To bypass the BBB, different delivery administration methods for anti-miRs were evaluated. Stereotaxic surgery was performed to administer anti-Let-7 through intratumoral (ITu), intrathecal (ITh), and intraventricular (ICV) routes, and each method's efficacy was determined by changes in the expression of anti-Let-7 target genes as well as by immunohistochemical analysis. ITu administration of anti-miRs led to a high rate of anti-miR delivery to tumors in the brain by both bolus and continuous administration. In addition, ICV administration, compared with ITu administration, showed a greater distribution of the miR across entire brain tissues. This study suggests that local administration methods are a promising strategy for anti-miR treatment and may overcome current limitations in the treatment of glioblastoma in preclinical animal models.
Collapse
Affiliation(s)
- Dong Geon Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Kang Ho Kim
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Yun Jee Seo
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Heekyoung Yang
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | - Eunju Son
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoungmin Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Jason K. Sa
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Hye Won Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Department of Urology, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, Korea
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
639
|
Calgani A, Vignaroli G, Zamperini C, Coniglio F, Festuccia C, Di Cesare E, Gravina GL, Mattei C, Vitale F, Schenone S, Botta M, Angelucci A. Suppression of SRC Signaling Is Effective in Reducing Synergy between Glioblastoma and Stromal Cells. Mol Cancer Ther 2016; 15:1535-44. [DOI: 10.1158/1535-7163.mct-15-1011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/15/2016] [Indexed: 11/16/2022]
|
640
|
Bioluminescence and MR Imaging of the Safety and Efficacy of Vascular Disruption in Gliomas. Mol Imaging Biol 2016; 18:860-869. [DOI: 10.1007/s11307-016-0963-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
641
|
Kovalevich J, Cornec AS, Yao Y, James M, Crowe A, Lee VMY, Trojanowski JQ, Smith AB, Ballatore C, Brunden KR. Characterization of Brain-Penetrant Pyrimidine-Containing Molecules with Differential Microtubule-Stabilizing Activities Developed as Potential Therapeutic Agents for Alzheimer's Disease and Related Tauopathies. J Pharmacol Exp Ther 2016; 357:432-50. [PMID: 26980057 PMCID: PMC4851320 DOI: 10.1124/jpet.115.231175] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/14/2016] [Indexed: 01/29/2023] Open
Abstract
The microtubule (MT)-stabilizing protein tau disengages from MTs and forms intracellular inclusions known as neurofibrillary tangles in Alzheimer's disease and related tauopathies. Reduced tau binding to MTs in tauopathies may contribute to neuronal dysfunction through decreased MT stabilization and disrupted axonal transport. Thus, the introduction of brain-penetrant MT-stabilizing compounds might normalize MT dynamics and axonal deficits in these disorders. We previously described a number of phenylpyrimidines and triazolopyrimidines (TPDs) that induce tubulin post-translational modifications indicative of MT stabilization. We now further characterize the biologic properties of these small molecules, and our results reveal that these compounds can be divided into two general classes based on the cellular response they evoke. One group composed of the phenylpyrimidines and several TPD examples showed a bell-shaped concentration-response effect on markers of MT stabilization in cellular assays. Moreover, these compounds induced proteasome-dependent degradation of α- and β-tubulin and caused altered MT morphology in both dividing cells and neuron cultures. In contrast, a second group comprising a subset of TPD molecules (TPD+) increased markers of stable MTs in a concentration-dependent manner in dividing cells and in neurons without affecting total tubulin levels or disrupting MT architecture. Moreover, an example TPD+ compound was shown to increase MTs in a neuron culture model with induced tau hyperphosphorylation and associated MT deficits. Several TPD+ compounds were shown to be both brain penetrant and orally bioavailable, and a TPD+ example increased MT stabilization in the mouse brain, making these compounds potential candidate therapeutics for neurodegenerative tauopathies such as Alzheimer's disease.
Collapse
Affiliation(s)
- Jane Kovalevich
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Anne-Sophie Cornec
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Yuemang Yao
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Michael James
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Alexander Crowe
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Virginia M-Y Lee
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Amos B Smith
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Carlo Ballatore
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| | - Kurt R Brunden
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania (J.K., A.-S.C., Y.Y., M.J., V.M.-Y.L., J.Q.T., C.B., K.R.B.); and Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania (A.-S.C., A.B.S., C.B.)
| |
Collapse
|
642
|
Torres A, Vargas Y, Uribe D, Carrasco C, Torres C, Rocha R, Oyarzún C, San Martín R, Quezada C. Pro-apoptotic and anti-angiogenic properties of the α /β-thujone fraction from Thuja occidentalis on glioblastoma cells. J Neurooncol 2016; 128:9-19. [PMID: 26900077 DOI: 10.1007/s11060-016-2076-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/09/2016] [Indexed: 10/22/2022]
Abstract
The most aggressive type of brain tumor is glioblastoma multiforme, which to date remains incurable. Thuja occidentalis is used in homeopathy for the treatment of cancer, however, its mechanism of action remains unknown. We set out to study the effects of thujone fractions of Thuja on glioblastoma using in vitro and in vivo models. We found that the α/ β-thujone fraction decrease the cell viability and exhibit a potent anti-proliferative, pro-apoptotic and anti-angiogenic effects in vitro. In vivo assays showed that α /β-thujone promotes the regression of neoplasia and inhibits the angiogenic markers VEGF, Ang-4 and CD31 into the tumor.
Collapse
Affiliation(s)
- Angelo Torres
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, P.O. Box 567, Valdivia, Chile
| | - Yosselyn Vargas
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, P.O. Box 567, Valdivia, Chile
| | - Daniel Uribe
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, P.O. Box 567, Valdivia, Chile
| | - Cristian Carrasco
- Departamento de Patología del Hospital Base de Valdivia (HBV), Valdivia, Chile
| | - Cristian Torres
- Departamento de Patología del Hospital Base de Valdivia (HBV), Valdivia, Chile
| | - René Rocha
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, P.O. Box 567, Valdivia, Chile
| | - Carlos Oyarzún
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, P.O. Box 567, Valdivia, Chile
| | - Rody San Martín
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, P.O. Box 567, Valdivia, Chile
| | - Claudia Quezada
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, P.O. Box 567, Valdivia, Chile.
| |
Collapse
|
643
|
Kuzmin NV, Wesseling P, Hamer PCDW, Noske DP, Galgano GD, Mansvelder HD, Baayen JC, Groot ML. Third harmonic generation imaging for fast, label-free pathology of human brain tumors. BIOMEDICAL OPTICS EXPRESS 2016; 7:1889-904. [PMID: 27231629 PMCID: PMC4871089 DOI: 10.1364/boe.7.001889] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/08/2016] [Accepted: 03/12/2016] [Indexed: 05/07/2023]
Abstract
In brain tumor surgery, recognition of tumor boundaries is key. However, intraoperative assessment of tumor boundaries by the neurosurgeon is difficult. Therefore, there is an urgent need for tools that provide the neurosurgeon with pathological information during the operation. We show that third harmonic generation (THG) microscopy provides label-free, real-time images of histopathological quality; increased cellularity, nuclear pleomorphism, and rarefaction of neuropil in fresh, unstained human brain tissue could be clearly recognized. We further demonstrate THG images taken with a GRIN objective, as a step toward in situ THG microendoscopy of tumor boundaries. THG imaging is thus a promising tool for optical biopsies.
Collapse
Affiliation(s)
- N. V. Kuzmin
- LaserLab Amsterdam, VU University, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - P. Wesseling
- Dept. of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Dept. of Pathology, Radboud University Medical Center, Geert Grooteplein Zuid, 6525 GA Nijmegen, The Netherlands
- Amsterdam Brain Tumor Center, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - P. C. de Witt Hamer
- Dept. of Neurosurgery, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Brain Tumor Center, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - D. P. Noske
- Dept. of Neurosurgery, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Brain Tumor Center, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - G. D. Galgano
- LaserLab Amsterdam, VU University, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
| | - H. D. Mansvelder
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - J. C. Baayen
- Dept. of Neurosurgery, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - M. L. Groot
- LaserLab Amsterdam, VU University, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
644
|
Jacus MO, Daryani VM, Harstead KE, Patel YT, Throm SL, Stewart CF. Pharmacokinetic Properties of Anticancer Agents for the Treatment of Central Nervous System Tumors: Update of the Literature. Clin Pharmacokinet 2016; 55:297-311. [PMID: 26293618 PMCID: PMC4761278 DOI: 10.1007/s40262-015-0319-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite significant improvement in outcomes for patients with hematologic malignancies and solid tumors over the past 10 years, patients with primary or metastatic brain tumors continue to have a poor prognosis. A primary reason for this is the inability of many chemotherapeutic drugs to penetrate into the brain and brain tumors at concentrations high enough to exert an antitumor effect because of unique barriers and efflux transporters. Several studies have been published recently examining the central nervous system pharmacokinetics of various anticancer drugs in patients with primary and metastatic brain tumors. To summarize recent advances in the field, this review critically presents studies published within the last 9 years examining brain and cerebrospinal fluid penetration of clinically available anticancer agents for patients with central nervous system tumors.
Collapse
Affiliation(s)
- Megan O Jacus
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Vinay M Daryani
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - K Elaine Harstead
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yogesh T Patel
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Stacy L Throm
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
645
|
The challenges associated with molecular targeted therapies for glioblastoma. J Neurooncol 2016; 127:427-34. [DOI: 10.1007/s11060-016-2080-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/15/2016] [Indexed: 01/06/2023]
|
646
|
Nanocarriers for the treatment of glioblastoma multiforme: Current state-of-the-art. J Control Release 2016; 227:23-37. [PMID: 26892752 DOI: 10.1016/j.jconrel.2016.02.026] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/12/2016] [Accepted: 02/13/2016] [Indexed: 01/26/2023]
Abstract
Glioblastoma multiforme, a grade IV glioma, is the most frequently occurring and invasive primary tumor of the central nervous system, which causes about 4% of cancer-associated-deaths, making it one of the most fatal cancers. With present treatments, using state-of-the-art technologies, the median survival is about 14 months and 2 year survival rate is merely 3-5%. Hence, novel therapeutic approaches are urgently necessary. However, most drug molecules are not able to cross the blood-brain barrier, which is one of the major difficulties in glioblastoma treatment. This review describes the features of blood-brain barrier, and its anatomical changes with different stages of tumor growth. Moreover, various strategies to improve brain drug delivery i.e. tight junction opening, chemical modification of the drug, efflux transporter inhibition, convection-enhanced delivery, craniotomy-based drug delivery and drug delivery nanosystems are discussed. Nanocarriers are one of the highly potential drug transport systems that have gained huge research focus over the last few decades for site specific drug delivery, including drug delivery to the brain. Properly designed nanocolloids are capable to cross the blood-brain barrier and specifically deliver the drug in the brain tumor tissue. They can carry both hydrophilic and hydrophobic drugs, protect them from degradation, release the drug for sustained period, significantly improve the plasma circulation half-life and reduce toxic effects. Among various nanocarriers, liposomes, polymeric nanoparticles and lipid nanocapsules are the most widely studied, and are discussed in this review. For each type of nanocarrier, a general discussion describing their composition, characteristics, types and various uses is followed by their specific application to glioblastoma treatment. Moreover, some of the main challenges regarding toxicity and standardized evaluation techniques are narrated in brief.
Collapse
|
647
|
Sminia P, Westerman BA. Blood-brain barrier crossing and breakthroughs in glioblastoma therapy. Br J Clin Pharmacol 2016; 81:1018-20. [PMID: 26744058 DOI: 10.1111/bcp.12881] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/23/2015] [Accepted: 01/01/2016] [Indexed: 10/22/2022] Open
Affiliation(s)
- P Sminia
- Department of Radiation Oncology, VU University Medical Center, Amsterdam
| | - B A Westerman
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
648
|
Mfsd2a-based pharmacological strategies for drug delivery across the blood–brain barrier. Pharmacol Res 2016; 104:124-31. [DOI: 10.1016/j.phrs.2015.12.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/20/2015] [Accepted: 12/20/2015] [Indexed: 12/20/2022]
|
649
|
Fan CH, Ting CY, Lin CY, Chan HL, Chang YC, Chen YY, Liu HL, Yeh CK. Noninvasive, Targeted, and Non-Viral Ultrasound-Mediated GDNF-Plasmid Delivery for Treatment of Parkinson's Disease. Sci Rep 2016; 6:19579. [PMID: 26786201 PMCID: PMC4726227 DOI: 10.1038/srep19579] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/16/2015] [Indexed: 01/30/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) supports the growth and survival of dopaminergic neurons. CNS gene delivery currently relies on invasive intracerebral injection to transit the blood-brain barrier. Non-viral gene delivery via systematic transvascular route is an attractive alternative because it is non-invasive, but a high-yield and targeted gene-expressed method is still lacking. In this study, we propose a novel non-viral gene delivery approach to achieve targeted gene transfection. Cationic microbubbles as gene carriers were developed to allow the stable formation of a bubble-GDNF gene complex, and transcranial focused ultrasound (FUS) exposure concurrently interacting with the bubble-gene complex allowed transient gene permeation and induced local GDNF expression. We demonstrate that the focused ultrasound-triggered GDNFp-loaded cationic microbubbles platform can achieve non-viral targeted gene delivery via a noninvasive administration route, outperform intracerebral injection in terms of targeted GDNF delivery of high-titer GDNF genes, and has a neuroprotection effect in Parkinson’s disease (PD) animal models to successfully block PD syndrome progression and to restore behavioral function. This study explores the potential of using FUS and bubble-gene complexes to achieve noninvasive and targeted gene delivery for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan
| | - Chien-Yu Ting
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan
| | - Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, 33302 Taiwan
| | - Hong-Lin Chan
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013 Taiwan
| | - Yuan-Chih Chang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529 Taiwan
| | - You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming University, Taipei, 11221 Taiwan
| | - Hao-Li Liu
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, 33302 Taiwan.,Department of Electrical Engineering, Chang-Gung University, Taoyuan, 33302 Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan
| |
Collapse
|
650
|
Frosina G. Nanoparticle-mediated drug delivery to high-grade gliomas. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1083-1093. [PMID: 26767516 DOI: 10.1016/j.nano.2015.12.375] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
Abstract
UNLABELLED High grade gliomas (HGGs) are fatal brain tumors due to their infiltration capacity and the presence of resistant cell populations. Further, the brain is naturally protected from many exogenous molecules by the brain blood barrier (BBB), which limits or cancels passage of cytotoxic drugs to the tumor sites. In order to cope with the latter problem, nanoparticle (NP)-based carriers are intensively investigated, due to multiple possibilities to drive them across the BBB to the tumor sites and drop cytotoxic molecules there. The current status of research on NP for drug delivery to HGGs has been analyzed. The results indicate gold, lipids and proteins as three main materials featuring NP formulations for HGG treatment. Albeit specific drug targeting to HGG cells may have not been so far significantly improved, NP may help drugs crossing the BBB and enter the brain thus potentially fixing at least one part of the problem. FROM THE CLINICAL EDITOR High grade gliomas (HGG) are very aggressive tumours and current therapy remains unsatisfactory. The advance in nanomedicine has allowed the development of novel treatment modalities. In this review article, the authors outlined the current status in using nanoparticle (NP)-based carriers for drug delivery to HGG. This should help readers to understand and develop ideas for further drug carrier designs.
Collapse
Affiliation(s)
- Guido Frosina
- Mutagenesis Unit, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy.
| |
Collapse
|