Current Challenges and Opportunities in Treating Glioblastomas

Total Page:16

File Type:pdf, Size:1020Kb

Current Challenges and Opportunities in Treating Glioblastomas Supplemental Material can be found at: /content/suppl/2018/04/23/70.3.412.DC1.html 1521-0081/70/3/412–445$35.00 https://doi.org/10.1124/pr.117.014944 PHARMACOLOGICAL REVIEWS Pharmacol Rev 70:412–445, July 2018 Copyright © 2018 by The Author(s) This is an open access article distributed under the CC BY-NC Attribution 4.0 International license. ASSOCIATE EDITOR: ERIC L. BARKER Current Challenges and Opportunities in Treating Glioblastomas Andrea Shergalis, Armand Bankhead, III, Urarika Luesakul, Nongnuj Muangsin, and Nouri Neamati Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.) Abstract ...................................................................................413 I. Introduction . ..............................................................................413 II. Current Treatment Options for Glioblastoma ...............................................414 III. Molecular Diagnostic Signature of Glioblastoma . ..........................................419 IV. Characteristics of Protein Expression in Glioblastoma . .....................................419 V. Emerging Targets in Glioblastoma . ......................................................422 A. Biomarker Identification . ............................................................422 B. Drug Discovery Targets ................................................................422 Downloaded from 1. Gene Expression Associated with Reduced Patient Survival . .........................423 a. BOC cell adhesion associated, oncogene regulated (BOC) . .........................426 b. C-Type lectin domain family 4 member G pseudogene 1 (CLEC4GP1) . .............426 c. ELOVL fatty acid elongase 6 (ELOVL6). ..........................................426 d. Epiregulin (EREG) ................................................................427 by guest on September 25, 2021 e. Estrogen receptor 2 (ESR2). ......................................................427 f. Follicular dendritic cell secreted protein (FDCSP) . ...............................427 g. Furin .............................................................................427 h. Fucosyltransferase 8 antisense RNA 1 (FUT8-AS1) . ...............................429 i. Granzyme B (GZMB) . ............................................................429 j. Iroquois homeobox 3 (IRX3). ......................................................429 k. Lipopolysaccharide-induced tissue-necrosis-factor factor (LITAF) ...................429 l. NudE neurodevelopment protein 1 like 1 (NDEL1)..................................429 m. NK3 homeobox 1 (NKX3-1) . ......................................................430 n. Podocan like 1 (PODNL1)..........................................................430 o. Protein tyrosine phosphatase, receptor type N (PTPRN). .........................430 p. Quiescin sulfhydryl oxidase 1 (QSOX1). ..........................................430 q. Semaphorin 4F (SEMA4F). ......................................................430 r. Tyrosine hydroxylase (TH). ......................................................431 s. Vascular endothelial growth factor C (VEGFC).....................................431 t. Chromosome 20 open reading frame 166 antisense RNA 1 (C20orf166AS1). .......431 2. Protein Targets Identified via Proteomic Approaches . ...............................431 3. Target Validation. ..................................................................432 C. Synthetic Lethality. ..................................................................432 D. Preclinical Models of Glioblastoma .....................................................432 VI. Blood-Brain Barrier........................................................................433 A. Characteristics of the Blood-Brain Barrier ...............................................433 B. Blood-Brain Barrier Computational Modeling for Drug Discovery ........................434 VII. Drug Discovery Challenges in GBM . ......................................................435 Address correspondence to: Dr. Nouri Neamati, North Campus Research Complex, Bldg. 520, Rm 1363, 1600 Huron Pkwy, Ann Arbor, MI 48109-2800. E-mail: [email protected] This work was supported by the National Institutes of Health, National Cancer Institute [Grant CA193690]. https://doi.org/10.1124/pr.117.014944. s This article has supplemental material available at pharmrev.aspetjournals.org. 412 Current Challenges and Opportunities in Treating GBM 413 VIII. Advances in Drug Delivery.................................................................437 A. Nanocarriers . ........................................................................437 1. General Properties of Nanoparticles. ................................................437 2. Nanocarriers in Clinical Trials . ......................................................437 B. Prodrugs. ..............................................................................438 IX. Conclusions and Future Directions . ......................................................439 Acknowledgments. ........................................................................441 References . ..............................................................................441 Abstract——Glioblastoma multiforme (GBM), the survival outcomes. Importantly, nine of these genes most common and aggressive primary brain tumor, exhibit higher expression in GBM versus low-grade has a high mortality rate despite extensive efforts glioma and may be involved in disease progression. In to develop new treatments. GBM exhibits both intra- this review, we discuss these proteins in the context and intertumor heterogeneity, lending to resistance and of GBM disease progression. We also conducted eventual tumor recurrence. Large-scale genomic and computational multi-parameter optimization to assess proteomic analysis of GBM tumors has uncovered the blood-brain barrier (BBB) permeability of small potential drug targets. Effective and “druggable” targets molecules in clinical trials for GBM treatment. Drug must be validated to embark on a robust medicinal delivery in the context of GBM is particularly chemistry campaign culminating in the discovery of challenging because the BBB hinders small molecule clinical candidates. Here, we review recent developments transport. Therefore, we discuss novel drug delivery in GBM drug discovery and delivery. To identify GBM drug methods, including nanoparticles and prodrugs. Given targets, we performed extensive bioinformatics analysis the aggressive nature of GBM and the complexity of usingdatafromTheCancerGenomeAtlasproject.We targeting the central nervous system, effective discovered 20 genes, BOC, CLEC4GP1, ELOVL6, EREG, treatment options are a major unmet medical need. ESR2, FDCSP, FURIN, FUT8-AS1, GZMB, IRX3, LITAF, Identification and validation of biomarkers and drug NDEL1, NKX3-1, PODNL1, PTPRN, QSOX1, SEMA4F, TH, targets associated with GBM disease progression VEGFC,andC20orf166AS1 that are overexpressed in a present an exciting opportunity to improve treatment subpopulation of GBM patients and correlate with poor of this devastating disease. I. Introduction an efficient approach to identify and vet potential drugs at the preclinical stage to prevent late stage failure. Glioblastoma multiforme (GBM) is the most malig- Genomic- and proteomic-scale analysis can identify nant primary central nervous system tumor, and the proteins and pathways involved in the development of prognosis for patients is often bleak. Currently, there are no curative treatment options for GBM, and despite chemotherapeutic resistance mechanisms responsible rigorous therapeutic research, the survival rate of for recurrent disease. patients diagnosed with GBM remains low. Median With the advent of TCGA (The Cancer Genome Atlas) overall survival is 15–23 months and 5-year survival is consortium and resources, genomic analysis of cancers less than 6%, which is the lowest long-term survival rate is at the forefront of drug discovery. Additionally, of malignant brain tumors (Ostrom et al., 2016). An proteomics is gaining widespread use in drug discovery estimated 79,270 new cases of primary brain and other efforts. Quantitative proteomics can measure the ex- central nervous system (CNS) tumors were expected to pression and, in some cases, posttranslational modifi- be diagnosed in 2017 (Ostrom et al., 2016). To improve cation status of up to and over 8000 proteins in the cell therapeutic options, studies to identify and validate at any given time. The advent of novel proteomic single protein targets are underway. However, in most techniques in the last decade, in tandem with the cases, targeted compounds that perform well in pre- resources allocated to address the lack of a cure for clinical studies have failed expensive Phase III clinical GBM, will accelerate the discovery of a treatment and trials in humans. Ultimately, several major factors are shed light on the feasibility of precision medicine. responsible for drug failure, including poor pharmaco- The target and mechanism of action of many Food and kinetic properties, emergence of resistance pathways, Drug Administration (FDA)-approved drugs are not complex intratumoral heterogeneity, and suboptimal fully established. Of the approximately
Recommended publications
  • A Human Multi-Lineage Hepatic Organoid Model for Liver Fibrosis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.01.278473; this version posted September 2, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. A Human Multi-Lineage Hepatic Organoid Model for Liver Fibrosis Yuan Guan1, Annika Enejder2, Meiyue Wang1, Zhuoqing Fang1, Lu Cui3, Shih-Yu Chen4, Jingxiao Wang1, Yalun Tan1, Manhong Wu1, Xinyu Chen1, Patrik K. Johansson2, Issra Osman1, Koshi Kunimoto3, Pierre Russo5, Sarah C. Heilshorn2 and Gary Peltz1* 1Department of Anesthesia, Stanford University School of Medicine, Stanford CA, 94305; 2Department of Materials Science and Engineering, Stanford University, Stanford CA, 94305; 3Department of Pathology, Institute of Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford University School of Medicine, Stanford, 94305, CA, USA; 4Shih-Yu Chen, Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan; 5Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104 The authors have declared that no conflict of interest exists. *Address Correspondence to: [email protected] 300 Pasteur Dr. Room L232 Stanford, CA 94305. bioRxiv preprint doi: https://doi.org/10.1101/2020.09.01.278473; this version posted September 2, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Summary Despite its devastating consequences, liver fibrosis has no treatments. Genome engineering and a hepatic organoid system was used to produce the first in vitro model for human liver fibrosis. Hepatic organoids engineered to express the most common causative mutation for Autosomal Recessive Polycystic Kidney Disease (ARPKD) developed the key features of ARPKD liver pathology (abnormal bile ducts and hepatic fibrosis) in only 21 days.
    [Show full text]
  • Deciphering Molecular Mechanisms and Prioritizing Therapeutic Targets in Cardio-Oncology
    Figure 1. This is a pilot view to explore the potential of EpiGraphDB to inform us about proteins that are linked to the pathophysiology of cancer and cardiovascular disease (CVD). For each cancer type (pink diamonds), we searched for cancer related proteins (light blue circles) that interact with other proteins identified as protein quantitative trait loci (pQTLs) for CVD (red diamonds for pathologies, orange triangles for risk factors). These pQTLs can be acting in cis (solid lines) or trans-acting (dotted lines). Proteins can interact either directly, a protein-protein interaction (dotted blue edges), or through the participation in the same pathway (red parallel lines). Shared pathways are represented with blue hexagons. We also queried which of these proteins are targeted by existing drugs. We found that the cancer drug cetuximab (yellow circle) inhibits EGFR. Other potential drugs are depicted in light brown hexagonal meta-nodes that are detailed below. Deciphering molecular mechanisms and prioritizing therapeutic targets in cardio-oncology Pau Erola1,2, Benjamin Elsworth1,2, Yi Liu2, Valeriia Haberland2 and Tom R Gaunt1,2,3 1 CRUK Integrative Cancer Epidemiology Programme; 2 MRC Integrative Epidemiology Unit, University of Bristol; 3 The Alan Turing Institute Cancer and cardiovascular disease (CVD) make by far the immense What is EpiGraphDB? contribution to the totality of human disease burden, and although mortality EpiGraphDB is an analytical platform and graph database that aims to is declining the number of those living with the disease shows little address the necessity of innovative and scalable approaches to harness evidence of change (Bhatnagar et al., Heart, 2016).
    [Show full text]
  • Therapeutic Inhibition of VEGF Signaling and Associated Nephrotoxicities
    REVIEW www.jasn.org Therapeutic Inhibition of VEGF Signaling and Associated Nephrotoxicities Chelsea C. Estrada,1 Alejandro Maldonado,1 and Sandeep K. Mallipattu1,2 1Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York; and 2Renal Section, Northport Veterans Affairs Medical Center, Northport, New York ABSTRACT Inhibition of vascular endothelial growth factor A (VEGFA)/vascular endothelial with hypertension and proteinuria. Re- growth factor receptor 2 (VEGFR2) signaling is a common therapeutic strategy in ports describe histologic changes in the oncology, with new drugs continuously in development. In this review, we consider kidney primarily as glomerular endothe- the experimental and clinical evidence behind the diverse nephrotoxicities associ- lial injury with thrombotic microangiop- ated with the inhibition of this pathway. We also review the renal effects of VEGF athy (TMA).8 Nephrotic syndrome has inhibition’s mediation of key downstream signaling pathways, specifically MAPK/ also been observed,9 with the clinical ERK1/2, endothelial nitric oxide synthase, and mammalian target of rapamycin manifestations varying according to (mTOR). Direct VEGFA inhibition via antibody binding or VEGF trap (a soluble decoy mechanism and direct target of VEGF receptor) is associated with renal-specific thrombotic microangiopathy (TMA). Re- inhibition. ports also indicate that tyrosine kinase inhibition of the VEGF receptors is prefer- Current VEGF inhibitors can be clas- entially associated with glomerulopathies such as minimal change disease and FSGS. sifiedbytheirtargetofactioninthe Inhibition of the downstream pathway RAF/MAPK/ERK has largely been associated VEGFA-VEGFR2 pathway: drugs that with tubulointerstitial injury. Inhibition of mTOR is most commonly associated with bind to VEGFA, sequester VEGFA, in- albuminuria and podocyte injury, but has also been linked to renal-specificTMA.In hibit receptor tyrosine kinases (RTKs), all, we review the experimentally validated mechanisms by which VEGFA-VEGFR2 or inhibit downstream pathways.
    [Show full text]
  • FLT3 Inhibitors in Acute Myeloid Leukemia Mei Wu1, Chuntuan Li2 and Xiongpeng Zhu2*
    Wu et al. Journal of Hematology & Oncology (2018) 11:133 https://doi.org/10.1186/s13045-018-0675-4 REVIEW Open Access FLT3 inhibitors in acute myeloid leukemia Mei Wu1, Chuntuan Li2 and Xiongpeng Zhu2* Abstract FLT3 mutations are one of the most common findings in acute myeloid leukemia (AML). FLT3 inhibitors have been in active clinical development. Midostaurin as the first-in-class FLT3 inhibitor has been approved for treatment of patients with FLT3-mutated AML. In this review, we summarized the preclinical and clinical studies on new FLT3 inhibitors, including sorafenib, lestaurtinib, sunitinib, tandutinib, quizartinib, midostaurin, gilteritinib, crenolanib, cabozantinib, Sel24-B489, G-749, AMG 925, TTT-3002, and FF-10101. New generation FLT3 inhibitors and combination therapies may overcome resistance to first-generation agents. Keywords: FMS-like tyrosine kinase 3 inhibitors, Acute myeloid leukemia, Midostaurin, FLT3 Introduction RAS, MEK, and PI3K/AKT pathways [10], and ultim- Acute myeloid leukemia (AML) remains a highly resist- ately causes suppression of apoptosis and differentiation ant disease to conventional chemotherapy, with a me- of leukemic cells, including dysregulation of leukemic dian survival of only 4 months for relapsed and/or cell proliferation [11]. refractory disease [1]. Molecular profiling by PCR and Multiple FLT3 inhibitors are in clinical trials for treat- next-generation sequencing has revealed a variety of re- ing patients with FLT3/ITD-mutated AML. In this re- current gene mutations [2–4]. New agents are rapidly view, we summarized the preclinical and clinical studies emerging as targeted therapy for high-risk AML [5, 6]. on new FLT3 inhibitors, including sorafenib, lestaurtinib, In 1996, FMS-like tyrosine kinase 3/internal tandem du- sunitinib, tandutinib, quizartinib, midostaurin, gilteriti- plication (FLT3/ITD) was first recognized as a frequently nib, crenolanib, cabozantinib, Sel24-B489, G-749, AMG mutated gene in AML [7].
    [Show full text]
  • Relevance Network Between Chemosensitivity and Transcriptome in Human Hepatoma Cells1
    Vol. 2, 199–205, February 2003 Molecular Cancer Therapeutics 199 Relevance Network between Chemosensitivity and Transcriptome in Human Hepatoma Cells1 Masaru Moriyama,2 Yujin Hoshida, topoisomerase II ␤ expression, whereas it negatively Motoyuki Otsuka, ShinIchiro Nishimura, Naoya Kato, correlated with expression of carboxypeptidases A3 Tadashi Goto, Hiroyoshi Taniguchi, and Z. Response to nimustine was associated with Yasushi Shiratori, Naohiko Seki, and Masao Omata expression of superoxide dismutase 2. Department of Gastroenterology, Graduate School of Medicine, Relevance networks identified several negative University of Tokyo, Tokyo 113-8655 [M. M., Y. H., M. O., N. K., T. G., H. T., Y. S., M. O.]; Cellular Informatics Team, Computational Biology correlations between gene expression and resistance, Research Center, Tokyo 135-0064 [S. N.]; and Department of which were missed by hierarchical clustering. Our Functional Genomics, Graduate School of Medicine, Chiba University, results suggested the necessity of systematically Chiba 260-8670 [N. S.], Japan evaluating the transporting systems that may play a major role in resistance in hepatoma. This may provide Abstract useful information to modify anticancer drug action in Generally, hepatoma is not a chemosensitive tumor, hepatoma. and the mechanism of resistance to anticancer drugs is not fully elucidated. We aimed to comprehensively Introduction evaluate the relationship between chemosensitivity and Hepatoma is a major cause of death even in developed gene expression profile in human hepatoma cells, by countries, and its incidence is increasing (1). Despite the using microarray analysis, and analyze the data by progress of therapeutic technique (2), the efficacy of radical constructing relevance networks. therapy is hampered by frequent recurrence and advance of In eight hepatoma cell lines (HLE, HLF, Huh7, Hep3B, the tumor (3).
    [Show full text]
  • Title Second-Line Chemotherapy for Small-Cell Lung Cancer
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Kyoto University Research Information Repository Second-line chemotherapy for small-Cell Lung Cancer Title (SCLC). Author(s) Kim, Young Hak; Mishima, Michiaki Citation Cancer treatment reviews (2011), 37(2): 143-150 Issue Date 2011-04 URL http://hdl.handle.net/2433/137220 Right © 2010 Elsevier Ltd Type Journal Article Textversion author Kyoto University Second-line Chemotherapy for Small-Cell Lung Cancer (SCLC) Young Hak Kim and Michiaki Mishima Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan For reprints and all correspondence: Young Hak Kim Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan Phone: +81-75-751-3830; Fax: +81-75-751-4643; E-mail: [email protected] Running title: Second-line Chemotherapy for SCLC Key words: small-cell lung cancer, relapsed, chemotherapy, second line, sensitive, refractory 1 Abstract Although small-cell lung cancer (SCLC) generally shows an excellent response to initial chemotherapy, most patients finally relapse and salvage chemotherapy is considered. Usually, the response to salvage chemotherapy significantly differs between sensitive and refractory relapse. Sensitive relapse is relatively chemosensitive and re-challenge with the same drugs as used in the initial chemotherapy has been used historically, while refractory relapse is extremely chemoresistant and its prognosis has been abysmal. To date, a number of clinical trials have been carried out for relapsed SCLC; however, the number of randomized trials is quite limited.
    [Show full text]
  • Leptomeningeal Metastases from Solid Tumors: Recent Advances in Diagnosis and Molecular Approaches
    cancers Review Leptomeningeal Metastases from Solid Tumors: Recent Advances in Diagnosis and Molecular Approaches Alessia Pellerino 1,* , Priscilla K. Brastianos 2, Roberta Rudà 1,3 and Riccardo Soffietti 1 1 Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; [email protected] (R.R.); riccardo.soffi[email protected] (R.S.) 2 Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02115, USA; [email protected] 3 Department of Neurology, Castelfranco Veneto and Brain Tumor Board Treviso Hospital, 31100 Treviso, Italy * Correspondence: [email protected]; Tel.: +39-011-633-4904 Simple Summary: Leptomeningeal metastases are a devastating complication of solid tumors with poor survival, regardless of the type of treatments. The limited efficacy of targeted agents is due to the molecular divergence between leptomeningeal recurrences and primary site, as well as the presence of a heterogeneous blood-brain barrier and blood-tumor barrier that interfere with the penetration of drugs into the brain. The diagnosis of leptomeningeal metastases is achieved by neurological examination, and/or brain and spinal magnetic resonance, and/or a positive cerebrospinal fluid cytology. The presence of neoplastic cells in the cerebrospinal fluid examination is the gold-standard for the diagnosis of leptomeningeal metastases; however, novel techniques known as “liquid biopsy” aim to improve the sensitivity and specificity in detecting circulating neoplastic cells or DNA in the cerebrospinal fluid. Targeted therapies and immunotherapies have changed the natural history Citation: Pellerino, A.; Brastianos, of metastatic solid tumors, including lung, breast cancer, and melanoma. Targeting actionable P.K.; Rudà, R.; Soffietti, R.
    [Show full text]
  • Cell-Cycle and DNA-Damage Response Pathway Is Involved In
    Published OnlineFirst October 13, 2017; DOI: 10.1158/1078-0432.CCR-17-1582 Biology of Human Tumors Clinical Cancer Research Cell-Cycle and DNA-Damage Response Pathway Is Involved in Leptomeningeal Metastasis of Non–Small Cell Lung Cancer Yun Fan1, Xuehua Zhu2, Yan Xu3, Xuesong Lu4, Yanjun Xu1, Mengzhao Wang3, Haiyan Xu4, Jingyan Ding2, Xin Ye2, Luo Fang5, Zhiyu Huang5, Lei Gong5, Hongyang Lu1, Weimin Mao1, and Min Hu2 Abstract Purpose: Leptomeningeal metastasis (LM) is a detrimental risk. Intriguingly, low overlapping of somatic protein-changing complication of non–small cell lung cancer (NSCLC) and asso- variants was observed between paired CSF and primary lesions, ciated with poor prognosis. However, the underlying mechanisms exhibiting tumor heterogeneity and genetic divergence. Moreover, of the metastasis process are still poorly understood. genes with CSF-recurrent genomic alterations were predominant- Experimental Design: We performed next-generation panel ly involved in cell-cycle regulation and DNA-damage response sequencing of primary tumor tissue, cerebrospinal fluid (CSF), (DDR), suggesting a role of the pathway in LM development. and matched normal controls from epidermal growth factor Conclusions: Our study has shed light on the genomic varia- receptor (EGFR) mutation-positive NSCLC patients with LM. tions of NSCLC-LM, demonstrated genetic heterogeneity and Results: The status of EGFR-activating mutations was highly divergence, uncovered involvement of cell-cycle and DDR path- concordant between primary tumor and CSF. PIK3CA aberrations way, and paved the way for potential therapeutic approaches to were high in these patients, implicating an association with LM this unmet medical need. Clin Cancer Res; 24(1); 209–16.
    [Show full text]
  • Nimustine Induces DNA Breaks and Crosslinks in NIH/3T3 Cells
    International Journal of Bioscience, Biochemistry and Bioinformatics, Vol. 3, No. 3, May 2013 Nimustine Induces DNA Breaks and Crosslinks in NIH/3T3 Cells Lin-Na Zhao, Xue-Chai Chen, Yan-Yan Zhong, Qin-Xia Hou, and Ru-Gang Zhong study of drug-induced DNA cross-linking to reveal the Abstract—The relationship between carcinogenicity and difference between the nitrosourea anticancer mechanism DNA interstrand cross-links of nitrosoureas is poorly defined. and carcinogenic role. 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3- (2-chloroethyl)- ACNU was discovered in 1974 as the first water-soluble 3- nitrosourea (ACNU, nimustine) is one of nitrosoureas used in nitrosourea compound [11], and mainly used in the clinic of the treatment of high-grade gliomas. It has the capability of causing DNA interstrand cross-links (ICLs) to kill cancer cells. glioblastoma patients before the introduction of But it can also cause the generation of secondary tumors with 8-carbamoyl-3-methylimidazo [5, 1-d]-1, 2, 3, carcinogenic side effects. In present study, we investigated DNA 5-tetrazin-4(3H) -one (temozolomide, TMZ) because of its interstrand cross-links, DNA double-strand breaks and cell high permeability across blood-brain barrier (BBB) and good cycle phase in NIH/3T3 cells from the primary mouse cytotoxic activity for gliomas [12]-[13]. BCNU always embryonic fibroblast cells induced by ACNU. This result induces two major types of genotoxic damage to the cell: indicated that the concentration of 60 and 75μg/ml of ACNU could be detected significantly ICLs, and the γ-H2AX has the DNA mono adducts and DNA interstrand cross-links.
    [Show full text]
  • Nanoconjugates Able to Cross the Blood-Brain Barrier Alexander H Stegh, Janina Paula Luciano, Samuel A
    (12) STANDARD PATENT (11) Application No. AU 2017216461 B2 (19) AUSTRALIAN PATENT OFFICE (54) Title Nanoconjugates Able To Cross The Blood-Brain Barrier (51) International Patent Classification(s) A61K 31/7088 (2006.01) A61K 48/00 (2006.0 1) A61K 9/00 (2006.01) A61P 35/00 (2006.01) (21) Application No: 2017216461 (22) Date of Filing: 2017.08.15 (43) Publication Date: 2017.08.31 (43) Publication Journal Date: 2017.08.31 (44) Accepted Journal Date: 2019.10.17 (62) Divisional of: 2012308302 (71) Applicant(s) NorthwesternUniversity (72) Inventor(s) Mirkin, Chad A.;Ko, Caroline H.;Stegh, Alexander;Giljohann, David A.;Luciano, Janina;Jensen, Sam (74) Agent / Attorney WRAYS PTY LTD, L7 863 Hay St, Perth, WA, 6000, AU (56) Related Art US 2010/0233084 Al LJUBIMOVA et al. "Nanoconjugate based on polymalic acid for tumor targeting", Chemico-Biological Interactions, 2008, Vol. 171, Pages 195-203. WO 2011/028847 Al BONOU et al. "Nanotechnology approach for drug addiction therapy: Gene silencing using delivery of gold nanorod-siRNA nanoplex in dopaminergic neurons", PNAS, 2009, Vol. 106, No. 14, Pages 5546-5550. PATIL et al. "Temozolomide Delivery to Tumor Cells by a Multifunctional Nano Vehicle Based on Poly(#-L-malic acid)", Pharmaceutical Research, 2010, Vol. 27, Pages 2317-2329. ABSTRACT Polyvalent nanoconjugates address the critical challenges in therapeutic use. The single-entity, targeted therapeutic is able to cross the blood-brain barrier (BBB) and is thus effective in the treatment of central nervous system (CNS) disorders. Further, despite the tremendously high 5 cellular uptake of nanoconjugates, they exhibit no toxicity in the cell types tested thus far.
    [Show full text]
  • Precision Medicine in Pediatric Neurooncology: a Review
    UCLA UCLA Previously Published Works Title Precision Medicine in Pediatric Neurooncology: A Review. Permalink https://escholarship.org/uc/item/6tv2b43d Journal ACS chemical neuroscience, 9(1) ISSN 1948-7193 Authors Mochizuki, Aaron Y Frost, Isaura M Mastrodimos, Melina B et al. Publication Date 2018 DOI 10.1021/acschemneuro.7b00388 Peer reviewed eScholarship.org Powered by the California Digital Library University of California HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author ACS Chem Manuscript Author Neurosci. Author Manuscript Author manuscript; available in PMC 2019 July 24. Published in final edited form as: ACS Chem Neurosci. 2018 January 17; 9(1): 11–28. doi:10.1021/acschemneuro.7b00388. Precision Medicine in Pediatric Neurooncology: A Review Aaron Y. Mochizuki∥, Isaura M. Frost∥, Melina B. Mastrodimos∥, Ashley S. Plant┴, Anthony C. Wang#, Theodore B. Moore∥, Robert M. Prins#,∇,○, Paul S. Weiss*,†,‡,§,∇, Steven J. Jonas*,†,∥,◆,¶ †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States §Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States ∥Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States ┴Division of Pediatric Oncology, Children’s Hospital of Orange
    [Show full text]
  • Patent Application Publication ( 10 ) Pub . No . : US 2019 / 0192440 A1
    US 20190192440A1 (19 ) United States (12 ) Patent Application Publication ( 10) Pub . No. : US 2019 /0192440 A1 LI (43 ) Pub . Date : Jun . 27 , 2019 ( 54 ) ORAL DRUG DOSAGE FORM COMPRISING Publication Classification DRUG IN THE FORM OF NANOPARTICLES (51 ) Int . CI. A61K 9 / 20 (2006 .01 ) ( 71 ) Applicant: Triastek , Inc. , Nanjing ( CN ) A61K 9 /00 ( 2006 . 01) A61K 31/ 192 ( 2006 .01 ) (72 ) Inventor : Xiaoling LI , Dublin , CA (US ) A61K 9 / 24 ( 2006 .01 ) ( 52 ) U . S . CI. ( 21 ) Appl. No. : 16 /289 ,499 CPC . .. .. A61K 9 /2031 (2013 . 01 ) ; A61K 9 /0065 ( 22 ) Filed : Feb . 28 , 2019 (2013 .01 ) ; A61K 9 / 209 ( 2013 .01 ) ; A61K 9 /2027 ( 2013 .01 ) ; A61K 31/ 192 ( 2013. 01 ) ; Related U . S . Application Data A61K 9 /2072 ( 2013 .01 ) (63 ) Continuation of application No. 16 /028 ,305 , filed on Jul. 5 , 2018 , now Pat . No . 10 , 258 ,575 , which is a (57 ) ABSTRACT continuation of application No . 15 / 173 ,596 , filed on The present disclosure provides a stable solid pharmaceuti Jun . 3 , 2016 . cal dosage form for oral administration . The dosage form (60 ) Provisional application No . 62 /313 ,092 , filed on Mar. includes a substrate that forms at least one compartment and 24 , 2016 , provisional application No . 62 / 296 , 087 , a drug content loaded into the compartment. The dosage filed on Feb . 17 , 2016 , provisional application No . form is so designed that the active pharmaceutical ingredient 62 / 170, 645 , filed on Jun . 3 , 2015 . of the drug content is released in a controlled manner. Patent Application Publication Jun . 27 , 2019 Sheet 1 of 20 US 2019 /0192440 A1 FIG .
    [Show full text]