346 results on '"Johns, Terrance G."'
Search Results
102. Abstract B234: Dual targeting of the tyrosine kinase receptors EGFRvIII and c‐Met in glioblastoma multiforme
- Author
-
Pillay, Vinochani, primary, Scott, Andrew M., additional, and Johns, Terrance G., additional
- Published
- 2009
- Full Text
- View/download PDF
103. Fyn and Src Are Effectors of Oncogenic Epidermal Growth Factor Receptor Signaling in Glioblastoma Patients
- Author
-
Lu, Kan V., primary, Zhu, Shaojun, additional, Cvrljevic, Anna, additional, Huang, Tiffany T., additional, Sarkaria, Shawn, additional, Ahkavan, David, additional, Dang, Julie, additional, Dinca, Eduard B., additional, Plaisier, Seema B., additional, Oderberg, Isaac, additional, Lee, Yohan, additional, Chen, Zugen, additional, Caldwell, Jeremy S., additional, Xie, Yongmin, additional, Loo, Joseph A., additional, Seligson, David, additional, Chakravari, Arnab, additional, Lee, Francis Y., additional, Weinmann, Roberto, additional, Cloughesy, Timothy F., additional, Nelson, Stanley F., additional, Bergers, Gabriele, additional, Graeber, Thomas, additional, Furnari, Frank B., additional, James, C. David, additional, Cavenee, Webster K., additional, Johns, Terrance G., additional, and Mischel, Paul S., additional
- Published
- 2009
- Full Text
- View/download PDF
104. Targeting a unique EGFR epitope with monoclonal antibody 806 activates NF-κB and initiates tumour vascular normalization
- Author
-
Gan, Hui K., primary, Lappas, Martha, additional, Cao, Diana X., additional, Cvrljevdic, Anna, additional, Scott, Andrew M., additional, and Johns, Terrance G., additional
- Published
- 2009
- Full Text
- View/download PDF
105. A truncated soluble epidermal growth factor receptor-Fc fusion ligand trap displays anti-tumour activityin vivo
- Author
-
Adams, Timothy E., primary, Koziolek, Eva J., additional, Hoyne, Peter H., additional, Bentley, John D., additional, Lu, Louis, additional, Lovrecz, George, additional, Ward, Colin W., additional, Lee, F. T., additional, Scott, Andrew M., additional, Nash, Andrew D., additional, Rothacker, Julie, additional, Nice, Edouard C., additional, Burgess, Antony W., additional, Johns, Terrance G., additional, and Adams, Timothy E., additional
- Published
- 2009
- Full Text
- View/download PDF
106. Heparin-Binding Epidermal Growth Factor–Like Growth Factor Signaling in Flow-Induced Arterial Remodeling
- Author
-
Zhang, Hua, primary, Sunnarborg, Susan W., additional, McNaughton, K. Kirk, additional, Johns, Terrance G., additional, Lee, David C., additional, and Faber, James E., additional
- Published
- 2008
- Full Text
- View/download PDF
107. The Efficacy of Epidermal Growth Factor Receptor–Specific Antibodies against Glioma Xenografts Is Influenced by Receptor Levels, Activation Status, and Heterodimerization
- Author
-
Johns, Terrance G., primary, Perera, Rushika M., additional, Vernes, Sonja C., additional, Vitali, Angela A., additional, Cao, Diana X., additional, Cavenee, Webster K., additional, Scott, Andrew M., additional, and Furnari, Frank B., additional
- Published
- 2007
- Full Text
- View/download PDF
108. Epidermal Growth Factor Receptor Activation: An Upstream Signal for Transition of Quiescent Astrocytes into Reactive Astrocytes after Neural Injury
- Author
-
Liu, Bin, primary, Chen, Huiyi, additional, Johns, Terrance G., additional, and Neufeld, Arthur H., additional
- Published
- 2006
- Full Text
- View/download PDF
109. An automated peptide and protein thiazolidine coupling chemistry for biosensor immobilization giving a unique N-terminal orientation
- Author
-
Wade, John D., primary, Hojo, Keiko, additional, Kawasaki, Koichi, additional, Johns, Terrance G., additional, Catimel, Bruno, additional, Rothacker, Julie, additional, and Nice, Edouard C., additional
- Published
- 2006
- Full Text
- View/download PDF
110. Functional Effects of Glycosylation at Asn-579 of the Epidermal Growth Factor Receptor
- Author
-
Whitson, Kristin B., primary, Whitson, Stefanie R., additional, Red-Brewer, Monica L., additional, McCoy, Austin J., additional, Vitali, Angela A., additional, Walker, Francesca, additional, Johns, Terrance G., additional, Beth, Albert H., additional, and Staros, James V., additional
- Published
- 2005
- Full Text
- View/download PDF
111. Enhanced Efficacy of Radioimmunotherapy with 90Y-CHX-A″-DTPA-hu3S193 by Inhibition of Epidermal Growth Factor Receptor (EGFR) Signaling with EGFR Tyrosine Kinase Inhibitor AG1478
- Author
-
Lee, Fook T., primary, Mountain, Angela J., additional, Kelly, Marcus P., additional, Hall, Cathrine, additional, Rigopoulos, Angela, additional, Johns, Terrance G., additional, Smyth, Fiona E., additional, Brechbiel, Martin W., additional, Nice, Edouard C., additional, Burgess, Antony W., additional, and Scott, Andrew M., additional
- Published
- 2005
- Full Text
- View/download PDF
112. Treatment of Human Tumor Xenografts with Monoclonal Antibody 806 in Combination with a Prototypical Epidermal Growth Factor Receptor–Specific Antibody Generates Enhanced Antitumor Activity
- Author
-
Perera, Rushika M., primary, Narita, Yoshitaka, additional, Furnari, Frank B., additional, Gan, Hui K., additional, Murone, Carmel, additional, Ahlkvist, Marika, additional, Luwor, Rodney B., additional, Burgess, Antony W., additional, Stockert, Elisabeth, additional, Jungbluth, Achim A., additional, Old, Lloyd J., additional, Cavenee, Webster K., additional, Scott, Andrew M., additional, and Johns, Terrance G., additional
- Published
- 2005
- Full Text
- View/download PDF
113. The antitumor monoclonal antibody 806 recognizes a high‐mannose form of the EGF receptor that reaches the cell surface when cells over‐express the receptor
- Author
-
Johns, Terrance G., primary, Mellman, Ira, additional, Cartwright, Glenn A., additional, Ritter, Gerd, additional, Old, Lloyd J., additional, Burgess, Antony W., additional, and Scott, Andrew M., additional
- Published
- 2005
- Full Text
- View/download PDF
114. Generation of Anti-Idiotype Antibodies for Application in Clinical Immunotherapy Laboratory Analyses
- Author
-
Liu, Zhanqi, primary, Panousis, Con, additional, Smyth, Fiona E., additional, Murphy, Roger, additional, Wirth, Veronika, additional, Cartwright, Glenn, additional, Johns, Terrance G., additional, and Scott, Andrew M., additional
- Published
- 2003
- Full Text
- View/download PDF
115. Novel monoclonal antibody specific for the de2-7 epidermal growth factor receptor (EGFR) that also recognizes the EGFR expressed in cells containing amplification of the EGFR gene
- Author
-
Johns, Terrance G., primary, Stockert, Elisabeth, additional, Ritter, Gerd, additional, Jungbluth, Achim A., additional, Huang, H-J. Su, additional, Cavenee, Webster K., additional, Smyth, Fiona E., additional, Hall, Cathrine M., additional, Watson, Nadine, additional, Nice, Edouard C., additional, Gullick, William J., additional, Old, Lloyd J., additional, Burgess, Antony W., additional, and Scott, Andrew M., additional
- Published
- 2002
- Full Text
- View/download PDF
116. Binding of complement component Clq to myelin oligodendrocyte glycoprotein: A novel mechanism for regulating CNS inflammation
- Author
-
Johns, Terrance G., primary and Bernard, Claude C.A., additional
- Published
- 1997
- Full Text
- View/download PDF
117. Antibody response in Lewis rats injected with myelin oligodendrocyte glycoprotein derived peptides
- Author
-
ichikawa, Motoki, primary, Johns, Terrance G., additional, Adalmann, Martin, additional, and Bernard, Claude C. A., additional
- Published
- 1996
- Full Text
- View/download PDF
118. The epidermal growth factor receptor variant III ( EGFRv III): where wild things are altered.
- Author
-
Gan, Hui K., Cvrljevic, Anna N., and Johns, Terrance G.
- Subjects
EPIDERMAL growth factor receptors ,GENE expression ,GENE amplification ,GENE rearrangement ,GENETIC mutation ,CANCER invasiveness ,PROTEIN-tyrosine kinases ,GLIOBLASTOMA multiforme - Abstract
The epidermal growth factor receptor ( EGFR) is overexpressed in a variety of human epithelial tumors, often as a consequence of gene amplification. Tumors with EGFR gene amplification frequently contain EGFR gene rearrangements, with the most common extracellular domain mutation being EGFRv III. This mutation leads to a deletion of exons 2-7 of the EGFR gene and renders the mutant receptor incapable of binding any known ligand. Despite this, EGFRv III displays low-level constitutive signaling that is augmented by reduced internalization and downregulation. Aberrant EGFRv III signaling has been shown to be important in driving tumor progression and often correlates with poor prognosis. It is clear that EGFRv III is expressed in a considerable proportion of patients with glioblastoma multiforme ( GBM). The presence of EGFRv III in other tumor types, however, remains controversial. In this review, we critically analyze the evidence for the expression of EGFRv III in a range of tumor types and discuss recent findings pertinent to its function and biology in GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
119. Glioma Surgical Aspirate: A Viable Source of Tumor Tissue for Experimental Research.
- Author
-
Day, Bryan W., Stringer, Brett W., Wilson, John, Jeffree, Rosalind L., Jamieson, Paul R., Ensbey, Kathleen S., Bruce, Zara C., Po Inglis, Allan, Suzanne, Winter, Craig, Tollesson, Gert, Campbell, Scott, Lucas, Peter, Findlay, Wendy, Kadrian, David, Johnson, David, Robertson, Thomas, Johns, Terrance G., Bartlett, Perry F., and Osborne, Geoffrey W.
- Subjects
ACADEMIC medical centers ,CELL lines ,CLINICAL medicine research ,FLOW cytometry ,GLIOMAS ,POLYMERASE chain reaction ,T-test (Statistics) ,DESCRIPTIVE statistics - Abstract
Brain cancer research has been hampered by a paucity of viable clinical tissue of sufficient quality and quantity for experimental research. This has driven researchers to rely heavily on long term cultured cells which no longer represent the cancers from which they were derived. Resection of brain tumors, particularly at the interface between normal and tumorigenic tissue, can be carried out using an ultrasonic surgical aspirator (CUSA) that deposits liquid (blood and irrigation fluid) and resected tissue into a sterile bottle for disposal. To determine the utility of CUSA-derived glioma tissue for experimental research, we collected 48 CUSA specimen bottles from glioma patients and analyzed both the solid tissue fragments and dissociated tumor cells suspended in the liquid waste fraction. We investigated if these fractions would be useful for analyzing tumor heterogeneity, using IHC and multi-parameter flow cytometry; we also assessed culture generation and orthotopic xenograft potential. Both cell sources proved to be an abundant, highly viable source of live tumor cells for cytometric analysis, animal studies and in-vitro studies. Our findings demonstrate that CUSA tissue represents an abundant viable source to conduct experimental research and to carry out diagnostic analyses by flow cytometry or other molecular diagnostic procedures. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
120. Amino Acid Substitutions Alter the Tissue Distribution of Murine Interferon-α1
- Author
-
KERRY, JULIE A., primary, JOHNS, TERRANCE G., additional, LINNANE, ANTHONY W., additional, and CHEETHAM, BRIAN F., additional
- Published
- 1993
- Full Text
- View/download PDF
121. Antiproliferative Potencies of Interferons on Melanoma Cell Lines and Xenografts: Higher Efficacy of Interferon β
- Author
-
Johns, Terrance G., primary, Mackay, Ian R., additional, Callister, Kerry A., additional, Hertzog, Paul J., additional, Devenish, Rodney J., additional, and Linnance, Anthony W., additional
- Published
- 1992
- Full Text
- View/download PDF
122. Phosphorylation of dedicator of cytokinesis 1 (Dock180) at tyrosine residue Y722 by Src family kinases mediates EGFRvIII-driven glioblastoma tumorigenesis.
- Author
-
Haizhong Feng, Bo Hu, Jarzynka, Michael J., Yanxin Li, Keezer, Susan, Johns, Terrance G., Tang, Careen K., Hamilton, Ronald L., Vuori, Kristiina, Nishikawa, Ryo, Sarkaria, Jann N., Tim Fenton, Tao Cheng, Furnari, Frank B., Cavenee, Webster K., and Shi-Yuan Cheng
- Subjects
BRAIN cancer research ,GLIOBLASTOMA multiforme ,EPIDERMAL growth factor ,EPIDERMAL growth factor receptors ,CANCER-related mortality ,GENE therapy - Abstract
Glioblastoma, the most common primary malignant cancer of the brain, is characterized by rapid tumor growth and infiltration of tumor cells throughout the brain. These traits cause glioblastomas to be highly resistant to current therapies with a resultant poor prognosis. Although aberrant oncogenic signaling driven by signature genetic alterations, such as EGF receptor (EGFR) gene amplification and mutation, plays a major role in glioblastoma pathogenesis, the responsible downstream mechanisms remain less clear. Here, we report that EGFRvIII (also known as ?EGFR and de2-7EGFR), a constitutively active EGFR mutant that is frequently co-overexpressed with EGFR in human glioblastoma, promotes tumorigenesis through Src family kinase (SFK)-dependent phosphorylation of Dock180, a guanine nucleotide exchange factor for Rac1. EGFRvIII induces phosphorylation of Dock180 at tyrosine residue 722 (Dock180
Y722 ) and stimulates Rac1-signaling, glioblastoma cell survival and migration. Consistent with this being causal, siRNA knockdown of Dock180 or expression of a Dock180Y722F mutant inhibits each of these EGFRvIII-stimulated activities. The SFKs, Src, Fyn, and Lyn, induce phosphorylation of Dock180Y722 and inhibition of these SFKs by pharmacological inhibitors or shRNA depletion markedly attenuates EGFRvIII-induced phosphorylation of Dock180Y722 , Rac1 activity, and glioblastoma cell migration. Finally, phosphorylated Dock180Y722 is coexpressed with EGFRvIII and phosphorylated SrcY418 in clinical specimens, and such coexpression correlates with an extremely poor survival in glioblastoma patients. These results suggest that targeting the SFK-p-Dock180Y722 -Rac1 signaling pathway may offer a novel therapeutic strategy for glioblastomas with EGFRvIII overexpression. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
123. Immuno-PET Quantitation of de2-7 Epidermal Growth Factor Receptor Expression in Glioma Using 124I-IMP-R4-Labeled Antibody ch806.
- Author
-
Lee, Fook T., O'Keefe, Graeme J., Gan, Hui K., Mountain, Angela J., Jones, Gareth R., Saunder, Timothy H., Sagona, Jessica, Rigopoulos, Angela, Smyth, Fiona E., Johns, Terrance G., Govindan, Serengulam V., Goldenberg, David M., Old, Lloyd J., and Scott, Andrew M.
- Published
- 2010
- Full Text
- View/download PDF
124. A truncated soluble epidermal growth factor receptor-Fc fusion ligand trap displays anti-tumour activity in vivo.
- Author
-
Adams, Timothy E., Koziolek, Eva J., Hoyne, Peter H., Bentley, John D., Lu, Louis, Lovrecz, George, Ward, Colin W., Lee, F. T., Scott, Andrew M., Nash, Andrew D., Rothacker, Julie, Nice, Edouard C., Burgess, Antony W., and Johns, Terrance G.
- Subjects
EPIDERMAL growth factor ,TUMOR growth prevention ,PROTEIN-tyrosine kinase inhibitors ,MONOCLONAL antibodies ,CANCER treatment ,LIGANDS (Biochemistry) ,AMINO acids ,TRANSFORMING growth factors - Abstract
A number of therapeutic strategies including small molecule tyrosine kinase inhibitors and monoclonal antibodies have been developed to target the epidermal growth factor receptor (EGFR) signalling axis for the treatment of cancer. To date, the focus of therapeutic intervention has been the EGFR itself. In the current study, we have assembled and expressed in mammalian cells a soluble, EGFR ligand trap comprising the first 501 amino acids of the mature EGFR sequence fused in-frame with a human IgG Fc domain. The fusion protein, designated sEGFR501.Fc, was secreted as a 220 kDa disulphide-linked homodimer that exhibited high affinity (0.4-8 nM) in competition assays for a number of EGFR ligands including EGF and transforming growth factor-α (TGF-α). sEGFR501.Fc inhibited EGF-stimulated tyrosine phosphorylation of the EGFR of the lung cancer cell lines A549 and H1437, and inhibited and blocked the proliferation of H1437 cells. Administration of sEGFR501.Fc to mice bearing human tumour xenografts derived from A431 (epidermoid carcinoma) and DU145 (androgen-independent prostate cancer) tumour cell lines resulted in modest retardation of tumour growth. These results provide proof-in-principle that using high affinity soluble receptors is a viable method for inhibiting multi-ligand systems, and the impetus to optimize this approach and develop reagents with greater affinity and broader specificity. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
125. Heparin-Binding Epidermal Growth Factor-Like Growth Factor Signaling in Flow-Induced Arterial Remodeling.
- Author
-
Hua Zhang, Sunnarborg, Susan W., McNaughton, K. Kirk, Johns, Terrance G., Lee, David C., and Faber, James E.
- Subjects
RESEARCH ,EPIDERMAL growth factor ,BLOOD flow ,VASCULAR smooth muscle ,CAROTID artery ,HEPARIN - Abstract
The article presents information on research testing whether heparin-binding epidermal growth factor (HB-EGF) contributes to low flow-induced negative hypertropic remodeling (FINR) of a mouse carotid artery. It cites that HB-EGF is activated by reduced endothelial shear stress and stimulates smooth muscle cell proliferation in vitro. The findings suggest that HB-EGF signaling is required for low-induced hypertropic remodeling and may participate in vascular wall disease and remodeling.
- Published
- 2008
- Full Text
- View/download PDF
126. Pre-Surgery Cognitive Performance and Voxel-Based Lesion-Symptom Mapping in Patients with Left High-Grade Glioma.
- Author
-
Guarracino, Ilaria, Ius, Tamara, Baiano, Cinzia, D'Agostini, Serena, Skrap, Miran, Tomasino, Barbara, and Johns, Terrance G.
- Subjects
SURGERY & psychology ,PREOPERATIVE care ,PHONOLOGICAL awareness ,GLIOMAS ,PATIENTS ,BRAIN mapping ,TASK performance ,COMPARATIVE grammar ,NEUROPSYCHOLOGICAL tests ,DESCRIPTIVE statistics ,COGNITIVE testing ,WRITTEN communication ,READING ,DISEASE complications - Abstract
Simple Summary: Studies on pre-surgery effects of high-grade glioma on cognition are few, and investigations mainly used general test batteries without specifically addressing selective neuropsychological functions. We studied the pre-surgery neuropsychological status of 85 patients with high-grade glioma, by administering several cognitive tasks to assess language, memory, executive functions, and praxis. We analyzed their lesion volumes to test anatomo-functional correlations. We found that high-grade glioma involving different sub-areas of the left temporal lobe selectively impacts cognitive functions, especially within the language domain. There was one small overlapping lesion area that was shared by all the tasks we examined, localized in the superior temporal cortex. (1) Background: The literature on the effects of high-grade glioma (HGG) growth on cognition is still scarce. (2) Method: A consecutive series of 85 patients with HGG involving the left hemisphere underwent an extended neuropsychological evaluation prior to surgery. Voxel-based lesion-symptom mapping (VLSM) was used to identify regions related to cognitive performance. (3) Results: The patients' mean level of pre-surgery accuracy was overall high. They showed the greatest difficulties in language with tasks such as naming (42.1% of patients impaired on nouns and 61.4% on verbs), reading (36.3% on words and 32.7% on pseudo-words), auditory lexical decisions (43.9%) and writing (41.3%) being most frequently impaired. VLSM analysis revealed anatomically separated areas along the temporal cortex and the white matter related to impairments on the different tasks, with voxels commonly shared by all tasks restricted to a small region in the ventral superior and middle temporal gyrus. (4) Conclusions: High-grade glioma affects cognition; nonetheless, lesions do not cause diffuse deficits but selectively impact the different language sub-domains along the ventral stream and the dorsal stream for language processing. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
127. Lemming and Vole Cycles: A New Intrinsic Model.
- Author
-
Levay, Elizabeth A., Nasser, Helen, Zelko, Matthew D., Penman, Jim, and Johns, Terrance G.
- Subjects
- *
ANIMAL populations , *PHENOTYPIC plasticity , *FIELD research , *POPULATION density , *LAGOMORPHA - Abstract
It is 100 years since the first paper described the multiannual cycles in Arctic rodents and lagomorphs. The mechanisms driving population cycles in animals like lemmings and voles are complex, often attributed to extrinsic factors, such as food availability and quality, pathogens, parasites and/or predators. While extrinsic factors provide insights into population cycles, none fully explain the phenomenon. We propose an underlying innate, intrinsic mechanism, based on epigenetic regulation, that drives population cycles under harsh arctic conditions. We propose that epigenetically driven phenotypic changes associated with sexual development, growth and behaviour accumulate over time in offspring, eventually producing a phase change from rising population density to eventual population collapse. Under this hypothesis, and unlike previous hypotheses, extrinsic factors modify population cycles but would not be primary drivers. The interaction between our intrinsic cycle and extrinsic factors explains established phenomena like delayed‐density dependence, whereby population growth is controlled by time‐dependent negative feedback. We advocate integrating a century of field research with the latest epigenetic analysis to better understand the drivers of population cycles. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
128. Q-Cell Glioblastoma Resource: Proteomics Analysis Reveals Unique Cell-States Are Maintained in 3D Culture.
- Author
-
D'Souza, Rochelle C. J., Offenhäuser, Carolin, Straube, Jasmin, Baumgartner, Ulrich, Kordowski, Anja, Li, Yuchen, Stringer, Brett W., Alexander, Hamish, Lwin, Zarnie, Inglis, Po-Ling, Jeffree, Rosalind L., Johns, Terrance G., Boyd, Andrew W., and Day, Bryan W.
- Subjects
GLIOBLASTOMA multiforme ,PROTEOMICS ,CENTRAL nervous system ,CENTRAL nervous system viral diseases - Abstract
Glioblastoma (GBM) is a treatment-refractory central nervous system (CNS) tumour, and better therapies to treat this aggressive disease are urgently needed. Primary GBM models that represent the true disease state are essential to better understand disease biology and for accurate preclinical therapy assessment. We have previously presented a comprehensive transcriptome characterisation of a panel (n = 12) of primary GBM models (Q-Cell). We have now generated a systematic, quantitative, and deep proteome abundance atlas of the Q-Cell models grown in 3D culture, representing 6167 human proteins. A recent study has highlighted the degree of functional heterogeneity that coexists within individual GBM tumours, describing four cellular states (MES-like, NPC-like, OPC-like and AC-like). We performed comparative proteomic analysis, confirming a good representation of each of the four cell-states across the 13 models examined. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis identified upregulation of a number of GBM-associated cancer pathway proteins. Bioinformatics analysis, using the OncoKB database, identified a number of functional actionable targets that were either uniquely or ubiquitously expressed across the panel. This study provides an in-depth proteomic analysis of the GBM Q-Cell resource, which should prove a valuable functional dataset for future biological and preclinical investigations. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
129. Activation of ERBB4 in Glioblastoma Can Contribute to Increased Tumorigenicity and Influence Therapeutic Response.
- Author
-
Donoghue, Jacqueline F., Kerr, Lauren T., Alexander, Naomi W., Greenall, Sameer A., Longano, Anthony B., Gottardo, Nicholas G., Wang, Rong, Tabar, Viviane, Adams, Timothy E., Mischel, Paul S., and Johns, Terrance G.
- Subjects
GLIOMAS ,BIOMARKERS ,IMMUNOHISTOCHEMISTRY ,NEOVASCULARIZATION ,ONCOGENES ,PHOSPHORYLATION ,SURVIVAL analysis (Biometry) ,TREATMENT effectiveness ,PREVENTION - Abstract
Glioblastoma (GBM) is often resistant to conventional and targeted therapeutics. ErbB2 Receptor Tyrosine Kinase 4 (ERBB4) is expressed throughout normal brain and is an oncogene in several pediatric brain cancers; therefore, we investigated ERBB4 as a prognostic marker and therapeutic target in GBM. Using RT-qPCR, we quantified mRNA encoding total ERBB4 and known ERBB4 variants in GBM and non-neoplastic normal brain (NNB) samples. Using immunohistochemistry, we characterized the localization of total and phosphorylated ERBB4 (p-ERBB4) and EGFR protein in archived GBM samples and assessed their association with patient survival. Furthermore, we evaluated the effect of ERBB4 phosphorylation on angiogenesis and tumorigenicity in GBM xenograft models. Total ERBB4 mRNA was significantly lower in GBM than NNB samples, with the juxtamembrane JM-a and cytoplasmic CYT-2 variants predominating. ERBB4 protein was ubiquitously expressed in GBM but was not associated with patient survival. However, high p-ERBB4 in 11% of archived GBM samples, independent of p-EGFR, was associated with shorter patient survival (12.0 ± 3.2 months) than was no p-ERBB4 (22.5 ± 9.5 months). Increased ERBB4 activation was also associated with increased proliferation, angiogenesis, tumorigenicity and reduced sensitivity to anti-EGFR treatment in xenograft models. Despite low ERBB4 mRNA in GBM, the functional effects of increased ERBB4 activation identify ERBB4 as a potential prognostic and therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
130. An Anti-VEGF-B Antibody Reduces Abnormal Tumor Vasculature and Enhances the Effects of Chemotherapy.
- Author
-
Janes, Peter W., Parslow, Adam C., Cao, Diana, Rigopoulos, Angela, Lee, Fook-Thean, Gong, Sylvia J., Cartwright, Glenn A., Burvenich, Ingrid J. G., Eriksson, Ulf, Johns, Terrance G., Scott, Fiona E., and Scott, Andrew M.
- Subjects
- *
VASCULAR endothelial growth factors , *BIOLOGICAL models , *RESEARCH funding , *TRANSPLANTATION of organs, tissues, etc. , *LIGANDS (Chemistry) , *OXYGEN , *BEVACIZUMAB , *COLORECTAL cancer , *NUTRITIONAL requirements , *DESCRIPTIVE statistics , *CANCER chemotherapy , *MICE , *ANIMAL experimentation , *GROWTH factors , *COMPARATIVE studies - Abstract
Simple Summary: In order to grow, tumors need nutrients and oxygen, which are supplied by new blood vessels that are often abnormal and leaky. The development of these blood vessels is driven by proteins called growth factors, which bind to receptor proteins on other cells. We show for the first time that an antibody against a vascular growth factor called VEGF-B, when used to treat mice with tumors, reduces the amount of abnormal tumor blood vessels, and inhibits tumor growth, as well as improving the response to chemotherapy. These results suggest that targeting VEGF-B could be a potential approach for anti-cancer therapy, particularly in combination with chemotherapy. The vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are key regulators of blood vessel formation, including in tumors, where their deregulated function can promote the production of aberrant, leaky blood vessels, supporting tumor development. Here we investigated the VEGFR1 ligand VEGF-B, which we demonstrate to be expressed in tumor cells and in tumor stroma and vasculature across a range of tumor types. We examined the anti-VEGF-B-specific monoclonal antibody 2H10 in preclinical xenograft models of breast and colorectal cancer, in comparison with the anti-VEGF-A antibody bevacizumab. Similar to bevacizumab, 2H10 therapy was associated with changes in tumor blood vessels and intra-tumoral diffusion consistent with normalization of the tumor vasculature. Accordingly, treatment resulted in partial inhibition of tumor growth, and significantly improved the response to chemotherapy. Our studies indicate the importance of VEGF-B in tumor growth, and the potential of specific anti-VEGF-B treatment to inhibit tumor development, alone or in combination with established chemotherapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
131. Epidermal Growth Factor Receptor Activation: An Upstream Signal for Transition of Quiescent Astrocytes into Reactive Astrocytes after Neural Injury.
- Author
-
Bin Liu, Huiyi Chen, Johns, Terrance G., and Neufeld, Arthur H.
- Subjects
ASTROCYTES ,EPIDERMAL growth factor ,NEUROPATHY ,PROTEIN-tyrosine kinases ,RETINAL ganglion cells - Abstract
Modulating the behaviors of reactive astrocytes is a potential therapeutic strategy for neurodegenerative diseases. We found that upregulation and activation of the epidermal growth factor receptor (EGFR) occur in astrocytes after different injuries in optic nerves in vivo. Activation of EGFR regulates genes and cellular processes representing most major markers of reactive astrocytes and genes related with glaucomatous optic neuropathy and other neural disorders. These results suggest that activation of EGFR is a common, regulatory pathway that triggers quiescent astrocytes into reactive astrocytes in response to neural injuries in the optic nerve, and perhaps other parts of the CNS. Targeting EGFR activation using an EGFR tyrosine kinase inhibitor prevents the loss of retinal ganglion cells in a model of glaucomatous optic neuropathy. Because these inhibitors are currently used clinically, our results present an approach to reactive astrocytes as a potential new target for the treatment of neurodegenerations. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
132. Antiproliferative Potencies of Interferons on Melanoma Cell Lines and Xenografts: Higher Efficacy of Interferon <IMG SRC="/math/beta.gif" ALT="beta" BORDER="0">
- Author
-
Johns, Terrance G., Mackay, Ian R., Callister, Kerry A., Hertzog, Paul J., Devenish, Rodney J., and Linnance, Anthony W.
- Abstract
Background: Human melanomas have shown only limited responsiveness to clinical therapy with interferon (IFN). Purpose: Our aim was to determine the most effective class of IFN for inhibiting growth of melanoma cells and to establish whether variation exists in response of various cell lines to different IFNs. Methods: We compared the direct antiproliferative effects of the type I IFN α-2b, IFN α-4a, and IFN-β and the type II IFN-γ on eight melanoma cell lines grown in vitro. We did this comparison by determining the concentration of each IFN that resulted in 50% growth inhibition, using the MTT [3-(4,5-dimethyi-2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide] dye uptake method. We also tested IFN α-2a and IFN-β for their ability to inhibit the growth of xenografts of the LiBr melanoma cell line in vivo in nude mice. Receptor binding was determined using [35S]methionine-labeled IFN α-4a, in competition with unlabeled IFN α-2b, IFN α-4a, and IFN-β. Results: The melanoma cell lines differed markedly in their sensitivity to the IFNs tested: Five were sensitive to low concentrations (>30 pM) of IFN-P, only one was sensitive to similar concentrations of IFN α-2b, and none were sensitive to IFN a-4a at concentrations up to 920 pM. For all cell lines, the antiproliferative potency of the type I IFNs was IFN-β > IFN α-2b > IFN α-4a. IFN-γ was less active than IFN-P on all except one of the cell lines. Similarly, IFN-β was more potent than IFN α-2a in inhibiting the growth of the LiBr xenograft in nude mice. Labeled IFN α-4a bound with high specificity in all four melanoma lines tested, and competitive binding experiments showed that the order of binding affinity (IFN-β ≶ IFN α-2b ≶ IFN α-4a) correlated with the order of antiproliferative potency. Conclusion: The finding that melanoma cell lines differ intrinsically in their sensitivity to IFNs may explain differences in clinical response. Our results suggest that IFN-P may be the most effective IFN in the treatment of melanoma, although confirmation will require clinical trials involving large numbers of patients. [J Natl Cancer Inst 84:1185–1190, 1992]
- Published
- 1992
- Full Text
- View/download PDF
133. The dystroglycan receptor maintains glioma stem cells in the vascular niche.
- Author
-
Day, Bryan W., Lathia, Justin D., Bruce, Zara C., D'Souza, Rochelle C. J., Baumgartner, Ulrich, Ensbey, Kathleen S., Lim, Yi Chieh, Stringer, Brett W., Akgül, Seçkin, Offenhäuser, Carolin, Li, Yuchen, Jamieson, Paul R., Smith, Fiona M., Jurd, Courtney L. R., Robertson, Thomas, Inglis, Po-Ling, Lwin, Zarnie, Jeffree, Rosalind L., Johns, Terrance G., and Bhat, Krishna P. L.
- Subjects
- *
STEM cell niches , *CENTRAL nervous system , *GLIOBLASTOMA multiforme , *EXTRACELLULAR matrix - Abstract
Glioblastomas (GBMs) are malignant central nervous system (CNS) neoplasms with a very poor prognosis. They display cellular hierarchies containing self-renewing tumourigenic glioma stem cells (GSCs) in a complex heterogeneous microenvironment. One proposed GSC niche is the extracellular matrix (ECM)-rich perivascular bed of the tumour. Here, we report that the ECM binding dystroglycan (DG) receptor is expressed and functionally glycosylated on GSCs residing in the perivascular niche. Glycosylated αDG is highly expressed and functional on the most aggressive mesenchymal-like (MES-like) GBM tumour compartment. Furthermore, we found that DG acts to maintain an MES-like state via tight control of MAPK activation. Antibody-based blockade of αDG induces robust ERK-mediated differentiation leading to reduced GSC potential. DG was shown to be required for tumour initiation in MES-like GBM, with constitutive loss significantly delaying or preventing tumourigenic potential in-vivo. These findings reveal a central role of the DG receptor, not only as a structural element, but also as a critical factor promoting MES-like GBM and the maintenance of GSCs residing in the perivascular niche. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
134. Changes in cell morphology guide identification of tubulin as the off-target for protein kinase inhibitors.
- Author
-
Hoque, Monira, Abbassi, Ramzi H., Froio, Danielle, Man, Jennifer, Johns, Terrance G., Stringer, Brett W., Day, Bryan W., Pajic, Marina, Kassiou, Michael, and Munoz, Lenka
- Subjects
- *
CELL morphology , *TUBULINS , *TARGETED drug delivery , *PROTEIN kinase inhibitors , *CANCER research - Abstract
In the field of kinase inhibitors for applications in cancer research, tubulin is emerging as a targeted cellular protein that can significantly contribute to their activities. However, investigation of kinase inhibitors beyond the kinome is an area often neglected. Herein, we describe the results of pharmacological studies using drugs targeting kinases, tubulin or both. A key finding is that if cells are treated with a kinase inhibitor unintentionally targeting tubulin, their characteristic shape will diminish within a short timeframe. These changes in cell morphology are not seen when cells are treated with bona fide kinase inhibitors that do not directly target tubulin. Thus, early changes in cell morphology upon treatments are a strong indication that the inhibitor is directly targeting tubulin. Recognizing tubulin as a target of kinase inhibitors will build confidence in the future mechanistic studies using kinase inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
135. Pharmacology of novel small-molecule tubulin inhibitors in glioblastoma cells with enhanced EGFR signalling.
- Author
-
Phoa, Athena F., Browne, Stephen, Gurgis, Fadi M.S., Åkerfeldt, Mia C., Döbber, Alexander, Renn, Christian, Peifer, Christian, Stringer, Brett W., Day, Bryan W., Wong, Chin, Chircop, Megan, Johns, Terrance G., Kassiou, Michael, and Munoz, Lenka
- Subjects
- *
PHARMACOLOGY , *TUBULINS , *GLIOBLASTOMA multiforme , *CANCER cells , *POLYMERIZATION , *APOPTOSIS , *EPIDERMAL growth factor receptors , *MITOGEN-activated protein kinases - Abstract
We recently reported that CMPD1, originally developed as an inhibitor of MK2 activation, primarily inhibits tubulin polymerisation and induces apoptosis in glioblastoma cells. In the present study we provide detailed pharmacological investigation of CMPD1 analogues with improved molecular properties. We determined their anti-cancer efficacy in glioblastoma cells with enhanced EGFR signalling, as deregulated EGFR often leads to chemoresistance. Eight analogues of CMPD1 with varying lipophilicity and basicity were synthesised and tested for efficacy in the cell viability assay using established glioblastoma cell lines and patient-derived primary glioblastoma cells. The mechanism of action for the most potent analogue 15 was determined using MK2 activation and tubulin polymerisation assays, together with the immunofluorescence analysis of the mitotic spindle formation. Apoptosis was analysed by Annexin V staining, imunoblotting analysis of bcl-2 proteins and PARP cleavage. The apoptotic activity of CMPD1 and analogue 15 was comparable across glioblastoma cell lines regardless of the EGFR status. Primary glioblastoma cells of the classical subtype that are characterized by enhanced EGFR activity were most sensitive to the treatment with CMPD1 and 15 . In summary, we present mechanism of action for a novel small molecule tubulin inhibitor, compound 15 that inhibits tubulin polymerisation and mitotic spindle formation, induces degradation of anti-apoptotic bcl-2 proteins and leads to apoptosis of glioblastoma cells. We also demonstrate that the enhanced EGFR activity does not decrease the efficacy of tubulin inhibitors developed in this study. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
136. The crystal structure of myelin oligodendrocyte glycoprotein, a key autoantigen in multiple sclerosis.
- Author
-
Clements, Craig S., Reid, Hugh H., Beddoe, Travis, Tynan, Fleur E., Perugini, Matthew A., Johns, Terrance G., Bernard, Claude C.A., and Rossjohn, Jamie
- Subjects
- *
GLYCOPROTEINS , *PROTEINS , *ANTIGENS - Abstract
Examines the crystal structure of myelin oligodendrocyte glycoprotein, a multiple sclerosis autoantigen. Expression, refolding and crystallization trials on protein; Automated structure comparisons; Dimeric species of the myelin oligodendrocyte glycoprotein.
- Published
- 2003
- Full Text
- View/download PDF
137. Antibody-Functionalized Porous Silicon Nanoparticles for Vectorization of Hydrophobic Drugs
- Author
-
Kevin S. Smith, Emilie Secret, Bahman Delalat, Frédérique Cunin, Nicolas H. Voelcker, Mary-Louise Rogers, Eli Moore, Valentina Dubljevic, Jean-Olivier Durand, Peter J. Macardle, Terrance Grant Johns, Institut Charles Gerhardt Montpellier - Institut de Chimie Moléculaire et des Matériaux de Montpellier (ICGM ICMMM), Ecole Nationale Supérieure de Chimie de Montpellier (ENSCM)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Université Montpellier 2 - Sciences et Techniques (UM2)-Institut de Chimie du CNRS (INC), Secret, Emilie, Smith, Kevin, Dubljevic, Valentina, Moore, Eli, Macardle, Peter, Delalat, Bahman, Rogers, Mary-Louise, Johns, Terrance G, Durand, Jean-Olivier, Cunin, Frédérique, and Voelcker, Nicolas H
- Subjects
neoplasms ,Pharmaceutical Science ,02 engineering and technology ,01 natural sciences ,Diffusion ,Materials Testing ,antineoplastic agents ,antibodies ,ComputingMilieux_MISCELLANEOUS ,medicine.diagnostic_test ,diffusion ,nanocapsules ,Antibodies, Monoclonal ,cell line ,[CHIM.MATE]Chemical Sciences/Material chemistry ,021001 nanoscience & nanotechnology ,3. Good health ,Treatment Outcome ,Drug delivery ,0210 nano-technology ,Hydrophobic and Hydrophilic Interactions ,Porosity ,medicine.drug ,Silicon ,Materials science ,porosity ,Immunocytochemistry ,Biomedical Engineering ,010402 general chemistry ,Flow cytometry ,Biomaterials ,Nanocapsules ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Viability assay ,materials testing ,Bioconjugation ,camptothecin ,Neoplasms, Experimental ,silicon/chemistry ,Antineoplastic Agents, Phytogenic ,Molecular biology ,0104 chemical sciences ,hydrophobic and hydrophilic interactions ,Cell culture ,Cancer cell ,Biophysics ,Camptothecin - Abstract
We describe the preparation of biodegradable porous silicon nanoparticles (pSiNP) functionalized with cancer cell targeting antibodies and loaded with the hydrophobic anti-cancer drug camptothecin. Orientated immobilization of the antibody on the pSiNP is achieved using novel semicarbazide based bioconjugate chemistry. To demonstrate the generality of this targeting approach, the three antibodies MLR2, mAb528 and Rituximab are used, which target neuroblastoma, glioblastoma and B lymphoma cells, respectively. Successful targeting is demonstrated by means of flow cytometry and immunocytochemistry both with cell lines and primary cells. Cell viability assays after incubation with pSiNPs show selective killing of cells expressing the receptor corresponding to the antibody attached on the pSiNP. Refereed/Peer-reviewed
- Published
- 2012
- Full Text
- View/download PDF
138. Ion channel modulator DPI-201-106 significantly enhances antitumor activity of DNA damage response inhibitors in glioblastoma.
- Author
-
Dewdney B, Miranda PJ, Kuchibhotla M, Palanisamy R, Richworth C, Milligan CJ, Ng ZY, Ursich L, Petrou S, Fletcher EV, Daly RJ, Lim Kam Sian TCC, Valvi S, Endersby R, and Johns TG
- Abstract
Background: Glioblastoma, a lethal high-grade glioma, has not seen improvements in clinical outcomes in nearly 30 years. Ion channels are increasingly associated with tumorigenesis, and there are hundreds of brain-penetrant drugs that inhibit ion channels, representing an untapped therapeutic resource. The aim of this exploratory drug study was to screen an ion channel drug library against patient-derived glioblastoma cells to identify new treatments for brain cancer., Methods: Seventy-two ion channel inhibitors were screened in patient-derived glioblastoma cells, and cell viability was determined using the ViaLight Assay. Cell cycle and apoptosis analysis were determined with flow cytometry using PI and Annexin V staining, respectively. Protein and phosphoprotein expression was determined using mass spectrometry and analyzed using gene set enrichment analysis. Kaplan-Meier survival analyses were performed using intracranial xenograft models of GBM6 and WK1 cells., Results: The voltage-gated sodium channel modulator, DPI-201-106, was revealed to reduce glioblastoma cell viability in vitro by inducing cell cycle arrest and apoptosis. Phosphoproteomics indicated that DPI-201-106 may impact DNA damage response (DDR) pathways. Combination treatment of DPI-201-106 with the CHK1 inhibitor prexasertib or the PARP inhibitor niraparib demonstrated synergistic effects in multiple patient-derived glioblastoma cells both in vitro and in intracranial xenograft mouse models, extending survival of glioblastoma-bearing mice., Conclusions: DPI-201-106 enhances the efficacy of DDR inhibitors to reduce glioblastoma growth. As these drugs have already been clinically tested in humans, repurposing DPI-201-106 in novel combinatorial approaches will allow for rapid translation into the clinic., Competing Interests: T.G.J., J.P.F., M.K., and E.V.F. are named inventors on an international patent for the invention of the use of an ion channel inhibitor that inhibits the cell cycle and an inhibitor of CHK1, ATM, Wee1, ATR, or PARP in the treatment or prevention of cancer (PCT/AU2023/050462)., (© The Author(s) 2024. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
139. From signalling pathways to targeted therapies: unravelling glioblastoma's secrets and harnessing two decades of progress.
- Author
-
Dewdney B, Jenkins MR, Best SA, Freytag S, Prasad K, Holst J, Endersby R, and Johns TG
- Subjects
- Adult, Humans, Child, Signal Transduction, Brain pathology, Precision Medicine, Glioblastoma drug therapy, Glioblastoma genetics, Brain Neoplasms drug therapy, Brain Neoplasms genetics
- Abstract
Glioblastoma, a rare, and highly lethal form of brain cancer, poses significant challenges in terms of therapeutic resistance, and poor survival rates for both adult and paediatric patients alike. Despite advancements in brain cancer research driven by a technological revolution, translating our understanding of glioblastoma pathogenesis into improved clinical outcomes remains a critical unmet need. This review emphasises the intricate role of receptor tyrosine kinase signalling pathways, epigenetic mechanisms, and metabolic functions in glioblastoma tumourigenesis and therapeutic resistance. We also discuss the extensive efforts over the past two decades that have explored targeted therapies against these pathways. Emerging therapeutic approaches, such as antibody-toxin conjugates or CAR T cell therapies, offer potential by specifically targeting proteins on the glioblastoma cell surface. Combination strategies incorporating protein-targeted therapy and immune-based therapies demonstrate great promise for future clinical research. Moreover, gaining insights into the role of cell-of-origin in glioblastoma treatment response holds the potential to advance precision medicine approaches. Addressing these challenges is crucial to improving outcomes for glioblastoma patients and moving towards more effective precision therapies., (© 2023. West China Hospital, Sichuan University.)
- Published
- 2023
- Full Text
- View/download PDF
140. A surgically optimized intraoperative poly(I:C)-releasing hydrogel prevents cancer recurrence.
- Author
-
Rwandamuriye FX, Evans CW, Wylie B, Norret M, Vitali B, Ho D, Nguyen D, Roper EA, Wang T, Hepburn MS, Sanderson RW, Pfirrmann M, Fear VS, Forbes CA, Wyatt K, Ryan AL, Johns TG, Phillips MB, Hodder R, Leslie C, Kennedy BF, Zemek RM, Iyer KS, and Lesterhuis WJ
- Subjects
- Mice, Animals, Dogs, Immunotherapy, Disease Models, Animal, Tumor Microenvironment, Hydrogels therapeutic use, Neoplasm Recurrence, Local prevention & control
- Abstract
Recurrences frequently occur following surgical removal of primary tumors. In many cancers, adjuvant therapies have limited efficacy. Surgery provides access to the tumor microenvironment, creating an opportunity for local therapy, in particular immunotherapy, which can induce local and systemic anti-cancer effects. Here, we develop a surgically optimized biodegradable hyaluronic acid-based hydrogel for sustained intraoperative delivery of Toll-like receptor 3 agonist poly(I:C) and demonstrate that it significantly reduces tumor recurrence after surgery in multiple mouse models. Mechanistically, poly(I:C) induces a transient interferon alpha (IFNα) response, reshaping the tumor/wound microenvironment by attracting inflammatory monocytes and depleting regulatory T cells. We demonstrate that a pre-existing IFN signature predicts response to the poly(I:C) hydrogel, which sensitizes tumors to immune checkpoint therapy. The safety, immunogenicity, and surgical feasibility are confirmed in a veterinary trial in canine soft tissue tumors. The surgically optimized poly(I:C)-loaded hydrogel provides a safe and effective approach to prevent cancer recurrence., Competing Interests: Declaration of interests We declare a patent application pertaining to aspects of this work. B.F.K. discloses a financial interest in OncoRes Medical, a company developing QME for use in surgery. W.J.L. declares consultancy for Douglas Pharmaceuticals and MSD and research funding from Douglas Pharmaceuticals, AstraZeneca, and ENA Therapeutics., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
141. Preventing recurrence in Sonic Hedgehog Subgroup Medulloblastoma using the OLIG2 inhibitor CT-179.
- Author
-
Li Y, Lim C, Dismuke T, Malawsky DS, Oasa S, Bruce ZC, Offenhäuser C, Baumgartner U, D'Souza RCJ, Edwards SL, French JD, Ock LSH, Nair S, Sivakumaran H, Harris L, Tikunov AP, Hwang D, Del Mar Alicea Pauneto C, Maybury M, Hassall T, Wainwright B, Kesari S, Stein G, Piper M, Johns TG, Sokolsky-Papkov M, Terenius L, Vukojević V, Gershon TR, and Day BW
- Abstract
Recurrence is the primary life-threatening complication for medulloblastoma (MB). In Sonic Hedgehog (SHH)-subgroup MB, OLIG2-expressing tumor stem cells drive recurrence. We investigated the anti-tumor potential of the small-molecule OLIG2 inhibitor CT-179, using SHH-MB patient-derived organoids, patient-derived xenograft (PDX) tumors and mice genetically-engineered to develop SHH-MB. CT-179 disrupted OLIG2 dimerization, DNA binding and phosphorylation and altered tumor cell cycle kinetics in vitro and in vivo , increasing differentiation and apoptosis. CT-179 increased survival time in GEMM and PDX models of SHH-MB, and potentiated radiotherapy in both organoid and mouse models, delaying post-radiation recurrence. Single cell transcriptomic studies (scRNA-seq) confirmed that CT-179 increased differentiation and showed that tumors up-regulated Cdk4 post-treatment. Consistent with increased CDK4 mediating CT-179 resistance, CT-179 combined with CDK4/6 inhibitor palbociclib delayed recurrence compared to either single-agent. These data show that targeting treatment-resistant MB stem cell populations by adding the OLIG2 inhibitor CT-179 to initial MB treatment can reduce recurrence., Competing Interests: Additional Declarations: Yes there is potential Competing Interest. G.S. is Chief Executive Officer, Chairman of the Board, and has equity ownership at Curtana Pharmaceuticals. S.K. is a member of the Board and has equity ownership at Curtana Pharmaceuticals. The other co-authors have no competing interests to report.
- Published
- 2023
- Full Text
- View/download PDF
142. Clinical Trials in the Brain Tumour Population: Challenges and Strategies for the Future.
- Author
-
Gan HK, Day BW, Harrup R, Johns TG, Lwin Z, Scott AM, Sim HW, and Koh ES
- Subjects
- Humans, Clinical Trials as Topic, Brain Neoplasms therapy
- Abstract
Purpose of Review: This review identifies challenges and barriers to successful development of drugs in neuro-oncology trials at the preclinical, clinical and translational stages that we believe has contributed to poor outcomes for patients over the last 30 years., Recent Findings: Several key strategies have been proposed by leading groups to address these and improve patient outcomes. Better preclinical testing using more sophisticated and clinically relevant models is needed. A greater focus on assessing blood-brain barrier penetrance and targeting key biological processes such as tumour heterogeneity and immune response is vital. Adopting innovative trial designs permitting faster results and addressing key issues (including molecular heterogeneity and combinatorial approaches) is highly desirable. A stronger translational focus is also clearly needed. Implementation of these strategies is already starting to occur. Maintaining and increasing these novel approaches will require coordinated efforts between clinicians, scientists, industry and funding/regulator bodies., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
143. Global phosphoproteomics reveals DYRK1A regulates CDK1 activity in glioblastoma cells.
- Author
-
Recasens A, Humphrey SJ, Ellis M, Hoque M, Abbassi RH, Chen B, Longworth M, Needham EJ, James DE, Johns TG, Day BW, Kassiou M, Yang P, and Munoz L
- Abstract
Both tumour suppressive and oncogenic functions have been reported for dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A). Herein, we performed a detailed investigation to delineate the role of DYRK1A in glioblastoma. Our phosphoproteomic and mechanistic studies show that DYRK1A induces degradation of cyclin B by phosphorylating CDC23, which is necessary for the function of the anaphase-promoting complex, a ubiquitin ligase that degrades mitotic proteins. DYRK1A inhibition leads to the accumulation of cyclin B and activation of CDK1. Importantly, we established that the phenotypic response of glioblastoma cells to DYRK1A inhibition depends on both retinoblastoma (RB) expression and the degree of residual DYRK1A activity. Moderate DYRK1A inhibition leads to moderate cyclin B accumulation, CDK1 activation and increased proliferation in RB-deficient cells. In RB-proficient cells, cyclin B/CDK1 activation in response to DYRK1A inhibition is neutralized by the RB pathway, resulting in an unchanged proliferation rate. In contrast, complete DYRK1A inhibition with high doses of inhibitors results in massive cyclin B accumulation, saturation of CDK1 activity and cell cycle arrest, regardless of RB status. These findings provide new insights into the complexity of context-dependent DYRK1A signalling in cancer cells.
- Published
- 2021
- Full Text
- View/download PDF
144. Simultaneous targeting of DNA replication and homologous recombination in glioblastoma with a polyether ionophore.
- Author
-
Lim YC, Ensbey KS, Offenhäuser C, D'souza RCJ, Cullen JK, Stringer BW, Quek H, Bruce ZC, Kijas A, Cianfanelli V, Mahboubi B, Smith F, Jeffree RL, Wiesmüeller L, Wiegmans AP, Bain A, Lombard FJ, Roberts TL, Khanna KK, Lavin MF, Kim B, Hamerlik P, Johns TG, Coster MJ, Boyd AW, and Day BW
- Subjects
- Animals, Autophagy drug effects, Drug Discovery, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Xenograft Model Antitumor Assays, Brain Neoplasms pathology, DNA Replication drug effects, Glioblastoma pathology, Pyrans pharmacology, Recombinational DNA Repair drug effects
- Abstract
Background: Despite significant endeavor having been applied to identify effective therapies to treat glioblastoma (GBM), survival outcomes remain intractable. The greatest nonsurgical benefit arises from radiotherapy, though tumors typically recur due to robust DNA repair. Patients could therefore benefit from therapies with the potential to prevent DNA repair and synergize with radiotherapy. In this work, we investigated the potential of salinomycin to enhance radiotherapy and further uncover novel dual functions of this ionophore to induce DNA damage and prevent repair., Methods: In vitro primary GBM models and ex vivo GBM patient explants were used to determine the mechanism of action of salinomycin by immunoblot, flow cytometry, immunofluorescence, immunohistochemistry, and mass spectrometry. In vivo efficacy studies were performed using orthotopic GBM animal xenograft models. Salinomycin derivatives were synthesized to increase drug efficacy and explore structure-activity relationships., Results: Here we report novel dual functions of salinomycin. Salinomycin induces toxic DNA lesions and prevents subsequent recovery by targeting homologous recombination (HR) repair. Salinomycin appears to target the more radioresistant GBM stem cell-like population and synergizes with radiotherapy to significantly delay tumor formation in vivo. We further developed salinomycin derivatives which display greater efficacy in vivo while retaining the same beneficial mechanisms of action., Conclusion: Our findings highlight the potential of salinomycin to induce DNA lesions and inhibit HR to greatly enhance the effect of radiotherapy. Importantly, first-generation salinomycin derivatives display greater efficacy and may pave the way for clinical testing of these agents., (© The Author(s) 2019. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2020
- Full Text
- View/download PDF
145. Publisher Correction: A reference collection of patient-derived cell line and xenograft models of proneural, classical and mesenchymal glioblastoma.
- Author
-
Stringer BW, Day BW, D'Souza RCJ, Jamieson PR, Ensbey KS, Bruce ZC, Lim YC, Goasdoué K, Offenhäuser C, Akgül S, Allan S, Robertson T, Lucas P, Tollesson G, Campbell S, Winter C, Do H, Dobrovic A, Inglis PL, Jeffree RL, Johns TG, and Boyd AW
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
146. Whole genome and biomarker analysis of patients with recurrent glioblastoma on bevacizumab: A subset analysis of the CABARET trial.
- Author
-
Olafson LR, Siddell AH, Field KM, Byrnes M, Rapkins RW, Ng B, Nixdorf S, Barnes EH, Johns TG, Yip S, Simes J, Nowak AK, Rosenthal MA, and McDonald KL
- Subjects
- Adult, Aged, Biomarkers, Tumor genetics, Brain Neoplasms metabolism, Chromosomes, Human, Pair 19 genetics, Clinical Trials, Phase II as Topic, Cohort Studies, Female, Glioblastoma metabolism, Humans, Male, Middle Aged, Randomized Controlled Trials as Topic, Retrospective Studies, Antineoplastic Agents therapeutic use, Bevacizumab therapeutic use, Biomarkers, Tumor analysis, Brain Neoplasms drug therapy, Glioblastoma drug therapy
- Abstract
The CABARET trial (ACTRN12610000915055) reported no difference in overall survival (OS) between patients with recurrent glioblastoma (GBM) randomized to either bevacizumab monotherapy or bevacizumab plus carboplatin. However, a subset of patients showed durable responses and prolonged survival, with recorded survival times of over 30 months in five of 122 patients (4%). Patient selection for bevacizumab therapy would be enhanced if a predictive biomarker of response or survival could be identified; this biomarker sub-study attempted to identify novel biomarkers. Patients who opted to participate in this sub-study and who had adequate biospecimens for analysis (n = 54) were retrospectively evaluated for the expression of a series of tumor proteins. Immunohistochemistry (IHC) was used to measure the expression of 19 proteins previously implicated in cancer treatment response to bevacizumab. MGMT promoter methylation was also assessed. Tumor DNA from five patients with outlying survival duration ('poor' and 'exceptional' survivors) was subjected to whole genome sequencing (WGS). No single protein expression level, including VEGF-A, predicted OS in the cohort. WGS of poor and exceptional survivors identified a gain in Chromosome 19 that was exclusive to the exceptional survivors. Validation of this finding requires examination of a larger independent cohort., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
147. A reference collection of patient-derived cell line and xenograft models of proneural, classical and mesenchymal glioblastoma.
- Author
-
Stringer BW, Day BW, D'Souza RCJ, Jamieson PR, Ensbey KS, Bruce ZC, Lim YC, Goasdoué K, Offenhäuser C, Akgül S, Allan S, Robertson T, Lucas P, Tollesson G, Campbell S, Winter C, Do H, Dobrovic A, Inglis PL, Jeffree RL, Johns TG, and Boyd AW
- Subjects
- Aged, Aged, 80 and over, Animals, Female, Humans, Male, Mice, Inbred NOD, Mice, SCID, Middle Aged, Brain Neoplasms pathology, Cell Line, Tumor, Glioblastoma pathology, Neoplasm Transplantation
- Abstract
Low-passage, serum-free cell lines cultured from patient tumour tissue are the gold-standard for preclinical studies and cellular investigations of glioblastoma (GBM) biology, yet entrenched, poorly-representative cell line models are still widely used, compromising the significance of much GBM research. We submit that greater adoption of these critical resources will be promoted by the provision of a suitably-sized, meaningfully-described reference collection along with appropriate tools for working with them. Consequently, we present a curated panel of 12 readily-usable, genetically-diverse, tumourigenic, patient-derived, low-passage, serum-free cell lines representing the spectrum of molecular subtypes of IDH-wildtype GBM along with their detailed phenotypic characterisation plus a bespoke set of lentiviral plasmids for bioluminescent/fluorescent labelling, gene expression and CRISPR/Cas9-mediated gene inactivation. The cell lines and all accompanying data are readily-accessible via a single website, Q-Cell (qimrberghofer.edu.au/q-cell/) and all plasmids are available from Addgene. These resources should prove valuable to investigators seeking readily-usable, well-characterised, clinically-relevant, gold-standard models of GBM.
- Published
- 2019
- Full Text
- View/download PDF
148. Differential Sensitivity of Human Hepatocellular Carcinoma Xenografts to an IGF-II Neutralizing Antibody May Involve Activated STAT3.
- Author
-
Greenall SA, Donoghue J, Johns TG, and Adams TE
- Abstract
Hepatocellular carcinoma (HCC) is highly refractory to current therapeutics used in the clinic. DX-2647, a recombinant human antibody, potently neutralizes the action of insulin-like growth factor-II (IGF-II), a ligand for three cell-surface receptors (IGF-IR, insulin receptor A and B isoforms, and the cation-independent mannose-6-phosphate receptor) which is overexpressed in primary human HCC. DX-2647 impaired the growth of tumor xenografts of the HCC cell line, Hep3B; however, xenografts of the HCC cell line, HepG2, were largely unresponsive to DX-2647 treatment. Analysis of a number of aspects of the IGF signaling axis in both cell lines did not reveal any significant differences between the two. However, while DX-2647 abolished phospho (p)-IGF-IR, p-IR and p-AKT signaling in both cell lines, HepG2 showed high levels of p-STAT3, which was unaffected by DX-2647 treatment and was absent from the Hep3B cell line. The driver of p-STAT3 was found to be a secreted cytokine, and treatment of HepG2 cells with a pan- JAK kinase inhibitor resulted in a loss of p-STAT3. These findings implicate the activation of STAT3 as one pathway that may mediate resistance to IGF-II-targeted therapy in HCC., (Copyright © 2018 BAYLOR COLLEGE OF MEDICINE. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
149. A Pre-Clinical Assessment of the Pan-ERBB Inhibitor Dacomitinib in Pediatric and Adult Brain Tumors.
- Author
-
Endersby R, Whitehouse J, Hii H, Greenall SA, Johns TG, and Gottardo NG
- Subjects
- Adult, Animals, Brain Neoplasms metabolism, Cell Line, Tumor, Child, Female, Glioblastoma drug therapy, Glioblastoma metabolism, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Signal Transduction drug effects, Brain Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors, Quinazolinones pharmacology
- Abstract
Glioblastoma in adults, and medulloblastoma and pineoblastoma that mainly affect children, are aggressive brain tumors. The survival for patients with glioblastoma remains dismal. While the cure rate for medulloblastoma exceeds 70%, this figure has stagnated over the past few decades and survivors still contend with significant long-term debilitating side effects. The prognosis for pineoblastoma is age-dependent, with little chance of a cure for children younger than three years. More effective molecularly targeted strategies are urgently required to treat these cancers. Hyper-activation of epidermal growth factor receptor (EGFR) signaling is characteristic of several different classes of human cancers, including a subset of glioblastoma and medulloblastoma. This has provided the impetus for the development of a suite of EGFR pathway blockers, including second generation irreversible inhibitors, such as dacomitinib. We have developed a comprehensive drug evaluation pipeline, including in vitro interaction analyses and orthotopic xenograft mouse models, to address the efficacy of drugs for brain tumor treatment, enabling the exclusion of potentially ineffective treatments and prioritization of truly beneficial novel treatments for clinical trial. We used this system to examine the effects of dacomitinib as a single agent, or in combination with conventional chemotherapeutics, on the growth of human adult and pediatric brain tumor cell lines. Dacomitinib inhibited EGFR or EGFRvIII activity in vitro in all three tumor types tested, and as a single agent induced a modest increase in survival time for mice bearing glioblastoma, which accurately predicted human clinical trial data. For pediatric medulloblastoma, dacomitinib blocked EGFR/HER signalling in orthotopic xenografts and extended median survival as a single agent, however was antagonistic when used in combination with standard frontline medulloblastoma chemotherapies. The findings caution against the use of dacomitinib for pediatric brain tumor clinical trials., (Crown Copyright © 2018. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
150. Dianthin-30 or gelonin versus monomethyl auristatin E, each configured with an anti-calcitonin receptor antibody, are differentially potent in vitro in high-grade glioma cell lines derived from glioblastoma.
- Author
-
Gilabert-Oriol R, Furness SGB, Stringer BW, Weng A, Fuchs H, Day BW, Kourakis A, Boyd AW, Hare DL, Thakur M, Johns TG, and Wookey PJ
- Subjects
- Antibodies, Monoclonal immunology, Cell Line, Tumor, Humans, Receptors, Calcitonin immunology, Tumor Cells, Cultured, Antibodies, Monoclonal pharmacology, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Oligopeptides pharmacology, Receptors, Calcitonin antagonists & inhibitors, Ribosome Inactivating Proteins, Type 1 pharmacology
- Abstract
We have reported that calcitonin receptor (CTR) is widely expressed in biopsies from the lethal brain tumour glioblastoma by malignant glioma and brain tumour-initiating cells (glioma stem cells) using anti-human CTR antibodies. A monoclonal antibody against an epitope within the extracellular domain of CTR was raised (mAb2C4) and chemically conjugated to either plant ribosome-inactivating proteins (RIPs) dianthin-30 or gelonin, or the drug monomethyl auristatin E (MMAE), and purified. In the high-grade glioma cell line (HGG, representing glioma stem cells) SB2b, in the presence of the triterpene glycoside SO1861, the EC
50 for mAb2C4:dianthin was 10.0 pM and for mAb2C4:MMAE [antibody drug conjugate (ADC)] 2.5 nM, 250-fold less potent. With the cell line U87MG, in the presence of SO1861, the EC50 for mAb2C4:dianthin was 20 pM, mAb2C4:gelonin, 20 pM, compared to the ADC (6.3 nM), which is >300 less potent. Several other HGG cell lines that express CTR were tested and the efficacies of mAb2C4:RIP (dianthin or gelonin) were similar. Co-administration of the enhancer SO1861 purified from plants enhances lysosomal escape. Enhancement with SO1861 increased potency of the immunotoxin (>3 log values) compared to the ADC (1 log). The uptake of antibody was demonstrated with the fluorescent conjugate mAb2C4:Alexa Fluor 568, and the release of dianthin-30:Alexa Fluor488 into the cytosol following addition of SO1861 supports our model. These data demonstrate that the immunotoxins are highly potent and that CTR is an effective target expressed by a large proportion of HGG cell lines representative of glioma stem cells and isolated from individual patients.- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.