36 results on '"Jeffrey Wojton"'
Search Results
2. Supplementary Figures from Bortezomib Treatment Sensitizes Oncolytic HSV-1–Treated Tumors to NK Cell Immunotherapy
- Author
-
Balveen Kaur, Matthew Old, Michael A. Caligiuri, Jianhua Yu, Carlo M. Croce, Jianying Zhang, Jun-Ge Yu, Michael T. Lotze, Tae Jin Lee, Theresa Relation, Brian S. Hurwitz, Jeffrey Wojton, Tejaswini Nallanagulagari, Hongsheng Dai, Chelsea Bolyard, Alena Cristina Jaime-Ramirez, and Ji Young Yoo
- Abstract
Supplementary Figure S1. Effects of combination treatment of both bortezomib and oHSV on cancer cell killing. Supplementary Figure S2. Necrostatin-1 (Nec-1) treatment and RIPK1 knock down did not reduce the bortezomib-induced increase in virus replication. Supplementary Figure S3. Single treatment with bortezomib or oHSV enhances NK cell mediated glioma killing. Supplementary Figure S4. Combination treatment with bortezomib and oHSV enhance mice survival in vivo.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 2 from BAI1 Orchestrates Macrophage Inflammatory Response to HSV Infection—Implications for Oncolytic Viral Therapy
- Author
-
Balveen Kaur, Jianhua Yu, Michael A. Caligiuri, Jonathan P. Godbout, Erwin G. Van Meir, Dan Zhu, Matthew Old, Jianying Zhang, Flavia Pichiorri, Pete Pow-anpongkul, Christopher Alvarez-Breckenridge, Jonathan Smith, Bo Xu, Maninder Khosla, Samuel Dubin, Jun-Ge Yu, Jeffrey Wojton, Eric S. Wohleb, Ji Young Yoo, Jayson Hardcastle, Yeshavanth Banasavadi-Siddegowda, W. Hans Meisen, and Chelsea Bolyard
- Abstract
Co-culture model of endogenous Vstat120 impact on microglia response to oHSV-infected glioma.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure Legend from Bortezomib Treatment Sensitizes Oncolytic HSV-1–Treated Tumors to NK Cell Immunotherapy
- Author
-
Balveen Kaur, Matthew Old, Michael A. Caligiuri, Jianhua Yu, Carlo M. Croce, Jianying Zhang, Jun-Ge Yu, Michael T. Lotze, Tae Jin Lee, Theresa Relation, Brian S. Hurwitz, Jeffrey Wojton, Tejaswini Nallanagulagari, Hongsheng Dai, Chelsea Bolyard, Alena Cristina Jaime-Ramirez, and Ji Young Yoo
- Abstract
Supplementary Figure Legend
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure Legend from Copper Chelation Enhances Antitumor Efficacy and Systemic Delivery of Oncolytic HSV
- Author
-
Balveen Kaur, Matthew Old, Joseph C. Glorioso, E. Antonio Chiocca, Theodoros N. Teknos, Andrew P. Mazar, Kimerly Powell, Jun-Ge Yu, Christopher A. Alvarez-Breckenridge, Anna Bratasz, Azeem Kaka, Jeffrey Wojton, Amy Haseley, Jason Pradarelli, and Ji Young Yoo
- Abstract
PDF file, 77K.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1. from BAI1 Orchestrates Macrophage Inflammatory Response to HSV Infection—Implications for Oncolytic Viral Therapy
- Author
-
Balveen Kaur, Jianhua Yu, Michael A. Caligiuri, Jonathan P. Godbout, Erwin G. Van Meir, Dan Zhu, Matthew Old, Jianying Zhang, Flavia Pichiorri, Pete Pow-anpongkul, Christopher Alvarez-Breckenridge, Jonathan Smith, Bo Xu, Maninder Khosla, Samuel Dubin, Jun-Ge Yu, Jeffrey Wojton, Eric S. Wohleb, Ji Young Yoo, Jayson Hardcastle, Yeshavanth Banasavadi-Siddegowda, W. Hans Meisen, and Chelsea Bolyard
- Abstract
Ewing sarcoma subcutaneous tumors stained for CD68+ macrophages, with hematoxylin counterstain.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table 2. from BAI1 Orchestrates Macrophage Inflammatory Response to HSV Infection—Implications for Oncolytic Viral Therapy
- Author
-
Balveen Kaur, Jianhua Yu, Michael A. Caligiuri, Jonathan P. Godbout, Erwin G. Van Meir, Dan Zhu, Matthew Old, Jianying Zhang, Flavia Pichiorri, Pete Pow-anpongkul, Christopher Alvarez-Breckenridge, Jonathan Smith, Bo Xu, Maninder Khosla, Samuel Dubin, Jun-Ge Yu, Jeffrey Wojton, Eric S. Wohleb, Ji Young Yoo, Jayson Hardcastle, Yeshavanth Banasavadi-Siddegowda, W. Hans Meisen, and Chelsea Bolyard
- Abstract
List of genes differentially induced ({greater than or equal to} 1.5 fold) in macrophages cultured with rHSVQ1 (Q) vs. RAMBO (R) infected glioma cells (UI, uninfected).
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 5 from Copper Chelation Enhances Antitumor Efficacy and Systemic Delivery of Oncolytic HSV
- Author
-
Balveen Kaur, Matthew Old, Joseph C. Glorioso, E. Antonio Chiocca, Theodoros N. Teknos, Andrew P. Mazar, Kimerly Powell, Jun-Ge Yu, Christopher A. Alvarez-Breckenridge, Anna Bratasz, Azeem Kaka, Jeffrey Wojton, Amy Haseley, Jason Pradarelli, and Ji Young Yoo
- Abstract
PDF file, 157K, Ex vivo serum rescue assay.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 3 from Copper Chelation Enhances Antitumor Efficacy and Systemic Delivery of Oncolytic HSV
- Author
-
Balveen Kaur, Matthew Old, Joseph C. Glorioso, E. Antonio Chiocca, Theodoros N. Teknos, Andrew P. Mazar, Kimerly Powell, Jun-Ge Yu, Christopher A. Alvarez-Breckenridge, Anna Bratasz, Azeem Kaka, Jeffrey Wojton, Amy Haseley, Jason Pradarelli, and Ji Young Yoo
- Abstract
PDF file, 304K, Effect of copper on virus therapy for glioma cells.
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Copper Chelation Enhances Antitumor Efficacy and Systemic Delivery of Oncolytic HSV
- Author
-
Balveen Kaur, Matthew Old, Joseph C. Glorioso, E. Antonio Chiocca, Theodoros N. Teknos, Andrew P. Mazar, Kimerly Powell, Jun-Ge Yu, Christopher A. Alvarez-Breckenridge, Anna Bratasz, Azeem Kaka, Jeffrey Wojton, Amy Haseley, Jason Pradarelli, and Ji Young Yoo
- Abstract
Purpose: Copper in serum supports angiogenesis and inhibits replication of wild-type HSV-1. Copper chelation is currently being investigated as an antiangiogenic and antineoplastic agent in patients diagnosed with cancer. Herpes simplex virus–derived oncolytic viruses (oHSV) are being evaluated for safety and efficacy in patients, but several host barriers limit their efficacy. Here, we tested whether copper inhibits oHSV infection and replication and whether copper chelation would augment therapeutic efficacy of oHSV.Experimental Design: Subcutaneous and intracranial tumor-bearing mice were treated with oHSV ± ATN-224 to evaluate tumor burden and survival. Virus replication and cell killing was measured in the presence or absence of the copper chelating agent ATN-224 and in the presence or absence of copper in vitro. Microvessel density and changes in perfusion were evaluated by immunohistochemistry and dynamic contrast enhanced MRI (DCE-MRI). Serum stability of oHSV was measured in mice fed with ATN-224. Tumor-bearing mice were injected intravenously with oHSV; tumor burden and amount of virus in tumor tissue were evaluated.Results: Combination of systemic ATN-224 and oHSV significantly reduced tumor growth and prolonged animal survival. Immunohistochemistry and DCE-MRI imaging confirmed that ATN-224 reduced oHSV-induced blood vessel density and vascular leakage. Copper at physiologically relevant concentrations inhibited oHSV replication and glioma cell killing, and this effect was rescued by ATN-224. ATN-224 increased serum stability of oHSV and enhanced the efficacy of systemic delivery.Conclusion: This study shows that combining ATN-224 with oHSV significantly increased serum stability of oHSV and greatly enhanced its replication and antitumor efficacy. Clin Cancer Res; 18(18); 4931–41. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 4 from Copper Chelation Enhances Antitumor Efficacy and Systemic Delivery of Oncolytic HSV
- Author
-
Balveen Kaur, Matthew Old, Joseph C. Glorioso, E. Antonio Chiocca, Theodoros N. Teknos, Andrew P. Mazar, Kimerly Powell, Jun-Ge Yu, Christopher A. Alvarez-Breckenridge, Anna Bratasz, Azeem Kaka, Jeffrey Wojton, Amy Haseley, Jason Pradarelli, and Ji Young Yoo
- Abstract
PDF file, 65K, ATN-224 by itself does not affect glioma cell viability or oHSV efficacy in the absence of copper.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table 1 from BAI1 Orchestrates Macrophage Inflammatory Response to HSV Infection—Implications for Oncolytic Viral Therapy
- Author
-
Balveen Kaur, Jianhua Yu, Michael A. Caligiuri, Jonathan P. Godbout, Erwin G. Van Meir, Dan Zhu, Matthew Old, Jianying Zhang, Flavia Pichiorri, Pete Pow-anpongkul, Christopher Alvarez-Breckenridge, Jonathan Smith, Bo Xu, Maninder Khosla, Samuel Dubin, Jun-Ge Yu, Jeffrey Wojton, Eric S. Wohleb, Ji Young Yoo, Jayson Hardcastle, Yeshavanth Banasavadi-Siddegowda, W. Hans Meisen, and Chelsea Bolyard
- Abstract
List of genes significantly up-regulated (>1.5 fold) in macrophages upon culture with glioma cells infected with RAMBO (R) or rHSVQ1 (Q) relative to uninfected (UI) cells.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 1 from Copper Chelation Enhances Antitumor Efficacy and Systemic Delivery of Oncolytic HSV
- Author
-
Balveen Kaur, Matthew Old, Joseph C. Glorioso, E. Antonio Chiocca, Theodoros N. Teknos, Andrew P. Mazar, Kimerly Powell, Jun-Ge Yu, Christopher A. Alvarez-Breckenridge, Anna Bratasz, Azeem Kaka, Jeffrey Wojton, Amy Haseley, Jason Pradarelli, and Ji Young Yoo
- Abstract
PDF file, 46K, Quantification of color coded Ktrans and ve parametric images following DCE-MRI.
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Bortezomib Treatment Sensitizes Oncolytic HSV-1–Treated Tumors to NK Cell Immunotherapy
- Author
-
Balveen Kaur, Matthew Old, Michael A. Caligiuri, Jianhua Yu, Carlo M. Croce, Jianying Zhang, Jun-Ge Yu, Michael T. Lotze, Tae Jin Lee, Theresa Relation, Brian S. Hurwitz, Jeffrey Wojton, Tejaswini Nallanagulagari, Hongsheng Dai, Chelsea Bolyard, Alena Cristina Jaime-Ramirez, and Ji Young Yoo
- Abstract
Purpose: Both the proteasome inhibitor bortezomib and an oncolytic herpes simplex virus-1 (oHSV)–expressing GM-CSF are currently FDA approved. Although proteasome blockade can increase oHSV replication, immunologic consequences, and consequent immunotherapy potential are unknown. In this study, we investigated the impact of bortezomib combined with oHSV on tumor cell death and sensitivity to natural killer (NK) cell immunotherapy.Experimental Design: Western blot, flow cytometry, and caspase 3/7 activity assays were used to evaluate the induction of apoptosis/autophagy and/or necroptotic cell death. Cellular and mitochondrial reactive oxygen species (ROS) production was measured using CellROX and MitoSOX. Inhibitors/shRNA–targeting ROS, JNK and RIP1 kinase (RIPK1) were used to investigate the mechanism of cell killing. The synergistic interaction between oHSV and bortezomib was calculated using a Chou–Talalay analysis. NK cells isolated from normal human blood were co-cultured with tumor cells to evaluate cellular interactions. Q-PCR, ELISA, and FACS analysis were used to evaluate NK cell activation. Intracranial tumor xenografts were used to evaluate antitumor efficacy.Results: Combination treatment with bortezomib- and oHSV-induced necroptotic cell death and increased the production of mitochondrial ROS and JNK phosphorylation. Inhibitors/shRNA of RIPK1 and JNK rescued synergistic cell killing. Combination treatment also significantly enhanced NK cell activation and adjuvant NK cell therapy of mice treated with bortezomib and oHSV improved antitumor efficacy.Conclusions: This study provides a significant rationale for triple combination therapy with bortezomib, oHSV, and NK cells to improve efficacy, in glioblastoma patients. Clin Cancer Res; 22(21); 5265–76. ©2016 AACR.See related commentary by Suryadevara et al., p. 5164
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Tables 1-2, Figures 1-7 from Extracellular Matrix Protein CCN1 Limits Oncolytic Efficacy in Glioma
- Author
-
Balveen Kaur, Michael A. Caligiuri, Joseph C. Glorioso, Kazuhiko Kurozumi, Jianhua Yu, Ji Young Yoo, Lianbo Yu, Jeffrey Wojton, Sean Boone, and Amy Haseley
- Abstract
PDF file - 441K
- Published
- 2023
- Full Text
- View/download PDF
16. Supplemental Figure S2 from Aurora-A Inhibition Offers a Novel Therapy Effective against Intracranial Glioblastoma
- Author
-
Norman L. Lehman, Balveen Kaur, Jeffery A. Ecsedy, David A. Kellough, Walter H. Meisen, Jeffrey Wojton, and James R. Van Brocklyn
- Abstract
Morphologic changes of GB neurosphere cells treated with different Aurora-A inhibitors.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental Figure Legend from Aurora-A Inhibition Offers a Novel Therapy Effective against Intracranial Glioblastoma
- Author
-
Norman L. Lehman, Balveen Kaur, Jeffery A. Ecsedy, David A. Kellough, Walter H. Meisen, Jeffrey Wojton, and James R. Van Brocklyn
- Abstract
Legend for Supplemental Figures S1-S3.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Materials, Figure Legends, and Tables from Genetic Validation of the Protein Arginine Methyltransferase PRMT5 as a Candidate Therapeutic Target in Glioblastoma
- Author
-
Robert A. Baiocchi, Balveen Kaur, Sean Lawler, Samson Jacob, Said Sif, Chenglong Li, E. Antonio Chiocca, John C. Byrd, Chang-Hyuk Kwon, Gerard Nuovo, Guido Marcucci, Xiaokui Mo, Xiaoli Zhang, John Ryu, Porsha L. Smith, John T. Patton, Amy Haseley, Kate Gordon, Bo Yu, Jharna Datta, Rosa Lapalombella, Xin Wu, Michal O. Nowicki, Arnab Chakravarti, Naduparambil K. Jacob, Jeffrey Wojton, Erica Hlavin Bell, Jill Barnholtz-Sloan, Selene Virk, Yeshavanth Banasavadi-Siddegowda, Hector M. Cordero-Nieves, Ludmila Katherine Martin, Mark E. Lustberg, Lapo Alinari, and Fengting Yan
- Abstract
PDF file - 636KB, Supplemental Table S1: information of the GBM lines used in this paper. Supplemental Table S2: information of the patient clinical data for Figure 1. Supplemental Table S3: Percentage of cells positive for staining with PRMT5 in spontaneous high grade astrocytoma from Mut3 strain (GFAP-cre; cisNf1-/+; P53-/-) and normal brain tissue from wild-type control mouse stained by PRMT5 and ki-67 antibody. Supplemental table S4. GBM cell line (U251) was transfected by scramble control or si-PRMT5. Affymetrix microarray genechip was performed in three independent experiments to evaluate genes up-regulated with PRMT5 silencing.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figures 1 - 7 from Genetic Validation of the Protein Arginine Methyltransferase PRMT5 as a Candidate Therapeutic Target in Glioblastoma
- Author
-
Robert A. Baiocchi, Balveen Kaur, Sean Lawler, Samson Jacob, Said Sif, Chenglong Li, E. Antonio Chiocca, John C. Byrd, Chang-Hyuk Kwon, Gerard Nuovo, Guido Marcucci, Xiaokui Mo, Xiaoli Zhang, John Ryu, Porsha L. Smith, John T. Patton, Amy Haseley, Kate Gordon, Bo Yu, Jharna Datta, Rosa Lapalombella, Xin Wu, Michal O. Nowicki, Arnab Chakravarti, Naduparambil K. Jacob, Jeffrey Wojton, Erica Hlavin Bell, Jill Barnholtz-Sloan, Selene Virk, Yeshavanth Banasavadi-Siddegowda, Hector M. Cordero-Nieves, Ludmila Katherine Martin, Mark E. Lustberg, Lapo Alinari, and Fengting Yan
- Abstract
PDF file - 1690KB, Supplemental Figure S1. PRMT5 is over-expressed in human GBM cell lines. Supplemental Figure S2: High grade astrocytomas Supplemental Figure S3: efficacy and specificity of PRMT5 knock-down by lead siRNA. Supplemental Figure S4: PRMT5 Knockdown promotes cell death of human GBM cells Supplemental Figure S5: si-PRMT5 induced cell death is P53-independent Supplemental Figure S6: Decreased ST7 expression correlates with worse survival in all glioma patients (grade I-IV). Supplemental Figure S7: Over-expression of ST7 in three GBM cell lines was confirmed by real-time PCR and western blot (top panels).
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure Legends 1-7 from Extracellular Matrix Protein CCN1 Limits Oncolytic Efficacy in Glioma
- Author
-
Balveen Kaur, Michael A. Caligiuri, Joseph C. Glorioso, Kazuhiko Kurozumi, Jianhua Yu, Ji Young Yoo, Lianbo Yu, Jeffrey Wojton, Sean Boone, and Amy Haseley
- Abstract
PDF file - 93K
- Published
- 2023
- Full Text
- View/download PDF
21. Data from Genetic Validation of the Protein Arginine Methyltransferase PRMT5 as a Candidate Therapeutic Target in Glioblastoma
- Author
-
Robert A. Baiocchi, Balveen Kaur, Sean Lawler, Samson Jacob, Said Sif, Chenglong Li, E. Antonio Chiocca, John C. Byrd, Chang-Hyuk Kwon, Gerard Nuovo, Guido Marcucci, Xiaokui Mo, Xiaoli Zhang, John Ryu, Porsha L. Smith, John T. Patton, Amy Haseley, Kate Gordon, Bo Yu, Jharna Datta, Rosa Lapalombella, Xin Wu, Michal O. Nowicki, Arnab Chakravarti, Naduparambil K. Jacob, Jeffrey Wojton, Erica Hlavin Bell, Jill Barnholtz-Sloan, Selene Virk, Yeshavanth Banasavadi-Siddegowda, Hector M. Cordero-Nieves, Ludmila Katherine Martin, Mark E. Lustberg, Lapo Alinari, and Fengting Yan
- Abstract
Glioblastoma is the most common and aggressive histologic subtype of brain cancer with poor outcomes and limited treatment options. Here, we report the selective overexpression of the protein arginine methyltransferase PRMT5 as a novel candidate theranostic target in this disease. PRMT5 silences the transcription of regulatory genes by catalyzing symmetric dimethylation of arginine residues on histone tails. PRMT5 overexpression in patient-derived primary tumors and cell lines correlated with cell line growth rate and inversely with overall patient survival. Genetic attenuation of PRMT5 led to cell-cycle arrest, apoptosis, and loss of cell migratory activity. Cell death was p53-independent but caspase-dependent and enhanced with temozolomide, a chemotherapeutic agent used as a present standard of care. Global gene profiling and chromatin immunoprecipitation identified the tumor suppressor ST7 as a key gene silenced by PRMT5. Diminished ST7 expression was associated with reduced patient survival. PRMT5 attenuation limited PRMT5 recruitment to the ST7 promoter, led to restored expression of ST7 and cell growth inhibition. Finally, PRMT5 attenuation enhanced glioblastoma cell survival in a mouse xenograft model of aggressive glioblastoma. Together, our findings defined PRMT5 as a candidate prognostic factor and therapeutic target in glioblastoma, offering a preclinical justification for targeting PRMT5-driven oncogenic pathways in this deadly disease. Cancer Res; 74(6); 1752–65. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
22. Dimer‐specific immunoprecipitation of active caspase‐2 identifies TRAF proteins as novel activators
- Author
-
Kenkyo Matsuura, Jeffrey Wojton, Alexander C. Robeson, Kelly R. Lindblom, and Sally Kornbluth
- Subjects
0301 basic medicine ,TRAF2 ,Programmed cell death ,DNA damage ,Immunoprecipitation ,Caspase 2 ,Interactome ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Bimolecular fluorescence complementation ,TRAF ,Protein Interaction Mapping ,Humans ,ubiquitylation ,Molecular Biology ,dimer purification ,Adaptor Proteins, Signal Transducing ,TNF Receptor-Associated Factor 3 ,General Immunology and Microbiology ,biology ,General Neuroscience ,apoptosis ,Articles ,TNF Receptor-Associated Factor 1 ,Cell biology ,030104 developmental biology ,biology.protein ,Autophagy & Cell Death ,Caspase‐2 ,Protein Multimerization ,Intracellular ,Protein Binding - Abstract
Caspase‐2 has been shown to initiate apoptotic cell death in response to specific intracellular stressors such as DNA damage. However, the molecular mechanisms immediately upstream of its activation are still poorly understood. We combined a caspase‐2 bimolecular fluorescence complementation (BiFC) system with fluorophore‐specific immunoprecipitation to isolate and study the active caspase‐2 dimer and its interactome. Using this technique, we found that tumor necrosis factor receptor‐associated factor 2 (TRAF2), as well as TRAF1 and 3, directly binds to the active caspase‐2 dimer. TRAF2 in particular is necessary for caspase‐2 activation in response to apoptotic cell death stimuli. Furthermore, we found that dimerized caspase‐2 is ubiquitylated in a TRAF2‐dependent manner at K15, K152, and K153, which in turn stabilizes the active caspase‐2 dimer complex, promotes its association with an insoluble cellular fraction, and enhances its activity to fully commit the cell to apoptosis. Together, these data indicate that TRAF2 positively regulates caspase‐2 activation and consequent cell death by driving its activation through dimer‐stabilizing ubiquitylation.
- Published
- 2018
- Full Text
- View/download PDF
23. Humanized Chondroitinase ABC Sensitizes Glioblastoma Cells to Temozolomide
- Author
-
Alena Cristina Jaime-Ramirez, Nina Dmitrieva, Jeffrey Wojton, Ji Young Yoo, Balveen Kaur, Yeshavanth Kumar Banasavadi-Siddegowda, Theresa Relation, Jianying Zhang, and Chelsea Bolyard
- Subjects
0301 basic medicine ,Gene Expression ,Apoptosis ,Herpesvirus 1, Human ,Pharmacology ,Chondroitin ABC Lyase ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Transduction, Genetic ,Drug Discovery ,Chlorocebus aethiops ,Tumor Cells, Cultured ,Genetics (clinical) ,Oncolytic Virotherapy ,Tumor Burden ,Dacarbazine ,Treatment Outcome ,030220 oncology & carcinogenesis ,Molecular Medicine ,medicine.drug ,Cell Survival ,Genetic Vectors ,Gene delivery ,Article ,03 medical and health sciences ,Glioma ,Neurosphere ,Cell Line, Tumor ,Genetics ,medicine ,Temozolomide ,Animals ,Humans ,Chondroitin sulfate ,Molecular Biology ,Protein kinase B ,Antineoplastic Agents, Alkylating ,Vero Cells ,Alleles ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Oncolytic virus ,Enzyme Activation ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Amino Acid Substitution ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,Glioblastoma - Abstract
BACKGROUND Malignant gliomas (glioblastomas; GBMs) are extremely aggressive and have a median survival of approximately 15 months. Current treatment modalities, which include surgical resection, radiation and chemotherapy, have done little to prolong the lives of GBM patients. Chondroitin sulfate proteoglycans (CSPG) are critical for cell-cell and cell-extracellular matrix (ECM) interactions and are implicated in glioma growth and invasion. Chondroitinase (Chase) ABC is a bacterial enzyme that cleaves chondroitin sulfate disaccharide chains from CSPGs in the tumor ECM. Wild-type Chase ABC has limited stability and/or activity in mammalian cells; therefore, we created a mutant humanized version (Chase M) with enhanced function in mammalian cells. METHODS We hypothesized that disruption of cell-cell and cell-ECM interactions by ChaseM and temozolomide (TMZ) will enhance the chemotherapeutic availability and sensitivity of glioma cells. RESULTS Utilizing primary patient-derived neurospheres, we found that ChaseM decreases glioma neurosphere aggregation in vitro. Furthermore, an oncolytic HSV-1 virus expressing secreted ChaseM (OV-ChaseM) enhanced viral spread and glioma cell killing compared to OV-Control, in vitro. OV-ChaseM plus TMZ combinatorial treatment resulted in a significant synergistic enhancement of glioma cell killing accompanied by an increase in apoptotic cell death. Intracellular flow cytometric analysis revealed a significant reduction in the phosphorylation of the pro-survival AKT protein following OV-ChaseM plus TMZ treatment. Lastly, in nude mice bearing intracranial GBM30 glioma xenografts, intratumoral OV-ChaseM plus TMZ (10 mg/kg by oral gavage) combination therapy resulted in a significant (p
- Published
- 2017
24. Systemic Delivery of SapC-DOPS Has Antiangiogenic and Antitumor Effects Against Glioblastoma
- Author
-
Sherry Thornton, Balveen Kaur, Haritha Mathsyaraja, Zhengtao Chu, Tristan Bourdeau, Mary B. Palascak, Nicholas L Denton, Michael C. Ostrowski, Chang-Hyuk Kwon, Jeffrey Wojton, Lionel M.L. Chow, Walter Hans Meisen, Robert S. Franco, and Xiaoyang Qi
- Subjects
Male ,Cell ,Brain tumor ,Neovascularization, Physiologic ,Angiogenesis Inhibitors ,Antineoplastic Agents ,Phosphatidylserines ,Pharmacology ,Biology ,Blood–brain barrier ,Saposins ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Brain Neoplasms ,Cell Membrane ,Phosphatidylserine ,medicine.disease ,Xenograft Model Antitumor Assays ,Cell Hypoxia ,Recombinant Proteins ,In vitro ,3. Good health ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,Blood-Brain Barrier ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Nanoparticles ,Molecular Medicine ,Female ,Original Article ,Glioblastoma - Abstract
Saposin C-dioleoylphosphatidylserine (SapC-DOPS) nanovesicles are a nanotherapeutic which effectively target and destroy cancer cells. Here, we explore the systemic use of SapC-DOPS in several models of brain cancer, including glioblastoma multiforme (GBM), and the molecular mechanism behind its tumor-selective targeting specificity. Using two validated spontaneous brain tumor models, we demonstrate the ability of SapC-DOPS to selectively and effectively cross the blood–brain tumor barrier (BBTB) to target brain tumors in vivo and reveal the targeting to be contingent on the exposure of the anionic phospholipid phosphatidylserine (PtdSer). Increased cell surface expression of PtdSer levels was found to correlate with SapC-DOPS–induced killing efficacy, and tumor targeting in vivo was inhibited by blocking PtdSer exposed on cells. Apart from cancer cell killing, SapC-DOPS also exerted a strong antiangiogenic activity in vitro and in vivo. Interestingly, unlike traditional chemotherapy, hypoxic cells were sensitized to SapC-DOPS–mediated killing. This study emphasizes the importance of PtdSer exposure for SapC-DOPS targeting and supports the further development of SapC-DOPS as a novel antitumor and antiangiogenic agent for brain tumors.
- Published
- 2013
- Full Text
- View/download PDF
25. BAI1 Orchestrates Macrophage Inflammatory Response to HSV Infection-Implications for Oncolytic Viral Therapy
- Author
-
Balveen Kaur, Christopher Alvarez-Breckenridge, Jianying Zhang, W. Hans Meisen, Jeffrey Wojton, Eric S. Wohleb, Matthew O. Old, Jonathan C. Smith, Michael A. Caligiuri, Samuel Dubin, Erwin G. Van Meir, Yeshavanth Kumar Banasavadi-Siddegowda, Jun Ge Yu, Ji Young Yoo, Maninder Khosla, Bo Xu, Jianhua Yu, Jonathan P. Godbout, Flavia Pichiorri, Dan Zhu, Jayson Hardcastle, Pete Pow-anpongkul, and Chelsea Bolyard
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Phagocytosis ,Inflammation ,Biology ,medicine.disease_cause ,Virus ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Simplexvirus ,Angiogenic Proteins ,Oncolytic Virotherapy ,Microglia ,Macrophages ,Brain ,Glioma ,Xenograft Model Antitumor Assays ,Oncolytic virus ,Oncolytic Viruses ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Herpes simplex virus ,Oncology ,Immunology ,Tumor necrosis factor alpha ,medicine.symptom - Abstract
Purpose: Brain angiogenesis inhibitor (BAI1) facilitates phagocytosis and bacterial pathogen clearance by macrophages; however, its role in viral infections is unknown. Here, we examined the role of BAI1, and its N-terminal cleavage fragment (Vstat120) in antiviral macrophage responses to oncolytic herpes simplex virus (oHSV). Experimental Design: Changes in infiltration and activation of monocytic and microglial cells after treatment of glioma-bearing mice brains with a control (rHSVQ1) or Vstat120-expressing (RAMBO) oHSV was analyzed using flow cytometry. Co-culture of infected glioma cells with macrophages or microglia was used to examine antiviral signaling. Cytokine array gene expression and Ingenuity Pathway Analysis (IPA) helped evaluate changes in macrophage signaling in response to viral infection. TNFα-blocking antibodies and macrophages derived from Bai1−/− mice were used. Results: RAMBO treatment of mice reduced recruitment and activation of macrophages/microglia in mice with brain tumors, and showed increased virus replication compared with rHSVQ1. Cytokine gene expression array revealed that RAMBO significantly altered the macrophage inflammatory response to infected glioma cells via altered secretion of TNFα. Furthermore, we showed that BAI1 mediated macrophage TNFα induction in response to oHSV therapy. Intracranial inoculation of wild-type/RAMBO virus in Bai1−/− or wild-type non–tumor-bearing mice revealed the safety of this approach. Conclusions: We have uncovered a new role for BAI1 in facilitating macrophage anti-viral responses. We show that arming oHSV with antiangiogenic Vstat120 also shields them from inflammatory macrophage antiviral response, without reducing safety. Clin Cancer Res; 23(7); 1809–19. ©2016 AACR.
- Published
- 2016
26. Bortezomib Treatment Sensitizes Oncolytic HSV-1-Treated Tumors to NK Cell Immunotherapy
- Author
-
Matthew O. Old, Tejaswini Nallanagulagari, Brian Hurwitz, Michael A. Caligiuri, Theresa Relation, Michael T. Lotze, Jianying Zhang, Ji Young Yoo, Balveen Kaur, Jianhua Yu, Alena Cristina Jaime-Ramirez, Jeffrey Wojton, Carlo M. Croce, Tae Jin Lee, Jun Ge Yu, Chelsea Bolyard, and Hongsheng Dai
- Subjects
0301 basic medicine ,Cancer Research ,MAP Kinase Kinase 4 ,medicine.medical_treatment ,Cell ,Mice, Nude ,Herpesvirus 1, Human ,Pharmacology ,Cell therapy ,Bortezomib ,03 medical and health sciences ,RIPK1 ,Mice ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Caspase 7 ,Oncolytic Virotherapy ,Cell Death ,business.industry ,Caspase 3 ,Immunotherapy ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Oncolytic virus ,Killer Cells, Natural ,Oncolytic Viruses ,030104 developmental biology ,Cell killing ,medicine.anatomical_structure ,Oncology ,Receptor-Interacting Protein Serine-Threonine Kinases ,Proteasome inhibitor ,Female ,business ,Reactive Oxygen Species ,medicine.drug - Abstract
Purpose: Both the proteasome inhibitor bortezomib and an oncolytic herpes simplex virus-1 (oHSV)–expressing GM-CSF are currently FDA approved. Although proteasome blockade can increase oHSV replication, immunologic consequences, and consequent immunotherapy potential are unknown. In this study, we investigated the impact of bortezomib combined with oHSV on tumor cell death and sensitivity to natural killer (NK) cell immunotherapy. Experimental Design: Western blot, flow cytometry, and caspase 3/7 activity assays were used to evaluate the induction of apoptosis/autophagy and/or necroptotic cell death. Cellular and mitochondrial reactive oxygen species (ROS) production was measured using CellROX and MitoSOX. Inhibitors/shRNA–targeting ROS, JNK and RIP1 kinase (RIPK1) were used to investigate the mechanism of cell killing. The synergistic interaction between oHSV and bortezomib was calculated using a Chou–Talalay analysis. NK cells isolated from normal human blood were co-cultured with tumor cells to evaluate cellular interactions. Q-PCR, ELISA, and FACS analysis were used to evaluate NK cell activation. Intracranial tumor xenografts were used to evaluate antitumor efficacy. Results: Combination treatment with bortezomib- and oHSV-induced necroptotic cell death and increased the production of mitochondrial ROS and JNK phosphorylation. Inhibitors/shRNA of RIPK1 and JNK rescued synergistic cell killing. Combination treatment also significantly enhanced NK cell activation and adjuvant NK cell therapy of mice treated with bortezomib and oHSV improved antitumor efficacy. Conclusions: This study provides a significant rationale for triple combination therapy with bortezomib, oHSV, and NK cells to improve efficacy, in glioblastoma patients. Clin Cancer Res; 22(21); 5265–76. ©2016 AACR. See related commentary by Suryadevara et al., p. 5164
- Published
- 2016
27. Impact of tumor microenvironment on oncolytic viral therapy
- Author
-
Balveen Kaur and Jeffrey Wojton
- Subjects
Stromal cell ,Angiogenesis ,Endocrinology, Diabetes and Metabolism ,Immunology ,Angiogenesis Inhibitors ,Biology ,Antibodies, Monoclonal, Humanized ,Article ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Immune system ,Immunity ,Neoplasms ,medicine ,Animals ,Humans ,Immunology and Allergy ,Oncolytic Virotherapy ,Clinical Trials as Topic ,Tumor microenvironment ,Antibodies, Monoclonal ,Cancer ,medicine.disease ,Combined Modality Therapy ,Immunity, Innate ,Extracellular Matrix ,Oncolytic virus ,Bevacizumab ,Oncolytic Viruses - Abstract
Interactions between tumor cells and their microenvironment have been shown to play a very significant role in the initiation, progression, and invasiveness of cancer. These tumor-stromal interactions are capable of altering the delivery and effectiveness of therapeutics into the tumor and are also known to influence future resistance and re-growth after treatment. Here we review recent advances in the understanding of the tumor microenvironment and its response to oncolytic viral therapy. The multifaceted environmental response to viral therapy can influence viral infection, replication, and propagation within the tumor. Recent studies have unveiled the complicated temporal changes in the tumor vasculature post-oncolytic virus (OV) treatment, and their impact on tumor biology. Similarly, the secreted extracellular matrix in solid tumors can affect both infection and spread of the therapeutic virus. Together, these complex changes in the tumor microenvironment also modulate the activation of the innate antiviral host immune response, leading to quick and efficient viral clearance. In order to combat these detrimental responses, viruses have been combined with pharmacological adjuvants and "armed" with therapeutic genes in order to suppress the pernicious environmental conditions following therapy. In this review we will discuss the impact of the tumor environment on viral therapy and examine some of the recent literature investigating methods of modulating this environment to enhance oncolysis.
- Published
- 2010
- Full Text
- View/download PDF
28. How to train glioma cells to die: molecular challenges in cell death
- Author
-
Walter Hans Meisen, Balveen Kaur, and Jeffrey Wojton
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Necrosis ,medicine.medical_treatment ,Apoptosis ,Disease ,Biology ,Article ,03 medical and health sciences ,Glioma ,medicine ,Humans ,Tumor microenvironment ,Brain Neoplasms ,Autophagy ,medicine.disease ,Radiation therapy ,030104 developmental biology ,Neurology ,Oncology ,Immunology ,Cancer research ,Neurology (clinical) ,medicine.symptom - Abstract
The five-year survival rate for patients with malignant glioma is less than 10%. Despite aggressive chemo/radiotherapy these tumors have remained resistant to almost every interventional strategy evaluated in patients. Resistance to these agents is attributed to extrinsic mechanisms such as the tumor microenvironment, poor drug penetration, and tumoral heterogeneity. In addition, genetic and molecular examination of these tumors has revealed defective apoptotic regulation, enhanced pro-survival autophagy signaling, and a propensity for necrosis that aids in the adaptation to environmental stress and resistance to treatment. The combination of extrinsic and intrinsic hallmarks in glioma contributes to the multifaceted resistance to traditional anti-tumor agents. Here we describe the biology of the disease relevant to therapeutic resistance, with a specific focus on molecular deregulation of cell death pathways. Emerging studies investigating the targeting of these pathways including BH3 mimetics and autophagy inhibitors that are being evaluated in both the preclinical and clinical settings are discussed. This review highlights the pathways exploited by glioblastoma cells that drive their hallmark pro-survival predisposition and makes therapy development such a challenge.
- Published
- 2015
29. Metabolic control of Ca2+/calmodulin-dependent protein kinase II (CaMKII)-mediated caspase-2 suppression by the B55β/protein phosphatase 2A (PP2A)
- Author
-
Jeffrey Wojton, Erik J. Soderblom, Liguo Zhang, Nai-Jia Huang, Sally Kornbluth, Chih-Sheng Yang, Chen Chen, and Bofu Huang
- Subjects
Protein subunit ,Phosphatase ,Glucose-6-Phosphate ,Pentose phosphate pathway ,Biology ,Xenopus Proteins ,Biochemistry ,Dephosphorylation ,Xenopus laevis ,Ca2+/calmodulin-dependent protein kinase ,Animals ,Humans ,Protein Phosphatase 2 ,Phosphorylation ,Molecular Biology ,chemistry.chemical_classification ,Caspase 2 ,Cell Biology ,Protein phosphatase 2 ,Cell biology ,Enzyme Activation ,Enzyme ,HEK293 Cells ,chemistry ,cardiovascular system ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Protein Processing, Post-Translational ,Protein Binding ,Signal Transduction - Abstract
High levels of metabolic activity confer resistance to apoptosis. Caspase-2, an apoptotic initiator, can be suppressed by high levels of nutrient flux through the pentose phosphate pathway. This metabolic control is exerted via inhibitory phosphorylation of the caspase-2 prodomain by activated Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). We show here that this activation of CaMKII depends, in part, on dephosphorylation of CaMKII at novel sites (Thr(393)/Ser(395)) and that this is mediated by metabolic activation of protein phosphatase 2A in complex with the B55β targeting subunit. This represents a novel locus of CaMKII control and also provides a mechanism contributing to metabolic control of apoptosis. These findings may have implications for metabolic control of the many CaMKII-controlled and protein phosphatase 2A-regulated physiological processes, because both enzymes appear to be responsive to alterations in glucose metabolized via the pentose phosphate pathway.
- Published
- 2014
30. SapC-DOPS-induced lysosomal cell death synergizes with TMZ in glioblastoma
- Author
-
Balveen Kaur, Zhengtao Chu, Amy Haseley Thorne, Jeffrey Wojton, Naduparambil K. Jacob, Jayson Hardcastle, Rachel L. Marsh, Erwin G. Van Meir, Xiaoyang Qi, Walter Hans Meisen, Arnab Chakravarti, Nina Dmitrieva, Chang-Hyuk Kwon, and Nicholas L Denton
- Subjects
Programmed cell death ,Cathepsin D ,Mice, Nude ,synergy ,Phosphatidylserines ,Pharmacology ,Biology ,Saposins ,lysosomal dysfunction ,chemistry.chemical_compound ,Mice ,Random Allocation ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Temozolomide ,Animals ,Humans ,Antineoplastic Agents, Alkylating ,Sphingosine ,LAMP1 ,Cell Death ,Brain Neoplasms ,Autophagy ,glioblastoma ,Drug Synergism ,Xenograft Model Antitumor Assays ,Nanostructures ,SapC-DOPS ,Dacarbazine ,Oncology ,chemistry ,Cell culture ,Apoptosis ,Cancer research ,Lysosomes ,TMZ ,medicine.drug ,Research Paper - Abstract
SapC-DOPS is a novel nanotherapeutic that has been shown to target and induce cell death in a variety of cancers, including glioblastoma (GBM). GBM is a primary brain tumor known to frequently demonstrate resistance to apoptosis-inducing therapeutics. Here we explore the mode of action for SapC-DOPS in GBM, a treatment being developed by Bexion Pharmaceuticals for clinical testing in patients. SapC-DOPS treatment was observed to induce lysosomal dysfunction of GBM cells characterized by decreased glycosylation of LAMP1 and altered proteolytic processing of cathepsin D independent of apoptosis and autophagic cell death. We observed that SapC-DOPS induced lysosomal membrane permeability (LMP) as shown by LysoTracker Red and Acridine Orange staining along with an increase of sphingosine, a known inducer of LMP. Additionally, SapC-DOPS displayed strong synergistic interactions with the apoptosis-inducing agent TMZ. Collectively our data suggest that SapC-DOPS induces lysosomal cell death in GBM cells, providing a new approach for treating tumors resistant to traditional apoptosis-inducing agents.
- Published
- 2014
31. How efficient are autophagy inhibitors as treatment for glioblastoma?
- Author
-
James B. Elder, Balveen Kaur, and Jeffrey Wojton
- Subjects
Programmed cell death ,business.industry ,Brain Neoplasms ,Autophagy ,Context (language use) ,Antineoplastic Agents ,General Medicine ,BECN1 ,Tumor initiation ,Article ,Wortmannin ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Tumor progression ,Lysosome ,Cancer research ,medicine ,Humans ,business ,Glioblastoma - Abstract
5 ISSN 2045-0907 10.2217/CNS.13.52 © 2014 Future Medicine Ltd CNS Oncol. (2014) 3(1), 5–7 Glioblastoma (GB) is the most common and most destructive form of primary brain tumor. Currently, the standard treatment for patients with GB consists of surgical resection, radiotherapy and chemotherapy with temozolomide (TMZ). These tumors have been well characterized for their resistance to standard therapeutic strategies and, despite decades of research, the median survival remains less than 15 months [1]. One of the proposed mechanisms for therapeutic resistance in GB is the activation of stressinduced autophagy. Autophagy is a form of self-degradation in which intracellular components are sequestered in double membrane vesicles called autophagosomes, which then fuse with lysosomes, allowing the recycling of cytoplasmic materials. Inhibition of this process is typically carried out using early-stage inhibitors, such as 3-MA and wortmannin, to prevent the initiation and elongation of the phagophore or late-stage inhibitors, such as bafilomycin-A1, chloroquine (CQ) and hydroxychloroquine (HCQ), which target the functionality of the lysosome in fusing and recycling autophagosomes [2]. The role of autophagy in the development of cancer and response to chemotherapy is highly controversial. The allelic loss of BECN1, the mammalian ortholog of yeast Atg6, results in a high incidence of spontaneous tumors in mice, suggesting BECN1 has a tumor suppressor role [3,4]. Autophagy is thought to play a significant role in the removal of damaged/ oncogenic proteins as well as damaged organelles, and mutations in autophagy genes have been associated with numerous cancers [2]. Interestingly, while the induction of autophagic-dependent cell death has been proposed as a mechanism of cytotoxicity for some chemotherapeutics, activation of autophagy in the context of most therapies has been associated with prosurvival signaling through the maintenance of cellular integrity in response to metabolic and hypoxic stress [5,6]. The currently accepted hypothesis for this paradox is that autophagy may suppress tumor initiation, but promote established tumor progression and resistance to therapeutics [2,7]. While numerous cytotoxic therapeutics have been shown to induce autophagosome formation, the consequences seem to
- Published
- 2014
32. Genetic validation of the protein arginine methyltransferase PRMT5 as a candidate therapeutic target in glioblastoma
- Author
-
Michał Nowicki, Xiaoli Zhang, Sean E. Lawler, Mark E. Lustberg, Gerard J. Nuovo, Erica Hlavin Bell, Naduparambil K. Jacob, Saïd Sif, John T. Patton, Rosa Lapalombella, Chang-Hyuk Kwon, Bo Yu, Chenglong Li, Kate Gordon, Jeffrey Wojton, Porsha Smith, Balveen Kaur, Fengting Yan, Hector M. Cordero-Nieves, Arnab Chakravarti, E. Antonio Chiocca, Xin Wu, Guido Marcucci, Yeshavanth Kumar Banasavadi-Siddegowda, Lapo Alinari, Samson T. Jacob, Jill S. Barnholtz-Sloan, Selene Virk, John Ryu, Robert A. Baiocchi, Xiaokui Mo, Ludmila Katherine Martin, John C. Byrd, Amy Haseley, and Jharna Datta
- Subjects
Cancer Research ,Programmed cell death ,Protein-Arginine N-Methyltransferases ,Cell cycle checkpoint ,Cell ,Gene Expression ,Mice, Nude ,Apoptosis ,Kaplan-Meier Estimate ,Biology ,Article ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,RNA, Small Interfering ,Cell Proliferation ,Regulation of gene expression ,Mice, Knockout ,Temozolomide ,Cell growth ,Brain Neoplasms ,Protein arginine methyltransferase 5 ,Tumor Suppressor Proteins ,Molecular biology ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Gene Knockdown Techniques ,Cancer research ,Tumor Suppressor Protein p53 ,Glioblastoma ,Chromatin immunoprecipitation ,Neoplasm Transplantation ,medicine.drug - Abstract
Glioblastoma is the most common and aggressive histologic subtype of brain cancer with poor outcomes and limited treatment options. Here, we report the selective overexpression of the protein arginine methyltransferase PRMT5 as a novel candidate theranostic target in this disease. PRMT5 silences the transcription of regulatory genes by catalyzing symmetric dimethylation of arginine residues on histone tails. PRMT5 overexpression in patient-derived primary tumors and cell lines correlated with cell line growth rate and inversely with overall patient survival. Genetic attenuation of PRMT5 led to cell-cycle arrest, apoptosis, and loss of cell migratory activity. Cell death was p53-independent but caspase-dependent and enhanced with temozolomide, a chemotherapeutic agent used as a present standard of care. Global gene profiling and chromatin immunoprecipitation identified the tumor suppressor ST7 as a key gene silenced by PRMT5. Diminished ST7 expression was associated with reduced patient survival. PRMT5 attenuation limited PRMT5 recruitment to the ST7 promoter, led to restored expression of ST7 and cell growth inhibition. Finally, PRMT5 attenuation enhanced glioblastoma cell survival in a mouse xenograft model of aggressive glioblastoma. Together, our findings defined PRMT5 as a candidate prognostic factor and therapeutic target in glioblastoma, offering a preclinical justification for targeting PRMT5-driven oncogenic pathways in this deadly disease. Cancer Res; 74(6); 1752–65. ©2014 AACR.
- Published
- 2014
33. NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors
- Author
-
Yan Wang, Soledad Fernandez, Hsiaoyin Mao, Sean E. Lawler, Shun He, Alessandro Moretta, Jianhua Yu, E. Antonio Chiocca, Jayson Hardcastle, Christopher Alvarez-Breckenridge, Eric Vivier, Jeffrey Wojton, Balveen Kaur, Ofer Mandelboim, Richard L. Price, Jason C. Pradarelli, Michael A. Caligiuri, and Min Wei
- Subjects
Adoptive cell transfer ,Cell ,Biology ,Real-Time Polymerase Chain Reaction ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Glioma ,Cell Line, Tumor ,Chlorocebus aethiops ,medicine ,Animals ,Antigens, Ly ,Humans ,Simplexvirus ,Virotherapy ,Cytotoxicity ,Vero Cells ,030304 developmental biology ,DNA Primers ,Mice, Knockout ,Oncolytic Virotherapy ,0303 health sciences ,Analysis of Variance ,Natural Cytotoxicity Triggering Receptor 3 ,Natural Cytotoxicity Triggering Receptor 1 ,Reverse Transcriptase Polymerase Chain Reaction ,General Medicine ,medicine.disease ,Flow Cytometry ,Adoptive Transfer ,3. Good health ,Oncolytic virus ,Killer Cells, Natural ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Glioblastoma - Abstract
Oncolytic virotherapy has been tested in cancer patients, but its efficacy is uncertain. Alvarez-Breckenridge et al. now report that in mouse models of glioblastoma, an antiviral response mediated by natural killer (NK) cells may impair the anticancer efficacy of oncolytic virotherapy. Their findings suggest that limiting the cytolytic activity of NK cells might enhance replication of oncolytic viruses and increase tumor cell killing. The role of the immune response to oncolytic Herpes simplex viral (oHSV) therapy for glioblastoma is controversial because it might enhance or inhibit efficacy. We found that within hours of oHSV infection of glioblastomas in mice, activated natural killer (NK) cells are recruited to the site of infection. This response substantially diminished the efficacy of glioblastoma virotherapy. oHSV-activated NK cells coordinated macrophage and microglia activation within tumors. In vitro, human NK cells preferentially lysed oHSV-infected human glioblastoma cell lines. This enhanced killing depended on the NK cell natural cytotoxicity receptors (NCRs) NKp30 and NKp46, whose ligands are upregulated in oHSV-infected glioblastoma cells. We found that HSV titers and oHSV efficacy are increased in Ncr1−/− mice and a Ncr1−/− NK cell adoptive transfer model of glioma, respectively. These results demonstrate that glioblastoma virotherapy is limited partially by an antiviral NK cell response involving specific NCRs, uncovering new potential targets to enhance cancer virotherapy.
- Published
- 2012
34. Copper chelation enhances antitumor efficacy and systemic delivery of oncolytic HSV
- Author
-
Andrew P. Mazar, Balveen Kaur, E. Antonio Chiocca, Theodoros N. Teknos, Jason C. Pradarelli, Christopher Alvarez-Breckenridge, Amy Haseley, Jeffrey Wojton, Matthew O. Old, Anna Bratasz, Ji Young Yoo, Kimerly A. Powell, Azeem Kaka, Joseph C. Glorioso, and Jun Ge Yu
- Subjects
Cancer Research ,Angiogenesis ,Genetic Vectors ,Mice, Nude ,Antineoplastic Agents ,Biology ,Virus Replication ,Virus ,Article ,Mice ,Cytopathogenic Effect, Viral ,Glioma ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Simplexvirus ,Chelating Agents ,Molybdenum ,Oncolytic Virotherapy ,Cancer ,Genetic Therapy ,medicine.disease ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Oncolytic virus ,Oncolytic Viruses ,Cell killing ,Oncology ,Viral replication ,Cell culture ,Immunology ,Cancer research ,Female ,Copper - Abstract
Purpose: Copper in serum supports angiogenesis and inhibits replication of wild-type HSV-1. Copper chelation is currently being investigated as an antiangiogenic and antineoplastic agent in patients diagnosed with cancer. Herpes simplex virus–derived oncolytic viruses (oHSV) are being evaluated for safety and efficacy in patients, but several host barriers limit their efficacy. Here, we tested whether copper inhibits oHSV infection and replication and whether copper chelation would augment therapeutic efficacy of oHSV. Experimental Design: Subcutaneous and intracranial tumor-bearing mice were treated with oHSV ± ATN-224 to evaluate tumor burden and survival. Virus replication and cell killing was measured in the presence or absence of the copper chelating agent ATN-224 and in the presence or absence of copper in vitro. Microvessel density and changes in perfusion were evaluated by immunohistochemistry and dynamic contrast enhanced MRI (DCE-MRI). Serum stability of oHSV was measured in mice fed with ATN-224. Tumor-bearing mice were injected intravenously with oHSV; tumor burden and amount of virus in tumor tissue were evaluated. Results: Combination of systemic ATN-224 and oHSV significantly reduced tumor growth and prolonged animal survival. Immunohistochemistry and DCE-MRI imaging confirmed that ATN-224 reduced oHSV-induced blood vessel density and vascular leakage. Copper at physiologically relevant concentrations inhibited oHSV replication and glioma cell killing, and this effect was rescued by ATN-224. ATN-224 increased serum stability of oHSV and enhanced the efficacy of systemic delivery. Conclusion: This study shows that combining ATN-224 with oHSV significantly increased serum stability of oHSV and greatly enhanced its replication and antitumor efficacy. Clin Cancer Res; 18(18); 4931–41. ©2012 AACR.
- Published
- 2012
35. Extracellular matrix protein CCN1 limits oncolytic efficacy in glioma
- Author
-
Lianbo Yu, Amy Haseley, Balveen Kaur, Kazuhiko Kurozumi, Joseph C. Glorioso, Michael A. Caligiuri, Sean Boone, Ji Young Yoo, Jianhua Yu, and Jeffrey Wojton
- Subjects
Cancer Research ,Integrin ,Mice, Nude ,Biology ,Article ,Mice ,Interferon ,Glioma ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Integrin Signaling Pathway ,Oncolytic Virotherapy ,Integrin alpha6beta1 ,Brain Neoplasms ,Gene Expression Profiling ,Interferon-alpha ,medicine.disease ,Oncolytic virus ,Oncology ,Viral replication ,Cell culture ,Interferon Type I ,Cancer research ,biology.protein ,Female ,Signal transduction ,medicine.drug ,Cysteine-Rich Protein 61 ,Signal Transduction - Abstract
Oncolytic viral therapy has been explored widely as an option for glioma treatment but its effectiveness has remained limited. Cysteine rich 61 (CCN1) is an extracellular matrix (ECM) protein elevated in cancer cells that modulates their adhesion and migration by binding cell surface receptors. In this study, we examined a hypothesized role for CCN1 in limiting the efficacy of oncolytic viral therapy for glioma, based on evidence of CCN1 induction that occurs in this setting. Strikingly, we found that exogenous CCN1 in glioma ECM orchestrated a cellular antiviral response that reduced viral replication and limited cytolytic efficacy. Gene expression profiling and real-time PCR analysis revealed a significant induction of type-I interferon responsive genes in response to CCN1 exposure. This induction was accompanied by activation of the Jak/Stat signaling pathway, consistent with induction of an innate antiviral cellular response. Both effects were mediated by the binding of CCN1 to the cell surface integrin α6β1, activating its signaling and leading to rapid secretion of interferon-α, which was essential for the innate antiviral effect. Together, our findings reveal how an integrin signaling pathway mediates activation of a type-I antiviral interferon response that can limit the efficacy of oncolytic viral therapy. Furthermore, they suggest therapeutic interventions to inhibit CCN1–integrin α6 interactions to sensitize gliomas to viral oncolysis. Cancer Res; 72(6); 1353–62. ©2012 AACR.
- Published
- 2012
36. Abstract 2169: SapC-DOPS induces lethal mitophagy in glioblastoma
- Author
-
Hiroshi Nakashima, Jeffrey Wojton, Chang-Hyuk Kwon, Xiaoyang Qi, Lionel M.L. Chow, Ennio Antonio Chiocca, Balveen Kaur, Arnab Chakravarti, Nicholas L Denton, Nina Dmitrieva, and Naduparambil K. Jacob
- Subjects
Cancer Research ,Programmed cell death ,Pathology ,medicine.medical_specialty ,ATG5 ,Autophagy ,Acridine orange ,Biology ,chemistry.chemical_compound ,Oncology ,chemistry ,Cell culture ,Mitophagy ,medicine ,Cancer research ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
The goal of this study is to evaluate SapC-DOPS, a novel cancer nanotherapeutic, for glioblastoma multiforme (GBM). SapC-DOPS delivered intravenously (i.v) was found to specifically target intracranial tumors in mice bearing spontaneous brain tumor, as well in nude mice intracranially implanted with human GBM cells. Treatment of tumor bearing mice with SapC-DOPS (i.v.) significantly increased survival: 25% and 75% long-term survivors in U87ΔEGFR-Luc and X12v2 implanted mice, respectively (P0.05). In contrast, SapC-DOPS treatment increased levels of an autophagic marker LC3-II via western blot. Autophagosome formation was also confirmed through transmission electron microscopy. Utilizing a stable GBM cell line expressing a GFP-LC3 fusion protein, we observed punctuated GFP expression following treatment, indicative of autophagosome formation. Quantification of GFP punctated cells showed a significant increase in SapC-DOPS treated cells compared to control (P Citation Format: Jeffrey Wojton, Naduparambil K. Jacob, Nicholas Denton, Nina Dmitrieva, Hiroshi Nakashima, Chang-Hyuk Kwon, Lionel Chow, Ennio A. Chiocca, Arnab Chakravarti, Balveen Kaur, Xiaoyang Qi. SapC-DOPS induces lethal mitophagy in glioblastoma. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2169. doi:10.1158/1538-7445.AM2013-2169
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.