86 results on '"Helena J. Mauceri"'
Search Results
2. Data from HMG-CoA Reductase Inhibition Delays DNA Repair and Promotes Senescence After Tumor Irradiation
- Author
-
Stephen J. Kron, Ralph R. Weichselbaum, Harold G. Sutton, Aishwarya Ramamurthy, Amy C. Flor, Helena J. Mauceri, Edwardine Labay, Natalia Ricco, and Elena V. Efimova
- Abstract
Despite significant advances in combinations of radiotherapy and chemotherapy, altered fractionation schedules and image-guided radiotherapy, many cancer patients fail to benefit from radiation. A prevailing hypothesis is that targeting repair of DNA double strand breaks (DSB) can enhance radiation effects in the tumor and overcome therapeutic resistance without incurring off-target toxicities. Unrepaired DSBs can block cancer cell proliferation, promote cancer cell death, and induce cellular senescence. Given the slow progress to date translating novel DSB repair inhibitors as radiosensitizers, we have explored drug repurposing, a proven route to improving speed, costs, and success rates of drug development. In a prior screen where we tracked resolution of ionizing radiation-induced foci (IRIF) as a proxy for DSB repair, we had identified pitavastatin (Livalo), an HMG-CoA reductase inhibitor commonly used for lipid lowering, as a candidate radiosensitizer. Here, we report that pitavastatin and other lipophilic statins are potent inhibitors of DSB repair in breast and melanoma models both in vitro and in vivo. When combined with ionizing radiation, pitavastatin increased persistent DSBs, induced senescence, and enhanced acute effects of radiation on radioresistant melanoma tumors. shRNA knockdown implicated HMG-CoA reductase, farnesyl diphosphate synthase, and protein farnesyl transferase in IRIF resolution, DSB repair, and senescence. These data confirm on-target activity of statins, although via inhibition of protein prenylation rather than cholesterol biosynthesis. In light of prior studies demonstrating enhanced efficacy of radiotherapy in patients taking statins, this work argues for clinical evaluation of lipophilic statins as nontoxic radiosensitizers to enhance the benefits of image-guided radiotherapy. Mol Cancer Ther; 17(2); 407–18. ©2017 AACR.See all articles in this MCT Focus section, “Developmental Therapeutics in Radiation Oncology.”
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Table I from HMG-CoA Reductase Inhibition Delays DNA Repair and Promotes Senescence After Tumor Irradiation
- Author
-
Stephen J. Kron, Ralph R. Weichselbaum, Harold G. Sutton, Aishwarya Ramamurthy, Amy C. Flor, Helena J. Mauceri, Edwardine Labay, Natalia Ricco, and Elena V. Efimova
- Abstract
Supplementary Table 1. shRNAs used
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figures S1-S8 and Legends from HMG-CoA Reductase Inhibition Delays DNA Repair and Promotes Senescence After Tumor Irradiation
- Author
-
Stephen J. Kron, Ralph R. Weichselbaum, Harold G. Sutton, Aishwarya Ramamurthy, Amy C. Flor, Helena J. Mauceri, Edwardine Labay, Natalia Ricco, and Elena V. Efimova
- Abstract
Supplementary Fig. S1. Identification of pitavastatin in IRIF persistence screen of NIH Clinical Collection in MCF7GFP-IBD and PANC02GFP-IBD cell lines; Supplementary Fig. S2. Mevalonic acid abrogates radiosensitization effects of pitavastatin in irradiated MCF7 GFP-IBD cells. Supplementary Fig. S3. Reduction in targeted protein expression by shRNA; Supplementary Fig. S4. Targeting mevalonate pathway affects protein prenylation; Supplementary Fig. S5. Farnesyl pyrophosphate abrogates radiosensitization effects of statins in irradiated MCF7 GFP-IBD cells; Supplementary Fig. S6. Silencing GGPS impacts DNA damage response; Supplementary Fig. S7. Clonogenic survival of B16.SIY cells treated with 5 μmol/L or 10 μmol/L veliparib 1 hour prior to the indicated irradiation dose; Supplementary Fig. S8. Flow cytometry co-staining assay for apoptosis and necrosis in tumor cells treated with pitavastatin and/or radiation.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Methods, Figure Legends 1-6 from Poly(ADP-Ribose) Polymerase Inhibitor Induces Accelerated Senescence in Irradiated Breast Cancer Cells and Tumors
- Author
-
Ralph R. Weichselbaum, Stephen J. Kron, Harold G. Sutton, Clayton Crawley, Juan Camilo Barreto-Andrade, Chaitali Chakraborty, Thomas E. Darga, Vytautas P. Bindokas, Edwardine Labay, Daniel W. Golden, Helena J. Mauceri, and Elena V. Efimova
- Abstract
Supplementary Methods, Figure Legends 1-6 from Poly(ADP-Ribose) Polymerase Inhibitor Induces Accelerated Senescence in Irradiated Breast Cancer Cells and Tumors
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Blockade of Tumor Necrosis Factor α Signaling in Tumor-Associated Macrophages as a Radiosensitizing Strategy
- Author
-
Ralph R. Weichselbaum, Kenneth S. Cohen, Nico van Rooijen, Helena J. Mauceri, Hua Liang, Michael A. Beckett, and Yuru Meng
- Abstract
Most cancer patients receive radiotherapy during the course of their disease. Improvements in the therapeutic index have been based mainly on physical improvements in delivery, as radiosensitizer development to target tumor cells has yet to yield effective agents. Recent investigations have focused on the tumor stroma as a target for radiosensitization. Here, we report that depletion of tumor-associated macrophages (TAMϕ) by systemic or local injection of the macrophage-depleting liposomal clodronate before radiotherapy can increase the antitumor effects of ionizing radiation (IR), either as a large single dose (20 Gy) or as a fractionated dose (2 Gy × 10). Coimplantation of tumor cells with bone marrow–derived macrophages (BMDMϕ) increased tumor radioresistance. Studies using mice with germline deletions in tumor necrosis factor receptors 1 and 2 (TNFR1,2−/−) or TNFα (TNF−/−), or treatment of wild-type mice with a soluble TNF receptor fusion protein (Enbrel), revealed that radioresistance mediated by BMDMϕ required intact TNFα signaling. Radiation exposure upregulated vascular endothelial growth factor (VEGF) in macrophages and VEGF-neutralizing antibodies enhanced the antitumor response to IR. Thus, the radioprotective effect of TNFα was mediated by TAM-produced VEGF. Our findings offer a mechanistic basis to target macrophage populations generally or TNFα-induced macrophage VEGF specifically as tractable strategies to improve the efficacy of radiotherapy. Cancer Res; 70(4); 1534–43
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Poly(ADP-Ribose) Polymerase Inhibitor Induces Accelerated Senescence in Irradiated Breast Cancer Cells and Tumors
- Author
-
Ralph R. Weichselbaum, Stephen J. Kron, Harold G. Sutton, Clayton Crawley, Juan Camilo Barreto-Andrade, Chaitali Chakraborty, Thomas E. Darga, Vytautas P. Bindokas, Edwardine Labay, Daniel W. Golden, Helena J. Mauceri, and Elena V. Efimova
- Abstract
Persistent DNA double-strand breaks (DSB) may determine the antitumor effects of ionizing radiation (IR) by inducing apoptosis, necrosis, mitotic catastrophe, or permanent growth arrest. IR induces rapid modification of megabase chromatin domains surrounding DSBs via poly-ADP-ribosylation, phosphorylation, acetylation, and protein assembly. The dynamics of these IR-induced foci (IRIF) have been implicated in DNA damage signaling and DNA repair. As an IRIF reporter, we tracked the relocalization of green fluorescent protein fused to a chromatin binding domain of the checkpoint adapter protein 53BP1 after IR of breast cancer cells and tumors. To block DSB repair in breast cancer cells and tumors, we targeted poly(ADP-ribose) polymerase (PARP) with ABT-888 (veliparib), one of several PARP inhibitors currently in clinical trials. PARP inhibition markedly enhanced IRIF persistence and increased breast cancer cell senescence both in vitro and in vivo, arguing for targeting IRIF resolution as a novel therapeutic strategy. Cancer Res; 70(15); 6277–82. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figures 1-7 from Blockade of Tumor Necrosis Factor α Signaling in Tumor-Associated Macrophages as a Radiosensitizing Strategy
- Author
-
Ralph R. Weichselbaum, Kenneth S. Cohen, Nico van Rooijen, Helena J. Mauceri, Hua Liang, Michael A. Beckett, and Yuru Meng
- Abstract
Supplementary Figures 1-7 from Blockade of Tumor Necrosis Factor α Signaling in Tumor-Associated Macrophages as a Radiosensitizing Strategy
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 3 from A Survivin-Associated Adaptive Response in Radiation Therapy
- Author
-
Ralph R. Weichselbaum, Gayle E. Woloschak, Jian Jian Li, Harold G. Sutton, Helena J. Mauceri, Richard C. Miller, Jeffrey S. Murley, and David J. Grdina
- Abstract
PDF - 186K, Representative TUNEL assay images of RKO36 apoptotic cells.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figures 1-6 from Poly(ADP-Ribose) Polymerase Inhibitor Induces Accelerated Senescence in Irradiated Breast Cancer Cells and Tumors
- Author
-
Ralph R. Weichselbaum, Stephen J. Kron, Harold G. Sutton, Clayton Crawley, Juan Camilo Barreto-Andrade, Chaitali Chakraborty, Thomas E. Darga, Vytautas P. Bindokas, Edwardine Labay, Daniel W. Golden, Helena J. Mauceri, and Elena V. Efimova
- Abstract
Supplementary Figures 1-6 from Poly(ADP-Ribose) Polymerase Inhibitor Induces Accelerated Senescence in Irradiated Breast Cancer Cells and Tumors
- Published
- 2023
- Full Text
- View/download PDF
11. Tumor-reprogrammed resident T cells resist radiation to control tumors
- Author
-
Jason J. Luke, Steven J. Chmura, Tasha N. Sims, Israel Lowy, Yang Xin Fu, Helena J. Mauceri, Ainhoa Arina, Wenxin Zheng, Christian A. Fernandez, Nikolai N. Khodarev, Sean P. Pitroda, Byron Burnette, Yuzhu Hou, Martin Forde, Ralph R. Weichselbaum, and Michael A. Beckett
- Subjects
0301 basic medicine ,Cell Survival ,medicine.medical_treatment ,T cell ,Science ,General Physics and Astronomy ,Translational immunology ,Mice, Transgenic ,Cytotoxic T cells ,02 engineering and technology ,Biology ,Radiation Tolerance ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Interferon-gamma ,Lymphocytes, Tumor-Infiltrating ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Interferon gamma ,lcsh:Science ,Mice, Knockout ,Tumor microenvironment ,Multidisciplinary ,Radiotherapy ,Gene Expression Profiling ,General Chemistry ,Immunotherapy ,Neoplasms, Experimental ,021001 nanoscience & nanotechnology ,Combined Modality Therapy ,Radiation therapy ,Mice, Inbred C57BL ,030104 developmental biology ,Lymphatic system ,medicine.anatomical_structure ,Cell culture ,Cancer research ,Tumour immunology ,Imaging the immune system ,lcsh:Q ,0210 nano-technology ,Reprogramming ,medicine.drug - Abstract
Successful combinations of radiotherapy and immunotherapy depend on the presence of live T cells within the tumor; however, radiotherapy is believed to damage T cells. Here, based on longitudinal in vivo imaging and functional analysis, we report that a large proportion of T cells survive clinically relevant doses of radiation and show increased motility, and higher production of interferon gamma, compared with T cells from unirradiated tumors. Irradiated intratumoral T cells can mediate tumor control without newly-infiltrating T cells. Transcriptomic analysis suggests T cell reprogramming in the tumor microenvironment and similarities with tissue-resident memory T cells, which are more radio-resistant than circulating/lymphoid tissue T cells. TGFβ is a key upstream regulator of T cell reprogramming and contributes to intratumoral Tcell radio-resistance. These findings have implications for the design of radio-immunotherapy trials in that local irradiation is not inherently immunosuppressive, and irradiation of multiple tumors might optimize systemic effects of radiotherapy., Lymphocytes are considered one of the most radiosensitive cell types in the body. Here the authors show that unlike circulating lymphocytes, tumor-infiltrating T cells survive therapeutic doses of irradiation, remaining functional and contributing to radiotherapy induced anti-tumor immunity.
- Published
- 2019
- Full Text
- View/download PDF
12. Tumor endothelial inflammation predicts clinical outcome in diverse human cancers.
- Author
-
Sean P Pitroda, Tong Zhou, Randy F Sweis, Matthew Filippo, Edwardine Labay, Michael A Beckett, Helena J Mauceri, Hua Liang, Thomas E Darga, Samantha Perakis, Sajid A Khan, Harold G Sutton, Wei Zhang, Nikolai N Khodarev, Joe G N Garcia, and Ralph R Weichselbaum
- Subjects
Medicine ,Science - Abstract
Vascular endothelial cells contribute to the pathogenesis of numerous human diseases by actively regulating the stromal inflammatory response; however, little is known regarding the role of endothelial inflammation in the growth of human tumors and its influence on the prognosis of human cancers.Using an experimental model of tumor necrosis factor-alpha (TNF-α)-mediated inflammation, we characterized inflammatory gene expression in immunopurified tumor-associated endothelial cells. These genes formed the basis of a multivariate molecular predictor of overall survival that was trained and validated in four types of human cancer.We report that expression of experimentally derived tumor endothelial genes distinguished pathologic tissue specimens from normal controls in several human diseases associated with chronic inflammation. We trained these genes in human cancer datasets and defined a six-gene inflammatory signature that predicted significantly reduced overall survival in breast cancer, colon cancer, lung cancer, and glioma. This endothelial-derived signature predicted outcome independently of, but cooperatively with, standard clinical and pathological prognostic factors. Consistent with these findings, conditioned culture media from human endothelial cells stimulated by pro-inflammatory cytokines accelerated the growth of human colon and breast tumors in immunodeficient mice as compared with conditioned media from untreated endothelial cells.This study provides the first prognostic cancer gene signature derived from an experimental model of tumor-associated endothelial inflammation. These findings support the notion that activation of inflammatory pathways in non-malignant tumor-infiltrating endothelial cells contributes to tumor growth and progression in multiple human cancers. Importantly, these results identify endothelial-derived factors that could serve as potential targets for therapy in diverse human cancers.
- Published
- 2012
- Full Text
- View/download PDF
13. STAT1 pathway mediates amplification of metastatic potential and resistance to therapy.
- Author
-
Nikolai N Khodarev, Paul Roach, Sean P Pitroda, Daniel W Golden, Mihir Bhayani, Michael Y Shao, Thomas E Darga, Mara G Beveridge, Ravi F Sood, Harold G Sutton, Michael A Beckett, Helena J Mauceri, Mitchell C Posner, and Ralph R Weichselbaum
- Subjects
Medicine ,Science - Abstract
Traditionally IFN/STAT1 signaling is connected with an anti-viral response and pro-apoptotic tumor-suppressor functions. Emerging functions of a constitutively activated IFN/STAT1 pathway suggest an association with an aggressive tumor phenotype. We hypothesized that tumor clones that constitutively overexpress this pathway are preferentially selected by the host microenvironment due to a resistance to STAT1-dependent cytotoxicity and demonstrate increased metastatic ability combined with increased resistance to genotoxic stress.Here we report that clones of B16F1 tumors grown in the lungs of syngeneic C57BL/6 mice demonstrate variable transcriptional levels of IFN/STAT1 pathway expression. Tumor cells that constitutively overexpress the IFN/STAT1 pathway (STAT1(H) genotype) are selected by the lung microenvironment. STAT1(H) tumor cells also demonstrate resistance to IFN-gamma (IFNgamma), ionizing radiation (IR), and doxorubicin relative to parental B16F1 and low expressors of the IFN/STAT1 pathway (STAT1(L) genotype). Stable knockdown of STAT1 reversed the aggressive phenotype and decreased both lung colonization and resistance to genotoxic stress.Our results identify a pathway activated by tumor-stromal interactions thereby selecting for pro-metastatic and therapy-resistant tumor clones. New therapies targeted against the IFN/STAT1 signaling pathway may provide an effective strategy to treat or sensitize aggressive tumor clones to conventional cancer therapies and potentially prevent distant organ colonization.
- Published
- 2009
- Full Text
- View/download PDF
14. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance
- Author
-
Xiangjiao Meng, Hua Liang, Meng Xu, Yang Xin Fu, Helena J. Mauceri, Ralph R. Weichselbaum, Yuzhu Hou, Liufu Deng, Matthias Mack, Xiaona Huang, Enyu Rao, and Wenxin Zheng
- Subjects
0301 basic medicine ,medicine.medical_treatment ,animal diseases ,General Physics and Astronomy ,Adaptive Immunity ,CD8-Positive T-Lymphocytes ,Radiation Tolerance ,Mice ,0302 clinical medicine ,Interferon ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Immunosuppression ,hemic and immune systems ,Acquired immune system ,3. Good health ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Interferon Type I ,Female ,medicine.symptom ,Signal Transduction ,medicine.drug ,Receptors, CCR2 ,Science ,Inflammation ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immunity ,Cell Line, Tumor ,Radioresistance ,parasitic diseases ,medicine ,Animals ,Humans ,business.industry ,Myeloid-Derived Suppressor Cells ,Membrane Proteins ,General Chemistry ,Immunity, Innate ,eye diseases ,Blockade ,Mice, Inbred C57BL ,Sting ,030104 developmental biology ,Cancer research ,lcsh:Q ,business - Abstract
Radiotherapy induces and promotes innate and adaptive immunity in which host STING plays an important role. However, radioresistance in irradiated tumors can also develop, resulting in relapse. Here we report a mechanism by which extrinsic resistance develops after local ablative radiation that relies on the immunosuppressive action of STING. The STING/type I interferon pathway enhances suppressive inflammation in tumors by recruiting myeloid cells in part via the CCR2 pathway. Germ-line knockouts of CCR2 or treatment with an anti-CCR2 antibody results in blockade of radiation-induced MDSC infiltration. Treatment with anti-CCR2 antibody alleviates immunosuppression following activation of the STING pathway, enhancing the anti-tumor effects of STING agonists and radiotherapy. We propose that radiation-induced STING activation is immunosuppressive due to (monocytic) M-MDSC infiltration, which results in tumor radioresistance. Furthermore, the immunosuppressive effects of radiotherapy and STING agonists can be abrogated in humans by a translational strategy involving anti-CCR2 antibody treatment to improve radiotherapy., Tumors often develop resistance to radiotherapy. Here the authors show that irradiation leads to a CCR2-dependent infiltration by myeloid derived suppressor cells that promote radio-resistance through inhibition of adaptive immune responses and that the use of CCR2 antibodies in mice reduces such resistance.
- Published
- 2017
- Full Text
- View/download PDF
15. HMG-CoA Reductase Inhibition Delays DNA Repair and Promotes Senescence After Tumor Irradiation
- Author
-
Amy Catherine Flor, Elena V. Efimova, Harold G. Sutton, Natalia Ricco, Helena J. Mauceri, Stephen J. Kron, Edwardine Labay, Aishwarya Ramamurthy, and Ralph R. Weichselbaum
- Subjects
0301 basic medicine ,Cancer Research ,Radiosensitizer ,DNA Repair ,DNA repair ,medicine.medical_treatment ,Biology ,Pharmacology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Radioresistance ,medicine ,Animals ,Humans ,Pitavastatin ,Cellular Senescence ,Radiation therapy ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,HMG-CoA reductase ,Cancer cell ,biology.protein ,Protein prenylation ,Female ,Acyl Coenzyme A ,medicine.drug - Abstract
Despite significant advances in combinations of radiotherapy and chemotherapy, altered fractionation schedules and image-guided radiotherapy, many cancer patients fail to benefit from radiation. A prevailing hypothesis is that targeting repair of DNA double strand breaks (DSB) can enhance radiation effects in the tumor and overcome therapeutic resistance without incurring off-target toxicities. Unrepaired DSBs can block cancer cell proliferation, promote cancer cell death, and induce cellular senescence. Given the slow progress to date translating novel DSB repair inhibitors as radiosensitizers, we have explored drug repurposing, a proven route to improving speed, costs, and success rates of drug development. In a prior screen where we tracked resolution of ionizing radiation-induced foci (IRIF) as a proxy for DSB repair, we had identified pitavastatin (Livalo), an HMG-CoA reductase inhibitor commonly used for lipid lowering, as a candidate radiosensitizer. Here, we report that pitavastatin and other lipophilic statins are potent inhibitors of DSB repair in breast and melanoma models both in vitro and in vivo. When combined with ionizing radiation, pitavastatin increased persistent DSBs, induced senescence, and enhanced acute effects of radiation on radioresistant melanoma tumors. shRNA knockdown implicated HMG-CoA reductase, farnesyl diphosphate synthase, and protein farnesyl transferase in IRIF resolution, DSB repair, and senescence. These data confirm on-target activity of statins, although via inhibition of protein prenylation rather than cholesterol biosynthesis. In light of prior studies demonstrating enhanced efficacy of radiotherapy in patients taking statins, this work argues for clinical evaluation of lipophilic statins as nontoxic radiosensitizers to enhance the benefits of image-guided radiotherapy. Mol Cancer Ther; 17(2); 407–18. ©2017 AACR. See all articles in this MCT Focus section, “Developmental Therapeutics in Radiation Oncology.”
- Published
- 2017
16. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors
- Author
-
Byron Burnette, Zhijian J. Chen, Michael A. Beckett, Thomas E. Darga, Yang Xin Fu, Helena J. Mauceri, Liufu Deng, Xiaona Huang, Xiao Dong Li, Meng Xu, Xuanming Yang, Ralph R. Weichselbaum, Hua Liang, Thomas F. Gajewski, and Ainhoa Arina
- Subjects
Xanthones ,Immunology ,Antineoplastic Agents ,chemical and pharmacologic phenomena ,Receptor, Interferon alpha-beta ,Adaptive Immunity ,Biology ,Mice ,Cross-Priming ,Immune system ,Interferon ,RNA interference ,Immunity ,Neoplasms ,Radiation, Ionizing ,medicine ,Animals ,Immunology and Allergy ,RNA, Small Interfering ,Cells, Cultured ,Mice, Knockout ,Membrane Proteins ,Signal transducing adaptor protein ,DNA ,Dendritic Cells ,Interferon-beta ,Acquired immune system ,Nucleotidyltransferases ,Immunity, Innate ,eye diseases ,3. Good health ,Mice, Inbred C57BL ,Adaptor Proteins, Vesicular Transport ,Sting ,Infectious Diseases ,Stimulator of interferon genes ,Myeloid Differentiation Factor 88 ,Cancer research ,RNA Interference ,Nucleotides, Cyclic ,Signal Transduction ,medicine.drug - Abstract
Summary Ionizing radiation-mediated tumor regression depends on type I interferon (IFN) and the adaptive immune response, but several pathways control I IFN induction. Here, we demonstrate that adaptor protein STING, but not MyD88, is required for type I IFN-dependent antitumor effects of radiation. In dendritic cells (DCs), STING was required for IFN-β induction in response to irradiated-tumor cells. The cytosolic DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS) mediated sensing of irradiated-tumor cells in DCs. Moreover, STING was essential for radiation-induced adaptive immune responses, which relied on type I IFN signaling on DCs. Exogenous IFN-β treatment rescued the cross-priming by cGAS or STING-deficient DCs. Accordingly, activation of STING by a second messenger cGAMP administration enhanced antitumor immunity induced by radiation. Thus radiation-mediated antitumor immunity in immunogenic tumors requires a functional cytosolic DNA-sensing pathway and suggests that cGAMP treatment might provide a new strategy to improve radiotherapy.
- Published
- 2014
- Full Text
- View/download PDF
17. A Manganese Superoxide Dismutase (SOD2)-Mediated Adaptive Response
- Author
-
Jeffrey S. Murley, Ralph R. Weichselbaum, Harold G. Sutton, David J. Grdina, Richard C. Miller, Jian Jian Li, Gayle E. Woloschak, Michael J. Thirman, and Helena J. Mauceri
- Subjects
Alkylating Agents ,Cell Survival ,Biophysics ,SOD2 ,Genomic Instability ,Article ,Superoxide dismutase ,Mice ,Pregnancy ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Receptor ,Radiation ,biology ,Superoxide Dismutase ,Dose-Response Relationship, Radiation ,Transfection ,Amifostine ,Adaptation, Physiological ,Molecular biology ,Mercaptoethylamines ,Mice, Inbred C57BL ,Cell culture ,Ethylnitrosourea ,Knockout mouse ,biology.protein ,Female ,Tumor necrosis factor alpha ,Signal Transduction ,medicine.drug - Abstract
Very low doses of ionizing radiation, 5 to 100 mGy, can induce adaptive responses characterized by elevation in cell survival and reduction in micronuclei formation. Utilizing these end points, RKO human colon carcinoma and transformed mouse embryo fibroblasts (MEF), wild-type or knockout cells missing TNF receptors 1 and 2 (TNFR1(-)R2(-)), and C57BL/6 and TNFR1(-)R2(-) knockout mice, we demonstrate that intact TNF signaling is required for induction of elevated manganese superoxide dismutase (SOD2) activity (P < 0.001) and the subsequent expression of these SOD2-mediated adaptive responses when cells are challenged at a later time with 2 Gy. In contrast, amifostine's free thiol form WR1065 can directly activate NF-κB giving rise to elevated SOD2 activity 24 h later and induce an adaptive response in both MEF wild-type and TNF signaling defective TNFR1(-)R2(-) cells. Transfection of cells with SOD2 siRNA completely abolishes both the elevation in SOD2 activity and expression of the adaptive responses. These results were confirmed in vivo using a micronucleus assay in splenocytes derived from C57BL/6 and TNFR1(-)R2(-) knockout mice that were exposed to 100 mGy or 400 mg/kg amifostine 24 h prior to exposure to a 2 Gy whole-body dose. A dose of 100 mGy also conferred enhanced protection to C57BL/6 mice exposed 24 h later to 100 mg/kg of N-Ethyl-N-nitrosourea (ENU). While very low radiation doses require an intact TNF signaling process to induce a SOD2-mediated adaptive response, amifostine can induce a similar adaptive response in both TNF receptor competent and knockout cells, respectively.
- Published
- 2013
- Full Text
- View/download PDF
18. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part two
- Author
-
Casey Ager, Matthew Reilley, Courtney Nicholas, Todd Bartkowiak, Ashvin Jaiswal, Michael Curran, Tina C. Albershardt, Anshika Bajaj, Jacob F. Archer, Rebecca S. Reeves, Lisa Y. Ngo, Peter Berglund, Jan ter Meulen, Caroline Denis, Hormas Ghadially, Thomas Arnoux, Fabien Chanuc, Nicolas Fuseri, Robert W. Wilkinson, Nicolai Wagtmann, Yannis Morel, Pascale Andre, Michael B. Atkins, Matteo S. Carlino, Antoni Ribas, John A. Thompson, Toni K. Choueiri, F. Stephen Hodi, Wen-Jen Hwu, David F. McDermott, Victoria Atkinson, Jonathan S. Cebon, Bernie Fitzharris, Michael B. Jameson, Catriona McNeil, Andrew G. Hill, Eric Mangin, Malidi Ahamadi, Marianne van Vugt, Mariëlle van Zutphen, Nageatte Ibrahim, Georgina V. Long, Robyn Gartrell, Zoe Blake, Ines Simoes, Yichun Fu, Takuro Saito, Yingzhi Qian, Yan Lu, Yvonne M. Saenger, Sadna Budhu, Olivier De Henau, Roberta Zappasodi, Kyle Schlunegger, Bruce Freimark, Jeff Hutchins, Christopher A. Barker, Jedd D. Wolchok, Taha Merghoub, Elena Burova, Omaira Allbritton, Peter Hong, Jie Dai, Jerry Pei, Matt Liu, Joel Kantrowitz, Venus Lai, William Poueymirou, Douglas MacDonald, Ella Ioffe, Markus Mohrs, William Olson, Gavin Thurston, Cristian Capasso, Federica Frascaro, Sara Carpi, Siri Tähtinen, Sara Feola, Manlio Fusciello, Karita Peltonen, Beatriz Martins, Madeleine Sjöberg, Sari Pesonen, Tuuli Ranki, Lukasz Kyruk, Erkko Ylösmäki, Vincenzo Cerullo, Fabio Cerignoli, Biao Xi, Garret Guenther, Naichen Yu, Lincoln Muir, Leyna Zhao, Yama Abassi, Víctor Cervera-Carrascón, Mikko Siurala, João Santos, Riikka Havunen, Suvi Parviainen, Akseli Hemminki, Angus Dalgleish, Satvinder Mudan, Mark DeBenedette, Ana Plachco, Alicia Gamble, Elizabeth W. Grogan, John Krisko, Irina Tcherepanova, Charles Nicolette, Pooja Dhupkar, Ling Yu, Eugenie S. Kleinerman, Nancy Gordon, Italia Grenga, Lauren Lepone, Sofia Gameiro, Karin M. Knudson, Massimo Fantini, Kwong Tsang, James Hodge, Renee Donahue, Jeffrey Schlom, Elizabeth Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Cathie Foster, Alan Howell, Leslie Balch, Alyssa Knapp, John E. Leonard, Mark Paris, Terry Fisher, Siwen Hu-Lieskovan, Ernest Smith, Maurice Zauderer, William Fogler, Marilyn Franklin, Matt Thayer, Dan Saims, John L. Magnani, Jian Gong, Michael Gray, George Fromm, Suresh de Silva, Louise Giffin, Xin Xu, Jason Rose, Taylor H. Schreiber, Sofia R. Gameiro, Paul E. Clavijo, Clint T. Allen, James W. Hodge, Kwong Y. Tsang, Jane Grogan, Nicholas Manieri, Eugene Chiang, Patrick Caplazi, Mahesh Yadav, Patrick Hagner, Hsiling Chiu, Michelle Waldman, Anke Klippel, Anjan Thakurta, Michael Pourdehnad, Anita Gandhi, Ian Henrich, Laura Quick, Rob Young, Margaret Chou, Andrew Hotson, Stephen Willingham, Po Ho, Carmen Choy, Ginna Laport, Ian McCaffery, Richard Miller, Kimberly A. Tipton, Kenneth R. Wong, Victoria Singson, Chihunt Wong, Chanty Chan, Yuanhiu Huang, Shouchun Liu, Jennifer H. Richardson, W. Michael Kavanaugh, James West, Bryan A. Irving, Ritika Jaini, Matthew Loya, Charis Eng, Melissa L. Johnson, Alex A. Adjei, Mateusz Opyrchal, Suresh Ramalingam, Pasi A. Janne, George Dominguez, Dmitry Gabrilovich, Laura de Leon, Jeannette Hasapidis, Scott J. Diede, Peter Ordentlich, Scott Cruickshank, Michael L. Meyers, Matthew D. Hellmann, Pawel Kalinski, Amer Zureikat, Robert Edwards, Ravi Muthuswamy, Nataša Obermajer, Julie Urban, Lisa H. Butterfield, William Gooding, Herbert Zeh, David Bartlett, Olga Zubkova, Larissa Agapova, Marina Kapralova, Liudmila Krasovskaia, Armen Ovsepyan, Maxim Lykov, Artem Eremeev, Vladimir Bokovanov, Olga Grigoryeva, Andrey Karpov, Sergey Ruchko, Alexandr Shuster, Danny N. Khalil, Luis Felipe Campesato, Yanyun Li, Adam S. Lazorchak, Troy D. Patterson, Yueyun Ding, Pottayil Sasikumar, Naremaddepalli Sudarshan, Nagaraj Gowda, Raghuveer Ramachandra, Dodheri Samiulla, Sanjeev Giri, Rajesh Eswarappa, Murali Ramachandra, David Tuck, Timothy Wyant, Jasmin Leshem, Xiu-fen Liu, Tapan Bera, Masaki Terabe, Birgit Bossenmaier, Gerhard Niederfellner, Yoram Reiter, Ira Pastan, Leiming Xia, Yang Xia, Yangyang Hu, Yi Wang, Yangyi Bao, Fu Dai, Shiang Huang, Elaine Hurt, Robert E. Hollingsworth, Lawrence G. Lum, Alfred E. Chang, Max S. Wicha, Qiao Li, Thomas Mace, Neil Makhijani, Erin Talbert, Gregory Young, Denis Guttridge, Darwin Conwell, Gregory B. Lesinski, Rodney JM Macedo Gonzales, Austin P. Huffman, Ximi K. Wang, Ran Reshef, Andy MacKinnon, Jason Chen, Matt Gross, Gisele Marguier, Peter Shwonek, Natalija Sotirovska, Susanne Steggerda, Francesco Parlati, Amani Makkouk, Mark K. Bennett, Ethan Emberley, Tony Huang, Weiqun Li, Silinda Neou, Alison Pan, Jing Zhang, Winter Zhang, Netonia Marshall, Thomas U. Marron, Judith Agudo, Brian Brown, Joshua Brody, Christopher McQuinn, Matthew Farren, Hannah Komar, Reena Shakya, Thomas Ludwug, Y. Maurice Morillon, Scott A. Hammond, John W. Greiner, Pulak R. Nath, Anthony L. Schwartz, Dragan Maric, David D. Roberts, Aung Naing, Kyriakos P. Papadopoulos, Karen A. Autio, Deborah J. Wong, Manish Patel, Gerald Falchook, Shubham Pant, Patrick A. Ott, Melinda Whiteside, Amita Patnaik, John Mumm, Filip Janku, Ivan Chan, Todd Bauer, Rivka Colen, Peter VanVlasselaer, Gail L. Brown, Nizar M. Tannir, Martin Oft, Jeffrey Infante, Evan Lipson, Ajay Gopal, Sattva S. Neelapu, Philippe Armand, Stephen Spurgeon, John P. Leonard, Rachel E. Sanborn, Ignacio Melero, Thomas F. Gajewski, Matthew Maurer, Serena Perna, Andres A. Gutierrez, Raphael Clynes, Priyam Mitra, Satyendra Suryawanshi, Douglas Gladstone, Margaret K. Callahan, James Crooks, Sheila Brown, Audrey Gauthier, Marc Hillairet de Boisferon, Andrew MacDonald, Laura Rosa Brunet, William T. Rothwell, Peter Bell, James M. Wilson, Fumi Sato-Kaneko, Shiyin Yao, Shannon S. Zhang, Dennis A. Carson, Cristina Guiducci, Robert L. Coffman, Kazutaka Kitaura, Takaji Matsutani, Ryuji Suzuki, Tomoko Hayashi, Ezra E. W. Cohen, David Schaer, Yanxia Li, Julie Dobkin, Michael Amatulli, Gerald Hall, Thompson Doman, Jason Manro, Frank Charles Dorsey, Lillian Sams, Rikke Holmgaard, Krishnadatt Persaud, Dale Ludwig, David Surguladze, John S. Kauh, Ruslan Novosiadly, Michael Kalos, Kyla Driscoll, Hardev Pandha, Christy Ralph, Kevin Harrington, Brendan Curti, Wallace Akerley, Sumati Gupta, Alan Melcher, David Mansfield, David R. Kaufman, Emmett Schmidt, Mark Grose, Bronwyn Davies, Roberta Karpathy, Darren Shafren, Katerina Shamalov, Cyrille Cohen, Naveen Sharma, James Allison, Tala Shekarian, Sandrine Valsesia-Wittmann, Christophe Caux, Aurelien Marabelle, Brian M. Slomovitz, Kathleen M. Moore, Hagop Youssoufian, Marshall Posner, Poonam Tewary, Alan D. Brooks, Ya-Ming Xu, Kithsiri Wijeratne, Leslie A. A. Gunatilaka, Thomas J. Sayers, John P. Vasilakos, Tesha Alston, Simon Dovedi, James Elvecrog, Iwen Grigsby, Ronald Herbst, Karen Johnson, Craig Moeckly, Stefanie Mullins, Kristen Siebenaler, Julius SternJohn, Ashenafi Tilahun, Mark A. Tomai, Katharina Vogel, Eveline E. Vietsch, Anton Wellstein, Martin Wythes, Stefano Crosignani, Joseph Tumang, Shilpa Alekar, Patrick Bingham, Sandra Cauwenberghs, Jenny Chaplin, Deepak Dalvie, Sofie Denies, Coraline De Maeseneire, JunLi Feng, Kim Frederix, Samantha Greasley, Jie Guo, James Hardwick, Stephen Kaiser, Katti Jessen, Erick Kindt, Marie-Claire Letellier, Wenlin Li, Karen Maegley, Reece Marillier, Nichol Miller, Brion Murray, Romain Pirson, Julie Preillon, Virginie Rabolli, Chad Ray, Kevin Ryan, Stephanie Scales, Jay Srirangam, Jim Solowiej, Al Stewart, Nicole Streiner, Vince Torti, Konstantinos Tsaparikos, Xianxian Zheng, Gregory Driessens, Bruno Gomes, Manfred Kraus, Chunxiao Xu, Yanping Zhang, Giorgio Kradjian, Guozhong Qin, Jin Qi, Xiaomei Xu, Bo Marelli, Huakui Yu, Wilson Guzman, Rober Tighe, Rachel Salazar, Kin-Ming Lo, Jessie English, Laszlo Radvanyi, Yan Lan, Michael Postow, Yasin Senbabaoglu, Billel Gasmi, Hong Zhong, Cailian Liu, Daniel Hirschhorhn-Cymerman, Yuanyuan Zha, Gregory Malnassy, Noreen Fulton, Jae-Hyun Park, Wendy Stock, Yusuke Nakamura, Hongtao Liu, Xiaoming Ju, Rachelle Kosoff, Kimberly Ramos, Brandon Coder, Robert Petit, Michael Princiotta, Kyle Perry, Jun Zou, Ainhoa Arina, Christian Fernandez, Wenxin Zheng, Michael A. Beckett, Helena J. Mauceri, Yang-Xin Fu, Ralph R. Weichselbaum, Whitney Lewis, Yanyan Han, Yeting Wu, Chou Yang, Jing Huang, Dongyun Wu, Jin Li, Xiaoling Liang, Xiangjun Zhou, Jinlin Hou, Raffit Hassan, Thierry Jahan, Scott J. Antonia, Hedy L. Kindler, Evan W. Alley, Somayeh Honarmand, Weiqun Liu, Meredith L. Leong, Chan C. Whiting, Nitya Nair, Amanda Enstrom, Edward E. Lemmens, Takahiro Tsujikawa, Sushil Kumar, Lisa M. Coussens, Aimee L. Murphy, Dirk G. Brockstedt, Sven D. Koch, Martin Sebastian, Christian Weiss, Martin Früh, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geissler, Frank Griesinger, Jens Kollmeier, Alexandros Papachristofilou, Fatma Doener, Mariola Fotin-Mleczek, Madeleine Hipp, Henoch S. Hong, Karl-Josef Kallen, Ute Klinkhardt, Claudia Stosnach, Birgit Scheel, Andreas Schroeder, Tobias Seibel, Ulrike Gnad-Vogt, Alfred Zippelius, Ha-Ram Park, Yong-Oon Ahn, Tae Min Kim, Soyeon Kim, Seulki Kim, Yu Soo Lee, Bhumsuk Keam, Dong-Wan Kim, Dae Seog Heo, Shari Pilon-Thomas, Amy Weber, Jennifer Morse, Krithika Kodumudi, Hao Liu, John Mullinax, Amod A. Sarnaik, Luke Pike, Andrew Bang, Tracy Balboni, Allison Taylor, Alexander Spektor, Tyler Wilhite, Monica Krishnan, Daniel Cagney, Brian Alexander, Ayal Aizer, Elizabeth Buchbinder, Mark Awad, Leena Ghandi, Jonathan Schoenfeld, Elizabeth Lessey-Morillon, Lisa Ridnour, Neil H. Segal, Manish Sharma, Dung T. Le, Robert L. Ferris, Andrew D. Zelenetz, Ronald Levy, Izidore S. Lossos, Caron Jacobson, Radhakrishnan Ramchandren, John Godwin, A. Dimitrios Colevas, Roland Meier, Suba Krishnan, Xuemin Gu, Jaclyn Neely, John Timmerman, Claire I. Vanpouille-Box, Silvia C. Formenti, Sandra Demaria, Erik Wennerberg, Aranzazu Mediero, Bruce N. Cronstein, Michael P. Gustafson, AriCeli DiCostanzo, Courtney Wheatley, Chul-Ho Kim, Svetlana Bornschlegl, Dennis A. Gastineau, Bruce D. Johnson, Allan B. Dietz, Cameron MacDonald, Mark Bucsek, Guanxi Qiao, Bonnie Hylander, Elizabeth Repasky, William J. Turbitt, Yitong Xu, Andrea Mastro, Connie J. Rogers, Sita Withers, Ziming Wang, Lam T. Khuat, Cordelia Dunai, Bruce R. Blazar, Dan Longo, Robert Rebhun, Steven K. Grossenbacher, Arta Monjazeb, William J. Murphy, Scott Rowlinson, Giulia Agnello, Susan Alters, David Lowe, Nicole Scharping, Ashley V. Menk, Ryan Whetstone, Xue Zeng, Greg M. Delgoffe, Patricia M. Santos, Jian Shi, Greg Delgoffe, Misako Nagasaka, Ammar Sukari, Miranda Byrne-Steele, Wenjing Pan, Xiaohong Hou, Brittany Brown, Mary Eisenhower, Jian Han, Natalie Collins, Robert Manguso, Hans Pope, Yashaswi Shrestha, Jesse Boehm, W. Nicholas Haining, Kyle R. Cron, Ayelet Sivan, Keston Aquino-Michaels, Marco Orecchioni, Davide Bedognetti, Wouter Hendrickx, Claudia Fuoco, Filomena Spada, Francesco Sgarrella, Gianni Cesareni, Francesco Marincola, Kostas Kostarelos, Alberto Bianco, Lucia Delogu, Jessica Roelands, Sabri Boughorbel, Julie Decock, Scott Presnell, Ena Wang, Franco M. Marincola, Peter Kuppen, Michele Ceccarelli, Darawan Rinchai, Damien Chaussabel, Lance Miller, Andrew Nguyen, J. Zachary Sanborn, Charles Vaske, Shahrooz Rabizadeh, Kayvan Niazi, Steven Benz, Shashank Patel, Nicholas Restifo, James White, Sam Angiuoli, Mark Sausen, Sian Jones, Maria Sevdali, John Simmons, Victor Velculescu, Luis Diaz, Theresa Zhang, Jennifer S. Sims, Sunjay M. Barton, Angela Kadenhe-Chiweshe, Filemon Dela Cruz, Andrew T. Turk, Christopher F. Mazzeo, Andrew L. Kung, Jeffrey N. Bruce, Darrell J. Yamashiro, Eileen P. Connolly, Jason Baird, Marka Crittenden, David Friedman, Hong Xiao, Rom Leidner, Bryan Bell, Kristina Young, Michael Gough, Zhen Bian, Koby Kidder, Yuan Liu, Emily Curran, Xiufen Chen, Leticia P. Corrales, Justin Kline, Ethan G. Aguilar, Jennifer Guerriero, Alaba Sotayo, Holly Ponichtera, Alexandra Pourzia, Sara Schad, Ruben Carrasco, Suzan Lazo, Roderick Bronson, Anthony Letai, Richard S. Kornbluth, Sachin Gupta, James Termini, Elizabeth Guirado, Geoffrey W. Stone, Christina Meyer, Laura Helming, Nicholas Wilson, Robert Hofmeister, Natalie J. Neubert, Laure Tillé, David Barras, Charlotte Soneson, Petra Baumgaertner, Donata Rimoldi, David Gfeller, Mauro Delorenzi, Silvia A. Fuertes Marraco, Daniel E. Speiser, Tara S. Abraham, Bo Xiang, Michael S. Magee, Scott A. Waldman, Adam E. Snook, Wojciech Blogowski, Ewa Zuba-Surma, Marta Budkowska, Daria Salata, Barbara Dolegowska, Teresa Starzynska, Leo Chan, Srinivas Somanchi, Kelsey McCulley, Dean Lee, Nico Buettner, Feng Shi, Paisley T. Myers, Stuart Curbishley, Sarah A. Penny, Lora Steadman, David Millar, Ellen Speers, Nicola Ruth, Gabriel Wong, Robert Thimme, David Adams, Mark Cobbold, Remy Thomas, Mariam Al-Muftah, Michael KK Wong, Michael Morse, Joseph I. Clark, Howard L. Kaufman, Gregory A. Daniels, Hong Hua, Tharak Rao, Janice P. Dutcher, Kai Kang, Yogen Saunthararajah, Vamsidhar Velcheti, Vikas Kumar, Firoz Anwar, Amita Verma, Zinal Chheda, Gary Kohanbash, John Sidney, Kaori Okada, Shruti Shrivastav, Diego A. Carrera, Shuming Liu, Naznin Jahan, Sabine Mueller, Ian F. Pollack, Angel M. Carcaboso, Alessandro Sette, Yafei Hou, Hideho Okada, Jessica J. Field, Weiping Zeng, Vincent FS Shih, Che-Leung Law, Peter D. Senter, Shyra J. Gardai, Nicole M. Okeley, Jennifer G. Abelin, Abu Z. Saeed, Stacy A. Malaker, Jeffrey Shabanowitz, Stephen T. Ward, Donald F. Hunt, Pam Profusek, Laura Wood, Dale Shepard, Petros Grivas, Kerstin Kapp, Barbara Volz, Detlef Oswald, Burghardt Wittig, Manuel Schmidt, Julian P. Sefrin, Lars Hillringhaus, Valeria Lifke, Alexander Lifke, Anna Skaletskaya, Jose Ponte, Thomas Chittenden, Yulius Setiady, Eva Sivado, Vincent Thomas, Meddy El Alaoui, Sébastien Papot, Charles Dumontet, Mike Dyson, John McCafferty, Said El Alaoui, Praveen K. Bommareddy, Andrew Zloza, Frederick Kohlhapp, Ann W. Silk, Sachin Jhawar, Tomas Paneque, Jenna Newman, Pedro Beltran, Felicia Cao, Bang-Xing Hong, Tania Rodriguez-Cruz, Xiao-Tong Song, Stephen Gottschalk, Hugo Calderon, Sam Illingworth, Alice Brown, Kerry Fisher, Len Seymour, Brian Champion, Emma Eriksson, Jessica Wenthe, Ann-Charlotte Hellström, Gabriella Paul-Wetterberg, Angelica Loskog, Ioanna Milenova, Magnus Ståhle, Justyna Jarblad-Leja, Gustav Ullenhag, Anna Dimberg, Rafael Moreno, Ramon Alemany, Sharad Goyal, Ann Silk, Janice Mehnert, Nashat Gabrail, Jennifer Bryan, Daniel Medina, Leah Mitchell, Kader Yagiz, Fernando Lopez, Daniel Mendoza, Anthony Munday, Harry Gruber, Douglas Jolly, Steven Fuhrmann, Sasa Radoja, Wei Tan, Aldo Pourchet, Alan Frey, Ian Mohr, Matthew Mulvey, Robert H. I. Andtbacka, Merrick Ross, Sanjiv Agarwala, Kenneth Grossmann, Matthew Taylor, John Vetto, Rogerio Neves, Adil Daud, Hung Khong, Stephanie M. Meek, Richard Ungerleider, Scott Welden, Maki Tanaka, Matthew Williams, Sigrun Hallmeyer, Bernard Fox, Zipei Feng, Christopher Paustian, Carlo Bifulco, Sadia Zafar, Otto Hemminki, Simona Bramante, Lotta Vassilev, Hongjie Wang, Andre Lieber, Silvio Hemmi, Tanja de Gruijl, Anna Kanerva, Tameem Ansari, Srividya Sundararaman, Diana Roen, Paul Lehmann, Anja C. Bloom, Lewis H. Bender, Ian B. Walters, Jay A. Berzofsky, Fanny Chapelin, Eric T. Ahrens, Jeff DeFalco, Michael Harbell, Amy Manning-Bog, Alexander Scholz, Danhui Zhang, Gilson Baia, Yann Chong Tan, Jeremy Sokolove, Dongkyoon Kim, Kevin Williamson, Xiaomu Chen, Jillian Colrain, Gregg Espiritu Santo, Ngan Nguyen, Wayne Volkmuth, Norman Greenberg, William Robinson, Daniel Emerling, Charles G. Drake, Daniel P. Petrylak, Emmanuel S. Antonarakis, Adam S. Kibel, Nancy N. Chang, Tuyen Vu, Dwayne Campogan, Heather Haynes, James B. Trager, Nadeem A. Sheikh, David I. Quinn, Peter Kirk, Murali Addepalli, Thomas Chang, Ping Zhang, Marina Konakova, Katsunobu Hagihara, Steven Pai, Laurie VanderVeen, Palakshi Obalapur, Peiwen Kuo, Phi Quach, Lawrence Fong, Deborah H. Charych, Jonathan Zalevsky, John L. Langowski, Yolanda Kirksey, Ravi Nutakki, Shalini Kolarkar, Rhoneil Pena, Ute Hoch, Stephen K. Doberstein, John Cha, Zach Mallon, Myra Perez, Amanda McDaniel, Snjezana Anand, Darrin Uecker, Richard Nuccitelli, Eva Wieckowski, Ravikumar Muthuswamy, Roshni Ravindranathan, Ariana N. Renrick, Menaka Thounaojam, Portia Thomas, Samuel Pellom, Anil Shanker, Duafalia Dudimah, Alan Brooks, Yu-Lin Su, Tomasz Adamus, Qifang Zhang, Sergey Nechaev, Marcin Kortylewski, Spencer Wei, Clark Anderson, Chad Tang, Jonathan Schoenhals, Efrosini Tsouko, John Heymach, Patricia de Groot, Joe Chang, Kenneth R. Hess, Adi Diab, Padmanee Sharma, David Hong, James Welsh, Andrea J. Parsons, Jardin Leleux, Stephane Ascarateil, Marie Eve Koziol, Dina Bai, Peihong Dai, Weiyi Wang, Ning Yang, Stewart Shuman, Liang Deng, Patrick Dillon, Gina Petroni, David Brenin, Kim Bullock, Walter Olson, Mark E. Smolkin, Kelly Smith, Carmel Nail, Craig L. Slingluff, Meenu Sharma, Faisal Fa’ak, Louise Janssen, Hiep Khong, Zhilan Xiao, Yared Hailemichael, Manisha Singh, Christina Vianden, Willem W. Overwijk, Andrea Facciabene, Pierini Stefano, Fang Chongyung, Stavros Rafail, Michael Nielsen, Peter Vanderslice, Darren G. Woodside, Robert V. Market, Ronald J. Biediger, Upendra K. Marathi, Kevin Hollevoet, Nick Geukens, Paul Declerck, Nathalie Joly, Laura McIntosh, Eustache Paramithiotis, Magnus Rizell, Malin Sternby, Bengt Andersson, Alex Karlsson-Parra, Rui Kuai, Lukasz Ochyl, Anna Schwendeman, James Moon, Weiwen Deng, Thomas E. Hudson, Bill Hanson, Chris S. Rae, Joel Burrill, Justin Skoble, George Katibah, Michele deVries, Peter Lauer, Thomas W. Dubensky, Xin Chen, Li Zhou, Xiubao Ren, Charu Aggarwal, Drishty Mangrolia, Roger Cohen, Gregory Weinstein, Matthew Morrow, Joshua Bauml, Kim Kraynyak, Jean Boyer, Jian Yan, Jessica Lee, Laurent Humeau, Sandra Oyola, Susan Duff, David Weiner, Zane Yang, Mark Bagarazzi, Douglas G. McNeel, Jens Eickhoff, Robert Jeraj, Mary Jane Staab, Jane Straus, Brian Rekoske, Glenn Liu, Marit Melssen, William Grosh, Nikole Varhegyi, Nadejda Galeassi, Donna H. Deacon, Elizabeth Gaughan, Maurizio Ghisoli, Minal Barve, Robert Mennel, Gladice Wallraven, Luisa Manning, Neil Senzer, John Nemunaitis, Masahiro Ogasawara, Shuichi Ota, Kaitlin M. Peace, Diane F. Hale, Timothy J. Vreeland, Doreen O. Jackson, John S. Berry, Alfred F. Trappey, Garth S. Herbert, Guy T. Clifton, Mark O. Hardin, Anne Toms, Na Qiao, Jennifer Litton, George E. Peoples, Elizabeth A. Mittendorf, Lila Ghamsari, Emilio Flano, Judy Jacques, Biao Liu, Jonathan Havel, Vladimir Makarov, Timothy A. Chan, Jessica B. Flechtner, John Facciponte, Stefano Ugel, Francesco De Sanctis, George Coukos, Sébastien Paris, Agnes Pottier, Laurent Levy, Bo Lu, Federica Cappuccini, Emily Pollock, Richard Bryant, Freddie Hamdy, Adrian Hill, Irina Redchenko, Hussein Sultan, Takumi Kumai, Valentyna Fesenkova, Esteban Celis, Ingrid Fernando, Claudia Palena, Justin M. David, Elizabeth Gabitzsch, Frank Jones, James L. Gulley, Mireia Uribe Herranz, Hiroshi Wada, Atsushi Shimizu, Toshihiro Osada, Satoshi Fukaya, Eiji Sasaki, Milad Abolhalaj, David Askmyr, Kristina Lundberg, Ann-Sofie Albrekt, Lennart Greiff, Malin Lindstedt, Dallas B. Flies, Tomoe Higuchi, Wojciech Ornatowski, Jaryse Harris, Sarah F. Adams, Todd Aguilera, Marjan Rafat, Laura Castellini, Hussein Shehade, Mihalis Kariolis, Dadi Jang, Rie vonEbyen, Edward Graves, Lesley Ellies, Erinn Rankin, Albert Koong, Amato Giaccia, Reham Ajina, Shangzi Wang, Jill Smith, Mariaelena Pierobon, Sandra Jablonski, Emanuel Petricoin, Louis M. Weiner, Lorcan Sherry, John Waller, Mark Anderson, Alison Bigley, Chantale Bernatchez, Cara Haymaker, Harriet Kluger, Michael Tetzlaff, Natalie Jackson, Ivan Gergel, Mary Tagliaferri, Patrick Hwu, Mario Snzol, Michael Hurwitz, Theresa Barberi, Allison Martin, Rahul Suresh, David Barakat, Sarah Harris-Bookman, Charles Drake, Alan Friedman, Sara Berkey, Stephanie Downs-Canner, Robert P. Edwards, Tyler Curiel, Kunle Odunsi, Tullia C. Bruno, Brandon Moore, Olivia Squalls, Peggy Ebner, Katherine Waugh, John Mitchell, Wilbur Franklin, Daniel Merrick, Martin McCarter, Brent Palmer, Jeffrey Kern, Dario Vignali, Jill Slansky, Anissa S. H. Chan, Xiaohong Qiu, Kathryn Fraser, Adria Jonas, Nadine Ottoson, Keith Gordon, Takashi O. Kangas, Steven Leonardo, Kathleen Ertelt, Richard Walsh, Mark Uhlik, Jeremy Graff, Nandita Bose, Ravi Gupta, Nitin Mandloi, Kiran Paul, Ashwini Patil, Rekha Sathian, Aparna Mohan, Malini Manoharan, Amitabha Chaudhuri, Yu Chen, Jing Lin, Yun-bin Ye, Chun-wei Xu, Gang Chen, Zeng-qing Guo, Andrey Komarov, Alex Chenchik, Michael Makhanov, Costa Frangou, Yi Zheng, Carla Coltharp, Darryn Unfricht, Ryan Dilworth, Leticia Fridman, Linying Liu, Milind Rajopadhye, Peter Miller, Fernando Concha-Benavente, Julie Bauman, Sumita Trivedi, Raghvendra Srivastava, James Ohr, Dwight Heron, Uma Duvvuri, Seungwon Kim, Heather Torrey, Toshi Mera, Yoshiaki Okubo, Eva Vanamee, Rosemary Foster, Denise Faustman, Edward Stack, Daisuke Izaki, Kristen Beck, Dan Tong Jia, Paul Armenta, Ashley White-Stern, Douglas Marks, Bret Taback, Basil Horst, Laura Hix Glickman, David B. Kanne, Kelsey S. Gauthier, Anthony L. Desbien, Brian Francica, Justin L. Leong, Leonard Sung, Ken Metchette, Shailaja Kasibhatla, Anne Marie Pferdekamper, Lianxing Zheng, Charles Cho, Yan Feng, Jeffery M. McKenna, John Tallarico, Steven Bender, Chudi Ndubaku, Sarah M. McWhirter, Elena Gonzalez Gugel, Charles J. M. Bell, Adiel Munk, Luciana Muniz, Nina Bhardwaj, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nathalie Scholler, Catherine Yin, Pien Van der Meijs, Andrew M. Prantner, Cecile M. Krejsa, Leia Smith, Brian Johnson, Daniel Branstetter, Paul L. Stein, Juan C. Jaen, Joanne BL Tan, Ada Chen, Timothy Park, Jay P. Powers, Holly Sexton, Guifen Xu, Steve W. Young, Ulrike Schindler, Wentao Deng, David John Klinke, Hannah M. Komar, Gregory Serpa, Omar Elnaggar, Philip Hart, Carl Schmidt, Mary Dillhoff, Ming Jin, Michael C. Ostrowski, Madhuri Koti, Katrina Au, Nichole Peterson, Peter Truesdell, Gillian Reid-Schachter, Charles Graham, Andrew Craig, Julie-Ann Francis, Beatrix Kotlan, Timea Balatoni, Emil Farkas, Laszlo Toth, Mihaly Ujhelyi, Akos Savolt, Zoltan Doleschall, Szabolcs Horvath, Klara Eles, Judit Olasz, Orsolya Csuka, Miklos Kasler, Gabriella Liszkay, Eytan Barnea, Collin Blakely, Patrick Flynn, Reid Goodman, Raphael Bueno, David Sugarbaker, David Jablons, V. Courtney Broaddus, Brian West, Paul R. Kunk, Joseph M. Obeid, Kevin Winters, Patcharin Pramoonjago, Edward B. Stelow, Todd W. Bauer, Osama E. Rahma, Adam Lamble, Yoko Kosaka, Fei Huang, Kate A. Saser, Homer Adams, Christina E. Tognon, Ted Laderas, Shannon McWeeney, Marc Loriaux, Jeffery W. Tyner, Brian J. Druker, Evan F. Lind, Zhuqing Liu, Shanhong Lu, Lawrence P. Kane, Gulidanna Shayan, Julia Femel, Ryan Lane, Jamie Booth, Amanda W. Lund, Anthony Rodriguez, Victor H. Engelhard, Alessandra Metelli, Bill X. Wu, Caroline W. Fugle, Rachidi Saleh, Shaoli Sun, Jennifer Wu, Bei Liu, Zihai Li, Zachary S. Morris, Emily I. Guy, Clinton Heinze, Jasdeep Kler, Monica M. Gressett, Lauryn R. Werner, Stephen D. Gillies, Alan J. Korman, Hans Loibner, Jacquelyn A. Hank, Alexander L. Rakhmilevich, Paul M. Harari, Paul M. Sondel, Erica Huelsmann, Joseph Broucek, Dorothee Brech, Tobias Straub, Martin Irmler, Johannes Beckers, Florian Buettner, Elke Schaeffeler, Matthias Schwab, Elfriede Noessner, Alison Wolfreys, Andre Da Costa, John Silva, Andrea Crosby, Ludovicus Staelens, Graham Craggs, Annick Cauvin, Sean Mason, Alison M. Paterson, Andrew C. Lake, Caroline M. Armet, Rachel W. O’Connor, Jonathan A. Hill, Emmanuel Normant, Ammar Adam, Detlev M. Biniszkiewicz, Scott C. Chappel, Vito J. Palombella, Pamela M. Holland, Annette Becker, Manmohan R. Leleti, Eric Newcomb, Joanne B. L. Tan, Suthee Rapisuwon, Arash Radfar, Kellie Gardner, Geoffrey Gibney, Michael Atkins, Keith R. Rennier, Robert Crowder, Ping Wang, Russell K. Pachynski, Rosa M. Santana Carrero, Sarai Rivas, Figen Beceren-Braun, Scott Anthony, Kimberly S. Schluns, Deepali Sawant, Maria Chikina, Hiroshi Yano, Creg Workman, Elise Salerno, Ileana Mauldin, Donna Deacon, Sofia Shea, Joel Pinczewski, Thomas Gajewski, Stefani Spranger, Brendan Horton, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Raj K. Puri, Randy F. Sweis, Riyue Bao, Jason Luke, Marie-Nicole Theodoraki, Frances-Mary Mogundo, Haejung Won, Dayson Moreira, Chan Gao, Xingli Zhao, Priyanka Duttagupta, Jeremy Jones, Massimo D’Apuzzo, and Sumanta Pal
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business - Abstract
O1 IL-15 primes an mTOR-regulated gene-expression program to prolong anti-tumor capacity of human natural killer cells #### Andreas Lundqvist1, Vincent van Hoef1, Xiaonan Zhang1, Erik Wennerberg2, Julie Lorent1, Kristina Witt1, Laia Masvidal Sanz1, Shuo Liang1, Shannon Murray3, Ola Larsson1
- Published
- 2016
- Full Text
- View/download PDF
19. Repurposing cephalosporin antibiotics as pro-senescent radiosensitizers
- Author
-
Amy Catherine Flor, Edwardine Labay, Helena J. Mauceri, Stephen J. Kron, Ralph R. Weichselbaum, Elena V. Efimova, and Harold G. Sutton
- Subjects
0301 basic medicine ,Radiosensitizer ,Radiation-Sensitizing Agents ,Time Factors ,senescence ,medicine.drug_class ,DNA damage ,medicine.medical_treatment ,Antibiotics ,cephalosporin ,Breast Neoplasms ,Toxicology ,03 medical and health sciences ,0302 clinical medicine ,Therapeutic index ,In vivo ,medicine ,Animals ,Humans ,Cellular Senescence ,Cephalosporin Antibiotic ,Cell Proliferation ,reactive oxygen species ,radiosensitizer ,Dose-Response Relationship, Drug ,drug repurposing ,business.industry ,Drug Repositioning ,Dose-Response Relationship, Radiation ,Xenograft Model Antitumor Assays ,3. Good health ,Anti-Bacterial Agents ,Cephalosporins ,Tumor Burden ,Radiation therapy ,Mice, Inbred C57BL ,Drug repositioning ,Oxidative Stress ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,MCF-7 Cells ,Female ,business ,DNA Damage ,Research Paper - Abstract
Radiation therapy remains a significant therapeutic modality in the treatment of cancer. An attractive strategy would be to enhance the benefits of ionizing radiation (IR)with radiosensitizers. A high-content drug repurposing screen of approved and investigational agents, natural products and other small molecules has identified multiple candidates that blocked repair of IR damage in vitro. Here, we validated a subset of these hits in vitro and then examined effects on tumor growth after IR in a murine tumor model. Based on robust radiosensitization in vivo and other favorable properties of cephalexin, we conducted additional studies with other beta-lactam antibiotics. When combined with IR, each cephalosporin tested increased DNA damage and slowed tumor growth without affecting normal tissue toxicity. Our data implicate reactive oxygen species in the mechanism by which cephalosporins augment the effects of IR. This work provides a rationale for using commonly prescribed beta-lactam antibiotics as non-toxic radiosensitizers to enhance the therapeutic ratio of radiotherapy.
- Published
- 2016
20. Response of Human Prostate Cancer Cells and Tumors to Combining PARP Inhibition with Ionizing Radiation
- Author
-
Elena V. Efimova, Everett E. Vokes, Juan Camilo Barreto-Andrade, Harold G. Sutton, Helena J. Mauceri, Michael A. Beckett, Mitchell C. Posner, Thomas E. Darga, Stephen J. Kron, and Ralph R. Weichselbaum
- Subjects
Male ,Senescence ,Aging ,Cancer Research ,Veliparib ,DNA damage ,medicine.medical_treatment ,Mice, Nude ,Poly(ADP-ribose) Polymerase Inhibitors ,Biology ,Poly (ADP-Ribose) Polymerase Inhibitor ,Article ,Mice ,chemistry.chemical_compound ,Prostate cancer ,Cell Line, Tumor ,Radiation, Ionizing ,medicine ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Enzyme Inhibitors ,Prostatic Neoplasms ,medicine.disease ,Xenograft Model Antitumor Assays ,Tumor Burden ,Radiation therapy ,Oncology ,chemistry ,Immunology ,Cancer cell ,PARP inhibitor ,Cancer research ,Benzimidazoles ,Female ,DNA Damage - Abstract
Radiation therapy remains a promising modality for curative treatment of localized prostate cancer, but dose-limiting toxicities significantly limit its effectiveness. Agents that enhance efficacy at lower radiation doses might have considerable value in increasing tumor control without compromising organ function. Here, we tested the hypothesis that the PARP inhibitor ABT-888 (veliparib) can enhance the response of prostate cancer cells and tumors to ionizing radiation (IR). Following exposure of DU-145 and PC-3 prostate cancer cell lines to the combination of 10 μmol/L ABT-888 and 6 Gy, we observed similar persistence between both cell lines of DNA damage foci and in vitro radiosensitization. We have previously observed that persistent DNA damage foci formed after ABT-888 plus IR efficiently promote accelerated cell senescence, but only PC-3 cells displayed the expected senescent response of G2–M arrest, induction of p21 and β-galactosidase expression, and accumulation as large flat cells. In turn, combining ABT-888 with 6 Gy resulted in delayed tumor regrowth compared with either agent alone only in PC-3 xenograft tumors, whereas DU-145 tumors continued to grow. By 7 days after treatment with ABT-888 plus IR, PC-3 tumors contained abundant senescent cells displaying persistent DNA damage foci, but no evidence of senescence was noted in the DU-145 tumors. That equivalent radiosensitization by ABT-888 plus IR in vitro failed to predict comparable results with tumors in vivo suggests that the efficacy of PARP inhibitors may partially depend on a competent senescence response to accumulated DNA damage. Mol Cancer Ther; 10(7); 1185–93. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
21. Simvastatin Attenuates Radiation-Induced Murine Lung Injury and Dysregulated Lung Gene Expression
- Author
-
Liliana Moreno-Vinasco, Patrick J. LaRiviere, Viswanathan Natarajan, Gabriel D. Lang, Yves A. Lussier, Biji Mathew, Yutong Zhao, Evegeny Berdyshev, Steven M. Dudek, Jeffrey R. Jacobson, Joe G.N. Garcia, Ting Wang, Saad Sammani, Helena J. Mauceri, Lynnette M. Gerhold, Aliya N. Husain, Chin-Tu Chen, Ralph R. Weichselbaum, and Yong Huang
- Subjects
Pulmonary and Respiratory Medicine ,VAV3 ,Simvastatin ,Pathology ,medicine.medical_specialty ,Transcription, Genetic ,Clinical Biochemistry ,Biology ,Lung injury ,MMP9 ,Bronchoalveolar Lavage ,Gene product ,Mice ,Protein Interaction Mapping ,Gene expression ,medicine ,Animals ,Radiation Injuries ,Lung ,Molecular Biology ,Regulation of gene expression ,CD44 ,Lung Injury ,Articles ,Cell Biology ,Mice, Inbred C57BL ,Radiation Pneumonitis ,Hyaluronan Receptors ,Gene Expression Regulation ,biology.protein ,Cancer research ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,medicine.drug - Abstract
Novel therapies are desperately needed for radiation-induced lung injury (RILI), which, despite aggressive corticosteroid therapy, remains a potentially fatal and dose-limiting complication of thoracic radiotherapy. We assessed the utility of simvastatin, an anti-inflammatory and lung barrier–protective agent, in a dose- and time-dependent murine model of RILI (18–(25 Gy). Simvastatin reduced multiple RILI indices, including vascular leak, leukocyte infiltration, and histological evidence of oxidative stress, while reversing RILI-associated dysregulated gene expression, including p53, nuclear factor–erythroid-2–related factor, and sphingolipid metabolic pathway genes. To identify key regulators of simvastatin-mediated RILI protection, we integrated whole-lung gene expression data obtained from radiated and simvastatin-treated mice with protein–protein interaction network analysis (single-network analysis of proteins). Topological analysis of the gene product interaction network identified eight top-prioritized genes (Ccna2a, Cdc2, fcer1 g, Syk, Vav3, Mmp9, Itgam, Cd44) as regulatory nodes within an activated RILI network. These studies identify the involvement of specific genes and gene networks in RILI pathobiology, and confirm that statins represent a novel strategy to limit RILI.
- Published
- 2011
- Full Text
- View/download PDF
22. Efficacy of the Multi-Kinase Inhibitor Enzastaurin Is Dependent on Cellular Signaling Context
- Author
-
Helena J. Mauceri, Ezra E.W. Cohen, Everett E. Vokes, Jing Liu, Wen-Liang Kuo, Marsha Rich Rosner, and Ralph R. Weichselbaum
- Subjects
Cancer Research ,Cell signaling ,Indoles ,Cell cycle checkpoint ,Transplantation, Heterologous ,Mice, Nude ,Context (language use) ,Biology ,Article ,Cell Line ,Mice ,chemistry.chemical_compound ,Cyclin D1 ,Enzastaurin ,Animals ,Humans ,RNA, Small Interfering ,Protein Kinase Inhibitors ,Cell Proliferation ,DNA Primers ,Oligonucleotide Array Sequence Analysis ,Cyclin ,Base Sequence ,Kinase ,Cell Cycle ,Molecular biology ,Cell Cycle Gene ,Oncology ,chemistry ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,Cancer research ,Female ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The number of targeted small molecules being developed in oncology is increasing rapidly. Many of these are designed to inhibit multiple kinases, and thus the mechanisms of responsiveness and predictive biomarkers can be difficult to discern. In fact, with few exceptions, multi-kinase inhibitors are developed with limited mechanism-based patient selection. Enzastaurin is a multi-kinase inhibitor being studied in several malignancies that we hypothesized would be active in squamous cell carcinoma of the head and neck, because it inhibits classic and novel protein kinase C isoforms. Indeed, enzastaurin reduced the growth of SQ-20B and CAL27 tumor xenografts, decreased proliferation in these cell lines, inhibited putative target phosphorylation, and induced cell cycle arrest. Gene expression arrays confirmed that expression of cell cycle genes, including cyclins D and E, were significantly altered by exposure to enzastaurin. However, testing a panel of squamous cell carcinoma of the head and neck cell lines revealed variable sensitivity to enzastaurin, which correlated significantly with baseline cyclin D1 protein expression. Moreover, sensitivity and resistance could be reversed, respectively, by expression or depletion of cyclin D1. Furthermore, analysis of sensitive and resistant cell lines revealed distinct differences in cyclin D1 regulation. Enzastaurin modulated cyclin D1 synthesis through an Akt-regulated pathway in the former, whereas high-level CCND1 gene amplification was present in the latter. These results underscore the critical relevance of cellular signaling context in developing cancer therapies in general and suggest that enzastaurin in particular would be most effective in tumors where baseline cyclin D1 expression is low to moderate and physiologically regulated. Mol Cancer Ther; 9(10); 2814–24. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
23. Non-canonical NF-κB Antagonizes STING Sensor-Mediated DNA Sensing in Radiotherapy
- Author
-
Hua Liang, Meng Xu, Xinshuang Yu, Yuan Zhang, Ralph R. Weichselbaum, Wenxin Zheng, Liufu Deng, Yang Xin Fu, Helena J. Mauceri, Xiaona Huang, Yuzhu Hou, Ainhoa Arina, and Enyu Rao
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Transcription Factor RelA ,Melanoma, Experimental ,Tumor initiation ,Biology ,Radiation Tolerance ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immunity ,Radiation, Ionizing ,Radioresistance ,medicine ,Animals ,Humans ,Immunology and Allergy ,Melanoma ,Immunity, Cellular ,Radiotherapy ,NF-kappa B ,Membrane Proteins ,NF-κB ,DNA ,Dendritic Cells ,Neoplasms, Experimental ,Xenograft Model Antitumor Assays ,Radiation therapy ,Disease Models, Animal ,Sting ,030104 developmental biology ,Infectious Diseases ,chemistry ,Receptors, Pattern Recognition ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Cancer research ,Signal Transduction - Abstract
The NF-κB pathway plays a crucial role in supporting tumor initiation, progression, and radioresistance of tumor cells. However, the role of the NF-κB pathway in radiation-induced anti-tumor host immunity remains unclear. Here we demonstrated that inhibiting the canonical NF-κB pathway dampened the therapeutic effect of ionizing radiation (IR), whereas non-canonical NF-κB deficiency promoted IR-induced anti-tumor immunity. Mechanistic studies revealed that non-canonical NF-κB signaling in dendritic cells (DCs) was activated by the STING sensor-dependent DNA-sensing pathway. By suppressing recruitment of the transcription factor RelA onto the Ifnb promoter, activation of the non-canonical NF-κB pathway resulted in decreased type I IFN expression. Administration of a specific inhibitor of the non-canonical NF-κB pathway enhanced the anti-tumor effect of IR in murine models. These findings reveal the potentially interactive roles for canonical and non-canonical NF-κB pathways in IR-induced STING-IFN production and provide an alternative strategy to improve cancer radiotherapy.
- Published
- 2018
- Full Text
- View/download PDF
24. Ad.Egr-TNF and Local Ionizing Radiation Suppress Metastases by Interferon-β-Dependent Activation of Antigen-specific CD8+ T Cells
- Author
-
Donald Kufe, Sean P. Pitroda, Thomas E. Darga, Helena J. Mauceri, Yuru Meng, Ralph R. Weichselbaum, Nikolai N. Khodarev, and Michael A. Beckett
- Subjects
Chemokine ,Genetic Vectors ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Adenoviridae ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,Radiation, Ionizing ,Pancreatic cancer ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Neoplasm Metastasis ,Molecular Biology ,Lymph node ,Cell Proliferation ,030304 developmental biology ,Mice, Knockout ,Pharmacology ,0303 health sciences ,Tumor microenvironment ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,Original Articles ,Interferon-beta ,medicine.disease ,Mice, Mutant Strains ,3. Good health ,Mice, Inbred C57BL ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Molecular Medicine ,Tumor necrosis factor alpha ,CD8 ,medicine.drug - Abstract
Ad.Egr-TNF is a radioinducible adenovector currently in phase 3 trials for inoperable pancreatic cancer. The combination of Ad.Egr-TNF and ionizing radiation (IR) contributes to local tumor control through the production of tumor necrosis factor-alpha (TNFalpha) in the tumor microenvironment. Moreover, clinical and preclinical studies with Ad.Egr-TNF/IR have suggested that this local approach suppresses the growth of distant metastatic disease; however, the mechanisms responsible for this effect remain unclear. These studies have been performed in wild-type (WT) and TNFR1,2(-/-) mice to assess the role of TNFalpha-induced signaling in the suppression of draining lymph node (DLN) metastases. The results demonstrate that production of TNFalpha in the tumor microenvironment induces expression of interferon (IFNbeta). In turn, IFNbeta stimulates the production of chemokines that recruit CD8(+) T cells to the tumor. The results further demonstrate that activation of tumor antigen-specific CD8(+) CTLs contributes to local antitumor activity and suppression of DLN metastases. These findings support a model in which treatment of tumors with Ad.Egr-TNF and IR is mediated by local and distant immune-mediated antitumor effects that suppress the development of metastases.
- Published
- 2010
- Full Text
- View/download PDF
25. Oral PEG 15–20 protects the intestine against radiation: role of lipid rafts
- Author
-
John C. Alverdy, Olga Zaborina, Helena J. Mauceri, Alexander Zaborin, Kenneth Drabik, Eugene B. Chang, Jeffrey B. Matthews, Christopher M. Holbrook, Mukta Katdare, Jason Long, Martin Hauer-Jensen, Junru Wang, Ka Yee C. Lee, Millicent A. Firestone, Ralph R. Weichselbaum, and Vesta Valuckaite
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Time Factors ,Physiology ,Administration, Oral ,Apoptosis ,Radiation-Protective Agents ,Ileum ,Pharmacology ,Biology ,Bacterial Adhesion ,Cell Line ,Polyethylene Glycols ,Rats, Sprague-Dawley ,Sepsis ,Mice ,Membrane Microdomains ,Intestinal mucosa ,Fibrosis ,Physiology (medical) ,PEG ratio ,medicine ,Animals ,Intestinal Mucosa ,Barrier function ,Dose-Response Relationship, Drug ,Virulence ,Hepatology ,Translational Physiology ,Gastroenterology ,medicine.disease ,Rats ,Mice, Inbred C57BL ,Molecular Weight ,Intestinal Diseases ,Radiation Injuries, Experimental ,Dose–response relationship ,Cholesterol ,medicine.anatomical_structure ,Pseudomonas aeruginosa - Abstract
Intestinal injury following abdominal radiation therapy or accidental exposure remains a significant clinical problem that can result in varying degrees of mucosal destruction such as ulceration, vascular sclerosis, intestinal wall fibrosis, loss of barrier function, and even lethal gut-derived sepsis. We determined the ability of a high-molecular-weight polyethylene glycol-based copolymer, PEG 15–20, to protect the intestine against the early and late effects of radiation in mice and rats and to determine its mechanism of action by examining cultured rat intestinal epithelia. Rats were exposed to fractionated radiation in an established model of intestinal injury, whereby an intestinal segment is surgically placed into the scrotum and radiated daily. Radiation injury score was decreased in a dose-dependent manner in rats gavaged with 0.5 or 2.0 g/kg per day of PEG 15–20 ( n = 9–13/group, P < 0.005). Complementary studies were performed in a novel mouse model of abdominal radiation followed by intestinal inoculation with Pseudomonas aeruginosa ( P. aeruginosa ), a common pathogen that causes lethal gut-derived sepsis following radiation. Mice mortality was decreased by 40% in mice drinking 1% PEG 15–20 ( n = 10/group, P < 0.001). Parallel studies were performed in cultured rat intestinal epithelial cells treated with PEG 15–20 before radiation. Results demonstrated that PEG 15–20 prevented radiation-induced intestinal injury in rats, prevented apoptosis and lethal sepsis attributable to P. aeruginosa in mice, and protected cultured intestinal epithelial cells from apoptosis and microbial adherence and possible invasion. PEG 15–20 appeared to exert its protective effect via its binding to lipid rafts by preventing their coalescence, a hallmark feature in intestinal epithelial cells exposed to radiation.
- Published
- 2009
- Full Text
- View/download PDF
26. Characterization of response to radiation mediated gene therapy by means of multimodality imaging
- Author
-
Marta Zamora, Rebecca Bell, Xiaobing Fan, Chad R. Haney, Charles A. Pelizzari, Ralph R. Weichselbaum, Adrian D. Parasca, Helena J. Mauceri, Gregory S. Karczmar, and Howard J. Halpern
- Subjects
Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Genetic enhancement ,Magnetic resonance imaging ,Perfusion scanning ,Oxygenation ,Viral vector ,In vivo ,Medicine ,Combined Modality Therapy ,Radiology, Nuclear Medicine and imaging ,business ,Perfusion - Abstract
Imaging techniques are under development to facilitate early analysis of spatial patterns of tumor response to combined radiation and antivascular gene therapy. A genetically modified, replication defective adenoviral vector (Ad.EGR-TNFalpha), injected intratumorally, mediates infected cells to express tumor necrosis factor alpha (TNFalpha), which is increased after exposure to radiation. The goal of this study was to characterize an image based "signature" for response to this combined radiation and gene therapy in mice with human prostate xenografts. This study is part of an imaged guided therapy project where such a signature would be useful in guiding subsequent treatments. Changes in the tumor micro-environment were assessed using MRI registered with electron paramagnetic resonance imaging which provides images of tissue oxygenation. Dynamic contrast-enhanced MRI was used to assess tissue perfusion. When compared with null vector (control) treatment, the ratio of contrast agent (Gd-DTPA-BMA) washout rate to uptake rate was lower (P = 0.001) after treatment, suggesting a more balanced perfusion. Concomitantly, oxygenation significantly increased in the treated animals and decreased or did not change in the control animals (P < 0.025). This is the first report of minimally invasive, quantitative, absolute oxygen measurements correlated with tissue perfusion in vivo.
- Published
- 2009
- Full Text
- View/download PDF
27. Translational strategies exploiting TNF-α that sensitize tumors to radiation therapy
- Author
-
Donald Kufe, Elena V. Efimova, Nikolai N. Khodarev, Eva Galka, Sean P. Pitroda, M.A. Beckett, Helena J. Mauceri, C R King, Samuel Hellman, Mitchell C. Posner, Hua Liang, Thomas E. Darga, Ralph R. Weichselbaum, and Harold G. Sutton
- Subjects
Radiation-Sensitizing Agents ,Cancer Research ,Pathology ,medicine.medical_specialty ,Endothelium ,medicine.medical_treatment ,Melanoma, Experimental ,Apoptosis ,X-Ray Therapy ,Biology ,Receptors, Tumor Necrosis Factor ,Article ,Etanercept ,Mice ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Humans ,Receptors, Tumor Necrosis Factor, Type II ,Radiosensitivity ,Receptor ,Molecular Biology ,Tumor Necrosis Factor-alpha ,Melanoma ,Endothelial Cells ,Genetic Therapy ,medicine.disease ,Radiation therapy ,medicine.anatomical_structure ,Receptors, Tumor Necrosis Factor, Type I ,Cell culture ,Immunoglobulin G ,Cancer research ,Molecular Medicine ,Tumor necrosis factor alpha ,Immunosuppressive Agents ,Neoplasm Transplantation - Abstract
TNFerade is a radioinducible adenoviral vector expressing tumor necrosis factor-alpha (TNF-alpha) (Ad.Egr-TNF) currently in a phase III trial for inoperable pancreatic cancer. We studied B16-F1 melanoma tumors in TNF receptor wild-type (C57BL/6) and deficient (TNFR1,2-/- and TNFR1-/-) mice. Ad.Egr-TNF+IR inhibited tumor growth compared with IR in C57BL/6 but not in receptor-deficient mice. Tumors resistant to TNF-alpha were also sensitive to Ad.Egr-TNF+IR in C57BL/6 mice. Ad.Egr-TNF+IR produced an increase in tumor-associated endothelial cell apoptosis not observed in receptor-deficient animals. Also, B16-F1 tumors in mice with germline deletions of TNFR1,2, TNFR1 or TNF-alpha, or in mice receiving anti-TNF-alpha exhibited radiosensitivity. These results show that tumor-associated endothelium is the principal target for Ad.Egr-TNF radiosensitization and implicate TNF-alpha signaling in tumor radiosensitivity.
- Published
- 2008
- Full Text
- View/download PDF
28. Resveratrol is an effective inducer of CArG-driven TNF-α gene therapy
- Author
-
Ralph R. Weichselbaum, Jula Veerapong, Mitchell C. Posner, Kai Bickenbach, Helena J. Mauceri, Stephen J. Kron, and Michael Y. Shao
- Subjects
Cancer Research ,Necrosis ,endocrine system diseases ,Transgene ,Genetic enhancement ,Blotting, Western ,Mice, Nude ,Enzyme-Linked Immunosorbent Assay ,Locus (genetics) ,Biology ,Resveratrol ,Adenoviridae ,Mice ,chemistry.chemical_compound ,Cell Line, Tumor ,Stilbenes ,medicine ,Animals ,Humans ,Sirtuins ,Inducer ,Molecular Biology ,Early Growth Response Protein 1 ,Etoposide ,Tumor Necrosis Factor-alpha ,food and beverages ,Acetylation ,Genetic Therapy ,Xenograft Model Antitumor Assays ,Molecular biology ,In vitro ,Rats ,Gene Expression Regulation, Neoplastic ,body regions ,chemistry ,Cancer research ,Molecular Medicine ,Cancer gene ,Female ,Tumor Suppressor Protein p53 ,medicine.symptom ,hormones, hormone substitutes, and hormone antagonists - Abstract
We report the anticarcinogenic, anti-aging polyphenol resveratrol activates the radio- and chemo-inducible cancer gene therapy vector Ad.Egr.TNF, a replication-deficient adenovirus that expresses human tumor necrosis factor alpha (TNF-alpha) under control of the Egr-1 promoter. Like ionizing radiation or chemotherapeutic agents previously shown to activate Ad.Egr.TNF, resveratrol also induces Egr-1 expression from its chromosomal locus with a possible role for Egr-1 promoter CC(A+T)richGG sequences in the expression of TNF-alpha. Resveratrol induction of TNF-alpha in Ad.Egr.TNF-infected tumor xenografts demonstrated antitumor response in human and rat tumor models comparable to that of radio- or chemotherapy-induced TNF-alpha. Although sirtuins are known targets of resveratrol, in vitro inhibition of SIRT1 activity did not abrogate resveratrol induction of Egr-1 expression. This suggests that SIRT1 is not essential to mediate resveratrol induction of Egr-1. Nevertheless, control of transgene expression via resveratrol activation of Egr-1 may extend use of Ad.Egr.TNF to patients intolerant of radiation or cytotoxic therapy and offer a novel tool for development of other inducible gene therapies.
- Published
- 2007
- Full Text
- View/download PDF
29. Local gene delivery of tumor necrosis factor alpha can impact primary tumor growth and metastases through a host-mediated response
- Author
-
Helena J. Mauceri, Thomas A. Davis, Jennifer Macko, Ralph R. Weichselbaum, C. Richter King, and Randall S. Macgill
- Subjects
Adult ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Axillary lymph nodes ,Genetic enhancement ,medicine.medical_treatment ,Genetic Vectors ,Receptors, Tumor Necrosis Factor ,Adenoviridae ,Mice ,Surgical oncology ,Neoplasms ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Melanoma ,Lymph node ,Mice, Knockout ,Chemotherapy ,Tumor Necrosis Factor-alpha ,business.industry ,Gene Transfer Techniques ,Dose fractionation ,Genetic Therapy ,General Medicine ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Primary tumor ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Lymphatic Metastasis ,Cancer research ,Female ,Dose Fractionation, Radiation ,business ,Neoplasm Transplantation ,Follow-Up Studies - Abstract
TNFerade is a replication incompetent adenovector designed to express human TNFalpha under control of the Egr-1 radiation and chemotherapy enhanced promoter, and is currently in Phase II/III clinical testing. Data from Phase I clinical testing of TNFerade in a limited set of melanoma patients suggested the potential to impact distal metastases following intratumoral injections of TNFerade. These clinical observations and the multiple potential mechanisms of TNFerade led us to hypothesize local treatment with TNFerade + radiation may impact metastatic disease. We explored this hypothesis in preclinical models using the spontaneously metastatic, syngeneic B16F10 murine melanoma model. Established subcutaneous B16F10 tumors were treated with intratumoral injections of TNFerade and localized 2 Gy fractionated radiation therapy, modeling the clinical treatment regimen. Following 10-14 days of treatment, mice were evaluated for metastases development in the iliac and axillary lymph nodes. Comparisons of metastatic burden to control groups indicated TNFerade +/- radiation suppressed the formation of metastases in the lymph nodes. Additional experiments in TNF receptor knockout mice, where the only possible effects are on tumor cells containing the TNFalpha receptor, indicate TNFerade's local and distal activities are critically dependent on a host-mediated response. These data provide direct preclinical evidence local therapy of a solid tumor with TNFerade can also reduce metastatic disease, in addition to effects on the treated lesion. Furthermore, our finding of a host dependant response(s) for TNFerade at both the treated tumor and on lymph node metastases suggest the potential for broad activity independent of tumor histology.
- Published
- 2007
- Full Text
- View/download PDF
30. Chemoinducible gene therapy: A strategy to enhance doxorubicin antitumor activity
- Author
-
Carlos A. Lopez, Eric T. Kimchi, Helena J. Mauceri, James O. Park, Neil Mehta, Kevin T. Murphy, Michael A. Beckett, Samuel Hellman, Mitchell C. Posner, Donald W. Kufe, and Ralph R. Weichselbaum
- Subjects
Cancer Research ,Oncology - Abstract
A replication-defective adenoviral vector, Ad.Egr-TNF.11D, was engineered by ligating the CArG (CC(A/T)6GG) elements of the Egr-1 gene promoter upstream to a cDNA encoding human tumor necrosis factor-α. We report here that Ad.Egr-TNF.11D is activated by the clinically important anticancer agents cisplatin, cyclophosphamide, doxorubicin, 5-fluorouracil, gemcitabine, and paclitaxel. N-acetylcysteine, a free radical scavenger, blocked induction of tumor necrosis factor-α by anticancer agents, supporting a role for reactive oxygen intermediates in activation of the CArG sequences. Importantly, resistance of PC-3 human prostate carcinoma and PROb rat colon carcinoma tumors to doxorubicin in vivo was reversed by combining doxorubicin with Ad.Egr-TNF and resulted in significant antitumor effects. Treatment with Ad.Egr-TNF.11D has been associated with inhibition of tumor angiogenesis. In this context, a significant decrease in tumor microvessel density was observed following combined treatment with doxorubicin and Ad.Egr-TNF.11D as compared with either agent alone. These data show that Ad.Egr-TNF.11D is activated by diverse anticancer drugs.
- Published
- 2004
- Full Text
- View/download PDF
31. Endothelial cells co-cultured with wild-type and dominant/negative p53-transfected glioblastoma cells exhibit differential sensitivity to radiation-induced apoptosis
- Author
-
Edwardine Labay, Jianqing Yu, Nalin Gupta, Helena J. Mauceri, Nikolai N. Khodarev, Ralph R. Weichselbaum, Yasushi Kataoka, and Thomas E. Darga
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Programmed cell death ,Cell ,Apoptosis ,Enzyme-Linked Immunosorbent Assay ,Biology ,Transfection ,Radiation Tolerance ,Cell Movement ,Radioresistance ,Tumor Cells, Cultured ,medicine ,Humans ,Radiosensitivity ,neoplasms ,Genes, Dominant ,Brain Neoplasms ,Gene Expression Profiling ,Coculture Techniques ,nervous system diseases ,Endothelial stem cell ,Phenotype ,medicine.anatomical_structure ,Oncology ,Cell culture ,Cancer research ,Endothelium, Vascular ,Tumor Suppressor Protein p53 ,Glioblastoma - Abstract
We performed expressional profiling of isogenic glioblastoma cell lines U87-Lux8 and U87-175.4. These cell lines differ in that U87-Lux8 expresses wild-type p53 and U87-175.4 expresses a dominant-negative p53 (175His mutation). DNA array analysis and real-time PCR measurements demonstrated that basal expression and response to irradiation were different in these isogenic glioblastoma cell lines. These differences included genes involved in growth regulation and genes associated with cell-to-cell and cell/ECM communications. Co-cultivation of U87-175.4 and U87-Lux8 with HUVE cells demonstrated that U87-175.4 cells suppress the angiogenic phenotype of HUVEC and increase their sensitivity to radiation-induced apoptosis compared to co-culture of U87-Lux8/HUVEC. These data suggest that blockade of p53 function may alter the communication between tumor cells and endothelial cells such that endothelial cells exhibit an increase in radiosensitivity. These findings may have important implications for the treatment of glioblastoma tumors and other human cancers. © 2003 Wiley-Liss, Inc.
- Published
- 2004
- Full Text
- View/download PDF
32. Dual Induction of the Epo-Egr-TNF-α Plasmid in Hypoxic Human Colon Adenocarcinoma Produces Tumor Growth Delay
- Author
-
Rabih M. Salloum, Helena J. Mauceri, Nader N. Hanna, David H. Gorski, Mitchell C. Posner, and Ralph R. Weichselbaum
- Subjects
General Medicine - Abstract
Gene therapy is a modality for the treatment of solid tumors that involves the introduction of a suicide gene into the tumor cells. Genetic radiotherapy involves the placement of a radiation-sensitive promoter upstream from a suicide gene. Because of their irregular vasculature some solid tumors are chronically hypoxic and hence are resistant to conventional treatment with chemotherapy and ionizing radiation (IR). The purpose of this study was to demonstrate that regional tumor hypoxia could be exploited to improve local tumor control. The cDNA coding the erythropoietin hypoxia-responsive element (EPO) was placed upstream from the Egr-TNF-α construct. WIDR human colon adenocarcinoma cells were injected into the right hind limb of nude mice and treated with Epo-Egr-TNF-α plasmid with or without IR. Tumor volumes were measured by calipers and tumor necrosis factor (TNF)-α content of the tumor was determined by enzyme-linked immunosorbent assay. Treatment with the combined regimen of Epo-Egr-TNF-α plasmid + IR resulted in significant tumor growth delay. Tumor TNF-α content was increased by 30 per cent in the combined treatment group compared with each treatment alone. Regional tumor hypoxia can be exploited successfully to induce tumor growth delay, enhance local control, and enhance the therapeutic ratio.
- Published
- 2003
- Full Text
- View/download PDF
33. Angiostatin potentiates cyclophosphamide treatment of metastatic disease
- Author
-
Vinay K. Gupta, Abdullah Manan, Ann Koons, Helena J. Mauceri, Anthony G. Montag, Saraswathy Seetharam, Ralph R. Weichselbaum, James O. Park, John Y. Lee, Donald Kufe, Michael A. Beckett, and Philip Schumm
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Angiogenesis ,medicine.medical_treatment ,Neovascularization, Physiologic ,Angiogenesis Inhibitors ,Toxicology ,Metastasis ,Carcinoma, Lewis Lung ,Mice ,chemistry.chemical_compound ,Cell Movement ,Antineoplastic Combined Chemotherapy Protocols ,Morphogenesis ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Single-Blind Method ,heterocyclic compounds ,Pharmacology (medical) ,Neoplasm Metastasis ,Angiostatins ,Cyclophosphamide ,Cells, Cultured ,Protein Synthesis Inhibitors ,Pharmacology ,Tube formation ,Chemotherapy ,Angiostatin ,business.industry ,Lewis lung carcinoma ,Drug Synergism ,Plasminogen ,medicine.disease ,Primary tumor ,Peptide Fragments ,Nitrogen mustard ,Mice, Inbred C57BL ,Oncology ,chemistry ,Linear Models ,cardiovascular system ,Cancer research ,Endothelium, Vascular ,Drug Screening Assays, Antitumor ,business - Abstract
Purpose. We examined the interaction between cyclophosphamide (CPA) and angiostatin (AS) on the growth of primary Lewis lung carcinoma (LLC) tumors and on the development of LLC pulmonary metastases. We studied the effects of AS and CPA on the stages of angiogenesis employing in vitro assays. Methods. Primary tumor growth and pulmonary metastases were measured to evaluate the effects of treatment with AS alone, CPA alone or the combination of CPA and AS. We examined the effects of CPA plus AS on endothelial cell (HUVEC) survival, migration and tube formation. Results. Combined treatment with CPA and AS did not significantly affect primary tumor growth when compared with CPA treatment alone. However, a significant decrease in the number of pulmonary metastases was observed following CPA plus AS treatment when compared with CPA treatment alone (P
- Published
- 2002
- Full Text
- View/download PDF
34. Tumor production of angiostatin is enhanced after exposure to TNF-?
- Author
-
Donald Kufe, Charles K. Brown, John Y. Lee, Helena J. Mauceri, Saraswathy Seetharam, M. Sharon Stack, Stephen T. Gately, Michael A. Beckett, Vinay K. Gupta, Ralph R. Weichselbaum, and Kirsten Swedberg
- Subjects
Male ,Cancer Research ,Angiogenesis ,Recombinant Fusion Proteins ,Genetic enhancement ,medicine.medical_treatment ,Genetic Vectors ,Transplantation, Heterologous ,Mice, Nude ,Adenocarcinoma ,Matrix Metalloproteinase Inhibitors ,Biology ,Models, Biological ,Adenoviridae ,Viral vector ,Mice ,Plasminogen Activators ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Protease Inhibitors ,Angiostatins ,Angiostatin ,Neovascularization, Pathologic ,Tumor Necrosis Factor-alpha ,Genetic transfer ,Defective Viruses ,Prostatic Neoplasms ,Plasminogen ,Genetic Therapy ,Peptide Fragments ,Neoplasm Proteins ,Cytokine ,Matrix Metalloproteinase 9 ,Oncology ,Culture Media, Conditioned ,Carcinoma, Squamous Cell ,Cancer research ,Matrix Metalloproteinase 2 ,Female ,Tumor necrosis factor alpha ,Plasminogen activator ,Neoplasm Transplantation - Abstract
Infection of tumors with an adenoviral vector expressing a chimeric gene composed of the CArG elements of the Egr-1 promoter and a cDNA encoding TNF-alpha (Ad.Egr-TNF) has previously been shown to result in the production of high intratumoral levels of TNF-alpha and thereby tumor regression. The antitumor effects of TNF-alpha were ascribed to vascular thrombosis. We and others, have reported that inhibition of tumor vessel thrombosis using anticoagulation therapy does not abrogate the antitumor effects after TNF-alpha treatment. To investigate the potential antiangiogenic effects of TNF-alpha, we studied the generation of angiostatin after intratumoral injection of Ad.Egr-TNF. We report an increase in plasma angiostatin levels both during and after treatment with Ad.Egr-TNF that parallel tumor regression. We also report that TNF-alpha enhances angiostatin production by inducing the activity of plasminogen activator and the release of MMP-9 by tumor cells. These studies support a model in which the antiangiogenic effects of TNF-alpha on the tumor microvasculature are mediated by generation of angiostatin.
- Published
- 2002
- Full Text
- View/download PDF
35. Combined effects of angiostatin and ionizing radiation in antitumour therapy
- Author
-
Michael A. Beckett, Donald Kufe, Kerri Anne Stellato, Mohanraj Dhanabal, Ralph R. Weichselbaum, Vikas P. Sukhatme, Gerald A. Soff, Ruth Heimann, Stephen T. Gately, M J Staba, Kevin Bigelow, Helena J. Mauceri, Nader Hanna, and David H. Gorski
- Subjects
Angiogenesis ,medicine.medical_treatment ,Cell ,Antineoplastic Agents ,Apoptosis ,Biology ,Ionizing radiation ,Neovascularization ,Carcinoma, Lewis Lung ,Mice ,In vivo ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Angiostatins ,Multidisciplinary ,Angiostatin ,Neovascularization, Pathologic ,Plasminogen ,Combined Modality Therapy ,Peptide Fragments ,Recombinant Proteins ,Mice, Inbred C57BL ,Radiation therapy ,medicine.anatomical_structure ,Immunology ,Cancer research ,Female ,Endothelium, Vascular ,medicine.symptom ,Neoplasm Transplantation - Abstract
Angiogenesis, the formation of new capillaries from pre-existing vessels, is essential for tumour progression1,2,3,4,5. Angiostatin, a proteolytic fragment of plasminogen6 that was first isolated from the serum and urine of tumour-bearing mice7, inhibits angiogenesis and thereby growth of primary8 and metastatic7,9,10 tumours. Radiotherapy is important in the treatment of many human cancers, but is often unsuccessful because of tumour cell radiation resistance11,12. Here we combine radiation with angiostatin to target tumour vasculature that is genetically stable and therefore less likely to develop resistance13,14,15. The results show an antitumour interaction between ionizing radiation and angiostatin for four distinct tumour types, at doses of radiation that are used in radiotherapy. The combination produced no increase in toxicity towards normal tissue. In vitro studies show that radiation and angiostatin have combined cytotoxic effects on endothelial cells, but not tumour cells. In vivo studies show that these agents, in combination, target the tumour vasculature. Our results provide support for combining ionizing radiation with angiostatin to improve tumour eradication without increasing deleterious effects.
- Published
- 1998
- Full Text
- View/download PDF
36. Increased injection number enhances adenoviral genetic radiotherapy
- Author
-
Helena J. Mauceri, Ralph R. Weichselbaum, Lisa P. Seung, Wendy L. Grdina, and Kirsten Swedberg
- Subjects
Radiation ,Radiological and Ultrasound Technology ,Genetic enhancement ,Biology ,medicine.disease_cause ,Molecular biology ,Immediate early protein ,Viral vector ,body regions ,Adenoviridae ,Titer ,chemistry.chemical_compound ,Oncology ,chemistry ,Radioresistance ,medicine ,Radiology, Nuclear Medicine and imaging ,Tumor necrosis factor alpha ,Growth inhibition ,hormones, hormone substitutes, and hormone antagonists - Abstract
Intratumoral injection of an adenoviral vector containing radiation-inducible DNA sequences of the early growth response gene (Egr-1) promoter ligated to a cDNA encoding tumor necrosis factor-alpha (TNF-alpha; Ad.Egr-TNF) increases the radiation killing of a human radioresistant xenograft (SQ-20B). Viral dose-escalation experiments demonstrated that SQ-20B growth inhibition correlated with viral titer. Injection of 5 x 10(8) PFU Ad.Egr-TNF produced regression to a mean volume of 22 +/- 13% of the original tumor volume, 1 x 10(8) PFU to a mean of 62 +/- 24%, and 5 x 10(7) PFU to a mean of 67 +/- 27%. No regression was observed when tumors were injected with 1 x 10(7) PFU Ad.Egr-TNF or with the null viral vector (Ad.null). When two injections of vector (2 x 10(8) PFU Ad.Egr-TNF) were combined with 50 Gy, a significant increase in tumor regression was observed compared with injection of buffer, Ad.Egr-TNF, or 50 Gy. The interactive killing between TNF and radiation was enhanced significantly (P = 0.05) when the number of injections was increased from two to five while maintaining a constant viral titer (2 x 10(8) PFU Ad.Egr-TNF) and a constant radiation dose (50 Gy). Significant TNF-alpha levels were present in irradiated vs. unirradiated tumors following injection with Ad.Egr-TNF. Taken together, these data suggest that the volumetric reduction produced by the combined effects of Ad.Egr-TNF and radiation is enhanced with increasing vector concentration and the number of vector injections.
- Published
- 1997
- Full Text
- View/download PDF
37. A survivin-associated adaptive response in radiation therapy
- Author
-
Richard C. Miller, Gayle E. Woloschak, Jian Jian Li, David J. Grdina, Helena J. Mauceri, Harold G. Sutton, Jeffrey S. Murley, and Ralph R. Weichselbaum
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Cell Survival ,medicine.medical_treatment ,Survivin ,Apoptosis ,Biology ,Inhibitor of apoptosis ,Radiation Dosage ,Radiation Tolerance ,Article ,Ionizing radiation ,Inhibitor of Apoptosis Proteins ,Mice ,Pregnancy ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Mice, Knockout ,Sarcoma ,Transfection ,medicine.disease ,Adaptation, Physiological ,Xenograft Model Antitumor Assays ,Radiation therapy ,Mice, Inbred C57BL ,Repressor Proteins ,Oncology ,Colonic Neoplasms ,Cancer research ,Tumor necrosis factor alpha ,Female ,Radiotherapy, Image-Guided - Abstract
Adaptive responses can be induced in cells by very low doses of ionizing radiation resulting in an enhanced resistance to much larger exposures. The inhibitor of apoptosis protein, survivin, has been implicated in many adaptive responses to cellular stress. Computerized axial tomography used in image-guided radiotherapy to position and monitor tumor response uses very low radiation doses ranging from 0.5 to 100 mGy. We investigated the ability of these very low radiation doses administered along with two 2 Gy doses separated by 24 hours, a standard conventional radiotherapy dosing schedule, to initiate adaptive responses resulting in the elevation of radiation resistance in exposed cells. Human colon carcinoma (RKO36), mouse sarcoma (SA-NH), along with transformed mouse embryo fibroblasts, wild type or cells lacking functional tumor necrosis factor receptors 1 and 2 were used to assess their relative ability to express an adaptive response when grown either to confluence in vitro or as tumors in the flank of C57BL/6 mice. The survival of each of these cells was elevated from 5% to 20% (P ≤ 0.05) as compared to cells not receiving a 100 mGy or lesser dose. In addition, the cells exposed to 100 mGy exhibited elevations in survivin levels, reductions in apoptosis frequencies, and loss of an adaptive response if transfected with survivin siRNA. This survivin-mediated adaptive response has the potential for affecting outcomes if regularly induced throughout a course of image guided radiation therapy. Cancer Res; 73(14); 4418–28. ©2013 AACR.
- Published
- 2013
38. The influence of ethanol exposure on insulin-like growth factor (IGF) type II receptors in fetal rat tissues
- Author
-
Kathryn Becker, Sonya Conway, and Helena J. Mauceri
- Subjects
medicine.medical_specialty ,Liquid diet ,Offspring ,medicine.medical_treatment ,Biology ,Receptor, IGF Type 2 ,General Biochemistry, Genetics and Molecular Biology ,Rats, Sprague-Dawley ,Insulin-like growth factor ,chemistry.chemical_compound ,Fetus ,Pregnancy ,Internal medicine ,medicine ,Animals ,General Pharmacology, Toxicology and Pharmaceutics ,Receptor ,reproductive and urinary physiology ,Kidney ,Ethanol ,Skeletal muscle ,General Medicine ,Rats ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Female - Abstract
The effect of ethanol (ETOH) exposure on the IGF type II receptor concentration was examined in 18 and 20 day fetal rat tissues. Pregnant dams were fed an ETOH (36% of calories derived from ETOH; 6.6% v/v) liquid diet. Control fetuses were offspring of dams either pair-fed a control liquid diet or ad libitum-fed a standard pelleted lab chow. Fetal brain, heart, kidney, liver, lung and skeletal muscle were removed and whole homogenates from individual animals were analyzed. Results of immunoquantification of IGF type II receptors in whole tissue homogenates show that there is a trend towards increased receptor concentration between 18 and 20 days in all tissue and this trend is significant for lung, liver and muscle. There were no significant differences in receptor concentration between treatment groups. These studies suggest that during the later stages of fetal development, there is an increase in total IGF type II receptors and this increase is undisturbed by ETOH exposure.
- Published
- 1996
- Full Text
- View/download PDF
39. Tumor size does not limit radiation-inducible gene therapy in a human xenograft model
- Author
-
Henry Lee, Helena J. Mauceri, Vikas P. Sukhatme, A B S Michael Beckett, Dennis E. Hallahan, and Ralph R. Weichselbaum
- Subjects
Radiation ,Radiological and Ultrasound Technology ,medicine.medical_treatment ,Genetic enhancement ,Clone (cell biology) ,Transfection ,Biology ,Molecular biology ,Radiation therapy ,Cytokine ,Oncology ,In vivo ,Cell culture ,Radioresistance ,medicine ,Radiology, Nuclear Medicine and imaging - Abstract
Tumor size has been reported to be a limiting factor in both radiotherapy (RT) and gene therapy. Therefore, we assessed the effects of tumor necrosis factor-alpha (TNF-α) gene therapy and radiation in relatively large tumors in a human xenograft system. We linked DNA sequences from the promoter region of the radiation-inducible gene Egr-1 to the cDNA sequence that encodes human TNF-α, a radiosensitizing cytokine. The Egr-TNF construct was transfected into the human hematopoietic cell line HL525. Stable transfectants, exhibiting radiation inducibility of TNF-α in vitro (clone 2 cells), were directly injected into xenografts of a radioresistant human squamous cell carcinoma cell line, SQ-20B. In a previous study, 100 mm3 tumors treated with radiation and clone 2 cells demonstrated significant tumor control (P < 0.0001) when compared with tumors treated with clone 2 alone or radiation alone. In the present study, 450 mm3 xenografts treated with clone 2 cells and radiation were significantly smaller (P < 0.01) at the nadir of regression than tumors treated with radiation alone. Because the radiation dose was held constant (40 Gy) in both 100 and 450 mm3 xenografts, only one cure was noted in the larger tumors. These data support an interactive anti-tumor effect in vivo between radiation and TNF-α gene therapy which is equal in both small and large human tumor xenograft models. © 1995 Wiley-Liss, Inc.
- Published
- 1995
- Full Text
- View/download PDF
40. Tumor endothelial inflammation predicts clinical outcome in diverse human cancers
- Author
-
Tong Zhou, Thomas E. Darga, Nikolai N. Khodarev, Michael A. Beckett, Samantha Perakis, Randy F. Sweis, Harold G. Sutton, Joe G.N. Garcia, Matthew Filippo, Edwardine Labay, Hua Liang, Ralph R. Weichselbaum, Wei Zhang, Sajid A. Khan, Sean P. Pitroda, and Helena J. Mauceri
- Subjects
Male ,Pathology ,Lung Neoplasms ,Colorectal cancer ,Tumor Physiology ,lcsh:Medicine ,Kaplan-Meier Estimate ,Lung and Intrathoracic Tumors ,Pathogenesis ,Mice ,0302 clinical medicine ,Neoplasms ,Molecular Cell Biology ,Basic Cancer Research ,Breast Tumors ,Medicine ,lcsh:Science ,Neurological Tumors ,Cells, Cultured ,Oligonucleotide Array Sequence Analysis ,0303 health sciences ,Multidisciplinary ,Neovascularization, Pathologic ,Glioma ,Genomics ,Middle Aged ,3. Good health ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Tumor necrosis factor alpha ,Female ,medicine.symptom ,Cellular Types ,Research Article ,Adult ,medicine.medical_specialty ,Stromal cell ,Endothelium ,Inflammation ,Breast Neoplasms ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,Gastrointestinal Tumors ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Lung cancer ,Biology ,030304 developmental biology ,Aged ,Immune Evasion ,business.industry ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,lcsh:R ,Endothelial Cells ,Cancers and Neoplasms ,medicine.disease ,Multivariate Analysis ,Cancer research ,lcsh:Q ,Endothelium, Vascular ,business ,Genome Expression Analysis - Abstract
Background Vascular endothelial cells contribute to the pathogenesis of numerous human diseases by actively regulating the stromal inflammatory response; however, little is known regarding the role of endothelial inflammation in the growth of human tumors and its influence on the prognosis of human cancers. Methods Using an experimental model of tumor necrosis factor-alpha (TNF-α)-mediated inflammation, we characterized inflammatory gene expression in immunopurified tumor-associated endothelial cells. These genes formed the basis of a multivariate molecular predictor of overall survival that was trained and validated in four types of human cancer. Results We report that expression of experimentally derived tumor endothelial genes distinguished pathologic tissue specimens from normal controls in several human diseases associated with chronic inflammation. We trained these genes in human cancer datasets and defined a six-gene inflammatory signature that predicted significantly reduced overall survival in breast cancer, colon cancer, lung cancer, and glioma. This endothelial-derived signature predicted outcome independently of, but cooperatively with, standard clinical and pathological prognostic factors. Consistent with these findings, conditioned culture media from human endothelial cells stimulated by pro-inflammatory cytokines accelerated the growth of human colon and breast tumors in immunodeficient mice as compared with conditioned media from untreated endothelial cells. Conclusions This study provides the first prognostic cancer gene signature derived from an experimental model of tumor-associated endothelial inflammation. These findings support the notion that activation of inflammatory pathways in non-malignant tumor-infiltrating endothelial cells contributes to tumor growth and progression in multiple human cancers. Importantly, these results identify endothelial-derived factors that could serve as potential targets for therapy in diverse human cancers.
- Published
- 2012
41. Radiation-inducible Immunotherapy for Cancer: Senescent Tumor Cells as a Cancer Vaccine
- Author
-
Khaled W. Hamzeh, Yang Xin Fu, Helena J. Mauceri, Ralph R. Weichselbaum, Elena V. Efimova, Yuru Meng, Stephen J. Kron, and Thomas E. Darga
- Subjects
Cytotoxicity, Immunologic ,Radiation-Sensitizing Agents ,Veliparib ,medicine.medical_treatment ,Melanoma, Experimental ,Biology ,Poly(ADP-ribose) Polymerase Inhibitors ,Lymphocyte Activation ,Poly (ADP-Ribose) Polymerase Inhibitor ,Cancer Vaccines ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Drug Discovery ,medicine ,Genetics ,Tumor Cells, Cultured ,Cytotoxic T cell ,Animals ,Humans ,Molecular Biology ,Cellular Senescence ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Melanoma ,Cancer ,Immunotherapy ,medicine.disease ,Combined Modality Therapy ,3. Good health ,Radiation therapy ,Survival Rate ,Pancreatic Neoplasms ,chemistry ,030220 oncology & carcinogenesis ,Immunology ,Commentary ,Molecular Medicine ,Cytokines ,Original Article ,Benzimidazoles ,Female ,Cancer vaccine ,Neoplasm Transplantation ,T-Lymphocytes, Cytotoxic - Abstract
Radiotherapy offers an effective treatment for advanced cancer but local and distant failures remain a significant challenge. Here, we treated melanoma and pancreatic carcinoma in syngeneic mice with ionizing radiation (IR) combined with the poly(ADP-ribose) polymerase inhibitor (PARPi) veliparib to inhibit DNA repair and promote accelerated senescence. Based on prior work implicating cytotoxic T lymphocytes (CTLs) as key mediators of radiation effects, we discovered that senescent tumor cells induced by radiation and veliparib express immunostimulatory cytokines to activate CTLs that mediate an effective antitumor response. When these senescent tumor cells were injected into tumor-bearing mice, an antitumor CTL response was induced which potentiated the effects of radiation, resulting in elimination of established tumors. Applied to human cancers, radiation-inducible immunotherapy may enhance radiotherapy responses to prevent local recurrence and distant metastasis.
- Published
- 2012
42. Effect of Ethanol on Insulin-Like Growth Factor-II Release from Fetal Organs
- Author
-
Wei‐Hua ‐H Lee, Sonya Conway, and Helena J. Mauceri
- Subjects
Male ,medicine.medical_specialty ,Liquid diet ,Fetal alcohol syndrome ,Medicine (miscellaneous) ,Gestational Age ,Biology ,Toxicology ,Rats, Sprague-Dawley ,Embryonic and Fetal Development ,Fetus ,Organ Culture Techniques ,Insulin-Like Growth Factor II ,Pregnancy ,Placenta ,Internal medicine ,medicine ,Animals ,Tissue Distribution ,reproductive and urinary physiology ,Ethanol ,Radioimmunoassay ,medicine.disease ,Rats ,Psychiatry and Mental health ,medicine.anatomical_structure ,Endocrinology ,Fetal Alcohol Spectrum Disorders ,In utero ,Insulin-like growth factor 2 ,embryonic structures ,biology.protein ,Gestation ,Female - Abstract
This study examines the effect of ethanol (ETOH) exposure and nutrient restriction on the release of insulin-like growth factor (IGF)-II from 18- and 20-day explanted fetal organs. Fetuses were exposed to ETOH (E) in utero by feeding dams a 36% (calories derived from ETOH: 6.6% v/v) ETOH liquid diet. Control fetuses were offsprings of dams either pair-fed (P) a control liquid diet or ad libitum (A) fed a standard pelleted lab chow. Brain, heart, kidney, liver, lung, muscle, and placenta of fetuses from the same litter were pooled and explanted, and IGF-II concentration in explanted media was analyzed by radioimmunoassay. Maternal and fetal weights were determined during pregnancy and at sacrifice, respectively, to evaluate the influence of ETOH on growth. Both maternal and fetal weights were substantially reduced by ETOH on 18 and 20 days of gestation compared with both A and P controls. At 18 days of gestation, E fetuses (1.33 +/- 0.03 g) weighed less than either A (1.47 +/- 0.03 g) or P (1.54 +/- 0.04 g) fetuses. By 20 days, A mean fetal weight (4.19 +/- 0.23 g) was significantly greater than both P (3.74 +/- 0.06 g) and E (3.28 +/- 0.06 g) fetuses. IGF-II concentration in media from 18-day fetal explants was highest from E (brain, heart, liver, and placenta) and P tissues (kidney, lung, and muscle). IGF-II in media from A tissues (except placenta) was lower than both E and P levels.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1994
- Full Text
- View/download PDF
43. Everolimus exhibits efficacy as a radiosensitizer in a model of non-small cell lung cancer
- Author
-
Thomas E. Darga, Everett E. Vokes, Masha Kocherginsky, Ralph R. Weichselbaum, Helena J. Mauceri, Harold G. Sutton, and Joel Kochanski
- Subjects
Cancer Research ,Radiosensitizer ,Radiation-Sensitizing Agents ,Lung Neoplasms ,medicine.medical_treatment ,Gene Expression ,Mice, Nude ,Pharmacology ,Mice ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Radiation, Ionizing ,medicine ,Animals ,Humans ,Everolimus ,Phosphorylation ,Lung cancer ,PI3K/AKT/mTOR pathway ,Cisplatin ,Sirolimus ,Chemotherapy ,Oncogene ,business.industry ,Cancer ,Ribosomal Protein S6 Kinases, 70-kDa ,General Medicine ,medicine.disease ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Oncology ,Female ,business ,medicine.drug - Abstract
Signaling pathways that activate mTOR (mammalian target of rapamycin) are altered in many human cancers and these alterations are associated with prognosis and treatment response. mTOR inhibition can restore sensitivity to DNA damaging agents such as cisplatin. The rapamycin derivative everolimus exhibits antitumor activity and is approved for patients with renal cell cancer. Clinically, everolimus has also been evaluated in patients with advanced non-small cell lung cancer (NSCLC) that were refractory to chemotherapy and epidermal growth factor receptor tyrosine kinase inhibitors. We tested the effects of combined treatment with everolimus (RAD001) and fractionated radiation using a xenograft model of human NSCLC (A549 cells). In growth studies, mean tumor volume was reduced in the everolimus plus 30 Gy cohort with significant tumor growth suppression compared to 30 Gy alone (p=0015), or everolimus alone (p
- Published
- 2011
44. Effect of Ethanol Exposure on Circulating Levels of Insulin-Like Growth Factor I and II, and Insulin-Like Growth Factor Binding Proteins in Fetal Rats
- Author
-
Sonya Conway, Terry G. Unterman, Sandra Dempsey, Wei-Hua Lee, and Helena J. Mauceri
- Subjects
Male ,medicine.medical_specialty ,Liquid diet ,medicine.medical_treatment ,Medicine (miscellaneous) ,Biology ,Toxicology ,Insulin-like growth factor-binding protein ,Rats, Sprague-Dawley ,Insulin-like growth factor ,Insulin-Like Growth Factor II ,Pregnancy ,Internal medicine ,medicine ,Animals ,Insulin-Like Growth Factor I ,Fetus ,Fetal Growth Retardation ,Ethanol ,Body Weight ,Radioimmunoassay ,Somatomedin ,Rats ,Insulin-Like Growth Factor Binding Proteins ,Psychiatry and Mental health ,Endocrinology ,Fetal Alcohol Spectrum Disorders ,Insulin-like growth factor 2 ,biology.protein ,Female ,Carrier Proteins ,Hormone - Abstract
Maternal ethanol (ETOH) exposure is associated with impaired fetal growth. Because insulin-like growth factors (IGFs) are thought to be important in the regulation of fetal somatic growth, we examined the influence of maternal ETOH exposure on fetal growth and plasma levels of IGF-I, IGF-II, and IGF binding proteins (IGFBPs) in the rat model. Control (A) dams were fed a standard rat chow ad libitum. ETOH (E) consuming dams were fed a 36% ETOH diet, and pair-fed (P) dams were fed isocaloric amounts of a control liquid diet. All animals were killed on day 20 of gestation. Plasma concentrations of IGF-I and -II were determined by radioimmunoassay after formic acid-acetone extraction and heat inactivation of IGFBPs. Levels of IGFBPs in fetal plasma were estimated by Western ligand blotting after protein separation by SDS-PAGE and electrotransfer to nitrocellulose. Membranes were probed with [125l]IGF-l, and IGFBPs were identified by autoradiography, quantified by scanning densitometry and results expressed relative to corresponding IGFBPs in control fetal plasma. Maternal weight gain from conception to 20 days of pregnancy was reduced for E compared to P and A dams (p < 0.05 E vs. P or A). The same pattern was reflected in fetal weight that tended to be lower in P compared with A pups, and was significantly reduced in E pups compared with both groups (p < 0.0001 E vs. P or A). Thus, fetal growth was more retarded in E animals despite equal caloric and protein intake by E and P dams. Radioimmunoassay of fetal plasma revealed that there was no difference in circulating levels of IGF-II in A (65.5 ± 8.7 ng/ml) and P (65.9 ± 9.8 ng/ml) plasma. However, levels of IGF-II were higher in E (87.1 ± 6.2 ng/ml) than in corresponding P animals. At the same time, there were no differences in IGF-I levels between any of the treatment groups. Western ligand blotting demonstrated that levels of 32–34 kDa IGFBPs were elevated in plasma of P compared with A fetal plasma, consistent with an effect of reduced maternal nutrition. In contrast, levels of32–34 kDa IGFBPs were reduced in E plasma compared with P and A (p < 0.04 and p < 0.02, respectively). These data suggest that circulating levels of IGF-II (but not IGF-I) and specific IGFBPs are altered in growth-retarded fetuses exposed to ETOH. Comparison to fetal plasma obtained from pair-fed litters demonstrate that these effects on IGF and IGFBP levels are specific to ETOH, and not simply due to maternal nutrient intake. Taken together, these observations indicate that ETOH has unique effects on fetal IGF physiology compared with previously studied models of fetal growth retardation. Alterations in the expression and levels of IGFs and IGFBPs may contribute to ETOH-induced fetal growth deficiency.
- Published
- 1993
- Full Text
- View/download PDF
45. Poly(ADP-ribose) polymerase inhibitor induces accelerated senescence in irradiated breast cancer cells and tumors
- Author
-
Chaitali Chakraborty, Daniel W. Golden, Vytautas P. Bindokas, Harold G. Sutton, Elena V. Efimova, Edwardine Labay, Helena J. Mauceri, Stephen J. Kron, Ralph R. Weichselbaum, Juan Camilo Barreto-Andrade, Clayton D. Crawley, and Thomas E. Darga
- Subjects
Cancer Research ,Veliparib ,DNA repair ,DNA damage ,Infrared Rays ,Poly ADP ribose polymerase ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Mice, Nude ,Breast Neoplasms ,Cell Growth Processes ,Biology ,Poly(ADP-ribose) Polymerase Inhibitors ,Poly (ADP-Ribose) Polymerase Inhibitor ,Article ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,Animals ,Humans ,Enzyme Inhibitors ,Cellular Senescence ,Chromatin binding ,Intracellular Signaling Peptides and Proteins ,Dose-Response Relationship, Radiation ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Chromatin ,Oncology ,Biochemistry ,chemistry ,Cancer research ,Benzimidazoles ,Female ,Tumor Suppressor p53-Binding Protein 1 ,Cell aging - Abstract
Persistent DNA double-strand breaks (DSB) may determine the antitumor effects of ionizing radiation (IR) by inducing apoptosis, necrosis, mitotic catastrophe, or permanent growth arrest. IR induces rapid modification of megabase chromatin domains surrounding DSBs via poly-ADP-ribosylation, phosphorylation, acetylation, and protein assembly. The dynamics of these IR-induced foci (IRIF) have been implicated in DNA damage signaling and DNA repair. As an IRIF reporter, we tracked the relocalization of green fluorescent protein fused to a chromatin binding domain of the checkpoint adapter protein 53BP1 after IR of breast cancer cells and tumors. To block DSB repair in breast cancer cells and tumors, we targeted poly(ADP-ribose) polymerase (PARP) with ABT-888 (veliparib), one of several PARP inhibitors currently in clinical trials. PARP inhibition markedly enhanced IRIF persistence and increased breast cancer cell senescence both in vitro and in vivo, arguing for targeting IRIF resolution as a novel therapeutic strategy. Cancer Res; 70(15); 6277–82. ©2010 AACR.
- Published
- 2010
46. Characterization of response to radiation mediated gene therapy by means of multimodality imaging
- Author
-
Chad R, Haney, Adrian D, Parasca, Xiaobing, Fan, Rebecca M, Bell, Marta A, Zamora, Gregory S, Karczmar, Helena J, Mauceri, Howard J, Halpern, Ralph R, Weichselbaum, and Charles A, Pelizzari
- Subjects
Male ,Perfusion Imaging ,Mice, Nude ,Prostatic Neoplasms ,Genetic Therapy ,Prognosis ,Combined Modality Therapy ,Magnetic Resonance Imaging ,Article ,Mice ,Treatment Outcome ,Cell Line, Tumor ,Subtraction Technique ,Animals ,Humans ,Female ,Radiotherapy, Conformal - Abstract
Imaging techniques are under development to facilitate early analysis of spatial patterns of tumor response to combined radiation and antivascular gene therapy. A genetically modified, replication defective adenoviral vector (Ad.EGR-TNFalpha), injected intratumorally, mediates infected cells to express tumor necrosis factor alpha (TNFalpha), which is increased after exposure to radiation. The goal of this study was to characterize an image based "signature" for response to this combined radiation and gene therapy in mice with human prostate xenografts. This study is part of an imaged guided therapy project where such a signature would be useful in guiding subsequent treatments. Changes in the tumor micro-environment were assessed using MRI registered with electron paramagnetic resonance imaging which provides images of tissue oxygenation. Dynamic contrast-enhanced MRI was used to assess tissue perfusion. When compared with null vector (control) treatment, the ratio of contrast agent (Gd-DTPA-BMA) washout rate to uptake rate was lower (P = 0.001) after treatment, suggesting a more balanced perfusion. Concomitantly, oxygenation significantly increased in the treated animals and decreased or did not change in the control animals (P0.025). This is the first report of minimally invasive, quantitative, absolute oxygen measurements correlated with tissue perfusion in vivo.
- Published
- 2009
47. The influence of acute ethanol exposure on growth hormone release in female rats
- Author
-
Helena J. Mauceri and Sonya Conway
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Health (social science) ,medicine.medical_treatment ,Hypothalamus ,Biology ,Growth Hormone-Releasing Hormone ,Toxicology ,Biochemistry ,Clonidine ,Behavioral Neuroscience ,Internal medicine ,mental disorders ,medicine ,Animals ,Adrenergic agonist ,Saline ,reproductive and urinary physiology ,Sex Characteristics ,Ethanol ,Immunization, Passive ,Rats, Inbred Strains ,General Medicine ,Rats ,Somatostatin ,Endocrinology ,Neurology ,Growth Hormone ,Pituitary Gland ,Toxicity ,Female ,Alpha-2 adrenergic receptor ,Blood sampling ,Hormone ,medicine.drug - Abstract
This study investigates the site (hypothalamic or pituitary) at which ethanol (ETOH) alters GH release in female rats. Both the hypothalamic response to clonidine (CLON), an alpha 2 -adrenergic agonist, and the pituitary response to growth-hormone releasing hormone (GRH) were tested. Jugular cannulae were inserted for drug administration and undisturbed blood sampling. ETOH was injected IP 24 and 1 h before experimentation. In animals receiving saline or ETOH (1, 2, or 3 g/kg), there was no response to CLON and no difference in GH levels between groups. On the other hand, there was a significant surge in GH release in response to a high dose of GRH (1000 ng/kg) in both saline controls and in ETOH (3 g/kg) animals. Although there was no difference in the height of the surge between groups, baseline GH levels were higher in animals that received ETOH. In response to a low dose of GRH (250 ng/kg) the GH surge was only significant in the ETOH animals. In animals receiving somatostatin antiserum (anti-SRIF; 0.5 ml) in combination with the low GRH dose, the surge in GH levels was significant in both saline and ETOH animals, however, the surge was higher in saline compared to ETOH animals. The results of this study suggest that: 1) ETOH alters the SRIF system (release or reception) in female rats and that this interaction is evident when GRH concentration is low, and 2) ETOH may also inhibit GH release by interfering with the GRH system, however, the site of this influence most likely does not involve an alpha 2 -GRH component.
- Published
- 1991
- Full Text
- View/download PDF
48. The effect of acute ethanol exposure on clonidine-induced growth hormone release in male rats
- Author
-
Helena J. Mauceri and Sonya Conway
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Health (social science) ,medicine.medical_treatment ,Adrenergic ,Stimulation ,Toxicology ,Biochemistry ,Clonidine ,Behavioral Neuroscience ,Internal medicine ,mental disorders ,medicine ,Animals ,Dose Reduced ,Saline ,reproductive and urinary physiology ,Dose-Response Relationship, Drug ,Ethanol ,Chemistry ,Rats, Inbred Strains ,General Medicine ,Growth hormone secretion ,Rats ,Endocrinology ,Neurology ,Growth Hormone ,Toxicity ,Alpha-2 adrenergic receptor ,Pituitary Hormone-Releasing Hormones ,medicine.drug - Abstract
The influence of acute ethanol (ETOH) on the regulation of GH release has not been clearly elucidated. In this study the effect of ETOH on clonidine (CLON)-induced GH secretion was examined. To test the effect of ETOH on CLON-induced GH release doses of 1, 2, and 3 g/kg ETOH in a 25% ETOH/saline solution or 10 ml/kg saline were injected IP 24 and 1 h before CLON (30 micrograms/kg BW IV). Blood samples were withdrawn through a chronic jugular cannula. ETOH was found to alter the GH surge, which occurred 15 min after CLON administration in controls, in a dose-dependent manner. The 1 g/kg dose reduced the GH surge slightly but not significantly. The 2 g/kg dose suppressed the GH surge which was significantly lower than in controls. The 3 g/kg dose eliminated the GH surge completely. To determine if pituitary GH release is directly influenced by ETOH, animals were injected with GRH (250 ng/kg IV) one hour after the second dose of ETOH (3 g/kg IP). There was no difference in the GH surge in control or ETOH-injected animals. Anti-SRIF administered 30 minutes before ETOH or saline did not alter the response to GRH. These results suggest that ETOH reduces the GH responsiveness to alpha 2-GRH stimulation.
- Published
- 1991
- Full Text
- View/download PDF
49. Signal transducer and activator of transcription 1 regulates both cytotoxic and prosurvival functions in tumor cells
- Author
-
Ralph R. Weichselbaum, Elena V. Efimova, Bernard Roizman, Nikolai N. Khodarev, Thomas E. Darga, Michael A. Beckett, Edwardine Labay, Helena J. Mauceri, and Andy J. Minn
- Subjects
Cancer Research ,Transplantation, Heterologous ,Gene Expression ,Mice, Nude ,Biology ,Radiation Tolerance ,Small hairpin RNA ,Interferon-gamma ,Mice ,Transcription (biology) ,Radioresistance ,Cytotoxic T cell ,Animals ,Humans ,STAT1 ,RNA, Small Interfering ,Activator (genetics) ,Interferon-alpha ,Cell biology ,STAT1 Transcription Factor ,Oncology ,Drug Resistance, Neoplasm ,Immunology ,STAT protein ,biology.protein ,Carcinoma, Squamous Cell ,Female ,Signal transduction ,Neoplasm Transplantation - Abstract
Elsewhere, we reported that multiple serial in vivo passage of a squamous cell carcinoma cells (SCC61) concurrent with ionizing radiation (IR) treatment resulted in the selection of radioresistant tumor (nu61) that overexpresses the signal transducer and activator of transcription 1 (Stat1)/IFN-dependent pathway. Here, we report that (a) the Stat1 pathway is induced by IR, (b) constitutive overexpression of Stat1 is linked with failure to transmit a cytotoxic signal by radiation or IFNs, (c) selection of parental cell line SCC61 against IFN-α and IFN-γ leads to the same IR- and IFN-resistant phenotype as was obtained by IR selection, and (d) suppression of Stat1 by short hairpin RNA renders the IR-resistant nu61 cells radiosensitive to IR. We propose a model that transient induction of Stat1 by IFN, IR, or other stress signals activates cytotoxic genes and cytotoxic response. Constitutive overexpression of Stat1 on the other hand leads to the suppression of the cytotoxic response and induces prosurvival genes that, at high levels of Stat1, render the cells resistant to IR or other inducers of cell death. [Cancer Res 2007;67(19):9214–20]
- Published
- 2007
50. Establishment of a syngeneic model of hepatic colorectal oligometastases
- Author
-
Carlos A. Lopez, Ralph R. Weichselbaum, Kerrington D. Smith, Samual Hellman, Eric T. Kimchi, M.A. Beckett, James J. Mezhir, Helena J. Mauceri, James O. Park, and Mitchell C. Posner
- Subjects
Male ,medicine.medical_specialty ,Colorectal cancer ,medicine.medical_treatment ,Gastroenterology ,Metastasis ,Animal model ,Internal medicine ,Laparotomy ,medicine ,Animals ,Thoracotomy ,Behavior, Animal ,business.industry ,Liver Neoplasms ,Cancer ,Rats, Inbred Strains ,medicine.disease ,Rats ,Disease Models, Animal ,Localized disease ,Surgery ,Hepatectomy ,business ,Colorectal Neoplasms ,Neoplasm Transplantation ,Spleen - Abstract
Background Regional and systemic therapies aimed at improving the outcome for patients with colorectal hepatic metastases have met with modest yet tangible success. Currently, liver resection remains the only curative treatment, but only a minority of patients are candidates for surgery. Animal models are an ideal way to study new treatments for patients with metastatic colorectal cancer. We propose a syngeneic animal model of hepatic colorectal metastases that simulates oligometastases, which is a clinical state considered amenable to regional therapeutic strategies. Materials and methods BDIX (BD-9) rats underwent intrasplenic injection of DHD/K12/TRb (Prob/K12) cells to create hepatic metastases via the portal system. After injection of 5 × 10 6 cells, rats underwent laparotomy to determine metastatic burden. Histological analysis confirmed the presence of metastases from resected tumors. Results Fifty-three animals were prospectively treated and observed for the development of oligometastases defined as between 1 and 10 hepatic lesions. Thirty-six (68%) of the animals developed detectable metastases while 32 (60%) developed oligometastases (average = 4.40 ± 2.67). Four animals had overwhelming metastatic liver and peritoneal disease. All animals underwent peritoneal examination and thoracotomy to ensure localized disease. Histological analysis of five hepatectomy specimens confirmed the presence of metastatic cancer. Animals with oligometastases were healthy as evidenced by normal feeding and grooming behavior. Conclusions An animal model of oligometastatic colorectal cancer to the liver can reproducibly mimic the stage IV state in humans conducive to regional therapy and can be used reliably to test novel treatments and mechanisms of metastatic colorectal cancer.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.