111 results on '"Bernhard EJ"'
Search Results
2. Modifying radiosensitivity in tumors: Targeting the Ras-PI-3 kinase pathway
- Author
-
Muschel, RJ, Gupta, A, Bernhard, EJ, and McKenna, G
- Published
- 2016
3. A Phase I trial of the farnesyltransferase inhibitor L-778,123 and radiotherapy for locally advanced lung and head and neck cancer
- Author
-
Hahn, SM, Bernhard, EJ, Regine, W, Mohiuddin, M, Haller, DG, Stevenson, JP, Smith, D, Pramanik, B, Tepper, J, DeLaney, TF, Kiel, KD, Morrison, B, Deutsch, P, Muschel, RJ, and Mckenna, WG
- Abstract
PURPOSE: Preclinical data have demonstrated that farnesyltransferaseinhibitors (FTIs) are radiation sensitizers in selected cell lines. The objective of this Phase I trial was to determine the maximally tolerated dose of the FTI L-778,123 in combination with radiotherapy in non-small cell lung cancer (NSCLC) and head and neck cancer (HNC). EXPERIMENTAL DESIGN: L-778,123 was given by continuous i.v. infusion and dose escalated in conjunction with standard radiotherapy. The presence of a ras mutation was not required for study entry. RESULTS: Nine patients (six NSCLC patients and three HNC patients) were enrolled on two dose levels of FTI. No dose-limiting toxicities were observed at the first dose level of 280 mg/m2/day during weeks 1, 2, 4, and 5 of radiotherapy. One episode of dose-limiting toxicity, grade IV neutropenia, was observed in one of three patients treated at 560 mg/m2/day during weeks 1, 2, 4, 5, and 7. No episodes of dose-limiting mucositis, esophagitis, or pneumonitis were observed. Of the four patients with NSCLC with evaluable disease, three patients had a complete response to treatment and one patient had a partial response. A complete clinical response to treatment was observed in two patients with HNC. In vitro studies in tumor cells obtained from a NSCLC patient on this trial showed radiosensitization with FTI and that tumor cells accumulated in G2-M after L-778,123 treatment. CONCLUSIONS: The combination of L-778,123 and radiotherapy at dose level 1 is associated with acceptable toxicity. Local responses have been observed in four NSCLC patients without a clear increase in radiotherapy-associated toxicities.
- Published
- 2016
4. Apoptosis: an early event in metastatic inefficiency
- Author
-
Wong, CW, Lee, A, Shientag, L, Yu, J, Dong, Y, Kao, G, Al-Mehdi, AB, Bernhard, EJ, and Muschel, RJ
- Abstract
Whereas large numbers of cells from a primary tumor may gain access to the circulation, few of them will give rise to metastases. The mechanism of elimination of these tumor cells, often termed "metastatic inefficiency," is poorly understood. In this study, we show that apoptosis in the lungs within 1-2 days of introduction of the cells is an important component of metastatic inefficiency. First, we show that death of transformed, metastatic rat embryo cells occurred via apoptosis in the lungs 24-48 h after injection into the circulation. Second, we show that Bcl-2 overexpression in these cells inhibited apoptosis in culture and also conferred resistance to apoptosis in vivo in the lungs 24-48 h after injection. This inhibition of apoptosis led to significantly more macroscopic metastases. Third, comparison between the extent of apoptosis by a poorly metastatic cell line to that by a highly metastatic cell line 24 h after injection in the lungs revealed more apoptosis by the poorly metastatic cell line. These results indicate that apoptosis, which occurs at 24-48 h after hematogenous dissemination in the lungs is an important determinant of metastatic inefficiency. Although prior work has shown an association between apoptosis in culture and metastasis in vivo, this work shows that apoptosis in vivo corresponds to decreased metastasis in vivo.
- Published
- 2016
5. Farnesyltransferase inhibitors: an overview of the results of preclinical and clinical investigations
- Author
-
Brunner, TB, Hahn, SM, Gupta, AK, Muschel, RJ, Mckenna, WG, and Bernhard, EJ
- Abstract
This article presents an overview of preclinical studies and clinical trials of a number of independently derived farnesyltransferase inhibitors (FTIs). Potential targets and biological modes of action of FTIs are discussed, and the results of clinical trials are summarized. The significant efficacy of FTIs as single or combined agents in preclinical studies stands in contrast with only moderate effects in clinical Phase II-III trials. These results reveal a substantial gap in the understanding of the complex activity of FTIs and their interactions with cytotoxic agents. We conclude that the rational combination of FTIs with other therapies, taking into account the biological activities of the individual agents, may improve the clinical results obtained with FTIs.
- Published
- 2016
6. Direct evidence for the contribution of activated N-ras and K-ras oncogenes to increased intrinsic radiation resistance in human tumor cell lines
- Author
-
Bernhard, EJ, Stanbridge, EJ, Gupta, S, Gupta, AK, Soto, D, Bakanauskas, VJ, Cerniglia, GJ, Muschel, RJ, and Mckenna, WG
- Abstract
Transformation with ras oncogenes results in increased radiation sur vival in many but not all cells. In addition, prenyltransferase inhibitors which inhibit ras proteins by blocking posttranslational modification radiosensitize cells with oncogenic ras. These findings suggest that oncogenic ras contributes to intrinsic radiation resistance. However, because introduction of ras oncogenes does not increase radiation survival in all cells and because prenyltransferase inhibitors target molecules other than ras, these studies left the conclusion that ras increases the intrinsic radi ation resistance of tumor cells in doubt. Here we show that genetic inactivation of K- or N-ras oncogenes in human tumor cells (DLD-1 and HT1080, respectively) leads to increased radiosensitivity. Reintroduction of the activated N-ras gene into the HT1080 line, having lost its mutant allele, resulted in increased radiation resistance. This study lends further support to the hypothesis that expression of activated ras can contribute to intrinsic radiation resistance in human tumor cells and extends this finding to the K- and N- members of the ras family. These findings support the development of strategies that target ras for inactivation in the treatment of cancer.
- Published
- 2016
7. POLQ (DNA polyermase theta) as a novel therapeutic target: preclinical and clinical data
- Author
-
Higgins, GS, Prevo, R, Lundin, C, Helleday, T, Buffa, FM, Harris, AL, Muschel, RJ, Bernhard, EJ, Hickson, ID, and Mckenna, WG
- Published
- 2016
8. Alternative Transcript Initiation and Splicing as a Response to DNA Damage
- Author
-
Bernhard, EJ, Sprung, CN, Li, J, Hovan, D, Mckay, MJ, Forrester, HB, Bernhard, EJ, Sprung, CN, Li, J, Hovan, D, Mckay, MJ, and Forrester, HB
- Abstract
Humans are exposed to the DNA damaging agent, ionizing radiation (IR), from background radiation, medical treatments, occupational and accidental exposures. IR causes changes in transcription, but little is known about alternative transcription in response to IR on a genome-wide basis. These investigations examine the response to IR at the exon level in human cells, using exon arrays to comprehensively characterize radiation-induced transcriptional expression products. Previously uncharacterized alternative transcripts that preferentially occur following IR exposure have been discovered. A large number of genes showed alternative transcription initiation as a response to IR. Dose-response and time course kinetics have also been characterized. Interestingly, most genes showing alternative transcript induction maintained these isoforms over the dose range and times tested. Finally, clusters of co-ordinately up- and down-regulated radiation response genes were identified at specific chromosomal loci. These data provide the first genome-wide view of the transcriptional response to ionizing radiation at the exon level. This study provides novel insights into alternative transcripts as a mechanism for response to DNA damage and cell stress responses in general.
- Published
- 2011
9. Impact of screening for mental health concerns on health service utilization and functional status in primary care patients.
- Author
-
Reifler DR, Kessler HS, Bernhard EJ, Leon AC, and Martin GJ
- Published
- 1996
- Full Text
- View/download PDF
10. Overview and Lessons From the Preclinical Chemoradiotherapy Testing Consortium.
- Author
-
Buchsbaum JC, Coleman CN, Bernhard EJ, Espey MG, and Vikram B
- Subjects
- Chemoradiotherapy, Humans, Antineoplastic Agents therapeutic use, Neoplasms drug therapy
- Abstract
Purpose: In the current molecular-targeted cancer treatment era, many new agents are being developed so that optimizing therapy with a combination of radiation and drugs is complex. The use of emerging laboratory technologies to further biological understanding of drug-radiation mechanisms of action will enhance the efficiency of the progression from preclinical studies to clinical trials. In 2017, the National Cancer Institute (NCI) solicited proposals through PAR 16-111 to conduct preclinical research combining targeted anticancer agents in the Cancer Therapy Evaluation Program's portfolio with chemoradiation., Methods and Materials: The Preclinical Chemo-Radiotherapy Testing Consortium (PCRTC) was formed with 4 U01 programs supported to generate validated high-quality preclinical data on the effects of molecular therapeutics when added to standard-of-care therapies with a concentration on cancers of the pancreas, lung, head and neck, gastrointestinal tract, and brain., Results: The PCRTC provides a rational basis for prioritizing NCI-supported investigational new drugs or agents most likely to have clinical activity with chemoradiotherapy and accelerate the pace at which combined modality treatments with greater efficacy are identified and incorporated into standard treatment practices., Conclusions: Herein, we introduce and summarize the course of the PCRTC to date and report 3 preliminary observations from the consortium's work to date., (Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
11. Circulating Tumor DNA Assays in Clinical Cancer Research.
- Author
-
Ossandon MR, Agrawal L, Bernhard EJ, Conley BA, Dey SM, Divi RL, Guan P, Lively TG, McKee TC, Sorg BS, and Tricoli JV
- Subjects
- Early Detection of Cancer, High-Throughput Nucleotide Sequencing, Humans, Liquid Biopsy methods, Liquid Biopsy standards, Neoplasm Metastasis, Neoplasm Staging, Neoplasms blood, Reproducibility of Results, Research, Biomarkers, Tumor, Circulating Tumor DNA, DNA, Neoplasm, Neoplasms diagnosis, Neoplasms genetics
- Abstract
The importance of circulating free DNA (cfDNA) in cancer clinical research was recognized in 1994 when a mutated RAS gene fragment was detected in a patient's blood sample. Up to 1% of the total circulating DNA in patients with cancer is circulating tumor DNA (ctDNA) that originates from tumor cells. As ctDNA is rapidly cleared from the blood stream and can be obtained by minimally invasive methods, it can be used as a dynamic cancer biomarker for cancer early detection, diagnosis, and treatment monitoring. Despite the potential for clinical use, few ctDNA assays have been cleared or approved by the US Food and Drug Administration. As tools for clinical and translational research, current ctDNA assays face some challenges, and more research is needed to advance use of these assays. On September 29-30, 2016, the Division of Cancer Treatment and Diagnosis at the National Cancer Institute convened a workshop entitled "Circulating Tumor DNA Assays in Clinical Cancer Research" to garner input from industry experts, academia, and government research and regulatory agencies to understand and promote the translation of ctDNA assays to clinical research, with potential to advance to use in clinical practice. This Commentary presents the topics of the workshop covered in the presentations and points made in the discussions that followed: 1) background on ctDNA, 2) potential clinical utility of ctDNA assays, 3) assay technology, 4) assay clinical and analytical validation, and 5) industry perspectives. Additional relevant information that has come to light since the workshop has been included.
- Published
- 2018
- Full Text
- View/download PDF
12. Accurate, Precision Radiation Medicine: A Meta-Strategy for Impacting Cancer Care, Global Health, and Nuclear Policy and Mitigating Radiation Injury From Necessary Medical Use, Space Exploration, and Potential Terrorism.
- Author
-
Coleman CN, Prasanna PGS, Bernhard EJ, Buchsbaum JC, Ahmed MM, Capala J, Obcemea C, Deye JA, Pistenmma DA, Vikram B, Bernier J, and Dosanjh M
- Subjects
- Decision Making, Global Health, Health Plan Implementation, Humans, Interdisciplinary Communication, International Cooperation, Radiation Exposure adverse effects, Radiation Exposure prevention & control, Radiation Oncology legislation & jurisprudence, Neoplasms radiotherapy, Organizational Objectives, Precision Medicine, Radiation Injuries prevention & control, Radiation Oncology organization & administration, Societies, Medical, Space Flight, Terrorism
- Published
- 2018
- Full Text
- View/download PDF
13. The Future of Radiobiology.
- Author
-
Kirsch DG, Diehn M, Kesarwala AH, Maity A, Morgan MA, Schwarz JK, Bristow R, Demaria S, Eke I, Griffin RJ, Haas-Kogan D, Higgins GS, Kimmelman AC, Kimple RJ, Lombaert IM, Ma L, Marples B, Pajonk F, Park CC, Schaue D, Tran PT, Willers, Wouters, and Bernhard EJ
- Subjects
- Animals, Humans, Signal Transduction, Biomedical Research, Neoplasms radiotherapy, Radiobiology
- Abstract
Innovation and progress in radiation oncology depend on discovery and insights realized through research in radiation biology. Radiobiology research has led to fundamental scientific insights, from the discovery of stem/progenitor cells to the definition of signal transduction pathways activated by ionizing radiation that are now recognized as integral to the DNA damage response (DDR). Radiobiological discoveries are guiding clinical trials that test radiation therapy combined with inhibitors of the DDR kinases DNA-dependent protein kinase (DNA-PK), ataxia telangiectasia mutated (ATM), ataxia telangiectasia related (ATR), and immune or cell cycle checkpoint inhibitors. To maintain scientific and clinical relevance, the field of radiation biology must overcome challenges in research workforce, training, and funding. The National Cancer Institute convened a workshop to discuss the role of radiobiology research and radiation biologists in the future scientific enterprise. Here, we review the discussions of current radiation oncology research approaches and areas of scientific focus considered important for rapid progress in radiation sciences and the continued contribution of radiobiology to radiation oncology and the broader biomedical research community.
- Published
- 2018
- Full Text
- View/download PDF
14. Radiation-Therapeutic Agent Clinical Trials: Leveraging Advantages of a National Cancer Institute Programmatic Collaboration.
- Author
-
Takebe N, Ahmed MM, Vikram B, Bernhard EJ, Zwiebel J, Norman Coleman C, and Kunos CA
- Subjects
- Clinical Trials, Phase I as Topic, Clinical Trials, Phase II as Topic, Humans, National Cancer Institute (U.S.), Radiobiology, Research Design, United States, Neoplasms radiotherapy, Radiation-Sensitizing Agents therapeutic use
- Abstract
A number of oncology phase II radiochemotherapy trials with promising results have been conducted late in the overall experimental therapeutic agent development process. Accelerated development and approval of experimental therapeutic agents have stimulated further interest in much earlier radiation-agent studies to increase the likelihood of success in phase III trials. To sustain this interest, more forward-thinking preclinical radiobiology experimental designs are needed to improve discovery of promising radiochemotherapy plus agent combinations for clinical trial testing. These experimental designs should better inform next-step radiation-agent clinical trial dose, schedule, exposure, and therapeutic effect. Recognizing the need for a better strategy to develop preclinical data supporting radiation-agent phase I or II trials, the National Cancer Institute (NCI)-Cancer Therapy Evaluation Program (CTEP) and the NCI-Molecular Radiation Therapeutics Branch of the Radiation Research Program have partnered to promote earlier radiobiology studies of CTEP portfolio agents. In this Seminars in Radiation Oncology article, four key components of this effort are discussed. First, we outline steps for accessing CTEP agents for preclinical testing. Second, we propose radiobiology studies that facilitate transition from preclinical testing to early phase trial activation. Third, we navigate steps that walk through CTEP agent strategic development paths available for radiation-agent testing. Fourth, we highlight a new NCI-sponsored cooperative agreement grant supporting in vitro and in vivo radiation-CTEP agent testing that informs early phase trial designs. Throughout the article, we include contemporary examples of successful radiation-agent development initiatives., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
15. Improving the Predictive Value of Preclinical Studies in Support of Radiotherapy Clinical Trials.
- Author
-
Coleman CN, Higgins GS, Brown JM, Baumann M, Kirsch DG, Willers H, Prasanna PG, Dewhirst MW, Bernhard EJ, and Ahmed MM
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, HeLa Cells, Humans, Mice, Neoplasms pathology, Neoplastic Stem Cells drug effects, Reproducibility of Results, Xenograft Model Antitumor Assays methods, Antineoplastic Agents pharmacology, Drug Evaluation, Preclinical methods, Molecular Targeted Therapy methods, Neoplasms therapy, Radiation Tolerance drug effects, Radiation-Sensitizing Agents pharmacology
- Abstract
There is an urgent need to improve reproducibility and translatability of preclinical data to fully exploit opportunities for molecular therapeutics involving radiation and radiochemotherapy. For in vitro research, the clonogenic assay remains the current state-of-the-art of preclinical assays, whereas newer moderate and high-throughput assays offer the potential for rapid initial screening. Studies of radiation response modification by molecularly targeted agents can be improved using more physiologic 3D culture models. Elucidating effects on the cancer stem cells (CSC, and CSC-like) and developing biomarkers for defining targets and measuring responses are also important. In vivo studies are necessary to confirm in vitro findings, further define mechanism of action, and address immunomodulation and treatment-induced modification of the microenvironment. Newer in vivo models include genetically engineered and patient-derived xenograft mouse models and spontaneously occurring cancers in domesticated animals. Selection of appropriate endpoints is important for in vivo studies; for example, regrowth delay measures bulk tumor killing, whereas local tumor control assesses effects on CSCs. The reliability of individual assays requires standardization of procedures and cross-laboratory validation. Radiation modifiers must be tested as part of clinical standard of care, which includes radiochemotherapy for most tumors. Radiation models are compatible with but also differ from those used for drug screening. Furthermore, the mechanism of a drug as a chemotherapeutic agent may be different from its interaction with radiation and/or radiochemotherapy. This provides an opportunity to expand the use of molecular-targeted agents. Clin Cancer Res; 22(13); 3138-47. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
16. Preclinical Data on Efficacy of 10 Drug-Radiation Combinations: Evaluations, Concerns, and Recommendations.
- Author
-
Stone HB, Bernhard EJ, Coleman CN, Deye J, Capala J, Mitchell JB, and Brown JM
- Abstract
Background: Clinical testing of new therapeutic interventions requires comprehensive, high-quality preclinical data. Concerns regarding quality of preclinical data have been raised in recent reports. This report examines the data on the interaction of 10 drugs with radiation and provides recommendations for improving the quality, reproducibility, and utility of future studies. The drugs were AZD6244, bortezomib, 17-DMAG, erlotinib, gefitinib, lapatinib, oxaliplatin/Lipoxal, sunitinib (Pfizer, Corporate headquarters, New York, NY), thalidomide, and vorinostat., Methods: In vitro and in vivo data were tabulated from 125 published papers, including methods, radiation and drug doses, schedules of administration, assays, measures of interaction, presentation and interpretation of data, dosimetry, and conclusions., Results: In many instances, the studies contained inadequate or unclear information that would hamper efforts to replicate or intercompare the studies, and that weakened the evidence for designing and conducting clinical trials. The published reports on these drugs showed mixed results on enhancement of radiation response, except for sunitinib, which was ineffective., Conclusions: There is a need for improved experimental design, execution, and reporting of preclinical testing of agents that are candidates for clinical use in combination with radiation. A checklist is provided for authors and reviewers to ensure that preclinical studies of drug-radiation combinations meet standards of design, execution, and interpretation, and report necessary information to ensure high quality and reproducibility of studies. Improved design, execution, common measures of enhancement, and consistent interpretation of preclinical studies of drug-radiation interactions will provide rational guidance for prioritizing drugs for clinical radiotherapy trials and for the design of such trials., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
17. Radioprotectors and Radiomitigators for Improving Radiation Therapy: The Small Business Innovation Research (SBIR) Gateway for Accelerating Clinical Translation.
- Author
-
Prasanna PG, Narayanan D, Hallett K, Bernhard EJ, Ahmed MM, Evans G, Vikram B, Weingarten M, and Coleman CN
- Subjects
- Genetic Therapy, Genistein therapeutic use, Humans, Megakaryocyte Progenitor Cells physiology, Neoplasms psychology, Peptide Fragments therapeutic use, Quality of Life, Radiotherapy adverse effects, Small Business, Thrombin therapeutic use, Toll-Like Receptor 5 agonists, Neoplasms radiotherapy, Radiation Protection
- Abstract
Although radiation therapy is an important cancer treatment modality, patients may experience adverse effects. The use of a radiation-effect modulator may help improve the outcome and health-related quality of life (HRQOL) of patients undergoing radiation therapy either by enhancing tumor cell killing or by protecting normal tissues. Historically, the successful translation of radiation-effect modulators to the clinic has been hindered due to the lack of focused collaboration between academia, pharmaceutical companies and the clinic, along with limited availability of support for such ventures. The U.S. Government has been developing medical countermeasures against accidental and intentional radiation exposures to mitigate the risk and/or severity of acute radiation syndrome (ARS) and the delayed effects of acute radiation exposures (DEARE), and there is now a drug development pipeline established. Some of these medical countermeasures could potentially be repurposed for improving the outcome of radiation therapy and HRQOL of cancer patients. With the objective of developing radiation-effect modulators to improve radiotherapy, the Small Business Innovation Research (SBIR) Development Center at the National Cancer Institute (NCI), supported by the Radiation Research Program (RRP), provided funding to companies from 2011 to 2014 through the SBIR contracts mechanism. Although radiation-effect modulators collectively refer to radioprotectors, radiomitigators and radiosensitizers, the focus of this article is on radioprotection and mitigation of radiation injury. This specific SBIR contract opportunity strengthened existing partnerships and facilitated new collaborations between academia and industry. In this commentary, we assess the impact of this funding opportunity, outline the review process, highlight the organ/site-specific disease needs in the clinic for the development of radiation-effect modulators, provide a general understanding of a framework for gathering preclinical and clinical evidence to obtain regulatory approval and provide a basis for broader venture capital needs and support from pharmaceutical companies to fully capitalize on the advances made thus far in this field.
- Published
- 2015
- Full Text
- View/download PDF
18. Lessons learned from radiation oncology clinical trials.
- Author
-
Liu FF, Okunieff P, Bernhard EJ, Stone HB, Yoo S, Coleman CN, Vikram B, Brown M, Buatti J, and Guha C
- Subjects
- Biomarkers, Tumor, Combined Modality Therapy, Humans, Quality Assurance, Health Care, Treatment Failure, Clinical Trials as Topic methods, Neoplasms radiotherapy
- Abstract
A workshop entitled "Lessons Learned from Radiation Oncology Trials" was held on December 7-8, 2011, in Bethesda, MD, to present and discuss some of the recently conducted radiation oncology clinical trials with a focus on those that failed to refute the null hypothesis. The objectives of this workshop were to summarize and examine the questions that these trials provoked, to assess the quality and limitations of the preclinical data that supported the hypotheses underlying these trials, and to consider possible solutions to these challenges for the design of future clinical trials. Several themes emerged from the discussions: (i) opportunities to learn from null-hypothesis trials through tissue and imaging studies; (ii) value of preclinical data supporting the design of combinatorial therapies; (iii) significance of validated biomarkers; (iv) necessity of quality assurance in radiotherapy delivery; (v) conduct of sufficiently powered studies to address the central hypotheses; and (vi) importance of publishing results of the trials regardless of the outcome. The fact that well-designed hypothesis-driven clinical trials produce null or negative results is expected given the limitations of trial design and complexities of cancer biology. It is important to understand the reasons underlying such null results, however, to effectively merge the technologic innovations with the rapidly evolving biology for maximal patient benefit through the design of future clinical trials., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
19. Harnessing the potential of radiation-induced immune modulation for cancer therapy.
- Author
-
Ahmed MM, Hodge JW, Guha C, Bernhard EJ, Vikram B, and Coleman CN
- Subjects
- Animals, Combined Modality Therapy, Humans, Neoplasms radiotherapy, Cancer Vaccines administration & dosage, Cancer Vaccines immunology, Neoplasms immunology, Neoplasms therapy
- Abstract
The conventional use of radiotherapy is for local tumor control. Radiotherapy of the primary tumor can prevent the development of distant metastases, but this modality is generally not effective for treating preexisting systemic disease. However, radiation-induced tumor destruction may be considered a novel strategy for in situ cancer vaccination, in which tumor antigens released from dying tumor cells may be presented in an immunostimulatory context. Moreover, radiation has been demonstrated to induce immunogenic modulation in various tumor types by altering the biology of surviving cells to render them more susceptible to T cell-mediated killing. Finally, radiotherapy typically has a favorable toxicity profile and is associated with the absence of systemic immunosuppression. Together, these properties suggest that radiotherapy may serve as an important component of combinatorial immunotherapies aimed at augmenting systemic antitumor immunity. Here, we provide an overview of the radiation-induced modulations of the immune system that may be harnessed for cancer therapy., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
20. G-CSF rescues tumor growth and neo-angiogenesis during liver metastasis under host angiopoietin-2 deficiency.
- Author
-
Im JH, Tapmeier T, Balathasan L, Gal A, Yameen S, Hill S, Smart S, Noterdaeme O, Kelly M, Brady M, Fu W, Kruse K, Bernhard EJ, Augustin HG, and Muschel RJ
- Subjects
- Adenocarcinoma blood supply, Adenocarcinoma metabolism, Angiopoietin-2 genetics, Animals, CD11b Antigen metabolism, Capillary Permeability, Cell Line, Tumor, Cell Proliferation, Cytokines blood, Cytokines physiology, Female, Granulocyte Colony-Stimulating Factor blood, Green Fluorescent Proteins biosynthesis, Liver immunology, Liver metabolism, Liver pathology, Liver Neoplasms blood supply, Liver Neoplasms metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Transplantation, Neoplasms, Experimental blood supply, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Neutrophils immunology, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptor, TIE-2, Statistics, Nonparametric, Vascular Endothelial Growth Factor A metabolism, Adenocarcinoma secondary, Angiopoietin-2 deficiency, Granulocyte Colony-Stimulating Factor physiology, Liver Neoplasms secondary, Neovascularization, Pathologic
- Abstract
Suppression of neo-angiogenesis is a clinically used anti-tumor strategy with new targets such as angiopoietin-2 (Ang2) being proposed. However, the functions of Ang2 in vascular remodeling, inflammation and tumor growth are not consistent. We examined effect of depletion of host Ang2 on liver colony formation using Ang2 deficient (Ang2(-/-)) mice. Surprisingly, the metastatic colonies formed in Ang2(-/-) mice were larger than those in the wild type. These colonies had greater vascular density with more pericyte coverage than the vessels in liver colonies in the wild type. Liver VEGF concentration in both genotypes was equivalent, and thus, the differences appeared VEGF independent. However, after colony formation, the serum concentration of granulocyte-colony stimulating factor (G-CSF) and CXCL1 in Ang2(-/-) mice was 12 and 6 times greater than after colony formation in wild type. Increase of these two cytokines was associated with two times greater numbers of neutrophils recruited to the liver. Two times more Tie2+/CD11b+/CD31- cells were present in the tumors in Ang2(-/-) than in the wild type livers. These results suggest that the depletion of host Ang2 induced compensatory VEGF-independent angiogenic mechanisms and thus enhanced liver metastatic colony growth and colony vascularity. They further indicate organotypic differences in response to tumor metastasis. In contrast, Ang2 deficiency inhibited tumor growth during metastatic colony formation in the lung, consistent with the reports of decreased pulmonary seeding of tumor cells after pharmacological inhibition of Ang2. Further studies are thus required to assess the effects of pharmacological Ang2 blockade for cancer patients particularly in the liver., (Copyright © 2012 UICC.)
- Published
- 2013
- Full Text
- View/download PDF
21. Normal tissue protection for improving radiotherapy: Where are the Gaps?
- Author
-
Prasanna PG, Stone HB, Wong RS, Capala J, Bernhard EJ, Vikram B, and Coleman CN
- Abstract
Any tumor could be controlled by radiation therapy if sufficient dose were delivered to all tumor cells. Although technological advances in physical treatment delivery have been developed to allow more radiation dose conformity, normal tissues are invariably included in any radiation field within the tumor volume and also as part of the exit and entrance doses relevant for particle therapy. Mechanisms of normal tissue injury and related biomarkers are now being investigated, facilitating the discovery and development of a next generation of radiation protectors and mitigators. Bringing recent research advances stimulated by development of radiation countermeasures for mass casualties, to clinical cancer care requires understanding the impact of protectors and mitigators on tumor response. These may include treatments that modify cellular damage and death processes, inflammation, alteration of normal flora, wound healing, tissue regeneration and others, specifically to counter cancer site-specific adverse effects to improve outcome of radiation therapy. Such advances in knowledge of tissue and organ biology, mechanisms of injury, development of predictive biomarkers and mechanisms of radioprotection have re-energized the field of normal tissue protection and mitigation. Since various factors, including organ sensitivity to radiation, cellular turnover rate, and differences in mechanisms of injury manifestation and damage response vary among tissues, successful development of radioprotectors/mitigators/treatments may require multiple approaches to address cancer site specific needs. In this review, we discuss examples of important adverse effects of radiotherapy (acute and intermediate to late occurring, when it is delivered either alone or in conjunction with chemotherapy, and important limitations in the current approaches of using radioprotectors and/or mitigators for improving radiation therapy. Also, we are providing general concepts for drug development for improving radiation therapy.
- Published
- 2012
22. NVP-BEZ235 and NVP-BGT226, dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitors, enhance tumor and endothelial cell radiosensitivity.
- Author
-
Fokas E, Yoshimura M, Prevo R, Higgins G, Hackl W, Maira SM, Bernhard EJ, McKenna WG, and Muschel RJ
- Subjects
- Cell Line, Tumor, Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells drug effects, Humans, Phosphoinositide-3 Kinase Inhibitors, TOR Serine-Threonine Kinases antagonists & inhibitors, Imidazoles pharmacology, Quinolines pharmacology, Radiation-Sensitizing Agents pharmacology
- Abstract
Background: The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is activated in tumor cells and promotes tumor cell survival after radiation-induced DNA damage. Because the pathway may not be completely inhibited after blockade of PI3K itself, due to feedback through mammalian target of rapamycin (mTOR), more effective inhibition might be expected by targeting both PI3K and mTOR inhibition., Materials and Methods: We investigated the effect of two dual PI3K/mTOR (both mTORC1 and mTORC2) inhibitors, NVP-BEZ235 and NVP-BGT226, on SQ20B laryngeal and FaDu hypopharyngeal cancer cells characterised by EGFR overexpression, on T24 bladder tumor cell lines with H-Ras mutation and on endothelial cells. Analysis of target protein phosphorylation, clonogenic survival, number of residual γH2AX foci, cell cycle and apoptosis after radiation was performed in both tumor and endothelial cells. In vitro angiogenesis assays were conducted as well., Results: Both compounds effectively inhibited phosphorylation of Akt, mTOR and S6 target proteins and reduced clonogenic survival in irradiated tumor cells. Persistence of DNA damage, as evidenced by increased number of γH2AX foci, was detected after irradiation in the presence of PI3K/mTOR inhibition, together with enhanced G2 cell cycle delay. Treatment with one of the inhibitors, NVP-BEZ235, also resulted in decreased clonogenicity after irradiation of tumor cells under hypoxic conditions. In addition, NVP-BEZ235 blocked VEGF- and IR-induced Akt phosphorylation and increased radiation killing in human umbilical venous endothelial cells (HUVEC) and human dermal microvascular dermal cells (HDMVC). NVP-BEZ235 inhibited VEGF-induced cell migration and capillary tube formation in vitro and enhanced the antivascular effect of irradiation. Treatment with NVP-BEZ235 moderately increased apoptosis in SQ20B and HUVEC cells but not in FaDu cells, and increased necrosis in both tumor and endothelial all cells tumor., Conclusions: The results of this study demonstrate that PI3K/mTOR inhibitors can enhance radiation-induced killing in tumor and endothelial cells and may be of benefit when combined with radiotherapy.
- Published
- 2012
- Full Text
- View/download PDF
23. Modulation of the tumor microvasculature by phosphoinositide-3 kinase inhibition increases doxorubicin delivery in vivo.
- Author
-
Qayum N, Im J, Stratford MR, Bernhard EJ, McKenna WG, and Muschel RJ
- Subjects
- Animals, Drug Delivery Systems, Female, Humans, Hypoxia drug therapy, Immunoenzyme Techniques, Mammary Neoplasms, Animal enzymology, Mice, Mice, Nude, Mice, Transgenic, Phosphatidylinositol 3-Kinases metabolism, Doxorubicin therapeutic use, Indazoles pharmacology, Mammary Neoplasms, Animal blood supply, Mammary Neoplasms, Animal drug therapy, Microvessels drug effects, Neovascularization, Pathologic prevention & control, Phosphoinositide-3 Kinase Inhibitors, Sulfonamides pharmacology
- Abstract
Purpose: Because effective drug delivery is often limited by inadequate vasculature within the tumor, the ability to modulate the tumor microenvironment is one strategy that may achieve better drug distribution. We have previously shown that treatment of mice bearing tumors with phosphoinositide-3 kinase (PI3K) inhibitors alters vascular structure in a manner analogous to vascular normalization and results in increased perfusion of the tumor. On the basis of that result, we asked whether inhibition of PI3K would improve chemotherapy delivery., Experimental Design: Mice with xenografts using the cell line SQ20B bearing a hypoxia marker or MMTV-neu transgenic mice with spontaneous breast tumors were treated with the class I PI3K inhibitor GDC-0941. The tumor vasculature was evaluated by Doppler ultrasound, and histology. The delivery of doxorubicin was assessed using whole animal fluorescence, distribution on histologic sections, high-performance liquid chromatography on tumor lysates, and tumor growth delay., Results: Treatment with GDC-0941 led to approximately three-fold increases in perfusion, substantially reduced hypoxia and vascular normalization by histology. Significantly increased amounts of doxorubicin were delivered to the tumors correlating with synergistic tumor growth delay. The GDC-0941 itself had no effect on tumor growth., Conclusion: Inhibition of PI3K led to vascular normalization and improved delivery of a chemotherapeutic agent. This study highlights the importance of the microvascular effects of some novel oncogenic signaling inhibitors and the need to take those changes into account in the design of clinical trials many of which use combinations of chemotherapeutic agents., (© 2011 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
24. Dual inhibition of the PI3K/mTOR pathway increases tumor radiosensitivity by normalizing tumor vasculature.
- Author
-
Fokas E, Im JH, Hill S, Yameen S, Stratford M, Beech J, Hackl W, Maira SM, Bernhard EJ, McKenna WG, and Muschel RJ
- Subjects
- Animals, Dose-Response Relationship, Drug, Humans, Mice, Neoplasms, Experimental blood supply, Xenograft Model Antitumor Assays, Neoplasms, Experimental radiotherapy, Neovascularization, Pathologic, Phosphoinositide-3 Kinase Inhibitors, Radiation Tolerance, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
The aberrant vascular architecture of solid tumors results in hypoxia that limits the efficacy of radiotherapy. Vascular normalization using antiangiogenic agents has been proposed as a means to improve radiation therapy by enhancing tumor oxygenation, but only short-lived effects for this strategy have been reported so far. Here, we show that NVP-BEZ235, a dual inhibitor of phosphoinositide-3-kinase (PI3K) and mTOR, can improve tumor oxygenation and vascular structure over a prolonged period that achieves the aim of effective vascular normalization. Because PI3K inhibition can radiosensitize tumor cells themselves, our experimental design explicitly distinguished effects on the blood vasculature versus tumor cells. Drug administration coincident with radiation enhanced the delay in tumor growth without changing tumor oxygenation, establishing that radiosensitization is a component of the response. However, the enhanced growth delay was substantially greater after induction of vascular normalization, meaning that this treatment enhanced the tumoral radioresponse. Importantly, changes in vascular morphology persisted throughout the entire course of the experiment. Our findings indicated that targeting the PI3K/mTOR pathway can modulate the tumor microenvironment to induce a prolonged normalization of blood vessels. The substantial therapeutic gain observed after combination of NVP-BEZ235 with irradiation has conceptual implications for cancer therapy and could be of broad translational importance., (©2011 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
25. New translational possibilities for microenvironmental modulation of radiosensitivity.
- Author
-
Glazer PM, Le QT, Bristow R, Helleday T, Pelroy R, and Bernhard EJ
- Subjects
- Animals, Cell Hypoxia, DNA Repair, Humans, Neoplasms genetics, Neoplasms immunology, Neoplasms pathology, Neoplasms radiotherapy, Signal Transduction, Protein Biosynthesis radiation effects, Radiation Tolerance
- Published
- 2011
- Full Text
- View/download PDF
26. Radiation Resistance in Cancer Therapy: meeting summary and research opportunities. Report of an NCI Workshop held September 1-3, 2010.
- Author
-
Glazer PM, Grandis J, Powell SN, Brown JM, Helleday T, Bristow R, Powis G, Hill RP, Le QT, Pelroy R, Mohla S, and Bernhard EJ
- Subjects
- Humans, Neoplasms radiotherapy, Radiation Tolerance
- Published
- 2011
27. Interventions that induce modifications in the tumor microenvironment.
- Author
-
Bernhard EJ
- Subjects
- Angiogenesis Inhibitors therapeutic use, Animals, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents pharmacology, Cell Adhesion drug effects, Cell Hypoxia, Clinical Trials, Phase III as Topic, Combined Modality Therapy, Cysteine analogs & derivatives, Cysteine therapeutic use, Cytotoxins therapeutic use, Drug Screening Assays, Antitumor, Extracellular Matrix drug effects, Genetic Therapy, Humans, Immunotherapy methods, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins physiology, Neoplasms metabolism, Neoplasms pathology, Neoplasms, Experimental drug therapy, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Neovascularization, Pathologic drug therapy, Organoselenium Compounds therapeutic use, Radiation Tolerance, Radiotherapy methods, Selenocysteine analogs & derivatives, Signal Transduction drug effects, Stromal Cells drug effects, Antineoplastic Agents therapeutic use, Molecular Targeted Therapy, Neoplasms therapy, Tumor Microenvironment drug effects, Tumor Microenvironment physiology, Tumor Microenvironment radiation effects
- Abstract
Non-surgical cancer therapeutic strategies have focused primarily on direct killing of cancer cells by chemotherapy and/or radiation therapy. However, it is becoming increasingly clear that the efficacy of these therapies can be significantly influenced by the tumor microenvironment. The microenvironment poses both obstacles and opportunities for new therapeutic interventions. New developments in this area are the topic of this review., (Published by Elsevier SAS.)
- Published
- 2011
- Full Text
- View/download PDF
28. A small interfering RNA screen of genes involved in DNA repair identifies tumor-specific radiosensitization by POLQ knockdown.
- Author
-
Higgins GS, Prevo R, Lee YF, Helleday T, Muschel RJ, Taylor S, Yoshimura M, Hickson ID, Bernhard EJ, and McKenna WG
- Subjects
- Antineoplastic Agents, Alkylating pharmacology, Cell Line, Tumor, Cell Survival genetics, Cell Survival radiation effects, DNA-Directed DNA Polymerase deficiency, DNA-Directed DNA Polymerase metabolism, Dacarbazine analogs & derivatives, Dacarbazine pharmacology, Gene Knockdown Techniques, HeLa Cells, Histones, Humans, Infrared Rays, Neoplasms drug therapy, Radiation Tolerance genetics, Temozolomide, Transfection, DNA Polymerase theta, DNA Repair genetics, DNA-Directed DNA Polymerase genetics, Neoplasms genetics, Neoplasms radiotherapy, RNA, Small Interfering genetics
- Abstract
The effectiveness of radiotherapy treatment could be significantly improved if tumor cells could be rendered more sensitive to ionizing radiation (IR) without altering the sensitivity of normal tissues. However, many of the key therapeutically exploitable mechanisms that determine intrinsic tumor radiosensitivity are largely unknown. We have conducted a small interfering RNA (siRNA) screen of 200 genes involved in DNA damage repair aimed at identifying genes whose knockdown increased tumor radiosensitivity. Parallel siRNA screens were conducted in irradiated and unirradiated tumor cells (SQ20B) and irradiated normal tissue cells (MRC5). Using gammaH2AX foci at 24 hours after IR, we identified several genes, such as BRCA2, Lig IV, and XRCC5, whose knockdown is known to cause increased cell radiosensitivity, thereby validating the primary screening end point. In addition, we identified POLQ (DNA polymerase ) as a potential tumor-specific target. Subsequent investigations showed that POLQ knockdown resulted in radiosensitization of a panel of tumor cell lines from different primary sites while having little or no effect on normal tissue cell lines. These findings raise the possibility that POLQ inhibition might be used clinically to cause tumor-specific radiosensitization.
- Published
- 2010
- Full Text
- View/download PDF
29. Modulating tumor vasculature through signaling inhibition to improve cytotoxic therapy.
- Author
-
Maity A and Bernhard EJ
- Subjects
- Humans, Neoplasms metabolism, Neoplasms pathology, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic therapy, Oxygen metabolism, Signal Transduction, Neoplasms blood supply, Neoplasms therapy
- Abstract
The tumor microenvironment is a key factor in cancer treatment response. Recent work has shown that changes in the tumor vasculature can be achieved by inhibiting tumor cell signaling, resulting in enhanced tumor oxygenation. These changes could promote responses to both chemo- and radiation therapy.
- Published
- 2010
- Full Text
- View/download PDF
30. Cell signalling and radiation survival: the impact of protein phosphatases.
- Author
-
Hamilton J and Bernhard EJ
- Subjects
- Animals, Apoptosis physiology, Apoptosis radiation effects, Cell Cycle physiology, Cell Cycle radiation effects, DNA Damage, Enzyme Inhibitors pharmacology, Humans, Models, Biological, Neoplasms drug therapy, Neoplasms pathology, Neoplasms physiopathology, Neoplasms radiotherapy, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases classification, Radiation Tolerance drug effects, Radiation Tolerance physiology, Cell Survival physiology, Cell Survival radiation effects, Phosphoprotein Phosphatases physiology, Signal Transduction physiology, Signal Transduction radiation effects
- Abstract
Purpose: This review will examine the role of phosphatases in cancer cell signalling and also outline emerging findings regarding the influence of phosphatases on tumor cell survival after ionising radiation., Conclusion: The exposure of tumour cells to clinically relevant doses of ionising radiation causes DNA damage and rapidly activates a series of signaling cascades involved in cell survival (reviewed in (Valerie et al. 2007 )). The role of kinases in this signalling has been extensively studied, but the role of phosphatases is less well defined. There is an abundance of literature implicating phosphatases in cell cycle control, cell growth and survival but there has been much less reported on the involvement of these enzymes as determinants of radiosensitivity. Recent studies, however, suggest that phosphatases may modulate tumor cell radiosensitivity and may be targets for the enhancement of radiotherapy.
- Published
- 2009
- Full Text
- View/download PDF
31. Phosphatase inhibition and cell survival after DNA damage induced by radiation.
- Author
-
Hamilton J, Grawenda AM, and Bernhard EJ
- Subjects
- Cell Line, Tumor, Humans, Marine Toxins, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins genetics, Nuclear Proteins metabolism, Okadaic Acid pharmacology, Oxazoles pharmacology, Phosphoprotein Phosphatases, Phosphoric Monoester Hydrolases genetics, Phosphoric Monoester Hydrolases metabolism, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering administration & dosage, RNA, Small Interfering genetics, Radiation Tolerance, Signal Transduction, Transfection, DNA Damage, DNA, Neoplasm radiation effects, Phosphoric Monoester Hydrolases antagonists & inhibitors
- Abstract
The activity of certain kinases can promote cell survival after DNA damage, but the role of phosphatases in determining cell fate, although documented, is much less well defined. We sought to define a role for phosphatases in radiation survival and identify potential targets for intervention. By using naturally occurring inhibitors and siRNA we have assessed inhibition of four serine/threonine phosphatases PP1, PP2A, PHLPP and PHLPPL in a panel of tumor cell lines with H-, K- or N-ras mutations or with EGFR activation for effects on tumor cell radiosensitivity. Calyculin A, which inhibits both PP1 and PP2A reduced radiation survival in SQ20B cells (overexpressing EGFR). Okadaic acid, which preferentially inhibits PP2A showed less effect in SQ20B cells suggesting a greater involvement of PP1 in modulating radiosensitivity of these cells. T24 cells (H-Ras mutant) appeared equally sensitive to both inhibitors. The suggestion from inhibitors that PP1 might be important in radiosensitivity was supported by the greater sensitization obtained after knocking down expression of the catalytic sub-unit of PP1 over that seen after PP2A knockdown. Knocking down the PP2C like phosphatase PHLPPL also increased radiosensitivity in all cell lines tested where a second isoform PHLPP had little effect. These data suggest that targeted inhibition of phosphatase activity may be an alternative to kinase inhibition to enhance radiosensitivity in tumors.
- Published
- 2009
- Full Text
- View/download PDF
32. Tumor vascular changes mediated by inhibition of oncogenic signaling.
- Author
-
Qayum N, Muschel RJ, Im JH, Balathasan L, Koch CJ, Patel S, McKenna WG, and Bernhard EJ
- Subjects
- Animals, Cell Hypoxia physiology, Cell Line, Tumor, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Female, Fibrosarcoma metabolism, Humans, Mice, Mice, SCID, Mice, Transgenic, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Oxygen metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, Xenograft Model Antitumor Assays, ras Proteins antagonists & inhibitors, ras Proteins metabolism, Enzyme Inhibitors pharmacology, Fibrosarcoma blood supply, Fibrosarcoma drug therapy
- Abstract
Many inhibitors of the epidermal growth factor receptor (EGFR)-RAS-phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway are in clinical use or under development for cancer therapy. Here, we show that treatment of mice bearing human tumor xenografts with inhibitors that block EGFR, RAS, PI3K, or AKT resulted in prolonged and durable enhancement of tumor vascular flow, perfusion, and decreased tumor hypoxia. The vessels in the treated tumors had decreased tortuosity and increased internodal length accounting for the functional alterations. Inhibition of tumor growth cannot account for these results, as the drugs were given at doses that did not alter tumor growth. The tumor cell itself was an essential target, as HT1080 tumors that lack EGFR did not respond to an EGFR inhibitor but did respond with vascular alterations to RAS or PI3K inhibition. We extended these observations to spontaneously arising tumors in MMTV-neu mice. These tumors also responded to PI3K inhibition with decreased tumor hypoxia, increased vascular flow, and morphologic alterations of their vessels, including increased vascular maturity and acquisition of pericyte markers. These changes are similar to the vascular normalization that has been described after the antiangiogenic treatment of xenografts. One difficulty in the use of vascular normalization as a therapeutic strategy has been its limited duration. In contrast, blocking tumor cell RAS-PI3K-AKT signaling led to persistent vascular changes that might be incorporated into clinical strategies based on improvement of vascular flow or decreased hypoxia. These results indicate that vascular alterations must be considered as a consequence of signaling inhibition in cancer therapy.
- Published
- 2009
- Full Text
- View/download PDF
33. Epigenetic modulation of radiation response in human cancer cells with activated EGFR or HER-2 signaling: potential role of histone deacetylase 6.
- Author
-
Kim IA, No M, Lee JM, Shin JH, Oh JS, Choi EJ, Kim IH, Atadja P, and Bernhard EJ
- Subjects
- Cell Line, Tumor, Down-Regulation, ErbB Receptors, Histone Deacetylase 6, Histone Deacetylases metabolism, Humans, Indoles, Neoplasms drug therapy, Panobinostat, Receptor, ErbB-2, Signal Transduction, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Hydroxamic Acids pharmacology, Radiation Tolerance drug effects, Radiation-Sensitizing Agents pharmacology
- Abstract
Background and Purpose: Histone deacetylase inhibitors (HDIs) are prototypes of agents targeting epigenetic modifications and have received considerable attention for their promise as targeted anticancer drugs. We examined the effects and potential mechanism(s) of combining LBH589 and irradiation in human cancer cells having activated EGFR or HER-2 signaling, focusing on the role of HDAC6., Methods and Materials: We evaluated whether the HDI, LBH589, would radiosensitize a panel of human tumor cell lines having activated EGFR or HER-2 signaling. A mechanistic role for the HDAC6 isotype was investigated using RNA interference and ectopic overexpression HDAC6., Results: The HDI, LBH589, enhanced the radiosensitivity of the human carcinoma cell lines we tested. Radiosensitization was accompanied by abrogation of radiation-induced G2/M arrest and was associated with aberrant mitotic features and prolonged gammaH2AX foci. Radiation-induced apoptosis was also increased. LBH589 radiosensitized cells with activated EGFR or HER-2 signaling to a greater degree than the HDIs SK7041 or TSA. However radiosensitization by the three HDI was equivalent in cells without activation of this signaling. LBH589 led acetylation of histone H3 and HSP90. This was associated with down-regulation of the client oncoproteins EGFR, HER-2, and decreased phosphorylation of Akt and ERK. Specific inhibition of HDAC6 by RNAi increased radiosensitivity as well as increasing acetylation of HSP90 and reducing the association of HSP90 with its client proteins. Conversely, ectopic overexpression of HDAC6 isotype increased the levels of p-EGFR and p-AKT expression, and reduced LBH589-mediated radiosensitization., Conclusions: These findings define a unique mechanism for counteracting pro-survival signaling from EGFR or HER-2 that is present in many tumor cells.
- Published
- 2009
- Full Text
- View/download PDF
34. Conventional radiotherapy or hypofractionation? A study of molecular changes resulting from different radiation fractionation schemes.
- Author
-
Hamilton J, Higgins G, and Bernhard EJ
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Dose Fractionation, Radiation, Dose-Response Relationship, Radiation, Female, Humans, Male, NF-kappa B biosynthesis, NF-kappa B genetics, NF-kappa B metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Breast Neoplasms radiotherapy, Prostatic Neoplasms radiotherapy
- Published
- 2009
- Full Text
- View/download PDF
35. Class I PI3 kinase inhibition by the pyridinylfuranopyrimidine inhibitor PI-103 enhances tumor radiosensitivity.
- Author
-
Prevo R, Deutsch E, Sampson O, Diplexcito J, Cengel K, Harper J, O'Neill P, McKenna WG, Patel S, and Bernhard EJ
- Subjects
- Cell Cycle, Cell Line, Tumor, DNA Damage, Drug Screening Assays, Antitumor, Flow Cytometry methods, G2 Phase, Histones metabolism, Humans, Phosphorylation, Pyrimidines antagonists & inhibitors, Radiation Tolerance, Enzyme Inhibitors pharmacology, Furans pharmacology, Phosphoinositide-3 Kinase Inhibitors, Pyridines pharmacology, Pyrimidines pharmacology, Radiation-Sensitizing Agents pharmacology
- Abstract
Cell signaling initiated at the epidermal growth factor receptor (EGFR), RAS oncoproteins, or PI3K contributes to a common pathway that promotes tumor survival after radiation-induced DNA damage. Inhibition of signaling at the level of EGFR, RAS, and PI3K has been tested, but clinical applicability has been shown only at the level of the EGFR or by inhibiting RAS indirectly with prenyltransferase inhibitors. Inhibition of PI3K with LY294002 or wortmannin lacks specificity and has shown unacceptable toxicity in preclinical studies. We previously showed that inhibiting class I PI3K expression with siRNA resulted in enhanced radiation killing of tumor cells. Here, we tested the possibility of achieving specific tumor cell radiosensitization with a pharmacologic inhibitor of class I PI3K, the pyridinylfuranopyrimidine inhibitor PI-103. Our results show that inhibiting PI3K activity reduces phosphorylation of AKT at serine 473. Reduced survival is seen in cells with AKT activation and seems preferential for tumor cells over cells in which AKT activity is not elevated. Reduced survival is accompanied by persistence of DNA damage as evidenced by persistence of gamma H2AX and Rad 51 foci after irradiation in the presence of the inhibitor. Reduced survival does not result from cell cycle redistribution during the PI-103 treatment intervals tested, although combining PI-103 treatment with radiation enhances the G(2)-M delay observed after irradiation. These results indicate that pharmacologic inhibitors with enhanced specificity for class I PI3K may be of benefit when combined with radiotherapy.
- Published
- 2008
- Full Text
- View/download PDF
36. Progress towards the use of HIV protease inhibitors in cancer therapy.
- Author
-
Bernhard EJ and Brunner TB
- Subjects
- Clinical Trials as Topic, Humans, Leukocytes, Mononuclear metabolism, Nelfinavir pharmacology, Radiation-Sensitizing Agents pharmacology, Biomarkers, Tumor metabolism, HIV Protease Inhibitors therapeutic use, HIV Protease Inhibitors toxicity, Leukocytes, Mononuclear virology, Neoplasms drug therapy, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Approaches to targeting the signaling pathways responsible for tumor survival are currently under intense investigation and there is ongoing rapid development of novel agents to target various signaling components that mediate tumor growth and survival. The development of agents as viable clinical interventions is, however, a relatively slow process when starting from novel compounds not previously tested in humans. An approach that could circumvent this delay in the development of molecular targeting agents for cancer therapy is to adopt drugs currently in clinical use for other diseases that have the desired effects on cell signaling and to test these for anti-cancer effects. This process is currently being followed with anti-retroviral drugs that inhibit AKT activation as candidates for the treatment of cancer in general, and is being pursued in particular for developing these agent as new means of sensitizing tumors to radiation.(1,2) Although such drugs have been approved for patient use, it is unclear whether or not these drugs can be safely combined with cytotoxic modalities such as radiation. It is this point that Plasteras et al. address in the current issue of Cancer Biology & Therapy, by examining the effects of combined treatment with radiation and HIV protease inhibitors.(3).
- Published
- 2008
- Full Text
- View/download PDF
37. Human pancreatic tumor cells are sensitized to ionizing radiation by knockdown of caveolin-1.
- Author
-
Cordes N, Frick S, Brunner TB, Pilarsky C, Grützmann R, Sipos B, Klöppel G, McKenna WG, and Bernhard EJ
- Subjects
- Blotting, Western, Cell Adhesion, Cell Cycle physiology, Cell Cycle radiation effects, Cell Proliferation radiation effects, Colony-Forming Units Assay, Fluorescent Antibody Technique, Focal Adhesion Kinase 1 antagonists & inhibitors, Focal Adhesion Kinase 1 genetics, Focal Adhesion Kinase 1 metabolism, Gene Expression Profiling, Humans, Integrin beta1 chemistry, Integrin beta1 genetics, Integrin beta1 metabolism, Mitogen-Activated Protein Kinases metabolism, Pancreatic Neoplasms metabolism, Paxillin metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins pp60(c-src) metabolism, RNA, Small Interfering pharmacology, Signal Transduction, X-Rays, Caveolin 1 metabolism, Pancreatic Neoplasms radiotherapy, Radiation Tolerance physiology
- Abstract
Caveolin-1 (Cav-1) is an integral transmembrane protein and a critical component in interactions of integrin receptors with cytoskeleton-associated and signaling molecules. Since integrin-mediated cell adhesion generates signals conferring radiation resistance, we examined the effects of small interfering RNA-mediated knockdown of Cav-1 alone or in combination with beta1-integrin or focal adhesion kinase (FAK) on radiation survival and proliferation of pancreatic carcinoma cell lines. Irradiation induced Cav-1 expression in PATU8902, MiaPaCa2 and Panc1 cell lines. The cell lines showed significant radiosensitization after knockdown of Cav-1, beta1-integrin or FAK and cholesterol depletion by beta-cyclodextrin relative to nonspecific controls. Under knockdown conditions, proliferation of non-irradiated and irradiated cells was significantly attenuated relative to controls. These findings correlated with changes in expression or phosphorylation of Akt, glycogen synthase kinase 3beta, Paxillin, Src, c-Jun N-terminal kinase and mitogen-activated protein kinase. Analysis of DNA microarray data revealed a Cav-1 overexpression in a subset of pancreatic ductal adenocarcinoma samples. The data presented show, for the first time, that disruption of interactions of Cav-1 with beta1-integrin or FAK affects radiation survival and proliferation of pancreatic carcinoma cells and suggest that Cav-1 is critical to these processes. These results indicate that strategies targeting Cav-1 may be useful as an approach to improve conventional therapies, including radiotherapy, for pancreatic cancer.
- Published
- 2007
- Full Text
- View/download PDF
38. Phosphatase and tensin homologue deficiency in glioblastoma confers resistance to radiation and temozolomide that is reversed by the protease inhibitor nelfinavir.
- Author
-
Jiang Z, Pore N, Cerniglia GJ, Mick R, Georgescu MM, Bernhard EJ, Hahn SM, Gupta AK, and Maity A
- Subjects
- Animals, Brain Neoplasms drug therapy, Brain Neoplasms enzymology, Brain Neoplasms radiotherapy, Cell Line, Tumor, Dacarbazine pharmacology, Drug Interactions, Drug Resistance, Neoplasm, Female, Glioblastoma drug therapy, Glioblastoma enzymology, Glioblastoma radiotherapy, Humans, Mice, Mice, Nude, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Radiation Tolerance, Temozolomide, Antineoplastic Agents, Alkylating pharmacology, Brain Neoplasms therapy, Dacarbazine analogs & derivatives, Glioblastoma therapy, Nelfinavir pharmacology, PTEN Phosphohydrolase deficiency, Protease Inhibitors pharmacology
- Abstract
Glioblastomas are malignant brain tumors that are very difficult to cure, even with aggressive therapy consisting of surgery, chemotherapy, and radiation. Glioblastomas frequently have loss of the phosphatase and tensin homologue (PTEN), leading to the activation of the phosphoinositide-3-kinase (PI3K)/Akt pathway. We examined whether PTEN deficiency leads to radioresistance and whether this can be reversed by nelfinavir, a protease inhibitor that decreases Akt signaling. Nelfinavir decreased Akt phosphorylation and enhanced radiosensitization in U251MG and U87MG glioblastoma cells, both of which are PTEN deficient. In the derivative line U251MG-PTEN, induction of wild-type PTEN with doxycycline decreased P-Akt expression and increased radiosensitivity to a similar extent as nelfinavir. Combining these two approaches had no greater effect on radiosensitivity than either alone. This epistasis-type analysis suggests that the nelfinavir acts along the Akt pathway to radiosensitize cells. However, nelfinavir neither decreased Akt phosphorylation in immortalized human astrocytes nor radiosensitized them. Radiosensitization was also assessed in vivo using a tumor regrowth delay assay in nude mice implanted with U87MG xenografts. The mean time to reach 1,000 mm(3) in the radiation + nelfinavir group was 71 days, as compared with 41, 34, or 45 days for control, nelfinavir alone, or radiation alone groups, respectively. A significant synergistic effect on tumor regrowth was detected between radiation and nelfinavir. (P = 0.01). Nelfinavir also increased the sensitivity of U251MG cells to temozolomide. These results support the clinical investigation of nelfinavir in combination with radiation and temozolomide in future clinical trials for patients with glioblastomas.
- Published
- 2007
- Full Text
- View/download PDF
39. Oncogenic K-Ras signals through epidermal growth factor receptor and wild-type H-Ras to promote radiation survival in pancreatic and colorectal carcinoma cells.
- Author
-
Cengel KA, Voong KR, Chandrasekaran S, Maggiorella L, Brunner TB, Stanbridge E, Kao GD, McKenna WG, and Bernhard EJ
- Subjects
- Cell Line, Tumor, Cell Survival genetics, Cell Survival radiation effects, Colorectal Neoplasms genetics, Colorectal Neoplasms radiotherapy, Gene Expression Regulation, Neoplastic radiation effects, Humans, Pancreatic Neoplasms genetics, Pancreatic Neoplasms radiotherapy, Colorectal Neoplasms pathology, ErbB Receptors physiology, ErbB Receptors radiation effects, Genes, ras physiology, Genes, ras radiation effects, Pancreatic Neoplasms pathology, Signal Transduction genetics, Signal Transduction radiation effects
- Abstract
Pancreatic and colorectal carcinomas frequently express oncogenic/mutant K-Ras that contributes to both tumorigenesis and clinically observed resistance to radiation treatment. We have previously shown that farnesyltransferase inhibitors (FTI) radiosensitize many pancreatic and colorectal cancer cell lines that express oncogenic K-ras at doses that inhibit the prenylation and activation of H-Ras but not K-Ras. In the present study, we have examined the mechanism of FTI-mediated radiosensitization in cell lines that express oncogenic K-Ras and found that wild-type H-Ras is a contributor to radiation survival in tumor cells that express oncogenic K-Ras. In these experiments, inhibiting the expression of oncogenic K-Ras, wild-type H-Ras, or epidermal growth factor receptor (EGFR) led to similar levels of radiosensitization as treatment with the FTI tipifarnib. Treatment with the EGFR inhibitor gefitinib led to similar levels of radiosensitization, and the combinations of tipifarnib or gefitinib plus inhibition of K-Ras, H-Ras, or EGFR expression did not provide additional radiosensitization compared with tipifarnib or gefitinib alone. Finally, supplementing culture medium with the EGFR ligand transforming growth factor alpha was able to reverse the radiosensitizing effect of inhibiting K-ras expression. Taken together, these findings suggest that EGFR-activated H-Ras signaling is initiated by oncogenic K-Ras to promote radiation survival in pancreatic and colorectal cancers.
- Published
- 2007
- Full Text
- View/download PDF
40. Pancreatic cancer inhibition by specific knockdown of K-ras mutant allele expression.
- Author
-
Bernhard EJ
- Subjects
- Alleles, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Humans, Mutation, Adenocarcinoma genetics, Genes, ras genetics, Pancreatic Neoplasms genetics
- Published
- 2007
- Full Text
- View/download PDF
41. Nelfinavir down-regulates hypoxia-inducible factor 1alpha and VEGF expression and increases tumor oxygenation: implications for radiotherapy.
- Author
-
Pore N, Gupta AK, Cerniglia GJ, Jiang Z, Bernhard EJ, Evans SM, Koch CJ, Hahn SM, and Maity A
- Subjects
- Animals, Carcinoma, Squamous Cell blood supply, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell radiotherapy, Cell Line, Tumor, Down-Regulation drug effects, Female, Head and Neck Neoplasms blood supply, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms radiotherapy, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Lung Neoplasms drug therapy, Lung Neoplasms radiotherapy, Mice, Mice, Inbred BALB C, Mice, Nude, Neovascularization, Pathologic drug therapy, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Radiation Tolerance drug effects, Random Allocation, Sp1 Transcription Factor metabolism, Vascular Endothelial Growth Factor A genetics, Xenograft Model Antitumor Assays, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms metabolism, Hypoxia-Inducible Factor 1, alpha Subunit biosynthesis, Lung Neoplasms metabolism, Nelfinavir pharmacology, Oxygen metabolism, Protease Inhibitors pharmacology, Vascular Endothelial Growth Factor A biosynthesis
- Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway can increase vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1alpha (HIF-1alpha) expression. We examined the effect of nelfinavir, an HIV protease inhibitor that inhibits Akt signaling, on VEGF and HIF-1alpha expression and on angiogenesis, tumor oxygenation, and radiosensitization. Nelfinavir decreases VEGF expression under normoxia via the transcription factor Sp1, which regulates the proximal core VEGF promoter. Nelfinavir decreased Sp1 phosphorylation and decreased Sp1 binding to a probe corresponding to the proximal VEGF promoter in a gel shift assay. Nelfinavir also decreased the hypoxic induction of HIF-1alpha, which also regulates the VEGF promoter, most likely by decreasing its translation. The effect of nelfinavir on VEGF expression had the functional consequence of decreasing angiogenesis in an in vivo Matrigel plug assay. To determine the effect this might have on tumor radiosensitization, we did tumor regrowth assays with xenografts in nude mice. The combination of nelfinavir and radiation increased time to regrowth compared with radiation alone whereas nelfinavir alone had little effect on tumor regrowth. This radiosensitizing effect was greater than suggested by in vitro clonogenic survival assays. One possible explanation for the discordance is that nelfinavir has an effect on tumor oxygenation. Therefore, we examined this with the hypoxia marker EF5 and found that nelfinavir leads to increased oxygenation within tumor xenografts. Our results suggest that nelfinavir decreases HIF-1alpha/VEGF expression and tumor hypoxia, which could play a role in its in vivo radiosensitizing effect. These data support the use of nelfinavir in combination with radiation in future clinical trials.
- Published
- 2006
- Full Text
- View/download PDF
42. Radiosensitizing effects of the prenyltransferase inhibitor AZD3409 against RAS mutated cell lines.
- Author
-
Cengel KA, Deutsch E, Stephens TC, Voong KR, Kao GD, and Bernhard EJ
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Mice, Mice, Nude, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Xenograft Model Antitumor Assays, ras Proteins biosynthesis, ras Proteins genetics, Dimethylallyltranstransferase antagonists & inhibitors, Genes, ras, Mutation, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms radiotherapy, Radiation-Sensitizing Agents pharmacology
- Abstract
Mutations at the H-, N- and K-ras loci are among the most frequent genetic alterations in human cancers. In this study, we have investigated the effect of AZD3409, a novel, peptidomimetic prenyltransferase inhibitor (PTI), on the radiosensitivity of cells with mutated ras alleles. AZD3409, developed by AstraZeneca, inhibits both farnesyl- and geranylgeranyl transferase in cell free systems. AZD3409 inhibits the growth of a variety of human cancer cell lines, including cells that express mutant alleles of either K- or H- ras and was well tolerated when administered orally to healthy volunteers in a phase I clinical trial. We have previously shown that PTI can radiosensitize human and rodent cancer cell lines that express activated RAS. Here we assessed the ability of AZD3409 to radiosensitize human cancer cell lines in vivo and in vitro and the activation state of RAS proteins in treated cells. Once daily oral administration of AZD3409 to nude mice bearing PSN-1 and MiaPaCa-2 human pancreatic cancer xenografts expressing mutant K-ras was well tolerated and resulted in a supra-additive reduction in clonogenic cell survival after irradiation. Similarly, AZD3409 reduced clonogenic survival in cells that express either mutant K- or H- ras in vitro. We next examined the effect of AZD3409 on the processing and activation of K- and H-RAS. AZD3409-mediated radiosensitization, both in vivo and in vitro, correlates with a decrease in H-RAS processing without detectable effect on K-RAS processing. RAS activation assays show that the decreased H-RAS processing is accompanied by decreased H-RAS activation in cell lines with mutations in either K- or H-ras. However, no decrease in K-RAS activation was detected. Thus, radiosensitization of human cancer cells that express mutated K-RAS occurred under conditions where AZD3409 inihibits the activation of farneyslated H-RAS, but did not inhibit K-RAS activation.
- Published
- 2006
- Full Text
- View/download PDF
43. Pancreatic cancer cell radiation survival and prenyltransferase inhibition: the role of K-Ras.
- Author
-
Brunner TB, Cengel KA, Hahn SM, Wu J, Fraker DL, McKenna WG, and Bernhard EJ
- Subjects
- Alkyl and Aryl Transferases antagonists & inhibitors, Carcinoma, Pancreatic Ductal enzymology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal radiotherapy, Cell Line, Tumor, Cell Survival physiology, Cell Survival radiation effects, Farnesyltranstransferase antagonists & inhibitors, Humans, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Radiation Tolerance physiology, Dimethylallyltranstransferase antagonists & inhibitors, Genes, ras physiology, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms radiotherapy
- Abstract
Activating K-ras mutations are found in approximately 90% of pancreatic carcinomas and may contribute to the poor prognosis of these tumors. Because radiotherapy is frequently used in pancreatic cancer treatment, we assessed the contribution of oncogenic K-ras signaling to pancreatic cancer radiosensitivity. Seven human pancreatic carcinoma lines with activated K-ras and two cell lines with wild-type ras were used to examine clonogenic cell survival after Ras inhibition. Ras inhibition was accomplished by small interfering RNA (siRNA) knockdown of K-ras expression and by blocking Ras processing using a panel of prenyltransferase inhibitors of differing specificity for the two prenyltransferases that modify K-Ras. K-ras knockdown by siRNA or inhibition of prenyltransferase activity resulted in radiation sensitization in vitro and in vivo in tumors with oncogenic K-ras mutations. Inhibition of farnesyltransferase alone was sufficient to radiosensitize most K-ras mutant tumors, although K-Ras prenylation was not blocked. These results show that inhibition of activated K-Ras can promote radiation killing of pancreatic carcinoma in a superadditive manner. The finding that farnesyltransferase inhibition alone radiosensitizes tumors with K-ras mutations implies that a farnesyltransferase inhibitor-sensitive protein other than K-Ras may contribute to survival in the context of mutant K-ras. Farnesyltransferase inhibitors could therefore be of use as sensitizers for pancreatic carcinoma radiotherapy.
- Published
- 2005
- Full Text
- View/download PDF
44. Selective inhibition of Ras, phosphoinositide 3 kinase, and Akt isoforms increases the radiosensitivity of human carcinoma cell lines.
- Author
-
Kim IA, Bae SS, Fernandes A, Wu J, Muschel RJ, McKenna WG, Birnbaum MJ, and Bernhard EJ
- Subjects
- Animals, Carcinoma genetics, Cell Survival radiation effects, Chromones pharmacology, Colonic Neoplasms enzymology, Colonic Neoplasms genetics, Colonic Neoplasms radiotherapy, Enzyme Inhibitors pharmacology, Flavonoids pharmacology, Humans, Isoenzymes antagonists & inhibitors, MAP Kinase Kinase Kinases metabolism, Mice, Morpholines pharmacology, Protein Serine-Threonine Kinases biosynthesis, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins biosynthesis, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-akt, RNA, Small Interfering genetics, Radiation Tolerance drug effects, Transfection, Urinary Bladder Neoplasms enzymology, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms radiotherapy, ras Proteins biosynthesis, ras Proteins genetics, Carcinoma enzymology, Carcinoma radiotherapy, Phosphoinositide-3 Kinase Inhibitors, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors, Radiation Tolerance physiology, ras Proteins antagonists & inhibitors
- Abstract
Ras activation promotes the survival of tumor cells after DNA damage. To reverse this survival advantage, Ras signaling has been targeted for inhibition. Other contributors to Ras-mediated DNA damage survival have been identified using pharmacologic inhibition of signaling, but this approach is limited by the specificity of the inhibitors used and their toxicity. To better define components of Ras signaling that could be inhibited in a clinical setting, RNA interference was used to selectively block expression of specific isoforms of Ras, phosphoinositide 3 (PI3) kinase, and Akt. Inhibition of oncogenic Ras expression decreased both phospho-Akt and phospho-p42/44 mitogen-activated protein (MAP) kinase levels and reduced clonogenic survival. Because pharmacologic inhibition of PI3 kinases and Akt radiosensitized cell lines with active Ras signaling, whereas inhibition of the MAP/extracellular signal-regulated kinase (ERK) kinase/ERK pathway did not, we examined the contribution of PI3 kinases and Akts to radiation survival. Selective inhibition the PI3 kinase P110alpha + p85beta isoforms reduced Akt phosphorylation and radiation survival. Similarly, inhibition of Akt-1 reduced tumor cell radiation survival. Inhibition of Akt-2 or Akt-3 had less effect. Retroviral transduction and overexpression of mouse Akt-1 was shown to rescue cells from inhibition of endogenous human Akt-1 expression. This study shows that Ras signaling to the PI3 kinase-Akt pathway is an important contributor to survival, whether Ras activation results from mutation of ras or overexpression of epidermal growth factor receptor. This study further shows that selective inhibition of the PI3 kinase P110alpha + p85beta isoforms or Akt-1 could be a viable approach to sensitizing many tumor cells to cytotoxic therapies.
- Published
- 2005
- Full Text
- View/download PDF
45. Effects of hyperbaric oxygen exposure on experimental head and neck tumor growth, oxygenation, and vasculature.
- Author
-
Shi Y, Lee CS, Wu J, Koch CJ, Thom SR, Maity A, and Bernhard EJ
- Subjects
- Animals, Carcinoma, Squamous Cell pathology, Cell Hypoxia, Dose-Response Relationship, Radiation, Head and Neck Neoplasms pathology, Immunohistochemistry, Male, Mice, Mice, Nude, Neoplasms, Experimental, Neovascularization, Pathologic, Transplantation, Heterologous, Vascular Endothelial Growth Factor A metabolism, Carcinoma, Squamous Cell blood supply, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms blood supply, Head and Neck Neoplasms metabolism, Hyperbaric Oxygenation
- Abstract
Background: Hyperbaric oxygen (HBO2) is used to promote healing in irradiated tissues, but concern persists about the possibility that it may promote residual tumor growth., Methods: The tumor growth of SQ20B and Detroit 562 head and neck squamous cell carcinoma xenografts were studied after single-dose irradiation and 5x/week HBO2 treatment at 2.4 atm absolute for 90 minutes. The effect of HBO2 treatment on tumor hypoxia and vasculature was also examined by immunohistochemical analysis., Results: HBO2 treatment increased tumor oxygenation during the treatment interval but did not promote the growth of either irradiated or unirradiated tumors. No increase in tumor vascular endothelial growth factor expression or vascularization was detected., Conclusions: This study found no evidence for persistent changes in tumor microenvironment or tumor growth promotion caused by hyperbaric oxygen exposure., (2005 Wiley Periodicals, Inc.)
- Published
- 2005
- Full Text
- View/download PDF
46. Farnesyltransferase inhibitor effects on prostate tumor micro-environment and radiation survival.
- Author
-
Shi Y, Wu J, Mick R, Cerniglia GJ, Cohen-Jonathan E, Rhim JS, Koch CJ, and Bernhard EJ
- Subjects
- Animals, Apoptosis drug effects, Apoptosis radiation effects, Cell Line, Tumor, Farnesyltranstransferase, Humans, Male, Matrix Metalloproteinase 9 metabolism, Mice, Mice, Nude, Mitogen-Activated Protein Kinases metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Radiation Tolerance drug effects, Signal Transduction, Xenograft Model Antitumor Assays, Alkyl and Aryl Transferases antagonists & inhibitors, Enzyme Inhibitors pharmacology, Genes, ras drug effects, Methionine analogs & derivatives, Methionine pharmacology, Prostatic Neoplasms radiotherapy, Radiation-Sensitizing Agents pharmacology
- Abstract
Background: Ras activation by mutation, overexpression, or receptor signaling can increase tumor cell survival after irradiation., Methods: We examined whether inhibiting Ras activity with farnesyltransferase inhibitors (FTI) altered the radiosensitivity and tumor micro-environment in prostate tumors., Results: Treatment with FTIs L-744,832 or FTI-277 reduced clonogenic survival of prostate tumor cells expressing oncogenic H-ras after irradiation. PI3-kinase/Akt and MAPK signaling pathways were downregulated by FTIs in these cells. FTI treatment reduced tumor hypoxia and also reduced MMP-9 expression in tumors with activated mutant H-ras. FTI treatment did not, however, increase apoptosis in irradiated intestine, demonstrating that acute radiation injury of this normal tissue was not enhanced by FTIs., Conclusions: FTIs can enhance the killing of prostate tumors with activated H-Ras. Together with the absence of increased acute toxicity to normal bowel, these results imply that FTI treatment should be further studied as a possible adjuvant to radiotherapy in the treatment of abdominal cancers with activated Ras signaling., (Copyright 2004 Wiley-Liss, Inc.)
- Published
- 2005
- Full Text
- View/download PDF
47. Bnip3L is induced by p53 under hypoxia, and its knockdown promotes tumor growth.
- Author
-
Fei P, Wang W, Kim SH, Wang S, Burns TF, Sax JK, Buzzai M, Dicker DT, McKenna WG, Bernhard EJ, and El-Deiry WS
- Subjects
- Animals, Apoptosis drug effects, Apoptosis genetics, Blotting, Western, CREB-Binding Protein, Caspase 3, Caspases metabolism, Cell Hypoxia, Cell Line, Tumor, Cell Proliferation, Chromatin Immunoprecipitation, Cisplatin pharmacology, Doxycycline pharmacology, Fluorouracil pharmacology, Gene Expression Regulation, Neoplastic drug effects, Genes, Reporter genetics, Glucose Transporter Type 1, Humans, Immunohistochemistry, In Situ Hybridization, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Mice, Nude, Monosaccharide Transport Proteins metabolism, Neoplasms genetics, Neoplasms metabolism, Nuclear Proteins metabolism, Oligonucleotide Array Sequence Analysis, Protein Binding drug effects, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, RNA Interference, RNA, Small Interfering genetics, Repressor Proteins genetics, Trans-Activators metabolism, Transfection, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Xenograft Model Antitumor Assays, Membrane Proteins physiology, Neoplasms pathology, Proto-Oncogene Proteins physiology, Tumor Suppressor Protein p53 physiology, Tumor Suppressor Proteins physiology
- Abstract
p53-dependent apoptosis is a major determinant of its tumor suppressor activity and can be triggered by hypoxia. No p53 target is known to be induced by p53 or to mediate p53-dependent apoptosis during hypoxia. We report that p53 can directly upregulate expression of Bnip3L, a cell death inducer. During hypoxia, Bnip3L is highly induced in wild-type p53-expressing cells, in part due to increased recruitment of p53 and CBP to Bnip3L. Apoptosis is reduced in hypoxia-exposed cells with functional p53 following Bnip3L knockdown. In vivo, Bnip3L knockdown promotes tumorigenicity of wild-type versus mutant p53-expressing tumors. Thus, Bnip3L, capable of attenuating tumorigenicity, mediates p53-dependent apoptosis under hypoxia, which provides a novel understanding of p53 in tumor suppression.
- Published
- 2004
- Full Text
- View/download PDF
48. Radiation sensitization by inhibition of activated Ras.
- Author
-
Brunner TB, Hahn SM, McKenna WG, and Bernhard EJ
- Subjects
- Animals, Enzyme Activation radiation effects, Gene Expression Regulation, Neoplastic radiation effects, Humans, Models, Biological, ras Proteins antagonists & inhibitors, Neoplasms metabolism, Neoplasms radiotherapy, Radiation Tolerance, Signal Transduction radiation effects, ras Proteins metabolism, ras Proteins radiation effects
- Abstract
Background and Purpose: Ras has been identified as a significant contributor to radiation resistance. This article reviews preclinical and phase I clinical studies that reported on combining inhibition of activated Ras and downstream effectors of Ras with radiotherapy., Material and Methods: Transfection studies and RNA interference were used to check the role of the Ras isoforms for intrinsic radiation sensibility. Western blotting was used to control for prenylation inhibition of the respective Ras isoforms and for changes in activity of downstream proteins. Clonogenic assays with human and rodent tumor cell lines served for testing radiosensitivity. In vivo, farnesyltransferase inhibitors (FTIs) and irradiation were used to treat xenograft tumors. Ex vivo plating efficiency measurements, regrowth of tumors, and EF5 staining for detection of hypoxia were endpoints in these studies. Simultaneous treatment with L-778,123 and irradiation was performed in non-small cell lung cancer, head and neck cancer, and pancreatic cancer patients., Results: Radiation sensitization was achieved in vitro and in vivo blocking the prenylation of Ras proteins in cell lines with Ras activated by mutations or receptor signaling. Among the many Ras downstream pathways the phosphoinositide 3 (PI3) kinase-Akt pathway was identified as a contributor to Ras-mediated radiation resistance. Furthermore, increased oxygenation was observed in xenograft tumors after FTI treatment. Combined treatment in a phase I study was safe and effective., Conclusion: The rational combination of FTIs with radiotherapy may improve the clinical results of patients with tumors who bear mutant or receptor-signaling activated Ras.
- Published
- 2004
- Full Text
- View/download PDF
49. Farnesyltransferase inhibition: who are the Aktors?
- Author
-
Bernhard EJ
- Subjects
- Down-Regulation, Farnesyltranstransferase, Humans, Lung Neoplasms metabolism, Lung Neoplasms pathology, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt, Signal Transduction, Alkyl and Aryl Transferases antagonists & inhibitors, Apoptosis drug effects, Cell Cycle drug effects, Enzyme Inhibitors pharmacology, Piperidines pharmacology, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Pyridines pharmacology
- Published
- 2004
- Full Text
- View/download PDF
50. A phase I trial of the dual farnesyltransferase and geranylgeranyltransferase inhibitor L-778,123 and radiotherapy for locally advanced pancreatic cancer.
- Author
-
Martin NE, Brunner TB, Kiel KD, DeLaney TF, Regine WF, Mohiuddin M, Rosato EF, Haller DG, Stevenson JP, Smith D, Pramanik B, Tepper J, Tanaka WK, Morrison B, Deutsch P, Gupta AK, Muschel RJ, McKenna WG, Bernhard EJ, and Hahn SM
- Subjects
- Adult, Aged, Cell Line, Tumor, Combined Modality Therapy, Dose-Response Relationship, Drug, Farnesyltranstransferase, Female, Humans, Imidazoles administration & dosage, Infusions, Intravenous, Male, Middle Aged, Survival Analysis, Time Factors, Alkyl and Aryl Transferases antagonists & inhibitors, Imidazoles toxicity, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms radiotherapy
- Abstract
Purpose: Preclinical and clinical studies have demonstrated that inhibition of prenylation can radiosensitize cell lines with activation of Ras and produce clinical response in patients with cancer. The aim of this study was to determine the maximally tolerated dose of the dual farnesyltransferase and geranylgeranyltransferase I inhibitor L-778,123 in combination with radiotherapy for patients with locally advanced pancreatic cancer., Experimental Design: L-778,123 was given by continuous intravenous infusion with concomitant radiotherapy to 59.4 Gy in standard fractions. Two L-778,123 dose levels were tested: 280 mg/m2/day over weeks 1, 2, 4, and 5 for dose level 1; and 560 mg/m2/day over weeks 1, 2, 4, 5, and 7 for dose level 2., Results: There were no dose-limiting toxicities observed in the eight patients treated on dose level 1. Two of the four patients on dose level 2 experienced dose-limiting toxicities consisting of grade 3 diarrhea in one case and grade 3 gastrointestinal hemorrhage associated with grade 3 thrombocytopenia and neutropenia in the other case. Other common toxicities were mild neutropenia, dehydration, hyperglycemia, and nausea/vomiting. One patient on dose level 1 showed a partial response of 6 months in duration. Both reversible inhibition of HDJ2 farnesylation and radiosensitization of a study patient-derived cell line were demonstrated in the presence of L-778,123. K-RAS mutations were found in three of the four patients evaluated., Conclusions: The combination of L-778,123 and radiotherapy at dose level 1 showed acceptable toxicity in patients with locally advanced pancreatic cancer. Radiosensitization of a patient-derived pancreatic cancer cell line was observed.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.