107 results on '"Frank, Pajonk"'
Search Results
2. Tumor necrosis factor receptor signaling modulates carcinogenesis in a mouse model of breast cancer
- Author
-
Ling He, Kruttika Bhat, Sara Duhacheck-Muggy, Angeliki Ioannidis, Le Zhang, Nhan T. Nguyen, Neda A. Moatamed, and Frank Pajonk
- Subjects
Breast cancer ,Mammary epithelial stem cells ,Tumor necrosis factor alpha receptor ,NF-κB signaling ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Pro-inflammatory conditions have long been associated with mammary carcinogenesis and breast cancer progression. The underlying mechanisms are incompletely understood but signaling of pro-inflammatory cytokine TNFα through its receptors TNFR1 and TNFR2 is a major mediator of inflammation in both obesity and in the response of tissues to radiation, 2 known risk factors for the development of breast cancer. Here, we demonstrated the loss of one TNFR2 allele led to ductal hyperplasia in the mammary gland with increased numbers of mammary epithelial stem cell and terminal end buds. Furthermore, loss of one TNFR2 allele increased the incidence of breast cancer in MMTV-Wnt1 mice and resulted in tumors with a more aggressive phenotype and metastatic potential. The underlying mechanisms include a preferential activation of canonical NF-κB signaling pathway and autocrine production of TNFα. Analysis of the TCGA dataset indicated inferior overall survival for patients with down-regulated TNFR2 expression. These findings unravel the imbalances in TNFR signaling promote the development and progression of breast cancer, indicating that selective agonists of TNFR2 could potentially modulate the risk for breast cancer in high-risk populations.
- Published
- 2021
- Full Text
- View/download PDF
3. Radiation mitigation of the intestinal acute radiation injury in mice by 1‐[(4‐nitrophenyl)sulfonyl]‐4‐phenylpiperazine
- Author
-
Sara Duhachek‐Muggy, Kruttika Bhat, Paul Medina, Fei Cheng, Ling He, Claudia Alli, Mohammad Saki, Sree Deepthi Muthukrishnan, Gregoire Ruffenach, Mansoureh Eghbali, Erina Vlashi, and Frank Pajonk
- Subjects
acute radiation syndrome ,developmental signaling ,intestinal stem cells ,radiation ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract The objective of the study was to identify the mechanism of action for a radiation mitigator of the gastrointestinal (GI) acute radiation syndrome (ARS), identified in an unbiased high‐throughput screen. We used mice irradiated with a lethal dose of radiation and treated with daily injections of the radiation mitigator 1‐[(4‐nitrophenyl)sulfonyl]‐4‐phenylpiperazine to study its effects on key pathways involved in intestinal stem cell (ISC) maintenance. RNASeq, quantitative reverse transcriptase‐polymerase chain reaction, and immunohistochemistry were performed to identify pathways engaged after drug treatment. Target validation was performed with competition assays, reporter cells, and in silico docking. 1‐[(4‐Nitrophenyl)sulfonyl]‐4‐phenylpiperazine activates Hedgehog signaling by binding to the transmembrane domain of Smoothened, thereby expanding the ISC pool, increasing the number of regenerating crypts and preventing the GI‐ARS. We conclude that Smoothened is a target for radiation mitigation in the small intestine that could be explored for use in radiation accidents as well as to mitigate normal tissue toxicity during and after radiotherapy of the abdomen.
- Published
- 2020
- Full Text
- View/download PDF
4. Serum erythropoietin levels, breast cancer and breast cancer-initiating cells
- Author
-
Kruttika Bhat, Kiri Sandler, Sara Duhachek-Muggy, Claudia Alli, Fei Cheng, Neda A. Moatamed, Clara E. Magyar, Lin Du, Gang Li, Susan McCloskey, Erina Vlashi, and Frank Pajonk
- Subjects
Breast cancer-initiating cells ,Erythropoietin ,Radiation therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Cancer is frequently associated with tumor-related anemia, and many chemotherapeutic agents impair hematopoiesis, leading to impaired quality of life for affected patients. The use of erythropoiesis-stimulating agents has come under scrutiny after prospective clinical trials using recombinant erythropoietin to correct anemia reported increased incidence of thromboembolic events and cancer-related deaths. Furthermore, previous preclinical reports indicated expansion of the pool of breast cancer-initiating cells when erythropoietin was combined with ionizing radiation. Methods Using four established breast cancer cell lines, we test the effects of recombinant human erythropoietin and the number of breast cancer-initiating cells in vitro and in vivo and study if recombinant human erythropoietin promotes the phenotype conversion of non-tumorigenic breast cancer cells into breast cancer-initiating cells. In a prospective study, we evaluate whether elevated endogenous serum erythropoietin levels correlate with increased numbers of tumor-initiating cells in a cohort of breast cancer patients who were scheduled to undergo radiation treatment. Results Our results indicate that recombinant erythropoietin increased the number of tumor-initiating cells in established breast cancer lines in vitro. Irradiation of breast cancer xenografts caused a phenotype conversion of non-stem breast cancer cells into induced breast cancer-initiating cells. This effect coincided with re-expression of the pluripotency factors c-Myc, Sox2, and Oct4 and was enhanced by recombinant erythropoietin. Hemoglobin levels were inversely correlated with serum erythropoietin levels, and the latter were correlated with disease stage. However, tumor sections revealed a negative correlation between serum erythropoietin levels and the number of ALDH1A3-positive cells, a marker for breast cancer-initiating cells. Conclusions We conclude that physiologically slow-rising serum erythropoietin levels in response to tumor-related or chemotherapy-induced anemia, as opposed to large doses of recombinant erythropoietin, do not increase the pool of breast cancer-initiating cells.
- Published
- 2019
- Full Text
- View/download PDF
5. Metabolic characterization of isocitrate dehydrogenase (IDH) mutant and IDH wildtype gliomaspheres uncovers cell type-specific vulnerabilities
- Author
-
Matthew Garrett, Jantzen Sperry, Daniel Braas, Weihong Yan, Thuc M. Le, Jack Mottahedeh, Kirsten Ludwig, Ascia Eskin, Yue Qin, Rachelle Levy, Joshua J. Breunig, Frank Pajonk, Thomas G. Graeber, Caius G. Radu, Heather Christofk, Robert M. Prins, Albert Lai, Linda M. Liau, Giovanni Coppola, and Harley I. Kornblum
- Subjects
2-hydroxyglutarate ,Metabolism ,Nucleotide ,Radiation ,Glioma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background There is considerable interest in defining the metabolic abnormalities of IDH mutant tumors to exploit for therapy. While most studies have attempted to discern function by using cell lines transduced with exogenous IDH mutant enzyme, in this study, we perform unbiased metabolomics to discover metabolic differences between a cohort of patient-derived IDH1 mutant and IDH wildtype gliomaspheres. Methods Using both our own microarray and the TCGA datasets, we performed KEGG analysis to define pathways differentially enriched in IDH1 mutant and IDH wildtype cells and tumors. Liquid chromatography coupled to mass spectrometry analysis with labeled glucose and deoxycytidine tracers was used to determine differences in overall cellular metabolism and nucleotide synthesis. Radiation-induced DNA damage and repair capacity was assessed using a comet assay. Differences between endogenous IDH1 mutant metabolism and that of IDH wildtype cells transduced with the IDH1 (R132H) mutation were also investigated. Results Our KEGG analysis revealed that IDH wildtype cells were enriched for pathways involved in de novo nucleotide synthesis, while IDH1 mutant cells were enriched for pathways involved in DNA repair. LC-MS analysis with fully labeled 13C-glucose revealed distinct labeling patterns between IDH1 mutant and wildtype cells. Additional LC-MS tracing experiments confirmed increased de novo nucleotide synthesis in IDH wildtype cells relative to IDH1 mutant cells. Endogenous IDH1 mutant cultures incurred less DNA damage than IDH wildtype cultures and sustained better overall growth following X-ray radiation. Overexpression of mutant IDH1 in a wildtype line did not reproduce the range of metabolic differences observed in lines expressing endogenous mutations, but resulted in depletion of glutamine and TCA cycle intermediates, an increase in DNA damage following radiation, and a rise in intracellular ROS. Conclusions These results demonstrate that IDH1 mutant and IDH wildtype cells are easily distinguishable metabolically by analyzing expression profiles and glucose consumption. Our results also highlight important differences in nucleotide synthesis utilization and DNA repair capacity that could be exploited for therapy. Altogether, this study demonstrates that IDH1 mutant gliomas are a distinct subclass of glioma with a less malignant, but also therapy-resistant, metabolic profile that will likely require distinct modes of therapy.
- Published
- 2018
- Full Text
- View/download PDF
6. Effects of Dopamine Receptor Antagonists and Radiation on Mouse Neural Stem/Progenitor Cells
- Author
-
Ling He, Kruttika Bhat, Angeliki Ioannidis, and Frank Pajonk
- Subjects
Article - Abstract
BackgroundDopamine receptor antagonists are psychotropic drugs that have been originally developed against psychiatric disorders. We recently identified dopamine receptor antagonists as potential anti-cancer agents and some have entered clinical trials against glioblastoma. Radiotherapy is known to cause cognitive impairment in patients receiving cranial irradiation through the elimination of neural stem/progenitor cells and subsequent loss of neurogenesis.MethodsUsing transgenic mice that report the presence of neural stem/progenitor cells through Nestin promoter-driven expression of enhanced green fluorescent protein, the effects of dopamine receptor antagonists alone or in combination with radiation on murine neural stem/progenitor cells were assessed in sphere-formation assays, flow cytometry and immunofluorescencein vitroandin vivo.ResultsWe report that several dopamine receptor antagonists show sex-dependent effects on neural stem/progenitor cells bothin vitroandin vivo. Hydroxyzine, trifluoperazine, amisulpride, nemonapride or quetiapine alone or in combination with radiation significantly increased the number of neural stem/progenitor cells in female neurospheres but not in male mice. Dopamine receptor antagonists either protected neural stem/progenitor cells from radiation or expanded the stem cell pool, thus indicating that this combination therapy against glioblastoma will not increase radiation-induced cognitive decline through increasing elimination of neural stem/progenitor cells and subsequent loss of neurogenesis.ConclusionsWe conclude that a therapeutic window for dopamine receptor antagonists in combination with radiation potentially exist, making it a novel combination therapy against glioblastoma. Normal tissue toxicity of this combination potentially differs depending on age and sex and should be taken into consideration when designing clinical trials.Key Points- Neural stem/progenitor cells show sex-dependent sensitivity to dopamine receptor antagonists- Dopamine receptor antagonists active against GBM increase Neural stem/progenitor cells countsImportance of the StudyCombination therapy of dopamine receptor antagonists with radiation have entered clinical trials against glioblastoma but the normal tissue toxicity of this combination has not been fully explored yet. Here we present evidence that some dopamine receptor antagonists show sex-dependent effects on neural stem/progenitor cells either by protecting neural stem/progenitor cells from radiation or inducing an expansion of the stem cell pool, suggesting that this combination therapy against glioblastoma will not increase radiation-induced cognitive decline through increasing elimination of neural stem/progenitor cells and subsequent loss of neurogenesis. Normal tissue toxicity of this combination potentially differs depending on age and sex and should be further explored in clinical trials.
- Published
- 2023
7. NADPH oxidase promotes glioblastoma radiation resistance in a PTEN-dependent manner
- Author
-
Kirsten Ludwig, Janel E. Le Belle, Sree Deepthi Muthukrishnan, Jantzen Sperry, Michael Condro, Erina Vlashi, Frank Pajonk, and Harley I. Kornblum
- Abstract
AimsThe goal of this study was to determine whether NADPH oxidase (NOX)-produced reactive oxygen species enhances brain tumor growth of glioblastoma (GBM) under hypoxic conditions and during radiation treatment.ResultsExogenous ROS promoted brain tumor growth in gliomasphere cultures that expressed functional PTEN, but not in tumors that were PTEN deficient. Hypoxia induced the production of endogenous cytoplasmic ROS and tumor cell growth via activation of NOX. NOX activation resulted in oxidation of PTEN and downstream Akt activation. Radiation also promoted ROS production via NOX which, in turn, resulted in cellular protection that could be abrogated by knockdown of the key NOX component, p22. Knockdown of p22 also inhibited tumor growth and enhanced the efficacy of radiation in PTEN-expressing GBM cells.InnovationWhile other studies have implicated NOX function in GBM models, these studies demonstrate NOX activation and function under physiological hypoxia and following radiation in GBM, two conditions that are seen in patients. NOX plays an important role in a PTEN-expressing GBM model system, but not in PTEN-non-functional systems and provide a potential, patient-specific therapeutic opportunity.ConclusionsThis study provides a strong basis for pursuing NOX inhibition in PTEN-expressing GBM cells as a possible adjunct to radiation therapy.
- Published
- 2022
8. Tumor necrosis factor receptor signaling modulates carcinogenesis in a mouse model of breast cancer
- Author
-
Kruttika Bhat, Le Zhang, Ling He, Neda A. Moatamed, Sara Duhacheck-Muggy, Angeliki Ioannidis, Nhan T. Nguyen, and Frank Pajonk
- Subjects
Proteomics ,0301 basic medicine ,Cancer Research ,c-Myc, myelocytoma ,Organogenesis ,medicine.medical_treatment ,GAPDH, glyceraldehyde 3-phosphate dehydrogenase ,Mammary gland ,IL-12, interleukin 12 ,medicine.disease_cause ,Receptors, Tumor Necrosis Factor ,Mice ,Breast cancer ,0302 clinical medicine ,Oct4, octamer-binding transcription factor 4 ,Receptor ,Mammary epithelial stem cells ,Tumor necrosis factor alpha receptor ,Klf4, Krüppel-like factor 4 ,Mice, Knockout ,NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells ,NF-kappa B ,ELISA, enzyme-linked immunosorbent assay ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunohistochemistry ,TNFR, tumor necrosis factor receptor ,3. Good health ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Cytokine ,030220 oncology & carcinogenesis ,NF-κB signaling ,iNOS, inducible nitric oxide synthase ,Cytokines ,Female ,Tumor necrosis factor alpha ,Disease Susceptibility ,Signal transduction ,Signal Transduction ,Original article ,IKK, the inhibitor of nuclear factor-κB (IκB) kinase ,TNFα, tumor necrosis factor alpha ,Breast Neoplasms ,lcsh:RC254-282 ,IL-1α, interleukin 1 alpha ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptors, Tumor Necrosis Factor, Type II ,Autocrine signalling ,Alleles ,IFN-γ, interferon gamma ,business.industry ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,PMSF, phenylmethylsulfonyl fluoride ,DTT, dithiothreitol ,CXCL1, C-X-C Motif chemokine ligand 1 ,Cancer research ,business ,Carcinogenesis ,Sox2, SRY (sex determining region Y)-box 2 ,Biomarkers - Abstract
Pro-inflammatory conditions have long been associated with mammary carcinogenesis and breast cancer progression. The underlying mechanisms are incompletely understood but signaling of pro-inflammatory cytokine TNFα through its receptors TNFR1 and TNFR2 is a major mediator of inflammation in both obesity and in the response of tissues to radiation, 2 known risk factors for the development of breast cancer. Here, we demonstrated the loss of one TNFR2 allele led to ductal hyperplasia in the mammary gland with increased numbers of mammary epithelial stem cell and terminal end buds. Furthermore, loss of one TNFR2 allele increased the incidence of breast cancer in MMTV-Wnt1 mice and resulted in tumors with a more aggressive phenotype and metastatic potential. The underlying mechanisms include a preferential activation of canonical NF-κB signaling pathway and autocrine production of TNFα. Analysis of the TCGA dataset indicated inferior overall survival for patients with down-regulated TNFR2 expression. These findings unravel the imbalances in TNFR signaling promote the development and progression of breast cancer, indicating that selective agonists of TNFR2 could potentially modulate the risk for breast cancer in high-risk populations.
- Published
- 2021
9. 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine treatment after brain irradiation preserves cognitive function in mice
- Author
-
Kruttika Bhat, Ling He, Harley I. Kornblum, Nhan T. Nguyen, Paul Medina, Mohammad Saki, Angeliki Ioannidis, David Sung, Linda M. Liau, Le Zhang, Clara E. Magyar, Sirajbir S. Sodhi, and Frank Pajonk
- Subjects
Cancer Research ,medicine.medical_treatment ,Inbred C57BL ,Piperazines ,Mice ,Cognition ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Medicine ,Aetiology ,Cognitive decline ,neural stem cells ,Cancer ,0303 health sciences ,medicine.diagnostic_test ,Microglia ,Brain ,Neural stem cell ,3. Good health ,Cytokine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Basic and Translational Investigations ,Toxicity ,Neurological ,Female ,Stem Cell Research - Nonembryonic - Non-Human ,Stem cell ,Oncology and Carcinogenesis ,Brain tumor ,Flow cytometry ,03 medical and health sciences ,Rare Diseases ,In vivo ,Neurosphere ,Behavioral and Social Science ,Animals ,Hedgehog Proteins ,Oncology & Carcinogenesis ,Progenitor cell ,cognitive function ,Neuroinflammation ,030304 developmental biology ,radiation mitigation ,business.industry ,Neurosciences ,Stem Cell Research ,medicine.disease ,Brain Disorders ,Mice, Inbred C57BL ,radiation ,Brain Cancer ,Cancer research ,Neurology (clinical) ,Cranial Irradiation ,business - Abstract
BackgroundNormal tissue toxicity to the CNS is an inevitable consequence of a successful radiotherapy of brain tumors or cancer metastases to the CNS. Cranial irradiation commonly leads to neurocognitive deficits that manifest months or years after treatment. Mechanistically, radiation-induced loss of neural stem/progenitor cells, neuro-inflammation and de-myelinization are contributing factors that lead to progressive cognitive decline.MethodsThe effects of Compound #5 on irradiated murine neurospheres, microglia cells and patients-derived gliomaspheres were assessed in sphere-formation assays, flow cytometry and IL-6 ELISAs, Activation of the Hedgehog pathway was studied by qRT-PCR. Thein vivoeffects of Compound #5 were analyzed using flow cytometry, sphere-formation assays, immune-histochemistry, behavioral testing and an intracranial mouse model of glioblastoma.ResultsWe report that 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine (Compound #5) mitigates radiation-induced normal tissue toxicity in the brains of mice. Compound #5 treatment significantly increased the number of neural stem/progenitor cells after brain irradiation in female animals, inhibited radiation-induced microglia activation and expression of the pro-inflammatory cytokine interleukin-6. Behavioral testing revealed that treatment with Compound #5 after radiotherapy successfully mitigates radiation-induced decline in motor, sensory and memory function of the brain. In mouse models of glioblastoma, Compound #5 showed no toxicity and did not interfere with the growth-delaying effects of radiation.ConclusionsWe conclude that Compound #5 has the potential to mitigate cognitive decline in patients undergoing partial or whole brain irradiation without promoting tumor growth and that the use of this compound as a radiation mitigator of radiation late effects on the CNS warrants further investigation.Importance of the StudySuccessful radiotherapy of CNS malignancies inevitably lead to cognitive decline in cancer survivors and treatment options to mitigate this side effect are limited. We present evidence that a piperazine compound can prevent cognitive decline in mice after total brain irradiation without compromising the antitumor effect of radiation, suggesting that this compound could be used to mitigate radiation side effects in brain tumor patients undergoing radiotherapy.
- Published
- 2020
10. Effects of Recombinant Erythropoietin on Breast Cancer-Initiating Cells
- Author
-
Tiffany M. Phillips, Kwanghee Kim, Erina Vlashi, William H. McBride, and Frank Pajonk
- Subjects
CD24ࢤ/low/CD44+ breast cancer cells ,breast cancer-initiating cells ,rhEpo ,Notch ,Epor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
BACKGROUND: Cancer anemia causes fatigue and correlates with poor treatment outcome. Erythropoietin has been introduced in an attempt to correct these defects. However, five recent clinical trials reported a negative impact of erythropoietin on survival and/or tumor control, indicating that experimental evaluation of a possible direct effect of erythropoietin on cancer cells is required. Cancer recurrence is thought to rely on the proliferation of cancer initiating cells (CICs). In breast cancer, CICs can be identified by phenotypic markers and their fate is controlled by the Notch pathway. METHODS: In this study, we investigated the effect of erythropoietin on CICs in breast cancer cell lines. Levels of erythropoietin receptor (EpoR), CD24, CD44, Jagged-1 expression, activation of Notch-1 were assessed by flow cytometry. Self-renewing capacity of CICs was investigated in sphere formation assays. RESULTS: EpoR expression was found on the surface of CICs. Recombinant human Epo (rhEpo) increased the numbers of CICs and self-renewing capacity in a Notch-dependent fashion by induction of Jagged-1. Inhibitors of the Notch pathway and P13-kinase blocked both effects. CONCLUSIONS: Erythropoietin functionally affects CICs directly. Our observation may explain the negative impact of recombinant Epo on local control and survival of cancer patients with EpoR-positive tumors.
- Published
- 2007
- Full Text
- View/download PDF
11. The Proteasome Inhibitor MG-132 Protects Hypoxic SiHa Cervical Carcinoma Cells after Cyclic Hypoxia/Reoxygenation from Ionizing Radiation
- Author
-
Frank Pajonk, Thorsten Grumann, and William H. McBride
- Subjects
Reoxygenation ,hypoxia ,radiation response ,MG-132 ,26S proteasome ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
INTRODUCTION: Transient hypoxia and subsequent reoxygenation are common phenomena in solid tumors that greatly influence the outcome of radiation therapy. This study was designed to determine how varying cycles of hypoxia/reoxygenation affect the response of cervical carcinoma cells irradiated under oxic and hypoxic conditions and whether this could be modulated by proteasome inhibition. MATERIALS AND METHODS: Plateau-phase SiHa cervical carcinoma cells in culture were exposed to varying numbers of 30-minute cycles of hypoxia/reoxygenation directly before irradiation under oxic or hypoxic conditions. 26S Proteasome activity was blocked by addition of MG-132. Clonogenic survival was measured by a colonyforming assay. RESULTS: Under oxic conditions, repeated cycles of hypoxia/reoxygenation decreased the clonogenic survival of SiHa cells. This effect was even more pronounced after the inhibition of 26S proteasome complex. In contrast, under hypoxic conditions, SiHa cells were radioresistant, as expected, but this was increased by proteasome inhibition. CONCLUSIONS: Proteasome inhibition radiosensitizes oxygenated tumor cells but may also protect tumor cells from ionizing radiation under certain hypoxic conditions.
- Published
- 2006
- Full Text
- View/download PDF
12. Dopamine Receptor Antagonists, Radiation, and Cholesterol Biosynthesis in Mouse Models of Glioblastoma
- Author
-
Kruttika Bhat, Le Zhang, Linda M. Liau, Frank Pajonk, Timothy F. Cloughesy, Steven J. Bensinger, Ling He, Mohammad Saki, Harley I. Kornblum, David Nathanson, Phioanh L. Nghiemphu, Angeliki Ioannidis, Fei Cheng, and Jonathan Tsang
- Subjects
Cancer Research ,Statin ,medicine.drug_class ,Atorvastatin ,Oncology and Carcinogenesis ,Brain tumor ,Pharmacology ,Inbred C57BL ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Rare Diseases ,In vivo ,Glioma ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,030304 developmental biology ,Cancer ,0303 health sciences ,Temozolomide ,Tumor ,business.industry ,Brain Neoplasms ,Hazard ratio ,Neurosciences ,Articles ,medicine.disease ,Xenograft Model Antitumor Assays ,Brain Disorders ,Mice, Inbred C57BL ,Brain Cancer ,Cholesterol ,Orphan Drug ,Oncology ,Dopamine receptor ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Dopamine Antagonists ,Development of treatments and therapeutic interventions ,business ,Glioblastoma ,medicine.drug ,Biotechnology - Abstract
Background Glioblastoma is the deadliest brain tumor in adults, and the standard of care consists of surgery followed by radiation and treatment with temozolomide. Overall survival times for patients suffering from glioblastoma are unacceptably low indicating an unmet need for novel treatment options. Methods Using patient-derived HK-157, HK-308, HK-374, and HK-382 glioblastoma lines, the GL261 orthotopic mouse models of glioblastoma, and HK-374 patient-derived orthotopic xenografts, we tested the effect of radiation and the dopamine receptor antagonist quetiapine on glioblastoma self-renewal in vitro and survival in vivo. A possible resistance mechanism was investigated using RNA-sequencing. The blood-brain-barrier–penetrating statin atorvastatin was used to overcome this resistance mechanism. All statistical tests were 2-sided. Results Treatment of glioma cells with the dopamine receptor antagonist quetiapine reduced glioma cell self-renewal in vitro, and combined treatment of mice with quetiapine and radiation prolonged the survival of glioma-bearing mice. The combined treatment induced the expression of genes involved in cholesterol biosynthesis. This rendered GL261 and HK-374 orthotopic tumors vulnerable to simultaneous treatment with atorvastatin and further statistically significantly prolonged the survival of C57BL/6 (n = 10 to 16 mice per group; median survival not reached; log-rank test, P Conclusions Our results indicate promising therapeutic efficacy with the triple combination of quetiapine, atorvastatin, and radiation treatment against glioblastoma without increasing the toxicity of radiation. With both drugs readily available for clinical use, our study could be rapidly translated into a clinical trial.
- Published
- 2021
13. Radiation-reprogrammed glioma stem cells generate vascular-like cells to build a trophic niche driving tumor recurrence
- Author
-
Michael C. Condro, Raymond Gau, Nathan VanderVeer-Harris, Q. Wang, Amy Pham, Yue Qin, Steven A. Goldman, Alvaro G. Alvarado, Maverick Johnson, Pooja Nair, Pedro R. Lowenstein, Rachna Prasad, Sree Deepthi Muthukrishnan, Arjun Deb, Maria G. Castro, Jason D Hinman, Daniel H. Geschwind, Riki Kawaguchi, Terry C. Burns, Frank Pajonk, and Harley I. Kornblum
- Subjects
Cell ,Histone acetyltransferase ,Biology ,medicine.disease ,Phenotype ,Chromatin ,medicine.anatomical_structure ,Glioma ,medicine ,Cancer research ,biology.protein ,Histone acetyltransferase activity ,Epigenetics ,Stem cell - Abstract
Treatment-refractory glioma stem and tumor cells exhibit phenotypic plasticity driving recurrence, but the underlying molecular mechanisms remain to be elucidated. Here, we employed single-cell and whole transcriptomic analyses to discover that radiation induces a dynamic shift in functional states of glioma cells allowing for acquisition of vascular endothelial-like and pericyte-like cell phenotypes. These vascular-like cells provide a trophic niche to promote proliferation of irradiated glioma cells, and their selective depletion results in reduced tumor growth post-treatment in vivo. Mechanistically, the acquisition of vascular-like phenotype is driven by increased chromatin accessibility and H3K27 acetylation in specific vascular gene regions post-treatment. Blocking P300 histone acetyltransferase activity reverses the epigenetic changes induced by radiation, and inhibits the phenotypic transition and tumor growth. Our findings highlight an important role for P300 histone acetyltransferase in treatment-induced plasticity and opens a new therapeutic avenue for preventing glioma recurrence.SignificanceOur study demonstrates that radiation therapy promotes glioma resistance by inducing vascular-like phenotypes in GSC that, in turn, aid in proliferation of the remaining tumor cells. This phenotype switch is mediated by P300 HAT, and inhibition of this enzyme is a potential therapeutic target for preventing glioma recurrence following radiation.
- Published
- 2021
14. Radiation mitigation of the intestinal acute radiation injury in mice by 1-[(4-nitrophenyl)sulfonyl]-4-phenylpiperazine
- Author
-
Sara Duhachek-Muggy, Gregoire Ruffenach, Fei Cheng, Paul Medina, Sree Deepthi Muthukrishnan, Claudia Alli, Kruttika Bhat, Frank Pajonk, Mohammad Saki, Erina Vlashi, Mansoureh Eghbali, and Ling He
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Medical Biotechnology ,Clinical Sciences ,Piperazines ,acute radiation syndrome ,Nitrophenols ,Vaccine Related ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Tissue‐specific Progenitor and Stem Cells ,Biodefense ,medicine ,Animals ,lcsh:QH573-671 ,intestinal stem cells ,lcsh:R5-920 ,Radiation ,lcsh:Cytology ,Chemistry ,Prevention ,Lethal dose ,Acute Radiation Syndrome ,Cell Biology ,General Medicine ,Stem Cell Research ,Small intestine ,Hedgehog signaling pathway ,3. Good health ,radiation ,Radiation therapy ,Emerging Infectious Diseases ,030104 developmental biology ,medicine.anatomical_structure ,Mechanism of action ,developmental signaling ,Cancer research ,Biochemistry and Cell Biology ,Stem cell ,medicine.symptom ,Digestive Diseases ,lcsh:Medicine (General) ,Smoothened ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The objective of the study was to identify the mechanism of action for a radiation mitigator of the gastrointestinal (GI) acute radiation syndrome (ARS), identified in an unbiased high‐throughput screen. We used mice irradiated with a lethal dose of radiation and treated with daily injections of the radiation mitigator 1‐[(4‐nitrophenyl)sulfonyl]‐4‐phenylpiperazine to study its effects on key pathways involved in intestinal stem cell (ISC) maintenance. RNASeq, quantitative reverse transcriptase‐polymerase chain reaction, and immunohistochemistry were performed to identify pathways engaged after drug treatment. Target validation was performed with competition assays, reporter cells, and in silico docking. 1‐[(4‐Nitrophenyl)sulfonyl]‐4‐phenylpiperazine activates Hedgehog signaling by binding to the transmembrane domain of Smoothened, thereby expanding the ISC pool, increasing the number of regenerating crypts and preventing the GI‐ARS. We conclude that Smoothened is a target for radiation mitigation in the small intestine that could be explored for use in radiation accidents as well as to mitigate normal tissue toxicity during and after radiotherapy of the abdomen., The intestinal stem cell niche responds to radiation with apoptosis of intestinal stem cells and subsequent loss of tissue integrity. Treatment with compound #5 activates Hedgehog signaling, expands the number of intestinal stem cells and regenerating crypts, and allows for survival of the animals after a lethal dose of radiation.
- Published
- 2019
15. Effects of the DRD2/3 antagonist ONC201 and radiation in glioblastoma
- Author
-
Phioanh L. Nghiemphu, Harley I. Kornblum, Angeliki Ioannidis, Le Zhang, Kruttika Bhat, Joshua E. Allen, Nhan T. Nguyen, Timothy F. Cloughesy, Ling He, Linda M. Liau, and Frank Pajonk
- Subjects
Pyridines ,medicine.medical_treatment ,Cell ,urologic and male genital diseases ,030218 nuclear medicine & medical imaging ,Transcriptome ,Mice ,0302 clinical medicine ,Receptors ,Dopamine receptor antagonist ,Cancer ,Tumor ,Brain Neoplasms ,Imidazoles ,Hematology ,female genital diseases and pregnancy complications ,Other Physical Sciences ,medicine.anatomical_structure ,Oncology ,Local ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Transcriptome analyses ,Stem cell ,Development of treatments and therapeutic interventions ,medicine.drug ,Adult ,Combination therapy ,Oncology and Carcinogenesis ,Article ,Cell Line ,03 medical and health sciences ,Rare Diseases ,Radioresistance ,Glioma ,Cell Line, Tumor ,Dopamine D2 ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Oncology & Carcinogenesis ,Temozolomide ,Radiotherapy ,urogenital system ,business.industry ,Receptors, Dopamine D2 ,Neurosciences ,medicine.disease ,Xenograft Model Antitumor Assays ,nervous system diseases ,Brain Disorders ,Radiation therapy ,Brain Cancer ,Neoplasm Recurrence ,Orphan Drug ,Pyrimidines ,Cancer research ,Neoplasm Recurrence, Local ,business ,Quiescent glioblastoma cells ,Glioblastoma - Abstract
Background Glioblastoma (GBM) is the deadliest of all brain cancers in adults. The current standard-of-care is surgery followed by radiotherapy and temozolomide, leading to a median survival time of only 15 months. GBM are organized hierarchically with a small number of glioma-initiating cells (GICs), responsible for therapy resistance and tumor recurrence, suggesting that targeting GICs could improve treatment response. ONC201 is a first-in-class anti-tumor agent with clinical efficacy in some forms of high-grade gliomas. Here we test its efficacy against GBM in combination with radiation. Methods Using patient-derived GBM lines and mouse models of GBM we test the effects of radiation and ONC201 on GBM self - renewal in vitro and survival in vivo. A possible resistance mechanism is investigated using RNA-Sequencing. Results Treatment of GBM cells with ONC201 reduced self-renewal, clonogenicity and cell viability in vitro. ONC201 exhibited anti-tumor effects on radioresistant GBM cells indicated by reduced self-renewal in secondary and tertiary glioma spheres. Combined treatment of ONC201 and radiation prolonged survival in syngeneic and patient-derived orthotopic xenograft mouse models of GBM. Subsequent transcriptome analyses after combined treatment revealed shifts in gene expression signatures related to quiescent GBM populations, GBM plasticity, and GBM stem cells. Conclusions Our findings suggest that combined treatment with the DRD2/3 antagonist ONC201 and radiation improves the efficacy of radiation against GBM in vitro and in vivo through suppression of GICs without increasing toxicity in mouse models of GBM. A clinical assessment of this novel combination therapy against GBM is further warranted.
- Published
- 2020
16. Synergistic Effects of the DRD2/3 Antagonist ONC201 and Radiation in Glioblastoma
- Author
-
Linda M. Liau, Le Zhang, Frank Pajonk, Kruttika Bhat, Angeliki Ioannidis, Timothy F. Cloughesy, Jashua E Allen, Ling He, Phioanh L. Nghiemphu, Harley I. Kornblum, and Nhan T. Nguyen
- Subjects
education.field_of_study ,Temozolomide ,Combination therapy ,business.industry ,medicine.medical_treatment ,Population ,medicine.disease ,Radiation therapy ,In vivo ,Glioma ,Radioresistance ,medicine ,Cancer research ,Stem cell ,education ,business ,medicine.drug - Abstract
BackgroundGlioblastoma (GBM) is the deadliest of all brain cancers in adults. The current standard-of-care is surgery followed by radiotherapy and temozolomide, leading to a median survival time of only 15 months. GBM are organized hierarchically with a small number of glioma-initiating cells, responsible for therapy resistance and tumor recurrence, suggesting that targeting glioma-initiating cells could improve treatment response. ONC201 is a first-in-class anti-tumor agent with clinical efficacy in some forms of high-grade gliomas. Here we test its efficacy against GBM in combination with radiation.MethodsUsing patient-derived GBM lines and mouse models of GBM we test the effects of radiation and ONC201 on GBM self - renewal in vitro and survival in vivo. A possible resistance mechanism is investigated using RNA-Sequencing.ResultsTreatment of GBM cells with ONC201 reduced self-renewal, clonogenicity and cell viability in vitro. ONC201 exhibited anti-tumor effects on radioresistant GBM cells indicated by reduced self-renewal in secondary and tertiary glioma spheres. Combined treatment with ONC201 and radiation prolonged survival in syngeneic and patient-derived orthotopic xenograft mouse models of GBM. Subsequent transcriptome analyses after combined treatment revealed shifts in gene expression signatures related to quiescent GBM populations, GBM plasticity, and GBM stem cells.ConclusionsOur findings suggest that combined treatment with the DRD2/3 antagonist ONC201 and radiation improves the efficacy of radiation against GBM in vitro and in vivo through suppression of GICs without increasing toxicity in mouse models of GBM. A clinical assessment of this novel combination therapy against GBM is further warranted.Key points- Combined treatment of ONC201 and radiation exhibit anti-tumor effects on cells from primary and recurrent GBM- Combined treatment significantly prolongs survival in vivo- Combined treatment potentially targets the quiescent GBM cell populationImportance of the StudyThe survival rates for patients with GBM are unacceptably low and novel treatment approaches are needed. This study provides evidence that a combination of radiation and the dopamine receptor antagonist ONC201 significantly prolongs survival in mouse models of glioma.
- Published
- 2020
17. Tumor Necrosis Factor Receptor Signaling Modulates Carcinogenesis in a Mouse Model of Breast Cancer
- Author
-
Le Zhang, Nhan T. Nguyen, Kruttika Bhat, Frank Pajonk, Sara Duhacheck-Muggy, Ling He, Neda A. Moatamed, and Angeliki Ioannidis
- Subjects
0303 health sciences ,business.industry ,Mammary gland ,medicine.disease_cause ,medicine.disease ,3. Good health ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Breast cancer ,030220 oncology & carcinogenesis ,Knockout mouse ,medicine ,Cancer research ,Tumor necrosis factor alpha ,Signal transduction ,Carcinogenesis ,Autocrine signalling ,business ,030304 developmental biology - Abstract
Proinflammatory conditions have long been associated with mammary carcinogenesis and breast cancer progression. The underlying mechanisms are incompletely understood but signaling of TNFα through its receptors TNFR1 and TNFR2 is a major mediator of inflammation in both, obesity and in the response of tissues to radiation, two known risk factors for the development of breast cancer. Using the MMTV-Wnt1 mouse model for spontaneous breast cancer and knockout mice for TNFR1 and TNFR2 we report that loss of a TNFR2 allele leads to ductal hyperplasia in the mammary gland with increased numbers of mammary epithelial stem cell and terminal endbuds. Furthermore, that loss of one TNFR2 allele increases the incidence of breast cancers in MMTV-Wnt1 mice and results in tumors with a more aggressive phenotype and metastatic potential. The underlying mechanisms include a preferential activation of canonical NF-κB signaling pathways and autocrine production of TNFα. Analysis of the TCGA dataset indicated inferior overall survival for patients with down-regulated TNFR2 expression.We conclude, that imbalances in TNFR signaling promote the development and progression of breast cancer, indicating that selective agonists of TNFR2 could potentially modulate the risk for breast cancer in high-risk populations.Significance StatementOver the past four decades the treatment results for patients suffering from breast cancer have constantly improved, leaving breast cancer prevention as an important frontier against the second leading cause of cancer death in the United States. Obesity has become a national health crisis and is a known proinflammatory risk factor for breast cancer. Our study describes a previously unknown aspect of proinflammatory signaling on breast cancer development and progression, thus suggesting novel potential targets to modulate the incidence of the disease.
- Published
- 2020
18. Combined Treatment with Dopamine Receptor Antagonists and Radiation Creates a Metabolic Vulnerability in Mouse Models of Glioblastoma
- Author
-
David Nathanson, Mohammad Saki, Fei Cheng, Phioanh L. Nghiemphu, Harley I. Kornblum, Kruttika Bhat, Linda M. Liau, Frank Pajonk, Timothy F. Cloughesy, Jonathan Tsang, Le Zhang, Angeliki Ioannidis, and Ling He
- Subjects
0303 health sciences ,Temozolomide ,business.industry ,Atorvastatin ,Brain tumor ,medicine.disease ,3. Good health ,nervous system diseases ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Dopamine receptor ,030220 oncology & carcinogenesis ,Glioma ,Toxicity ,Cancer research ,medicine ,Quetiapine ,business ,neoplasms ,030304 developmental biology ,medicine.drug - Abstract
BackgroundGlioblastoma is the deadliest brain tumor in adults and the standard-of-care consists of surgery followed by radiation and treatment with temozolomide. Overall survival times for patients suffering from glioblastoma are unacceptably low indicating an unmet need for novel treatment options.MethodsUsing patient-derived glioblastoma lines and mouse models of glioblastoma we test the effect of radiation and the dopamine receptor antagonist on glioblastoma self-renewal in vitro and survival in vivo. A possible resistance mechanism is investigated using RNA-Sequencing.ResultsTreatment of glioma cells with the dopamine receptor antagonist quetiapine reduced glioma cell self-renewal in vitro and combined treatment of mice with quetiapine and radiation prolonged the survival of glioma-bearing animals. The combined treatment induced the expression of genes involved in cholesterol biosynthesis. This rendered the tumors vulnerable to simultaneous treatment with atorvastatin and further significantly prolonged the survival of the animals.ConclusionsOur results indicate high efficacy of a triple combination of quetiapine, atorvastatin and radiation against glioblastoma without increasing the toxicity of radiation. With both drugs readily available for clinical use our study could be rapidly translated into a clinical trial.
- Published
- 2020
- Full Text
- View/download PDF
19. Oxygen levels do not determine radiation survival of breast cancer stem cells.
- Author
-
Chann Lagadec, Carmen Dekmezian, Lucile Bauché, and Frank Pajonk
- Subjects
Medicine ,Science - Abstract
For more than a century oxygen has been known to be one of the most powerful radiosensitizers. However, despite decades of preclinical and clinical research aimed at overcoming tumor hypoxia, little clinical progress has been made so far. Ionizing radiation damages DNA through generation of free radicals. In the presence of oxygen these lesions are chemically modified, and thus harder to repair while hypoxia protects cells from radiation (Oxygen enhancement ratio (OER)). Breast cancer stem cells (BSCSs) are protected from radiation by high levels of free radical scavengers even in the presence of oxygen. This led us to hypothesize that BCSCs exhibit an OER of 1. Using four established breast cancer cell lines (MCF-7, T47D, MDA-MB-231, SUM159PT) and primary breast cancer samples, we determined the number of BCSCs using cancer stem cell markers (ALDH1, low proteasome activity), compared radiation clonogenic survival and mammosphere formation under normoxic and hypoxic conditions, and correlated these results to the expression levels of key members of the free radical scavenging systems. The number of BCSCs increased with increased aggressiveness of the cancer. This correlated with increased radioresistance (SF(8Gy)), and decreasing OERs. When cultured as mammospheres, breast cancer cell lines and primary samples were highly radioresistant and not further protected by hypoxia (OER∼1).We conclude that because BCSCs are protected from radiation through high expression levels of free radical scavengers, hypoxia does not lead to additional radioprotection of BCSCs.
- Published
- 2012
- Full Text
- View/download PDF
20. The Dopamine Receptor Antagonist TFP Prevents Phenotype Conversion and Improves Survival in Mouse Models of Glioblastoma
- Author
-
Phioanh L. Nghiemphu, Erina Vlashi, Sara Duhachek-Muggy, Kruttika Bhat, Frank Pajonk, Timothy F. Cloughesy, Claudia Alli, Mohammad Saki, Matteo Pellegrini, Garrett Yu, Harley I. Kornblum, Ling He, Fei Cheng, Robert Damoiseaux, Paul Medina, Nathan R. Zemke, and Linda M. Liau
- Subjects
Homeobox protein NANOG ,business.industry ,medicine.medical_treatment ,Trifluoperazine ,medicine.disease ,Phenotype ,Radiation therapy ,Radiation sensitivity ,SOX2 ,In vivo ,medicine ,Cancer research ,business ,Glioblastoma ,medicine.drug - Abstract
Glioblastoma (GBM) is the deadliest adult brain cancer and all patients ultimately succumb to the disease. Radiation therapy (RT) provides survival benefit of 6 months over surgery alone but these results have not improved in decades. We report that radiation induces a glioma-initiating cell phenotype and we have identified trifluoperazine (TFP) as a compound that interferes with this phenotype conversion. TFP caused loss of radiation-induced Nanog mRNA expression, activation of GSK3 with consecutive post-translational reduction in p-Akt, Sox2 and β-catenin protein levels. TFP did not alter the intrinsic radiation sensitivity of glioma-initiating cells (GICs). Continuous treatment with TFP and a single dose of radiation reduced the number of GICs in vivo and prolonged survival in syngeneic and patient-derived orthotopic xenograft (PDOX) mouse models of GBM. Our findings suggest that combination of a dopamine receptor antagonist with radiation enhances the efficacy of RT in GBM by preventing radiation-induced phenotype conversion of radiosensitive non-GICs into treatment resistant, induced GICs.SignificanceGBM is the most common and most deadly adult brain cancer. The current standard-of-care is surgery followed by RT and temozolomide, which results in a median survival time of only 15 months. The efficacy of chemotherapies and targeted therapies in GBM is very limited because most of these drugs do not pass the blood-brain-barrier. Ultimately, all patients succumb to the disease. Our study describes radiation-induced cellular plasticity as a novel resistance mechanism in GBM. We identified a dopamine receptor antagonist as a readily available, FDA-approved drug known to penetrate the blood-brain-barrier which prevents phenotype conversion of glioma cells into glioma-initiating cells and prologs survival in mouse models of GBM, thus suggesting that it will improve the efficacy of RT without increasing toxicity.
- Published
- 2019
21. Metabolic characterization of isocitrate dehydrogenase (IDH) mutant and IDH wildtype gliomaspheres uncovers cell type-specific vulnerabilities
- Author
-
Thomas G. Graeber, Linda M. Liau, Giovanni Coppola, Weihong Yan, Daniel Braas, Harley I. Kornblum, Jack Mottahedeh, Joshua J. Breunig, Kirsten Ludwig, Matthew C. Garrett, Thuc Le, Ascia Eskin, Albert Lai, Yue Qin, Robert M. Prins, Heather R. Christofk, Caius G. Radu, Rachelle Levy, Frank Pajonk, and Jantzen Sperry
- Subjects
0301 basic medicine ,DNA damage ,DNA repair ,Mutant ,2-hydroxyglutarate ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,03 medical and health sciences ,Rare Diseases ,medicine ,Genetics ,Cancer ,Mutation ,Radiation ,Research ,Wild type ,Glioma ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular biology ,Brain Disorders ,Comet assay ,Brain Cancer ,Psychiatry and Mental health ,030104 developmental biology ,Isocitrate dehydrogenase ,Metabolism ,Cell culture ,Nucleotide - Abstract
Background There is considerable interest in defining the metabolic abnormalities of IDH mutant tumors to exploit for therapy. While most studies have attempted to discern function by using cell lines transduced with exogenous IDH mutant enzyme, in this study, we perform unbiased metabolomics to discover metabolic differences between a cohort of patient-derived IDH1 mutant and IDH wildtype gliomaspheres. Methods Using both our own microarray and the TCGA datasets, we performed KEGG analysis to define pathways differentially enriched in IDH1 mutant and IDH wildtype cells and tumors. Liquid chromatography coupled to mass spectrometry analysis with labeled glucose and deoxycytidine tracers was used to determine differences in overall cellular metabolism and nucleotide synthesis. Radiation-induced DNA damage and repair capacity was assessed using a comet assay. Differences between endogenous IDH1 mutant metabolism and that of IDH wildtype cells transduced with the IDH1 (R132H) mutation were also investigated. Results Our KEGG analysis revealed that IDH wildtype cells were enriched for pathways involved in de novo nucleotide synthesis, while IDH1 mutant cells were enriched for pathways involved in DNA repair. LC-MS analysis with fully labeled 13C-glucose revealed distinct labeling patterns between IDH1 mutant and wildtype cells. Additional LC-MS tracing experiments confirmed increased de novo nucleotide synthesis in IDH wildtype cells relative to IDH1 mutant cells. Endogenous IDH1 mutant cultures incurred less DNA damage than IDH wildtype cultures and sustained better overall growth following X-ray radiation. Overexpression of mutant IDH1 in a wildtype line did not reproduce the range of metabolic differences observed in lines expressing endogenous mutations, but resulted in depletion of glutamine and TCA cycle intermediates, an increase in DNA damage following radiation, and a rise in intracellular ROS. Conclusions These results demonstrate that IDH1 mutant and IDH wildtype cells are easily distinguishable metabolically by analyzing expression profiles and glucose consumption. Our results also highlight important differences in nucleotide synthesis utilization and DNA repair capacity that could be exploited for therapy. Altogether, this study demonstrates that IDH1 mutant gliomas are a distinct subclass of glioma with a less malignant, but also therapy-resistant, metabolic profile that will likely require distinct modes of therapy. Electronic supplementary material The online version of this article (10.1186/s40170-018-0177-4) contains supplementary material, which is available to authorized users.
- Published
- 2018
22. If It Seems Too Good to Be True…
- Author
-
Michael L. Steinberg, Erina Vlashi, William H. McBride, and Frank Pajonk
- Subjects
Cancer Research ,Radiation ,Text mining ,Oncology ,business.industry ,Medicine ,Radiology, Nuclear Medicine and imaging ,business ,Data science - Published
- 2019
23. The Future of Radiobiology
- Author
-
David G Kirsch, Max Diehn, Aparna H Kesarwala, Amit Maity, Meredith A Morgan, Julie K Schwarz, Robert Bristow, Sandra Demaria, Iris Eke, Robert J Griffin, Daphne Haas-Kogan, Geoff S Higgins, Alec C Kimmelman, Randall J Kimple, Isabelle M Lombaert, Li Ma, Brian Marples, Frank Pajonk, Catherine C Park, Dörthe Schaue, Phuoc T. Tran, Henning Willers, Brad G. Wouters, and Eric J Bernhard
- Subjects
0301 basic medicine ,Scientific enterprise ,Cancer Research ,Radiobiology ,business.industry ,medicine.medical_treatment ,medicine.disease ,Radiation therapy ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Research community ,Ataxia-telangiectasia ,Radiation oncology ,medicine ,Ataxia telangiectasia mutated ,Engineering ethics ,business - 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
- 2017
24. PK-M2-mediated metabolic changes in breast cancer cells induced by ionizing radiation
- Author
-
Justine Bailleul, Sara Duhachek-Muggy, Claudia Alli, David Nathanson, Le Zhang, Kruttika Bhat, Laura Gosa, Taha Yazal, Erina Vlashi, Milana Bochkur Dratver, Kevin Dong, Frank Pajonk, David Sung, and Amy Dao
- Subjects
0301 basic medicine ,Cancer Research ,Ionizing ,medicine.medical_treatment ,Triple Negative Breast Neoplasms ,0302 clinical medicine ,Breast cancer ,Radiation, Ionizing ,2.1 Biological and endogenous factors ,Aetiology ,Pyruvate kinase ,Cellular localization ,Cancer ,Tumor ,Radiation ,Chemistry ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Radiation therapy ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Female ,Stem Cell Research - Nonembryonic - Non-Human ,Stem cell ,Thyroid Hormones ,Clinical Sciences ,Oncology and Carcinogenesis ,Article ,Cell Line ,03 medical and health sciences ,Cancer stem cell ,Cell Line, Tumor ,Radioresistance ,medicine ,Humans ,Lactic Acid ,Oncology & Carcinogenesis ,Cell Nucleus ,Neoplastic ,Membrane Proteins ,medicine.disease ,Stem Cell Research ,030104 developmental biology ,Glucose ,Metabolism ,Gene Expression Regulation ,Cancer cell ,Cancer research ,Carrier Proteins - Abstract
PURPOSE: Radiotherapy (RT) constitutes an important part of breast cancer treatment. However, triple negative breast cancers (TNBC) exhibit remarkable resistance to most therapies, including RT. Developing new ways to radiosensitize TNBC cells could result in improved patient outcomes. The M2 isoform of pyruvate kinase (PK-M2) is believed to be responsible for the re-wiring of cancer cell metabolism after oxidative stress. The aim of the study was to determine the effect of ionizing radiation (IR) on PK-M2-mediated metabolic changes in TNBC cells, and their survival. In addition, we determine the effect of PK-M2 activators on breast cancer stem cells, a radioresistant subpopulation of breast cancer stem cells. METHODS: Glucose uptake, lactate production and glutamine consumption were assessed. The cellular localization of PK-M2 was evaluated by western blot and confocal microscopy. The small molecule activator of PK-M2, TEPP46, was used to promote its pyruvate kinase function. Finally, effects on cancer stem cell were evaluated via sphere forming capacity. RESULTS: Exposure of TNBC cells to IR increased their glucose uptake and lactate production. As expected, PK-M2 expression levels also increased, especially in the nucleus, although overall pyruvate kinase activity was decreased. PK-M2 nuclear localization was shown to be associated with breast cancer stem cells, and activation of PK-M2 by TEPP46 depleted this population. CONCLUSIONS: Radiotherapy can induce metabolic changes in TNBC cells, and these changes seem to be mediated, at least in part by PK-M2. Importantly, our results show that activators of PKM2 can deplete breast cancer stem cells in vitro. This study supports the idea of combining PK-M2 activators with radiation to enhance the effect of radiotherapy in resistant cancers, such as TNBC.
- Published
- 2019
25. Mebendazole Potentiates Radiation Therapy in Triple-Negative Breast Cancer
- Author
-
Kruttika Bhat, Michael Bochkur Dratver, Amy Dao, Milana Bochkur Dratver, Le Zhang, Claudia Alli, Kevin Dong, Garrett Yu, Erina Vlashi, Andrea Nguyen, Frank Pajonk, Taha Yazal, and Sara Duhachek-Muggy
- Subjects
Cancer Research ,medicine.medical_treatment ,Apoptosis ,Triple Negative Breast Neoplasms ,030218 nuclear medicine & medical imaging ,chemistry.chemical_compound ,Mice ,Double-Stranded ,0302 clinical medicine ,Stem Cell Research - Nonembryonic - Human ,Medicine ,DNA Breaks, Double-Stranded ,Triple-negative breast cancer ,Cancer ,education.field_of_study ,Radiation ,Tumor ,Cell cycle ,Other Physical Sciences ,Mebendazole ,Oncology ,030220 oncology & carcinogenesis ,Female ,Reprogramming ,Biotechnology ,Population ,Clinical Sciences ,Oncology and Carcinogenesis ,Article ,Cell Line ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,Breast Cancer ,Genetics ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Propidium iodide ,Oncology & Carcinogenesis ,education ,business.industry ,Prevention ,DNA Breaks ,Cell Dedifferentiation ,medicine.disease ,Stem Cell Research ,High-Throughput Screening Assays ,Radiation therapy ,chemistry ,Cancer research ,business - Abstract
Purpose The lack of a molecular target in triple-negative breast cancer (TNBC) makes it one of the most challenging breast cancers to treat. Radiation therapy (RT) is an important treatment modality for managing breast cancer; however, we previously showed that RT can also reprogram a fraction of the surviving breast cancer cells into breast cancer–initiating cells (BCICs), which are thought to contribute to disease recurrence. In this study, we characterize mebendazole (MBZ) as a drug with potential to prevent the occurrence of radiation-induced reprogramming and improve the effect of RT in patients with TNBC. Methods and Materials A high-throughput screen was used to identify drugs that prevented radiation-induced conversion of TNBC cells into cells with a cancer-initiating phenotype and exhibited significant toxicity toward TNBC cells. MBZ was one of the drug hits that fulfilled these criteria. In additional studies, we used BCIC markers and mammosphere-forming assays to investigate the effect of MBZ on the BCIC population. Staining with propidium iodide, annexin-V, and γ-H2AX was used to determine the effect of MBZ on cell cycle, apoptosis, and double-strand breaks. Finally, the potential for MBZ to enhance the effect of RT in TNBC was evaluated in vitro and in vivo. Results MBZ efficiently depletes the BCIC pool and prevents the ionizing radiation–induced conversion of breast cancer cells into therapy-resistant BCICs. In addition, MBZ arrests cells in the G2/M phase of the cell cycle and causes double-strand breaks and apoptosis. MBZ sensitizes TNBC cells to ionizing radiation in vitro and in vivo, resulting in improved tumor control in a human xenograft model of TNBC. Conclusions The data presented in this study support the repurposing of MBZ as a combination treatment with RT in patients with TNBC.
- Published
- 2019
26. Abstract P3-03-07: TNFR-signaling in breast cancer development
- Author
-
Fei Cheng, Mohammad Saki, Paul Medina, Frank Pajonk, Kruttika Bhat, Ling He, and Claudia Alli
- Subjects
Cancer Research ,education.field_of_study ,animal structures ,Mammary gland ,Population ,Inflammation ,Biology ,medicine.disease ,Breast cancer ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine ,Immunohistochemistry ,Tumor necrosis factor alpha ,medicine.symptom ,Stem cell ,education ,CD8 - Abstract
Introduction: Breast cancer (BC) is the 2nd leading cause of cancer death among women in the US. Known risk factors for the development of BC include thoracic radiotherapy during puberty and obesity with the latter turning into a nation-wide health crisis. The underlying mechanisms that lead to increased BC incidences in these two scenarios are incompletely understood. However, exposure to ionizing radiation (IR) or obesity generate pro-inflammatory conditions that have been linked to mammary carcinogenesis. As inflammation involves signaling through TNF receptors (TNFR), we have therefore employed crosses between mice that spontaneously develop BC (MMTV-Wnt1) and TNFR KO mice to study the impact of TNFR downstream signaling on BC development. Material and Methods: MMTV-Wnt1 were crossed with TNFR1 or TNFR2 KO mice to explore the BC incidence during the first year post-partum in both genders. Mammary gland area and ductal outgrowth were evaluated by whole mount histological staining with 6-week-old female (end of puberty) and 14-week-old mice (adult). Tumor cells were harvested from tumor-developed MMTV-Wnt1 and MMTV-Wnt1 x TNFR2 KO female mice, the population of tumor initiating cells (EpCAMlow/CD49fhi) was analyzed by FACS. Vasculature formation (CD31), cell proliferation (Ki67) and immune system (CD4, CD8, F4/80) were evaluated within tumors by immunohistochemistry staining. Tumor cells were digested out from primary tumors and cultured in spheres condition. In-vitro sphere formation assay was performed w/o IR or TNFα treatment. All quantitative results were treated by ANOVA and Bonferroni analysis, upon verification of normal data distribution with α value at 0.05. Results: TNFR2KO drastically accelerates BC in female animals but not in males. In contrary, TNFR1 KOhad no effect on female BC development but completely prevented BC in males. TNFR2 KO led to elongation of mammary gland duct system, expansion of mammary epithelial stem cell pool and increase of BC initiating cells, as well as more vasculatures and proliferating cells in developed tumors. T-cellsand macrophage content in established tumors don’t correlate with differences in BC incidence between the crosses. MMTV-Wnt1 x TNFR2 KO mammospheres were more resistant to IR compared to Wnt1 x TNFR1 KO and MMTV-Wnt1. Pro-inflammatory TNFα ligand increased the sphere formation in a dose-dependent manner within Wnt1 x TNFR1 KO and MMTV-Wnt1 spheres, but not in Wnt1 x TNFR2 KO ones. Conclusion: Heterozygous TNFR2 KO in female MMTV-Wnt1 Tg mice increases the BC incidence, along with more resistance to IR, suggesting the downstream of TNFR2 may potentially suppress breast carcinogenesis. Also TNFR2 KO leads to more mammary gland branching and expansion of epithelial stem cells during development, which imply the potential use of TNFR2 agonist in thoracic radiotherapy and obese scenarios to reduce the BC incidence. Citation Format: Ling He, Kruttika Bhat, Paul Medina, Claudia Alli, Mohammad Saki, Fei Cheng, Frank Pajonk. TNFR-signaling in breast cancer development [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P3-03-07.
- Published
- 2020
27. 1-(4-nitrobenzenesulfonyl)-4-penylpiperazine increases the number of Peyer's patch-associated regenerating crypts in the small intestines after radiation injury
- Author
-
Mohammad Saki, Kruttika Bhat, Renuka Ramanathan, Paul Medina, Sara Duhachek-Muggy, Frank Pajonk, Dörthe Schaue, William H. McBride, Claudia Alli, Erina Vlashi, Julian P. Whitelegge, and Robert Damoiseaux
- Subjects
Crypt ,Apoptosis ,Radiation-Protective Agents ,Biology ,Piperazines ,Article ,030218 nuclear medicine & medical imaging ,Flow cytometry ,03 medical and health sciences ,Mice ,Peyer's Patches ,Random Allocation ,0302 clinical medicine ,Immune system ,Intestine, Small ,medicine ,Animals ,Regeneration ,Radiology, Nuclear Medicine and imaging ,Intestinal Mucosa ,Radiation Injuries ,Nitrobenzenes ,Mice, Inbred C3H ,medicine.diagnostic_test ,Peyer's patch ,Germinal center ,Hematology ,Total body irradiation ,Molecular biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Lymphatic system ,Oncology ,030220 oncology & carcinogenesis ,Female ,Whole-Body Irradiation - Abstract
OBJECTIVE: Exposure to lethal doses of radiation has severe effects on normal tissues. Exposed individuals experience a plethora of symptoms in different organ systems including the gastrointestinal (GI) tract, summarized as Acute Radiation Syndrome (ARS). There are currently no approved drugs for mitigating GI-ARS. A recent high-throughput screen performed at the UCLA Center for Medical Countermeasures against Radiation identified compounds containing sulfonylpiperazine groups with radiation mitigation properties to the hematopoietic system and the gut. Among these 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine (Compound #5) efficiently mitigated gastrointestinal ARS. However, the mechanism of action and target cells of this drug is still unknown. In this study we examined if Compound #5 affects gut-associated lymphoid tissue (GALT) with its subepithelial domes called Peyer’s patches. METHODS: C3H mice were irradiated with 0 or 12 Gy total body irradiation (TBI). A single dose of Compound #5 or solvent was administered subcutaneously 24 hours later. 48 hours after irradiation the mice were sacrificed, and the guts examined for changes in the number of visible Peyer’s patches. In some experiments the mice received 4 daily injections of treatment and were sacrificed 96 hours after TBI. For immune histochemistry gut tissues were fixed in formalin and embedded in paraffin blocks. Sections were stained with H&E, anti-Ki67 or a TUNEL assay to assess the number of regenerating crypts, mitotic and apoptotic indices. Cells isolated from Peyer’s patches were subjected to immune profiling using flow cytometry. RESULTS: Compound #5 significantly increased the number of visible Peyer’s patches when compared to its control in non-irradiated and irradiated mice. Additionally, assessment of total cells per Peyer’s patch isolated from these mice demonstrated an overall increase in the total number of Peyer’s patch cells per mouse in Compound #5-treated mice. In non-irradiated animals the number of CD11b(high) in Peyer’s patches increased significantly. These Compound #5-driven increases did not coincide with a decrease in apoptosis or an increase in proliferation in the germinal centers inside the Peyer’s patches 24 hours after drug treatment. A single dose of Compound #5 significantly increased the number of CD45(+) cells after 12 Gy TBI. Importantly, 96 hours after 12 Gy TBI Compound #5 induced a significant rise in the number of visible Peyer’s patches and the number of Peyer’s patch-associated regenerating crypts. CONCLUSION: In summary, our study provides evidence that Compound #5 leads to an influx of immune cells into GALT, thereby supporting crypt regeneration preferentially in the proximity of Peyer’s patches.
- Published
- 2018
28. Incorporating Cancer Stem Cells in Radiation Therapy Treatment Response Modeling and the Implication in Glioblastoma Multiforme Treatment Resistance
- Author
-
Michael T. Selch, Victoria Y. Yu, Daniel A. Low, Dan Nguyen, Frank Pajonk, Patrick A. Kupelian, Tania Kaprealian, and Ke Sheng
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Cell Survival ,medicine.medical_treatment ,Uterine Cervical Neoplasms ,Bone Neoplasms ,Breast Neoplasms ,Radiation Tolerance ,Breast cancer ,Cancer stem cell ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Radioresistance ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Radiosensitivity ,Melanoma ,Osteosarcoma ,Radiation ,Brain Neoplasms ,business.industry ,Prostatic Neoplasms ,medicine.disease ,Radiation therapy ,Oncology ,Cancer cell ,Linear Models ,Neoplastic Stem Cells ,Cancer research ,Female ,Stem cell ,Glioblastoma ,business - Abstract
Purpose To perform a preliminary exploration with a simplistic mathematical cancer stem cell (CSC) interaction model to determine whether the tumor-intrinsic heterogeneity and dynamic equilibrium between CSCs and differentiated cancer cells (DCCs) can better explain radiation therapy treatment response with a dual-compartment linear-quadratic (DLQ) model. Methods and Materials The radiosensitivity parameters of CSCs and DCCs for cancer cell lines including glioblastoma multiforme (GBM), non–small cell lung cancer, melanoma, osteosarcoma, and prostate, cervical, and breast cancer were determined by performing robust least-square fitting using the DLQ model on published clonogenic survival data. Fitting performance was compared with the single-compartment LQ (SLQ) and universal survival curve models. The fitting results were then used in an ordinary differential equation describing the kinetics of DCCs and CSCs in response to 2- to 14.3-Gy fractionated treatments. The total dose to achieve tumor control and the fraction size that achieved the least normal biological equivalent dose were calculated. Results Smaller cell survival fitting errors were observed using DLQ, with the exception of melanoma, which had a low α/β = 0.16 in SLQ. Ordinary differential equation simulation indicated lower normal tissue biological equivalent dose to achieve the same tumor control with a hypofractionated approach for 4 cell lines for the DLQ model, in contrast to SLQ, which favored 2 Gy per fraction for all cells except melanoma. The DLQ model indicated greater tumor radioresistance than SLQ, but the radioresistance was overcome by hypofractionation, other than the GBM cells, which responded poorly to all fractionations. Conclusion The distinct radiosensitivity and dynamics between CSCs and DCCs in radiation therapy response could perhaps be one possible explanation for the heterogeneous intertumor response to hypofractionation and in some cases superior outcome from stereotactic ablative radiation therapy. The DLQ model also predicted the remarkable GBM radioresistance, a result that is highly consistent with clinical observations. The radioresistance putatively stemmed from accelerated DCC regrowth that rapidly restored compartmental equilibrium between CSCs and DCCs.
- Published
- 2015
29. The metabolic state of cancer stem cells—a valid target for cancer therapy?
- Author
-
Frank Pajonk and Erina Vlashi
- Subjects
Biochemistry & Molecular Biology ,Pathology ,medicine.medical_specialty ,Free radicals ,Context (language use) ,Medical Biochemistry and Metabolomics ,Biology ,Carbohydrate metabolism ,Biochemistry ,Article ,Medicinal and Biomolecular Chemistry ,Cancer stem cell ,Neoplasms ,Physiology (medical) ,medicine ,Humans ,Oxidative phosphorylation ,Glycolysis ,Cancer ,Radiation ,Cancer stem cells ,Metabolism ,Stem Cell Research ,medicine.disease ,Oxygen ,Anaerobic glycolysis ,Cancer cell ,Neoplastic Stem Cells ,Cancer research ,Biomedical Imaging ,Biochemistry and Cell Biology - Abstract
In the 1920s Otto Warburg first described high glucose uptake, aerobic glycolysis, and high lactate production in tumors. Since then high glucose uptake has been utilized in the development of PET imaging for cancer. However, despite a deepened understanding of the molecular underpinnings of glucose metabolism in cancer, this fundamental difference between normal and malignant tissue has yet to be employed in targeted cancer therapy in the clinic. In this review, we highlight attempts in the recent literature to target cancer cell metabolism and elaborate on the challenges and controversies of these strategies in general and in the context of tumor cell heterogeneity in cancer.
- Published
- 2015
30. Radioresistance of the breast tumor is highly correlated to its level of cancer stem cell and its clinical implication for breast irradiation
- Author
-
Frank Pajonk, Patrick A. Kupelian, Michael L. Steinberg, Ke Sheng, Xiangrong Sharon Qi, Daniel A. Low, and Susan A. McCloskey
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Cell Survival ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Tumor cells ,Breast Neoplasms ,Biology ,Radiation Tolerance ,Article ,Breast tumor ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Breast cancer cell line ,Cancer stem cell ,Cell Line, Tumor ,Radioresistance ,Internal medicine ,Tumor cell ,Breast Cancer ,medicine ,Humans ,Linear-quadratic model ,Radiology, Nuclear Medicine and imaging ,Radiosensitivity ,Breast ,Oncology & Carcinogenesis ,Dose Fractionation ,Cancer ,Tumor ,Radiation ,Hematology ,medicine.disease ,Stem Cell Research ,Radiation therapy ,Other Physical Sciences ,030104 developmental biology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Female ,Dual-compartment model ,Stem Cell Research - Nonembryonic - Non-Human ,Dose Fractionation, Radiation - Abstract
Background and purpose Growing evidence suggested the coexistence of cancer stem cells (CSCs) within solid tumors. We aimed to study radiosensitivity parameters for the CSCs and differentiated tumor cells (TCs) and the correlation of the fractions of CSCs to the overall tumor radioresistance. Material and methods Surviving fractions of breast cancer cell lines were analyzed using a dual-compartment Linear-quadratic model with independent fitting parameters: radiosensitive αTC, βTC, αCSC, βCSC, and fraction of CSCs f. The overall tumor radio-resistance, the biological effective doses and tumor control probability were estimated as a function of CSC fraction for different fractionation regimens. The pooled clinical outcome data were fitted to the single- and dual-compartment linear-quadric models. Results CSCs were more radioresistant characterized by smaller α compared to TCs: αTC = 0.1 ± 0.2, αCSC = 0.04 ± 0.07 for MCF-7 (f = 0.1%), αTC = 0.08 ± 0.25, αCSC = 0.04 ± 0.18 for SUM159PT (f = 2.46%). Higher f values were correlated with increasing radioresistance in cell lines. Analysis of clinical outcome data is in accordance of a dual-compartment CSC model prediction. Higher percentage of BCSCs resulted in more overall tumor radioresistance and less biological effectiveness. Conclusions Percentage of CSCs strongly correlated to overall tumor radioresistance. This observation suggested potential individualized radiotherapy to account for heterogeneous population of CSCs and their distinct radiosensitivity for breast cancer.
- Published
- 2017
31. The RNA-Binding Protein Musashi-1 Regulates Proteasome Subunit Expression in Breast Cancer- and Glioma-Initiating Cells
- Author
-
Mabel Chan, Chann Lagadec, Frank Pajonk, Erina Vlashi, Yazeed Alhiyari, and Patricia Frohnen
- Subjects
Proteasome Endopeptidase Complex ,Notch signaling pathway ,Down-Regulation ,Breast Neoplasms ,Nerve Tissue Proteins ,Cell Growth Processes ,Biology ,Transfection ,Article ,Downregulation and upregulation ,Cancer stem cell ,Cell Line, Tumor ,Humans ,RNA, Messenger ,RNA, Small Interfering ,RNA-Binding Proteins ,Glioma ,Cell Biology ,Cell biology ,CCAAT-Binding Factor ,Notch proteins ,Hes3 signaling axis ,Cancer cell ,Neoplastic Stem Cells ,NUMB ,Molecular Medicine ,Female ,Stem cell ,Signal Transduction ,Developmental Biology - Abstract
Cancer stem cells (CSCs) or tumor-initiating cells, similar to normal tissue stem cells, rely on developmental pathways, such as the Notch pathway, to maintain their stem cell state. One of the regulators of the Notch pathway is Musashi-1, a mRNA-binding protein. Musashi-1 promotes Notch signaling by binding to the mRNA of Numb, the negative regulator of Notch signaling, thus preventing its translation. CSCs have also been shown to downregulate their 26S proteasome activity in several types of solid tumors, thus making them resistant to proteasome-inhibitors used as anticancer agents in the clinic. Interestingly, the Notch pathway can be inhibited by proteasomal degradation of the Notch intracellular domain (Notch-ICD); therefore, downregulation of the 26S proteasome activity can lead to stabilization of Notch-ICD. Here, we present evidence that the downregulation of the 26S proteasome in CSCs constitutes another level of control by which Musashi-1 promotes signaling through the Notch pathway and maintenance of the stem cell phenotype of this subpopulation of cancer cells. We demonstrate that Musashi-1 mediates the downregulation of the 26S proteasome by binding to the mRNA of NF-YA, the transcriptional factor regulating 26S proteasome subunit expression, thus providing an additional route by which the degradation of Notch-ICD is prevented, and Notch signaling is sustained. Stem Cells 2014;32:135–144
- Published
- 2014
32. Effects of Brain Irradiation in Immune-Competent and Immune-Compromised Mouse Models
- Author
-
Kruttika Bhat, Nhan T. Nguyen, Mohammad Saki, Harley I. Kornblum, Frank Pajonk, and Sirajbir S. Sodhi
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Combination therapy ,Biophysics ,Article ,Luxol fast blue stain ,030218 nuclear medicine & medical imaging ,Ionizing radiation ,Immunocompromised Host ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Radiation ,biology ,Brain ,Dose-Response Relationship, Radiation ,Histology ,030220 oncology & carcinogenesis ,Toxicity ,biology.protein ,NeuN ,Antibody ,Immunocompetence - Abstract
Patient-derived orthotopic xenografts (PDOXs) closely recapitulate primary human glioblastoma (GBM) tumors in terms of histology and genotype. Compared to other mouse strains, NOD-scid IL2Rgamma(null) (NSG) mice show excellent tumor take rates, which makes them an ideal host for PDOXs. However, NSG mice harbor a mutation in the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs), which renders them relatively radiosensitive. This has been a frequently voiced concern in studies involving ionizing radiation. In this study, we assessed brain toxicity in NSG mice compared to three other different mouse strains frequently used in radiation studies at radiation doses commonly used in experimental combination therapy studies. C3H/Sed/Kam, C57Bl/6, nude and NOD-scid IL2Rgamma(null) mice received a single dose of 4 Gy to the right brain hemispheres using an image-guided small animal irradiator. Brains were stained using H&E, luxol fast blue, and antibodies against IBA1 and GFAP one, two, four or six months postirradiation. Additional animals of all four strains were exposed to five daily fractions of 2 Gy (5 × 2 Gy), and tissue sections were stained 72 h later against gH2AX, NeuN, GFAP and IBA1. None of the mouse strains displayed radiation-induced toxicity at any of the time points tested. Radiation doses relevant for testing combination therapies can be safely applied to the brains of NSG mice without the occurrence of radiation-induced normal tissue toxicity.
- Published
- 2019
33. Targeted elimination of breast cancer cells with low proteasome activity is sufficient for tumor regression
- Author
-
Chann Lagadec, Erina Vlashi, Mabel Chan, Alexandra Jean McDonald, Patricia Frohnen, and Frank Pajonk
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Down-Regulation ,Breast Neoplasms ,Biology ,Article ,Mice ,Breast cancer ,Cancer stem cell ,In vivo ,Cell Line, Tumor ,Spheroids, Cellular ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Cancer ,medicine.disease ,Fusion protein ,Molecular biology ,In vitro ,Tumor Burden ,Enzyme Activation ,Disease Models, Animal ,Oncology ,Proteasome ,Neoplastic Stem Cells ,Cancer research ,Heterografts ,Female ,Stem cell - Abstract
Breast cancers are thought to be organized hierarchically with a small number of breast cancer stem cells (BCSCs), able to regrow a tumor after sublethal treatment while their progeny lack this feature. Furthermore, BCSCs are highly resistant to conventional anticancer treatments. According to the cancer stem cell hypothesis, all cancer stem cells in a tumor have to be eliminated to achieve cancer cure. In this study we tested if targeted elimination of BCSCs leads to tumor regression. Specific targeting of BCSCs was achieved via a unique imaging and targeting system that relies on their low proteasome activity. In our system breast cancer cells stably express a fluorescent fusion protein, thymidine kinase-ZsGreen-cODC, which is readily degraded after translation in cells with normal 26S proteasome activity. However, cells with low proteasome activity accumulate this fluorescent fusion protein, thus allowing for their identification, tracking, and specific elimination. Here, we show that the activity of the 26S proteasome was significantly down-regulated in MCF-7, T47D, and MDA-MB-231 cultures enriched for BCSCs. Treatment with ganciclovir resulted in abrogation of sphere formation in vitro, and tumor regression in vivo, thus demonstrating that targeted elimination of BCSCs leads to loss of self-renewal in vitro and tumor regression in vivo. We conclude that specific targeting of BCSCs could be a useful strategy to improve treatment outcome.
- Published
- 2013
34. National Institutes of Health Funding in Radiation Oncology: A Snapshot
- Author
-
Frank Pajonk, Michael L. Steinberg, Erina Vlashi, and William H. McBride
- Subjects
Cancer Research ,medicine.medical_specialty ,Medical education ,Radiation ,Descriptive statistics ,business.industry ,education ,Alternative medicine ,MEDLINE ,Work force ,Small field ,Fiscal year ,Oncology ,Radiation oncology ,medicine ,Radiology, Nuclear Medicine and imaging ,business ,health care economics and organizations ,Health funding - Abstract
Currently, pay lines for National Institutes of Health (NIH) grants are at a historical low. In this climate of fierce competition, knowledge about the funding situation in a small field like radiation oncology becomes very important for career planning and recruitment of faculty. Unfortunately, these data cannot be easily extracted from the NIH's database because it does not discriminate between radiology and radiation oncology departments. At the start of fiscal year 2013 we extracted records for 952 individual grants, which were active at the time of analysis from the NIH database. Proposals originating from radiation oncology departments were identified manually. Descriptive statistics were generated using the JMP statistical software package. Our analysis identified 197 grants in radiation oncology. These proposals came from 134 individual investigators in 43 academic institutions. The majority of the grants (118) were awarded to principal investigators at the full professor level, and 122 principal investigators held a PhD degree. In 79% of the grants, the research topic fell into the field of biology, 13% in the field of medical physics. Only 7.6% of the proposals were clinical investigations. Our data suggest that the field of radiation oncology is underfunded by the NIH and that the current level of support does not match the relevance of radiation oncology for cancer patients or the potential of its academic work force.
- Published
- 2013
35. P1-04-06: Ionizing Radiation Reprograms Non-Tumorigenic Cancer Cells into Cancer Stem Cells
- Author
-
Erina Vlashi, L Chann, Carmen Dekmezian, DD Lorenza, and Frank Pajonk
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Oncology ,business.industry ,Cancer stem cell ,Cancer cell ,Medicine ,business ,Ionizing radiation - Abstract
Breast cancers are thought to be organized hierarchically with a small number of breast cancer stem cells (BCSCs) able to re-grow a tumor while their progeny lack this feature. BCSCs in breast cancer have been found to be relatively resistant to radiation and several groups reported enrichment for BCSCs when breast cancers are subjected to classical anticancer treatment. Differentiation of BCSCs is thought to be unidirectional but an alternative model assumes that stemness can be obtained by clonal evolution. In this study, we quantified the number of BCSCs surviving after radiation treatment. We compared the number of surviving BCSCs to the expected number and found an increase in BCSCs after irradiation that could not be explained by current models. We propose that radiation induces a BCSC phenotype in previously non-BCSCs and show that this transition is Notch-dependent and coincided with up-regulation of the transcription factors Oct4, Sox-2, Nanog, and Klf4. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P1-04-06.
- Published
- 2011
36. Metabolic state of glioma stem cells and nontumorigenic cells
- Author
-
Erina Vlashi, Lorenza Della Donna, Kenta Masui, Carmen Dekmezian, Paul S. Mischel, Ruxandra Popescu, Tomoo Matsutani, Patrick D. Evers, Heather R. Christofk, Maria Poulou, Karen Reue, Frank Pajonk, Chann Lagadec, and Laurent Vergnes
- Subjects
Proteasome Endopeptidase Complex ,endocrine system ,Cellular differentiation ,Blotting, Western ,Deoxyglucose ,Biology ,PKM2 ,Adenosine Triphosphate ,Oxygen Consumption ,Cell Line, Tumor ,Glioma ,medicine ,Humans ,Glycolysis ,Progenitor cell ,Multidisciplinary ,Uncoupling Agents ,Stem Cells ,fungi ,Biological Sciences ,Cell cycle ,medicine.disease ,Immunohistochemistry ,Clone Cells ,Cell biology ,Glucose ,Tissue Array Analysis ,Positron-Emission Tomography ,Cancer cell ,Lactates ,Neoplastic Stem Cells ,Oligomycins ,Stem cell ,Energy Metabolism ,Reactive Oxygen Species - Abstract
Gliomas contain a small number of treatment-resistant glioma stem cells (GSCs), and it is thought that tumor regrowth originates from GSCs, thus rendering GSCs an attractive target for novel treatment approaches. Cancer cells rely more on glycolysis than on oxidative phosphorylation for glucose metabolism, a phenomenon used in 2-[ 18 F]fluoro-2-deoxy- d -glucose positron emission tomography imaging of solid cancers, and targeting metabolic pathways in cancer cells has become a topic of considerable interest. However, if GSCs are indeed important for tumor control, knowledge of the metabolic state of GSCs is needed. We hypothesized that the metabolism of GSCs differs from that of their progeny. Using a unique imaging system for GSCs, we assessed the oxygen consumption rate, extracellular acidification rate, intracellular ATP levels, glucose uptake, lactate production, PKM1 and PKM2 expression, radiation sensitivity, and cell cycle duration of GSCs and their progeny in a panel of glioma cell lines. We found GSCs and progenitor cells to be less glycolytic than differentiated glioma cells. GSCs consumed less glucose and produced less lactate while maintaining higher ATP levels than their differentiated progeny. Compared with differentiated cells, GSCs were radioresistant, and this correlated with a higher mitochondrial reserve capacity. Glioma cells expressed both isoforms of pyruvate kinase, and inhibition of either glycolysis or oxidative phosphorylation had minimal effect on energy production in GSCs and progenitor cells. We conclude that GSCs rely mainly on oxidative phosphorylation. However, if challenged, they can use additional metabolic pathways. Therefore, targeting glycolysis in glioma may spare GSCs.
- Published
- 2011
37. Marizomib, a Proteasome Inhibitor for All Seasons: Preclinical Profile and a Framework for Clinical Trials
- Author
-
Irene M. Ghobrial, KC Anderson, Saskia T. C. Neuteboom, E. Valashi, G. K. Lloyd, William H. McBride, Paul G. Richardson, Barbara C. M. Potts, Joya Chandra, Huib Ovaa, Michael A. Palladino, Callum M. Sloss, Michael Groll, William Fenical, Aldo M. Roccaro, Frank Pajonk, Anas Younes, Y. Oki, James C. Cusack, Paul R. Jensen, Celia R. Berkers, Claudia P. Miller, M. X. Albitar, Stavroula Baritaki, Kin S. Lam, David J. McConkey, Ben Bonavida, D Chauhan, and Matthew A. Spear
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Combination therapy ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,Pharmacology ,Article ,Lactones ,chemistry.chemical_compound ,Therapeutic index ,Neoplasms ,hemic and lymphatic diseases ,Drug Discovery ,medicine ,Animals ,Humans ,Protease Inhibitors ,Pyrroles ,Multiple myeloma ,Lenalidomide ,Bortezomib ,business.industry ,medicine.disease ,Oncology ,chemistry ,Cancer research ,Proteasome inhibitor ,Mantle cell lymphoma ,business ,Proteasome Inhibitors ,Salinosporamide A ,medicine.drug - Abstract
The proteasome has emerged as an important clinically relevant target for the treatment of hematologic malignancies. Since the Food and Drug Administration approved the first-in-class proteasome inhibitor bortezomib (Velcade®) for the treatment of relapsed/refractory multiple myeloma (MM) and mantle cell lymphoma, it has become clear that new inhibitors are needed that have a better therapeutic ratio, can overcome inherent and acquired bortezomib resistance and exhibit broader anti-cancer activities. Marizomib (NPI-0052; salinosporamide A) is a structurally and pharmacologically unique β-lactone-γ-lactam proteasome inhibitor that may fulfill these unmet needs. The potent and sustained inhibition of all three proteolytic activities of the proteasome by marizomib has inspired extensive preclinical evaluation in a variety of hematologic and solid tumor models, where it is efficacious as a single agent and in combination with biologics, che-motherapeutics and targeted therapeutic agents. Specifically, marizomib has been evaluated in models for multiple myeloma, mantle cell lymphoma, Waldenstrom’s macroglobulinemia, chronic and acute lymphocytic leukemia, as well as glioma, colorectal and pancreatic cancer models, and has exhibited synergistic activities in tumor models in combination with bortezomib, the immunomodulatory agent lenalidomide (Revlimid®), and various histone deacetylase inhibitors. These and other studies provided the framework for ongoing clinical trials in patients with MM, lymphomas, leukemias and solid tumors, including those who have failed bortezomib treatment, as well as in patients with diagnoses where other proteasome inhibitors have not demonstrated significant efficacy. This review captures the remarkable translational studies and contributions from many collaborators that have advanced marizomib from seabed to bench to bedside.
- Published
- 2011
38. Abstract PD02-10: Radiation Induces Notch-Dependent De Novo Generation of Breast Cancer Stem Cells
- Author
-
Frank Pajonk, Erina Vlashi, Carmen Dekmezian, L. Della Donna, and Chann Lagadec
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Wnt signaling pathway ,Notch signaling pathway ,Cancer ,Biology ,medicine.disease ,Notch proteins ,SOX2 ,Cancer stem cell ,Internal medicine ,medicine ,Cancer research ,Stem cell ,Notch 2 - Abstract
Like in normal tissues, the self-renewal of cancer stem cells (CSCs) might be also under tight control of developmental pathway like the Notch, Wnt, Sonic Hedgehog or TGFβ pathways. The Notch pathway plays an important role in normal breast development, cell fate, and normal stem cell self-renewal, and its deregulation has been shown to play a role in cancer. Aberrant Notch signaling has been implicated in the development and progression of both preinvasive ductal carcinomas in situ and invasive. Interestingly, in breast cancer, the Notch pathway plays major role for CSCs maintenance. We previously published that BCSCs (Breast Cancer Stem Cells) are more resistant to radiation. Here we demonstrate a link between BCSCs radio-resistance and the Notch pathway. We show that irradiation of MCF-7 cells increases CSC numbers and that this correlates with an induction of Notch signaling proteins expression in a dose and time specific manner. Jagged 1 was quickly (1h) increased 28-fold after 2Gy, DLL1 was increased 15-fold after 3 to 6h of 2 and 4 Gy irradiation, and Notch 2 was increased 16-fold 6h after 2, 4, 6 or 8 Gy, while DLL3 was increased 10-fold after the highest doses (6 to 12 Gy). Inhibition of Notch signaling pathway by the γ-secretase inhibitor prevents enrichment for CSCs and reduces radiation-induced overexpression of Notch proteins. More interestingly, we demonstrated that radiation-induced Notch signaling contributes to the phenotype plasticity of BCSCs and their progeny. Therefore, ionizing radiation induces de novo generation of BCSCs from non-tumorigenic cells. Moreover, we also identified, by FACS analysis, an induction of polyploid CSCs. expressed a relatively higher level of Oct4 and Sox2 than the unirradiated non-CSCs (two key transcription factors involved in stemness maintenance). In summary, we provide evidence that radiation-induced Notch signaling contributes to the plasticity of the BCSC phenotype, thereby generating BCSCs de novo from non-tumorigenic cells. Our data suggest that stochastic as well hierarchical CSC models apply to breast cancer. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr PD02-10.
- Published
- 2010
39. Radiation Resistance of Cancer Stem Cells: The 4 R's of Radiobiology Revisited
- Author
-
William H. McBride, Erina Vlashi, and Frank Pajonk
- Subjects
education.field_of_study ,Hypoxic tumor ,Radiobiology ,DNA Repair ,DNA repair ,DNA damage ,Population ,Cell Biology ,Cell cycle ,Biology ,Radiation Tolerance ,Article ,Toxicology ,Cancer stem cell ,Neoplastic Stem Cells ,Cancer research ,Animals ,Humans ,Molecular Medicine ,Repopulation ,education ,Oxidation-Reduction ,DNA Damage ,Developmental Biology - Abstract
There is compelling evidence that many solid cancers are organized hierarchically and contain a small population of cancer stem cells (CSCs). It seems reasonable to suggest that a cancer cure can be achieved only if this population is eliminated. Unfortunately, there is growing evidence that CSCs are inherently resistant to radiation, and perhaps other cancer therapies. In general, success or failure of standard clinical radiation treatment is determined by the 4 R's of radiobiology: repair of DNA damage, redistribution of cells in the cell cycle, repopulation, and reoxygenation of hypoxic tumor areas. We relate recent findings on CSCs to these four phenomena and discuss possible consequences.
- Published
- 2010
40. Postoperative Pterygium Prevention by Radiotherapy with Strontium-90 Beta-Rays
- Author
-
J Slanina, H Mittelviefhaus, Frank Pajonk, and H Flick
- Subjects
Disease free survival ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Brachytherapy ,Follow up studies ,Strontium Radioisotopes ,Retrospective cohort study ,medicine.disease ,Pterygium ,Surgery ,Radiation therapy ,Medicine ,business - Published
- 2015
41. The Response of CD24 −/low /CD44 + Breast Cancer–Initiating Cells to Radiation
- Author
-
Tiffany M. Phillips, Frank Pajonk, and William H. McBride
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Cell Survival ,medicine.medical_treatment ,Population ,Breast Neoplasms ,Radiation Tolerance ,Flow cytometry ,Histones ,Andrology ,Breast cancer ,Spheroids, Cellular ,Radioresistance ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Humans ,Serrate-Jagged Proteins ,Phosphorylation ,Receptor, Notch1 ,education ,education.field_of_study ,biology ,medicine.diagnostic_test ,Calcium-Binding Proteins ,CD44 ,Dose fractionation ,CD24 Antigen ,Membrane Proteins ,Cancer ,Flow Cytometry ,medicine.disease ,Radiation therapy ,Hyaluronan Receptors ,Oncology ,Neoplastic Stem Cells ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Female ,Dose Fractionation, Radiation ,Reactive Oxygen Species ,Signal Transduction - Abstract
Background: If cancer arises and is maintained by a small population of cancer-initiating cells within every tumor, understanding how these cells react to cancer treatment will facilitate improvement of cancer treatment in the future. Cancer-initiating cells can now be prospectively isolated from breast cancer cell lines and tumor samples and propagated as mammospheres in vitro under serum-free conditions. Methods: CD24 − /low /CD44 + cancer-initiating cells were isolated from MCF-7 and MDA-MB-231 breast cancer monolayer cultures and propagated as mammospheres. Their response to radiation was investigated by assaying clonogenic survival and by measuring reactive oxygen species (ROS) levels, phosphorylation of the replacement histone H2AX, CD44 levels, CD24 levels, and Notch-1 activation using fl ow cytometry. All statistical tests were two-sided. Results: Cancer-initiating cells were more resistant to radiation than cells grown as monolayer cultures (MCF-7: monolayer cultures, mean surviving fraction at 2 Gy [SF 2Gy ] = 0.2, versus mammospheres, mean SF 2Gy = 0.46, difference = 0.26, 95% confi dence interval [CI] = 0.05 to 0.47; P = .026; MDA-MB-231: monolayer cultures, mean SF 2Gy = 0.5, versus mammospheres, mean SF 2Gy = 0.69, difference = 0.19, 95% CI = − 0.07 to 0.45; P = .09). Levels of ROS increased in both mammospheres and monolayer cultures after irradiation with a single dose of 10 Gy but were lower in mammospheres than in monolayer cultures (MCF-7 monolayer cultures: 0 Gy, mean = 1.0, versus 10 Gy, mean = 3.32, difference = 2.32, 95% CI = 0.67 to 3.98; P = .026; mammospheres: 0 Gy, mean = 0.58, versus 10 Gy, mean = 1.46, difference = 0.88, 95% CI = 0.20 to 1.56; P = .031); phosphorylation of H2AX increased in irradiated monolayer cultures, but no change was observed in mammospheres. Fractionated doses of irradiation increased activation of Notch-1 (untreated, mean = 10.7, versus treated, mean = 15.1, difference = 4.4, 95% CI = 2.7 to 6.1, P = .002) and the percentage of the cancer stem/initiating cells in the nonadherent cell population of MCF-7 monolayer cultures (untreated, mean = 3.52%, versus treated, mean = 7.5%, difference = 3.98%, 95% CI = 1.67% to 6.25%, P = .009). Conclusions: Breast cancer – initiating cells are a relatively radioresistant subpopulation of breast cancer cells and increase in numbers after short courses of fractionated irradiation. These fi ndings offer a possible mechanism for the accelerated repopulation of tumor cells observed during gaps in radiotherapy. [J Natl Cancer Inst 2006;98: 1777 – 85 ]
- Published
- 2006
42. Safety and Efficacy of Hyperbaric Oxygen Therapy for the Treatment of Interstitial Cystitis: A Randomized, Sham Controlled, Double-Blind Trial
- Author
-
Gordon Rossbach, Lothar Hertle, Frank Pajonk, and Arndt van Ophoven
- Subjects
Adult ,Male ,Visual analogue scale ,Urology ,Cystitis, Interstitial ,law.invention ,Patient satisfaction ,Double-Blind Method ,Randomized controlled trial ,law ,medicine ,Humans ,Aged ,Hyperbaric Oxygenation ,business.industry ,Pelvic pain ,Interstitial cystitis ,Middle Aged ,medicine.disease ,Clinical trial ,Treatment Outcome ,Complex regional pain syndrome ,Patient Satisfaction ,Anesthesia ,Breathing ,Feasibility Studies ,Female ,medicine.symptom ,business ,Follow-Up Studies - Abstract
We conducted a double-blind, sham controlled study to evaluate the safety, efficacy and feasibility of hyperbaric oxygenation for interstitial cystitis.A total of 21 patients with interstitial cystitis were randomized to 90 minutes treatment in a hyperbaric chamber pressurized with 100% O2 to 2.4 atmosphere absolute for 30 treatments sessions or 1.3 atmosphere absolute, breathing normal air in the control group. Moderate or marked improvement in a global response assessment questionnaire was defined as treatment response (primary outcomes). Secondary measurements included changes of pain and urgency evaluated by visual analog scales, functional bladder capacity and frequency. Changes in the O'Leary-Sant Interstitial Cystitis Index and rating of overall satisfaction with the therapeutic outcome were also reported.There were 3 of 14 patients on verum and no control patients who were identified as responders (p0.52) [corrected] At 12-month followup 3 patients (21.4%) still reported treatment response. Hyperbaric oxygenation resulted in a decrease of baseline urgency intensity from 60.2 +/- 15.0 to 49.9 +/- 35.2 mm at 3 months and decrease of pain intensity from 43.1 +/- 20.5 to 31.2 +/- 19.8 mm, respectively (p0.05). The Interstitial Cystitis Symptom Index score sum decreased from 25.7 to 19.9 points in patients on verum. Sham treatment did not result in improvement of the baseline parameters.A total of 30 treatment sessions of hyperbaric oxygenation appear to be a safe, effective and feasible therapeutic approach to interstitial cystitis. In the treatment responders application of hyperbaric oxygenation resulted in a sustained decrease of interstitial cystitis symptoms with a discordant profile regarding the peak amelioration of the various interstitial cystitis symptoms compared with a normobaric, normoxic sham treatment.
- Published
- 2006
43. Hyperthermia-Induced Proteasome Inhibition and Loss of Androgen Receptor Expression in Human Prostate Cancer Cells
- Author
-
Frank Pajonk, William H. McBride, and Arndt van Ophoven
- Subjects
Male ,Hyperthermia ,Proteasome Endopeptidase Complex ,Cancer Research ,medicine.medical_specialty ,Programmed cell death ,Down-Regulation ,Apoptosis ,Biology ,Heat Stress Disorders ,Mice ,Prostate cancer ,Cell Line, Tumor ,Internal medicine ,LNCaP ,medicine ,Animals ,Humans ,NF-kappa B ,Prostatic Neoplasms ,Hyperthermia, Induced ,medicine.disease ,Androgen receptor ,Endocrinology ,Oncology ,Proteasome ,Receptors, Androgen ,Cancer cell ,Cancer research ,Signal transduction ,Proteasome Inhibitors ,Signal Transduction - Abstract
Prostate cancer is the second leading cause of death in men in western countries and is usually treated by surgery and/or radiotherapy. More recently, hyperthermia has been introduced into clinical trials investigating a possible effect in the first-line treatment of prostate cancer. However, the molecular mechanisms of hyperthermia are not completely understood. In this study, we investigated the effects of hyperthermia on proteasome function and its significance for signal transduction, cell death and androgen receptor (AR) expression in PC-3, LnCaP, and DU-145 human and TRAMP-C2 murine prostate cancer cells. Hyperthermia caused apoptosis and radiosensitization and decreased 26S proteasome activity in all three human cell lines to about 40% of untreated control cells. 20S proteasome activity was not affected by heat. Heat treatment inhibited constitutive and radiation-induced activation of nuclear factor κB caused by stabilization of IκB. Although stabilization of AR by proteasome inhibitors has been reported previously, AR protein levels in LnCaP cells decreased dramatically after heat. Our data suggest that inhibition of proteasome function and dependent signal transduction pathways might be a major molecular mechanisms of heat-induced apoptosis and radiosensitization. Hyperthermia abrogates AR expression in androgen-dependent cells and might thus promote malignant progression of prostate cancer.
- Published
- 2005
44. Hypericin?an inhibitor of proteasome function
- Author
-
Frank Pajonk, Jutta Scholber, and B Fiebich
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Programmed cell death ,Electrophoretic Mobility Shift Assay ,Mammary Neoplasms, Animal ,Pharmacology ,Toxicology ,chemistry.chemical_compound ,Tumor Cells, Cultured ,Humans ,Pharmacology (medical) ,Perylene ,Caspase ,Anthracenes ,biology ,NF-kappa B ,Hypericum perforatum ,NF-κB ,Glioma ,Antidepressive Agents ,Hypericin ,IκBα ,Oncology ,Proteasome ,chemistry ,Biochemistry ,Apoptosis ,biology.protein ,Proteasome Inhibitors - Abstract
Hypericin is the presumed active moiety within Saint John's wort. Extracts of Saint John's wort are widely used as an effective treatment for depression. Available as "over-the-counter" drugs, they are frequently part of the self-medication of patients undergoing radiation therapy for malignant diseases. In addition to antidepressive properties, hypericin has been shown to be able to induce apoptosis and radiosensitize tumor cells, and to have antiinflammatory and phototoxic skin effects. However, the underlying mechanisms are not clear. In this study, we investigated possible inhibitory effects of hypericin on proteasome function and related pathways. Extracts from U373 human glioma cells were incubated with different concentrations of hypericin. Three proteasome activities were monitored using a fluorogenic peptide assay. Activity of the transcription factor NF-kappaB and protein levels of p65, p50, IkappaBalpha and caspase-3 were investigated by EMSA and Western blotting, respectively. Hypericin caused a dose-dependent and photoactivation-independent inhibition of proteasome function. Hypericin treatment (6.25-50 microM) inhibited NF-kappaB, caused accumulation of phosphorylated IkappaBalpha, decreased p50 protein levels and induced cleavage of p65 protein in U373 cells. These effects were observed in MCF-7 cells only at higher concentrations of hypericin (12.5-50 microM). Additionally, inhibition of NF-kappaB activity in U373 cells by hypericin was prevented by caspase inhibition. Although hypericin clearly inhibits proteasome function, its effect NF-kappaB DNA-binding activity was not exclusively proteasome-dependent. The underlying mechanism might also involve caspase activation, a consequence of proteasome inhibition.
- Published
- 2005
45. A Sense of Danger from Radiation1
- Author
-
Chun-Chieh Wang, Graeme J. Dougherty, Jennifer L. Olson, William H. McBride, Yu-Pei Liao, Ji-Hong Hong, Chi-Shiun Chiang, Frank Pajonk, Milena Pervan, and Keisuke S. Iwamoto
- Subjects
Radiation exposure ,Intrusion ,Radiation ,Physical agents ,Bone marrow transplantation ,Radiation tolerance ,Tissue damage ,Immunology ,Biophysics ,Radiology, Nuclear Medicine and imaging ,Biology ,Tissue repair ,Ionizing radiation - Abstract
McBride, W. H., Chiang, C-S., Olson, J. L., Wang, C-C., Hong, J-H., Pajonk, F., Dougherty, G. J., Iwamoto, K. S., Pervan, M. and Liao, Y-P. A Sense of Danger from Radiation. Radiat. Res. 162, 1–19 (2004). Tissue damage caused by exposure to pathogens, chemicals and physical agents such as ionizing radiation triggers production of generic “danger” signals that mobilize the innate and acquired immune system to deal with the intrusion and effect tissue repair with the goal of maintaining the integrity of the tissue and the body. Ionizing radiation appears to do the same, but less is known about the role of “danger” signals in tissue responses to this agent. This review deals with the nature of putative “danger” signals that may be generated by exposure to ionizing radiation and their significance. There are a number of potential consequences of “danger” signaling in response to radiation exposure. “Danger” signals could mediate the pathogenesis of, or recovery from, radiation damage. They could alte...
- Published
- 2004
46. The role of the ubiquitin/proteasome system in cellular responses to radiation
- Author
-
William H. McBride, Keisuke S. Iwamoto, Randi G. Syljuåsen, Milena Pervan, and Frank Pajonk
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Programmed cell death ,Cell cycle checkpoint ,DNA repair ,Apoptosis ,Biology ,medicine.disease_cause ,Adjuvants, Immunologic ,Ubiquitin ,Multienzyme Complexes ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Cycle ,Cell cycle ,Cell biology ,Cysteine Endopeptidases ,Oxidative Stress ,Proteasome ,Biochemistry ,biology.protein ,Signal transduction ,Carcinogenesis - Abstract
In the last few years, the ubiquitin(Ub)/proteasome system has become increasingly recognized as a controller of numerous physiological processes, including signal transduction, DNA repair, chromosome maintenance, transcriptional activation, cell cycle progression, cell survival, and certain immune cell functions. This is in addition to its more established roles in the removal of misfolded, damaged, and effete proteins. This review examines the role of the Ub/proteasome system in processes underlying the classical effects of irradiation on cells, such as radiation-induced gene expression, DNA repair and chromosome instability, oxidative damage, cell cycle arrest, and cell death. Furthermore, recent evidence suggests that the proteasome is a redox-sensitive target for ionizing radiation and other oxidative stress signals. In other words, the Ub/proteasome system may not simply be a passive player in radiation-induced responses, but may modulate them. The extent of the modulation will be influenced by the functional and structural diversity that is expressed by the system. Cell types vary in the Ub/proteasome structures they possess and the level at which they function, and this changes as they go from the normal to the cancerous condition. Cancer-related functional changes within the Ub/proteasome system may therefore present unique targets for cancer therapy, especially when targeting agents are used in combination with radio- or chemotherapy. The peptide boronic acid compound PS-341, which was designed to inhibit proteasome chymotryptic activity, is in clinical trials for the treatment of solid and hematogenous tumors. It has shown some efficacy on its own and in combination with chemotherapy. Preclinical studies have shown that PS-341 will also potentiate the cytotoxic effects of radiation therapy. In addition, other drugs in common clinical use have been shown to affect proteasome function, and their activities may be valuably reconsidered from this perspective.
- Published
- 2003
47. Vitiligo at the Sites of Irradiation in a Patient with Hodgkin's Disease
- Author
-
Christian Weissenberger, Michael Henke, Frank Pajonk, and Gerlo Witucki
- Subjects
Adult ,medicine.medical_specialty ,Side effect ,medicine.medical_treatment ,Vitiligo ,Melanin ,Depigmentation ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,External beam radiotherapy ,skin and connective tissue diseases ,Pigmentation disorder ,Neoplasm Staging ,integumentary system ,business.industry ,Dose-Response Relationship, Radiation ,medicine.disease ,Hodgkin Disease ,Dermatology ,Radiation therapy ,Oncology ,Colorimetry ,Female ,Radiodermatitis ,medicine.symptom ,business ,Complication ,Follow-Up Studies - Abstract
Vitiligo is one of the most common skin disorders. However, the etiology of vitiligo is still unknown. Current hypotheses discuss autoimmune, autotoxic and neuronal mechanisms. Here we report the case of radiation-induced depigmentation of the skin of a patient with Hodgkin's disease and 25-year history of vitiligo. Patient and Method: We compared possible differences in skin color, skin moisture, microcirculation and skin elasticity between normal skin and skin exhibiting persistent depigmentation in a 37-year-old patient 40 months after completion of external beam radiotherapy. Results: Colormetrically we found a dose-dependent decrease of the red/green and yellow/blue saturation combined with an overall increase in brightness in depigmented skin when compared with normal skin. This was in agreement with a loss of melanocytes in vitiligo. Depigmentation was complete in areas receiving 40 Gy. Areas which received 30 Gy showed depigmentation only if the skin dose was increased by the loss of depth of the build-up dose region in areas with direct contact with the irradiation table. We could not show any change in skin moisture, microcirculation or skin elasticity. Conclusions: Complete radiation-induced depigmentation of skin from patients suffering from vitiligo is a side effect of radiation therapy. Patients should be informed about this side effect by the radiooncologist. Preventing the loss of depth of the build-up dose region might improve the cosmetic results of radiation therapy in patients with history of vitiligo.
- Published
- 2002
48. RBIO-06. NADPH OXIDASE (NOX) PROMOTES RADIATION RESISTANCE THROUGH OXIDATION OF PTEN IN GLIOBLASTOMA
- Author
-
Erina Vlashi, Kirsten Ludwig, Harley I. Kornblum, Frank Pajonk, Jantzen Sperry, and Janel E. Le Belle
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Cancer Research ,Programmed cell death ,Reactive oxygen species ,NADPH oxidase ,biology ,Cell growth ,Chemistry ,Mitochondrion ,Abstracts ,03 medical and health sciences ,030104 developmental biology ,Oncology ,biology.protein ,Cancer research ,PTEN ,Neurology (clinical) ,Radiation resistance ,NOx - Abstract
Radiation is a mainstay of treatment for glioblastoma (GBM). While all the effects of radiation are not known, it is thought to induce cell death by increasing the production of reactive oxygen species (ROS). High amounts of ROS are known to induce cell death, however recent data suggests that a moderate increase in ROS can promote proliferation, migration, tumorigenesis, and stem cell maintenance. We have found that in primary patient-derived gliomasphere lines both exogenous and endogenous ROS oxidizes PTEN, resulting in Akt activation and increased proliferation. Under hypoxic conditions, endogenous ROS is produced primarily by a complex called NADPH oxidase (NOX), whose formation results in PTEN oxidation and downstream activation of Akt and cell proliferation. Prior studies demonstrate that radiation itself can induce activation of Akt, however the mechanism is still unclear. In the present study, we tested the hypothesis that NOX-produced ROS plays a role in the response to radiation in GBM. We find that following radiation, NOX4 expression and activation increases, and chemical or genetic inhibition of this complex decreases radiation induced ROS, and rescues PTEN oxidation as well as Akt activation. Most importantly, NOX inhibition decreases cell viability and survival following radiation, suggesting that activation of this complex helps promote survival following radiation. While NOX inhibition did decrease ROS levels in PTEN null lines, no significant difference in survival or Akt activation was found, suggesting that NOX protection functions through the inactivation of PTEN. Finally, NOX-produced ROS occurs primarily in the cytosol, as no significant decrease in mitochondrial ROS levels were detected following NOX inhibition. Taken together, these data indicate that NOX may be a viable target to promote the sensitization of GBM to radiation, especially in PTEN-intact tumors.
- Published
- 2017
49. Cancer Stem Cells, Cancer Cell Plasticity and Radiation Therapy
- Author
-
Frank Pajonk and Erina Vlashi
- Subjects
Cancer Research ,Cell Survival ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Context (language use) ,Biology ,Regenerative Medicine ,Article ,Metastasis ,Clonal Evolution ,Stem Cell Research - Nonembryonic - Human ,Cancer stem cell ,Neoplasms ,medicine ,Biomarkers, Tumor ,Humans ,Tumor growth ,Oncology & Carcinogenesis ,Cancer ,Neoplastic ,Tumor ,Radiotherapy ,Cancer stem cells ,Reprogramming ,Cell Differentiation ,Stem Cell Research ,medicine.disease ,Tumor Burden ,Radiation therapy ,Gene Expression Regulation, Neoplastic ,Gene Expression Regulation ,Immunology ,Cancer cell ,Cancer research ,Neoplastic Stem Cells ,Stem Cell Research - Nonembryonic - Non-Human ,Repopulation ,Biomarkers - Abstract
Since the first prospective identification of cancer stem cells in solid cancers the cancer stem cell hypothesis has reemerged as a research topic of increasing interest. It postulates that solid cancers are organized hierarchically with a small number of cancer stem cells driving tumor growth, repopulation after injury and metastasis. They give rise to differentiated progeny, which lack these features. The model predicts that for any therapy to provide cure, all cancer stem cells have to be eliminated while the survival of differentiated progeny is less critical. In this review we discuss recent reports challenging the idea of a unidirectional differentiation of cancer cells. These reports provide evidence supporting the idea that non-stem cancer cells exhibit a remarkable degree of plasticity that allows them to re-acquire cancer stem cell traits, especially in the context of radiation therapy. We summarize conditions under which differentiation is reversed and discuss the current knowledge of the underlying mechanisms.
- Published
- 2014
50. Molecular Pathways That Modify Tumor Radiation Response
- Author
-
William H. McBride, H. Rodney Withers, Ji-Rong Sun, Frank Pajonk, and Milena Pervan
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Programmed cell death ,Leupeptins ,DNA repair ,Antineoplastic Agents ,Radiation Tolerance ,Bortezomib ,Ubiquitin ,Multienzyme Complexes ,In vivo ,Tumor Cells, Cultured ,Animals ,Humans ,Medicine ,Protease Inhibitors ,Ubiquitins ,biology ,business.industry ,Cell growth ,Drug Synergism ,Cell cycle ,Boronic Acids ,Cysteine Endopeptidases ,Oncology ,Proteasome ,Pyrazines ,Immunology ,biology.protein ,Cancer research ,Signal transduction ,business ,Signal Transduction - Abstract
Aberrant expression of signal transduction molecules in pathways controlling cell survival, proliferation, death, or differentiation are a common feature of all tumors. The identification of the molecules that are involved allows the development of novel tumor-specific strategies. Not surprisingly, targeting these pathways often also results in radiosensitization. The efficacy of such directed therapies may, however, be limited by the heterogeneity and the multiple mutations that are associated with the cancerous state. A more robust alternative may be to target global mechanisms of cellular control. The ubiquitin/proteasome degradation pathway is one candidate for such therapeutic intervention. This pathway is the main posttranscriptional mechanism that controls levels of many short-lived proteins involved in regulation of cell cycle progression, DNA transcription, DNA repair, and apoptosis. Many of these proteins are involved in various malignancies and/or radiation responses. In recent years, proteasome inhibitors have gained interest as a promising new group of antitumor drugs. PS-341, a reversible inhibitor of proteasome chymotryptic activity, is currently being tested in phase I clinical trials. In this study, we show that proteasome inhibition by PS-341 can alter cellular radiosensitivity in vitro and in vivo, in addition to having direct antitumor effects.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.