162 results on '"Koritzinsky M"'
Search Results
2. SP-0352 Targeting mitochondrial activity with metformin to reduce tumor hypoxia and increase radiation response in non-diabetic patients: Importance of patient selection
- Author
-
Koritzinsky, M., primary, Han, K., additional, Bruce, J., additional, and Milosevic, M., additional
- Published
- 2022
- Full Text
- View/download PDF
3. A pilot ‘window of opportunity’ neoadjuvant study of metformin in localised prostate cancer
- Author
-
Joshua, A M, Zannella, V E, Downes, M R, Bowes, B, Hersey, K, Koritzinsky, M, Schwab, M, Hofmann, U, Evans, A, van der Kwast, T, Trachtenberg, J, Finelli, A, Fleshner, N, Sweet, J, and Pollak, M
- Published
- 2014
- Full Text
- View/download PDF
4. Chemoradiation With or Without Metformin in Locally Advanced Cervical Cancer: Phase II Randomized Trial
- Author
-
Han, K., primary, Fyles, T.W., additional, Shek, T., additional, Croke, J.M., additional, Dhani, N., additional, D'Souza, D.P., additional, Lee, T.Y., additional, Chaudary, N., additional, Cairns, R., additional, Pintilie, M., additional, Vines, D., additional, Jaffray, D.A., additional, Metser, U., additional, Milosevic, M., additional, and Koritzinsky, M., additional
- Published
- 2021
- Full Text
- View/download PDF
5. Strategic Training in Transdisciplinary Radiation Science for the 21st Century (STARS21): 5-Year Prospective Evaluation of an Innovative Curriculum in Radiation Science
- Author
-
Patel, P., primary, Mustafa, S., additional, Liu, Z., additional, Harding, S., additional, Koritzinsky, M., additional, and Koch, C.A., additional
- Published
- 2021
- Full Text
- View/download PDF
6. SP-0458 Targeting metabolism to sensitize hypoxic tumor cells
- Author
-
Koritzinsky, M., primary
- Published
- 2019
- Full Text
- View/download PDF
7. Hypoxia as a target for combined modality treatments
- Author
-
Wouters, B.G, Weppler, S.A, Koritzinsky, M, Landuyt, W, Nuyts, S, Theys, J, Chiu, R.K, and Lambin, P
- Published
- 2002
- Full Text
- View/download PDF
8. Hypoxic Activation of the PERK/eIF2? Arm of the Unfolded Protein Response Promotes Metastasis through Induction of LAMP3
- Author
-
Mujcic, H., Nagelkerke, A., Rouschop, K. M. A., Chung, S., Chaudary, N., Span, P. N., Clarke, B., Milosevic, M., Sykes, J., Hill, R. P., Koritzinsky, M., and Wouters, B. G.
- Published
- 2013
- Full Text
- View/download PDF
9. Targeting Lactate Transporters MCT-1 and MCT-4 to Inhibit the Growth of Hypoxic HNSCC Cells In Vitro
- Author
-
Boasquevisque, P.H., primary, Schoeneberger, V., additional, Caporiccio, L., additional, Vellanki, R.N., additional, Koritzinsky, M., additional, and Wouters, B.G., additional
- Published
- 2017
- Full Text
- View/download PDF
10. SP-0514: A radiobiologist’s perspective
- Author
-
Koritzinsky, M., primary
- Published
- 2015
- Full Text
- View/download PDF
11. 204 High-throughput functional screening identifies the flavoreductase POR as a principal determinant of sensitivity to the hypoxia-targeting prodrug SN30000
- Author
-
Hunter, F.W., primary, Shalev, Z., additional, Wang, J., additional, Moffat, J., additional, Katella, T., additional, Koritzinsky, M., additional, Wilson, W.R., additional, and Wouters, B.G., additional
- Published
- 2014
- Full Text
- View/download PDF
12. Hypoxia Signaling and the Metastatic Phenotype
- Author
-
Mujcic, H., primary, Hill, R.P., additional, Koritzinsky, M., additional, and Wouters, B.G., additional
- Published
- 2014
- Full Text
- View/download PDF
13. 105: Reprogramming metabolism with metformin to improve radiation response
- Author
-
Koritzinsky, M., primary
- Published
- 2014
- Full Text
- View/download PDF
14. PERK/eIF2alpha signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS
- Author
-
Rouschop, K.M., Dubois, L.J., Keulers, T.G., Beucken, T. van den, Lambin, P., Bussink, J., Kogel, A.J. van der, Koritzinsky, M., Wouters, B.G., Rouschop, K.M., Dubois, L.J., Keulers, T.G., Beucken, T. van den, Lambin, P., Bussink, J., Kogel, A.J. van der, Koritzinsky, M., and Wouters, B.G.
- Abstract
Item does not contain fulltext, Hypoxia is a common feature of tumors and an important contributor to malignancy and treatment resistance. The ability of tumor cells to survive hypoxic stress is mediated in part by hypoxia-inducible factor (HIF)-dependent transcriptional responses. More severe hypoxia activates endoplasmatic reticulum stress responses, including the double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2alpha (eIF2alpha)-dependent arm of the unfolded protein response (UPR). Although several studies implicate important roles for HIF and UPR in adaption to hypoxia, their importance for hypoxic cells responsible for therapy resistance in tumors is unknown. By using isogenic models, we find that HIF and eIF2alpha signaling contribute to the survival of hypoxic cells in vitro and in vivo. However, the eIF2alpha-dependent arm of the UPR is uniquely required for the survival of a subset of hypoxic cells that determine tumor radioresistance. We demonstrate that eIF2alpha signaling induces uptake of cysteine, glutathione synthesis, and protection against reactive oxygen species produced during periods of cycling hypoxia. Together these data imply that eIF2alpha signaling is a critical contributor to the tolerance of therapy-resistant cells that arise as a consequence of transient changes in oxygenation in solid tumors and thus a therapeutic target in curative treatments for solid cancers.
- Published
- 2013
15. Deregulation of cap-dependent mRNA translation increases tumour radiosensitivity through reduction of the hypoxic fraction
- Author
-
Rouschop, K.M., Dubois, L., Schaaf, M.B., Beucken, T. van den, Lieuwes, N., Keulers, T.G., Savelkouls, K.G., Bussink, J., Kogel, A.J. van der, Koritzinsky, M., Wouters, B.G., Rouschop, K.M., Dubois, L., Schaaf, M.B., Beucken, T. van den, Lieuwes, N., Keulers, T.G., Savelkouls, K.G., Bussink, J., Kogel, A.J. van der, Koritzinsky, M., and Wouters, B.G.
- Abstract
Item does not contain fulltext, BACKGROUND AND PURPOSE: Tumour hypoxia is an important limiting factor in the successful treatment of cancer. Adaptation to hypoxia includes inhibition of mTOR, causing scavenging of eukaryotic initiation factor 4E (eIF4E), the rate-limiting factor for cap-dependent translation. The aim of this study was to determine the effect of preventing mTOR-dependent translation inhibition on hypoxic cell survival and tumour sensitivity towards irradiation. MATERIAL AND METHODS: The effect of eIF4E-overexpression on cell proliferation, hypoxia-tolerance, and radiation sensitivity was assessed using isogenic, inducible U373 and HCT116 cells. RESULTS: We found that eIF4E-overexpression significantly enhanced proliferation of cells under normal conditions, but not during hypoxia, caused by increased cell death during hypoxia. Furthermore, eIF4E-overexpression stimulated overall rates of tumour growth, but resulted in selective loss of hypoxic cells in established tumours and increased levels of necrosis. This markedly increased overall tumour sensitivity to irradiation. CONCLUSIONS: Our results demonstrate that hypoxia induced inhibition of translational control through regulation of eIF4E is an important mediator of hypoxia tolerance and radioresistance of tumours. These data also demonstrate that deregulation of metabolic pathways such as mTOR can influence the proliferation and survival of tumour cells experiencing metabolic stress in opposite ways of nutrient replete cells.
- Published
- 2011
16. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5.
- Author
-
Rouschop, K.M., Beucken, T. van den, Dubois, L., Niessen, H., Bussink, J., Savelkouls, K., Keulers, T., Mujcic, H., Landuyt, W., Voncken, J.W., Lambin, P., Kogel, A.J. van der, Koritzinsky, M., Wouters, B.G., Rouschop, K.M., Beucken, T. van den, Dubois, L., Niessen, H., Bussink, J., Savelkouls, K., Keulers, T., Mujcic, H., Landuyt, W., Voncken, J.W., Lambin, P., Kogel, A.J. van der, Koritzinsky, M., and Wouters, B.G.
- Abstract
Contains fulltext : 87690.pdf (publisher's version ) (Open Access), Tumor hypoxia is a common microenvironmental factor that adversely influences tumor phenotype and treatment response. Cellular adaptation to hypoxia occurs through multiple mechanisms, including activation of the unfolded protein response (UPR). Recent reports have indicated that hypoxia activates a lysosomal degradation pathway known as autophagy, and here we show that the UPR enhances the capacity of hypoxic tumor cells to carry out autophagy, and that this promotes their survival. In several human cancer cell lines, hypoxia increased transcription of the essential autophagy genes microtubule-associated protein 1 light chain 3beta (MAP1LC3B) and autophagy-related gene 5 (ATG5) through the transcription factors ATF4 and CHOP, respectively, which are regulated by PKR-like ER kinase (PERK, also known as EIF2AK3). MAP1LC3B and ATG5 are not required for initiation of autophagy but mediate phagophore expansion and autophagosome formation. We observed that transcriptional induction of MAP1LC3B replenished MAP1LC3B protein that was turned over during extensive hypoxia-induced autophagy. Correspondingly, cells deficient in PERK signaling failed to transcriptionally induce MAP1LC3B and became rapidly depleted of MAP1LC3B protein during hypoxia. Consistent with these data, autophagy and MAP1LC3B induction occurred preferentially in hypoxic regions of human tumor xenografts. Furthermore, pharmacological inhibition of autophagy sensitized human tumor cells to hypoxia, reduced the fraction of viable hypoxic tumor cells, and sensitized xenografted human tumors to irradiation. Our data therefore demonstrate that the UPR is an important mediator of the hypoxic tumor microenvironment and that it contributes to resistance to treatment through its ability to facilitate autophagy.
- Published
- 2010
17. Hypoxia-induced expression of carbonic anhydrase 9 is dependent on the unfolded protein response.
- Author
-
Beucken, T. van den, Koritzinsky, M., Niessen, H., Dubois, L., Savelkouls, K., Mujcic, H., Jutten, B., Kopacek, J., Pastorekova, S., Kogel, A.J. van der, Lambin, P., Voncken, W., Rouschop, K.M., Wouters, B.G., Beucken, T. van den, Koritzinsky, M., Niessen, H., Dubois, L., Savelkouls, K., Mujcic, H., Jutten, B., Kopacek, J., Pastorekova, S., Kogel, A.J. van der, Lambin, P., Voncken, W., Rouschop, K.M., and Wouters, B.G.
- Abstract
Contains fulltext : 80413.pdf (Publisher’s version ) (Open Access), Adaptation to tumor hypoxia is mediated in large part by changes in protein expression. These are driven by multiple pathways, including activation of the hypoxia inducible factor-1 (HIF-1) transcription factor and the PKR-like endoplasmic reticulum kinase PERK, a component of the unfolded protein response. Through gene expression profiling we discovered that induction of the HIF-1 target gene CA9 was defective in mouse embryo fibroblasts derived from mice harboring an eIF2alpha S51A knock-in mutation. This finding was confirmed in two isogenic human cell lines with an engineered defect in eIF2alpha phosphorylation. We show that impaired CA9 expression was not due to changes in HIF activity or CA9 mRNA stability. Using chromatin immunoprecipitation we show that the eIF2alpha-dependent translationally regulated gene ATF4 binds directly to the CA9 promoter and is associated with loss of the transcriptional repressive histone 3 lysine 27 tri-methylation mark. Loss or overexpression of ATF4 confirmed its role in CA9 induction during hypoxia. Our data indicate that expression of CA9 is regulated through both the HIF-1 and unfolded protein response hypoxia response pathways in vitro and in vivo.
- Published
- 2009
18. Metformin and Prostate Cancer Radiation Therapy: Improved Outcomes Due to Enhanced Tumor Oxygenation
- Author
-
Dal Pra, A., primary, Zannella, V., additional, Glicksman, R., additional, Sykes, J., additional, Muaddi, H., additional, Joshua, A.M., additional, Wouters, B.G., additional, Milosevic, M., additional, Koritzinsky, M., additional, and Bristow, R.G., additional
- Published
- 2013
- Full Text
- View/download PDF
19. SP-0628: Treatment strategies targeting tumor hypoxia
- Author
-
Koritzinsky, M., primary
- Published
- 2013
- Full Text
- View/download PDF
20. 355 HYPOXIA INHIBITS DISULFIDE BOND FORMATION AND PROTEIN FOLDING IN THE ENDOPLASMIC RETICULUM
- Author
-
Koritzinsky, M., primary, Van den Beucken, T., additional, Chu, K., additional, Boutros, P.C., additional, Braakman, I., additional, and Wouters, B.G., additional
- Published
- 2012
- Full Text
- View/download PDF
21. 347 NOVEL MECHANISMS OF GENE REGULATION BY HYPOXIA
- Author
-
Prickaerts, P., primary, Adriaens, M., additional, Chan-Seng-Yue, M., additional, Beck, T., additional, Koritzinsky, M., additional, Voncken, W., additional, and Wouters, B., additional
- Published
- 2012
- Full Text
- View/download PDF
22. Modulation of cell death in the tumor micro environment
- Author
-
Wouters, BG, Koritzinsky, M, Chin, RK, Theys, J, Buijsen, J, Lambin, P, Wouters, BG, Koritzinsky, M, Chin, RK, Theys, J, Buijsen, J, and Lambin, P
- Abstract
The microenvironment of solid human tumors is characterized by heterogeneity in oxygenation. Hypoxia arises early in the process of tumor development because rapidly proliferating tumor cells outgrow the capacity of the host vasculature. Formation of solid tumors thus requires coordination of angiogenesis with continued tumor cell proliferation. However, despite such neovascularization, hypoxia is persistent and frequently found in tumors at the time of diagnosis. Tumors with low oxygenation have a poor prognosis, and strong evidence suggests this is because of the effects of hypoxia on malignant progression, angiogenesis, metastasis, and therapy resistance. The presence of viable hypoxic cells is likely a reflection of the development of hypoxia tolerance resulting from modulation of cell death in the microenvironment. This acquired feature has been explained on the basis of clonal selection-the hypoxic microenvironment selects cells capable of surviving in the absence of normal oxygen availability. However, the persistence and frequency of hypoxia in solid tumors raises a second potential explanation. We suggest that stable microregions of hypoxia may play a positive role in tumor growth. Although hypoxia inhibits cell proliferation and in tumor cells will eventually induce cell death, hypoxia also provides angiogenic and metastatic signals. The development of hypoxia tolerance will thus allow prolonged survival in the absence of oxygen and generation of a persistent angiogenic signal. We will discuss the concept of hypoxia tolerance and review mechanisms used by cancer cells to acquire this phenotype. The concept of hypoxia tolerance has important implications for current and future therapeutic approaches. Most therapeutic efforts to combat hypoxia have focused on targeting the presence of hypoxia itself. Our hypothesis predicts that targeting the biological responses to hypoxia and the pathways leading to hypoxia tolerance may also be attractive therapeutic stra
- Published
- 2003
23. Hypoxia as a target for combined modality treatments
- Author
-
Wouters, BG, Weppler, SA, Koritzinsky, M, Landuyt, W, Nuyts, S, Theys, J, Chiu, RK, Lambin, P, Wouters, BG, Weppler, SA, Koritzinsky, M, Landuyt, W, Nuyts, S, Theys, J, Chiu, RK, and Lambin, P
- Abstract
There is overwhelming evidence that solid human tumours grow within a unique micro-environment. This environment is characterised by an abnormal vasculature, which leads to an insufficient supply of oxygen and nutrients to the tumour cells. These characteristics of the environment limit the effectiveness of both radiotherapy and chemotherapy. Measurement of the oxygenation status of human tumours has unequivocally demonstrated the importance of this parameter on patient prognosis. Tumour hypoxia has been shown to be an independent prognostic indicator of poor outcome in prostate, head and neck and cervical cancers. Recent laboratory and clinical data have shown that hypoxia is also associated with a more malignant phenotype, affecting genomic stability, apoptosis, angiogenesis and metastasis. Several years ago, scientists realised that the unique properties within the tumour micro-environment could provide the basis for tumour-specific therapies. Efforts that are underway to develop therapies that exploit the tumour micro-environment can be categorised into three groups. The first includes agents that exploit the environmental changes that occur within the micro-environment such as hypoxia and reduced pH. This includes bioreductive drugs that are specifically toxic to hypoxic cells, as well as hypoxia-specific gene delivery systems. The second category includes therapies designed to exploit the unique properties of the tumour vasculature and include both angiogenesis inhibitors and vascular targeting agents. The final category includes agents that exploit the molecular and cellular responses to hypoxia. For example, many genes are induced by hypoxia and promoter elements from these genes can be used for the selective expression of therapeutic proteins in hypoxic tumour cells. An overview of the various properties ascribed to tumour hypoxia and the current efforts underway to exploit hypoxia for improving cancer treatment will be discussed. (C) 2002 Published by E
- Published
- 2002
24. Hypoxia strongly upregulate the expression of EGFRvIII in glioma cells
- Author
-
Li, Y., primary, Lieuwes, N., additional, Jutten, B., additional, Chiu, R., additional, Theys, J., additional, Koritzinsky, M., additional, Lambin, P., additional, Wouters, B.G., additional, and Lammering, G., additional
- Published
- 2008
- Full Text
- View/download PDF
25. Dose- and Time-Dependent Changes in Gene Expression in Human Glioma Cells after Low Radiation Doses
- Author
-
Short, S. C., primary, Buffa, F. M., additional, Bourne, S., additional, Koritzinsky, M., additional, Wouters, B. G., additional, and Bentzen, S. M., additional
- Published
- 2007
- Full Text
- View/download PDF
26. 9 Hypoxic regulation of translation initiation and its impact on gene expression
- Author
-
Wouters, B.G., primary, Koritzinsky, M., additional, Magagnin, M., additional, van den Beucken, T., additional, Koumenis, C., additional, and Lambin, P., additional
- Published
- 2006
- Full Text
- View/download PDF
27. 31 Radiation responsive genes in glioma cells
- Author
-
Short, S.C., primary, Bourne, S., additional, Buffa, F., additional, Koritzinsky, M., additional, and Wouters, B., additional
- Published
- 2006
- Full Text
- View/download PDF
28. Lack of inverse dose‐rate effect and binding of the retinoblastoma gene product in the nucleus of human cancer T‐47D cells arrested in G2 by ionizing radiation
- Author
-
Furre, T., primary, Eggen Furre, I., additional, Koritzinsky, M., additional, Åmellem, Ø., additional, and Pettersen, E. O., additional
- Published
- 2003
- Full Text
- View/download PDF
29. Cell cycle progression and radiation survival following prolonged hypoxia and re-oxygenation
- Author
-
Koritzinsky, M., primary, Wouters, B. G., additional, Åmellem, Ø., additional, and Pettersen, E. O., additional
- Published
- 2001
- Full Text
- View/download PDF
30. Inverse dose-rate effect due to pre-mitotic accumulation during continuous low dose-rate irradiation of cervix carcinoma cells.
- Author
-
Furre, T., Koritzinsky, M., Olsen, D. R., and Pettersen, E. O.
- Subjects
- *
EFFECT of radiation on cells , *RADIATION dosimetry - Abstract
Purpose: To investigate the radiation sensitivity of asynchronous and synchronized cancer cervix cells irradiated with low dose rates. Materials and methods: Cells were exposed to 60Co gamma-rays at dose rates ranging from 0.33 to 0.94Gy/h. Synchronized cells were obtained by collecting detached mitotic cells after a shaking procedure. Cell survival was measured as the ability of cells to form colonies. Cell-cycle distributions were calculated by computer analysis of a DNA histogram recorded by flow cytometry. Results: Irradiation of asynchronous cells at either 0.33 or 0.86Gy/h resulted in exponential dose-survival curves with equal alpha-values, i.e. same radiation sensitivity, when dose-survival data for irradiation periods less than 20h were considered. However, the radiation sensitivity was higher by a factor of two when analysing dose-survival data for irradiation periods exceeding 20h. This increase in radiation sensitivity occurred when 80% of the cells accumulated in a pre-mitotic stage of the cell cycle. Irradiation of synchronized cell populations confirmed that these cells were a factor of two more sensitive to radiation in G2 than in G1. Conclusions: An inverse dose-rate effect, i.e. more efficient inactivation of cells at lower rather than at higher dose rates, was observed for radiation doses exceeding 7Gy due to pre-mitotic accumulation of cells during low dose-rate irradiation. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
31. Survival of synchronized human NHIK 3025 cells irradiated aerobically following a prolonged treatment with extremely hypoxic conditions.
- Author
-
Koritzinsky, M., Furre, T., Amellem, O., and Pettersen, E. O.
- Subjects
- *
IRRADIATION , *HYPOXEMIA - Abstract
Purpose : To investigate whether radiation survival of cells irradiated aerobically in the oxygen-sensitive restriction point in late G1 is dependent on where in the cell cycle the cells first were rendered hypoxic. Materials and methods : Human cervix carcinoma, NHIK 3025 cells, were synchronized and rendered hypoxic while in early-, midor late G1 or in early G2. Cell-cycle progression during the treatment was monitored by flow cytometry, and cell survival following either hypoxia alone or hypoxia with subsequent reoxygenation and irradiation was measured by the ability of the cells to form macroscopic colonies. Results : During prolonged hypoxia, all surviving cells accumulated in an oxygen-sensitive restriction point in late G1. Cells rendered hypoxic in G2 initiated DNA synthesis following reoxygenation and irradiation several hours later than cells rendered hypoxic in G1. Radiation survival of cells accumulated in the oxygensensitive restriction point was independent of where in the cell cycle the cells first were rendered hypoxic. The hypoxia-treated cells had lower radiation survival probability than untreated cells in late G1. Conclusions : Although cells accumulated in the oxygen-sensitive restriction point from different parts of the cell cycle are not biologically identical, they are radiobiologically similar. The radiosensitizing effect of prolonged hypoxia was not merely due to cell-cycle redistribution. [ABSTRACT FROM AUTHOR]
- Published
- 1998
- Full Text
- View/download PDF
32. Post-transcriptional regulation of MRE11 expression in muscle-invasive bladder tumours
- Author
-
Rm, Martin, Kerr M, Mt, Teo, Sj, Jevons, Koritzinsky M, Bg, Wouters, Bhattarai S, and Anne Kiltie
33. Hypoxia-induced metastasis : the role of the unfolded protein response
- Author
-
Mujcic, H., Wouters, Brad, Lambin, Philippe, Koritzinsky, M., Rouschop, Kasper, and Radiotherapie
- Published
- 2013
34. Three discipline collaborative radiation therapy (3DCRT) special debate: Systemic radiotherapy using targeted isotopes is the best hope for advancing curative radiation therapy.
- Author
-
Koontz BF, Koritzinsky M, Zoberi JE, Brown SL, Ding X, Wong J, Joiner MC, Dominello MM, and Burmeister J
- Published
- 2024
- Full Text
- View/download PDF
35. Cysteamine dioxygenase (ADO) governs cancer cell mitochondrial redox homeostasis through proline metabolism.
- Author
-
Lee SCS, Pyo AHA, Mohammadi H, Zhang J, Dvorkin-Gheva A, Malbeteau L, Chung S, Khan S, Ciudad MT, Rondeau V, Cairns RA, Kislinger T, McGaha TL, Wouters BG, Reisz JA, Culp-Hill R, D'Alessandro A, Jones CL, and Koritzinsky M
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Neoplasms metabolism, Neoplasms pathology, Neoplasms genetics, Polyamines metabolism, Dioxygenases metabolism, Mice, Knockout, Reactive Oxygen Species metabolism, Proline Oxidase metabolism, Proline Oxidase genetics, Cysteamine metabolism, Cell Proliferation, Proline metabolism, Mitochondria metabolism, Oxidation-Reduction, Homeostasis
- Abstract
2-Aminoethanethiol dioxygenase (ADO) is a thiol dioxygenase that sulfinylates cysteamine and amino-terminal cysteines in polypeptides. The pathophysiological roles of ADO remain largely unknown. Here, we demonstrate that ADO expression represents a vulnerability in cancer cells, as ADO depletion led to loss of proliferative capacity and survival in cancer cells and reduced xenograft growth. In contrast, generation of the ADO knockout mouse revealed high tolerance for ADO depletion in adult tissues. To understand the mechanism underlying ADO's essentiality in cancer cells, we characterized the cell proteome and metabolome following depletion of ADO. This revealed that ADO depletion leads to toxic levels of polyamines which can be driven by ADO's substrate cysteamine. Polyamine accumulation in turn stimulated expression of proline dehydrogenase (PRODH) which resulted in mitochondrial hyperactivity and ROS production, culminating in cell toxicity. This work identifies ADO as a unique vulnerability in cancer cells, due to its essential role in maintenance of redox homeostasis through restraining polyamine levels and proline catabolism.
- Published
- 2024
- Full Text
- View/download PDF
36. Oral pimonidazole unveils clinicopathologic and epigenetic features of hypoxic tumour aggressiveness in localized prostate cancer.
- Author
-
Ci X, Chen S, Zhu R, Zarif M, Jain R, Guo W, Ramotar M, Gong L, Xu W, Singh O, Mansouri S, Zadeh G, Wei GH, Xu W, Bristow R, Berlin A, Koritzinsky M, van der Kwast T, and He HH
- Subjects
- Humans, Male, Aged, Middle Aged, Tumor Hypoxia genetics, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Prognosis, Administration, Oral, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Prostatic Neoplasms surgery, Prostatic Neoplasms metabolism, Epigenesis, Genetic, Prostatectomy, DNA Methylation, Nitroimidazoles
- Abstract
Background: Tumor hypoxia is associated with prostate cancer (PCa) treatment resistance and poor prognosis. Pimonidazole (PIMO) is an investigational hypoxia probe used in clinical trials. A better understanding of the clinical significance and molecular alterations underpinning PIMO-labeled tumor hypoxia is needed for future clinical application. Here, we investigated the clinical significance and molecular alterations underpinning PIMO-labeled tumor hypoxia in patients with localized PCa, in order to apply PIMO as a prognostic tool and to identify potential biomarkers for future clinical translation., Methods: A total of 39 patients with localized PCa were recruited and administered oral PIMO before undergoing radical prostatectomy (RadP). Immunohistochemical staining for PIMO was performed on 37 prostatectomy specimens with staining patterns evaluated and clinical association analyzed. Whole genome bisulfite sequencing was performed using laser-capture of microdissected specimen sections comparing PIMO positive and negative tumor areas. A hypoxia related methylation molecular signature was generated by integrating the differentially methylated regions with previously established RNA-seq datasets., Results: Three PIMO staining patterns were distinguished: diffuse, focal, and comedo-like. The comedo-like staining pattern was more commonly associated with adverse pathology. PIMO-defined hypoxia intensity was positively correlated with advanced pathologic stage, tumor invasion, and cribriform and intraductal carcinoma morphology. The generated DNA methylation signature was found to be a robust hypoxia biomarker, which could risk-stratify PCa patients across multiple clinical datasets, as well as be applicable in other cancer types., Conclusions: Oral PIMO unveiled clinicopathologic features of disease aggressiveness in localized PCa. The generated DNA methylation signature is a novel and robust hypoxia biomarker that has the potential for future clinical translation., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
37. Dabrafenib Alters MDSC Differentiation and Function by Activation of GCN2.
- Author
-
Ciudad MT, Quevedo R, Lamorte S, Jin R, Nzirorera N, Koritzinsky M, and McGaha TL
- Subjects
- Animals, Mice, Cell Line, Tumor, Proto-Oncogene Proteins B-raf metabolism, DNA-Binding Proteins metabolism, Transcription Factors metabolism, Myeloid-Derived Suppressor Cells, Imidazoles, Oximes
- Abstract
The effect of targeted therapeutics on anticancer immune responses is poorly understood. The BRAF inhibitor dabrafenib has been reported to activate the integrated stress response (ISR) kinase GCN2, and the therapeutic effect has been partially attributed to GCN2 activation. Because ISR signaling is a key component of myeloid-derived suppressor cell (MDSC) development and function, we measured the effect of dabrafenib on MDSC differentiation and suppressive activity. Our data showed that dabrafenib attenuated MDSC ability to suppress T-cell activity, which was associated with a GCN2-dependent block of the transition from monocytic progenitor to polymorphonuclear (PMN)-MDSCs and proliferative arrest resulting in PMN-MDSC loss. Transcriptional profiling revealed that dabrafenib-driven GCN2 activation altered metabolic features in MDSCs enhancing oxidative respiration, and attenuated transcriptional programs required for PMN development. Moreover, we observed a broad downregulation of transcriptional networks associated with PMN developmental pathways, and increased activity of transcriptional regulons driven by Atf5, Mafg, and Zbtb7a. This transcriptional program alteration underlies the basis for PMN-MDSC developmental arrest, skewing immature MDSC development toward monocytic lineage cells. In vivo, we observed a pronounced reduction in PMN-MDSCs in dabrafenib-treated tumor-bearing mice suggesting that dabrafenib impacts MDSC populations systemically and locally, in the tumor immune infiltrate. Thus, our data reveal transcriptional networks that govern MDSC developmental programs, and the impact of GCN2 stress signaling on the innate immune landscape in tumors, providing novel insight into potentially beneficial off-target effects of dabrafenib., Significance: An important, but poorly understood, aspect of targeted therapeutics for cancer is the effect on antitumor immune responses. This article shows that off-target effects of dabrafenib activating the kinase GCN2 impact MDSC development and function reducing PMN-MDSCs in vitro and in vivo. This has important implications for our understanding of how this BRAF inhibitor impacts tumor growth and provides novel therapeutic target and combination possibilities., (© 2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
38. Longitudinal dynamics of the tumor hypoxia response: From enzyme activity to biological phenotype.
- Author
-
Lee SCS, Pyo AHA, and Koritzinsky M
- Subjects
- Humans, Hypoxia, Oxygen metabolism, Phenotype, Tumor Hypoxia, Neoplasms metabolism
- Abstract
Poor oxygenation (hypoxia) is a common spatially heterogeneous feature of human tumors. Biological responses to tumor hypoxia are orchestrated by the decreased activity of oxygen-dependent enzymes. The affinity of these enzymes for oxygen positions them along a continuum of oxygen sensing that defines their roles in launching reactive and adaptive cellular responses. These responses encompass regulation of all steps in the central dogma, with rapid perturbation of the metabolome and proteome followed by more persistent reprogramming of the transcriptome and epigenome. Core hypoxia response genes and pathways are commonly regulated at multiple inflection points, fine-tuning the dependencies on oxygen concentration and hypoxia duration. Ultimately, shifts in the activity of oxygen-sensing enzymes directly or indirectly endow cells with intrinsic hypoxia tolerance and drive processes that are associated with aggressive phenotypes in cancer including angiogenesis, migration, invasion, immune evasion, epithelial mesenchymal transition, and stemness.
- Published
- 2023
- Full Text
- View/download PDF
39. Dabrafenib alters MDSC differentiation and function by activation of GCN2.
- Author
-
Ciudad MT, Quevedo R, Lamorte S, Jin R, Nzirorera N, Koritzinsky M, and McGaha TL
- Abstract
The effect of targeted therapeutics on anti-cancer immune responses is poorly understood. The BRAF inhibitor dabrafenib has been reported to activate the integrated stress response (ISR) kinase GCN2, and the therapeutic effect has been partially attributed to GCN2 activation. Since ISR signaling is a key component of myeloid-derived suppressor cell (MDSC) development and function, we measured the effect of dabrafenib on MDSC differentiation and suppressive activity. Our data showed that dabrafenib attenuated MDSC ability to suppress T cell activity, which was associated with a GCN2-dependent block of the transition from monocytic progenitor to polymorphonuclear (PMN)-MDSCs and proliferative arrest resulting in PMN-MDSC loss. Transcriptional profiling revealed that dabrafenib-driven GCN2 activation altered metabolic features in MDSCs enhancing oxidative respiration, and attenuated transcriptional programs required for PMN development. Moreover, we observed a broad downregulation of transcriptional networks associated with PMN developmental pathways, and increased activity of transcriptional regulons driven by Atf5 , Mafg , and Zbtb7a . This transcriptional program alteration underlies the basis for PMN-MDSC developmental arrest, skewing immature MDSC development towards monocytic lineage cells. In vivo , we observed a pronounced reduction in PMN-MDSCs in dabrafenib-treated tumor-bearing mice suggesting that dabrafenib impacts MDSC populations systemically and locally, in the tumor immune infiltrate. Thus, our data reveals transcriptional networks that govern MDSC developmental programs, and the impact of GCN2 stress signaling on the innate immune landscape in tumors, providing novel insight into potentially beneficial off target effects of dabrafenib.
- Published
- 2023
- Full Text
- View/download PDF
40. Distinct shared and compartment-enriched oncogenic networks drive primary versus metastatic breast cancer.
- Author
-
Jiang Z, Ju Y, Ali A, Chung PED, Skowron P, Wang DY, Shrestha M, Li H, Liu JC, Vorobieva I, Ghanbari-Azarnier R, Mwewa E, Koritzinsky M, Ben-David Y, Woodgett JR, Perou CM, Dupuy A, Bader GD, Egan SE, Taylor MD, and Zacksenhaus E
- Subjects
- Female, Humans, Animals, Mice, Signal Transduction, Neoplasm Metastasis, Breast Neoplasms pathology
- Abstract
Metastatic breast-cancer is a major cause of death in women worldwide, yet the relationship between oncogenic drivers that promote metastatic versus primary cancer is still contentious. To elucidate this relationship in treatment-naive animals, we hereby describe mammary-specific transposon-mutagenesis screens in female mice together with loss-of-function Rb, which is frequently inactivated in breast-cancer. We report gene-centric common insertion-sites (gCIS) that are enriched in primary-tumors, in metastases or shared by both compartments. Shared-gCIS comprise a major MET-RAS network, whereas metastasis-gCIS form three additional hubs: Rho-signaling, Ubiquitination and RNA-processing. Pathway analysis of four clinical cohorts with paired primary-tumors and metastases reveals similar organization in human breast-cancer with subtype-specific shared-drivers (e.g. RB1-loss, TP53-loss, high MET, RAS, ER), primary-enriched (EGFR, TGFβ and STAT3) and metastasis-enriched (RHO, PI3K) oncogenic signaling. Inhibitors of RB1-deficiency or MET plus RHO-signaling cooperate to block cell migration and drive tumor cell-death. Thus, targeting shared- and metastasis- but not primary-enriched derivers offers a rational avenue to prevent metastatic breast-cancer., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
41. Identification of acquired Notch3 dependency in metastatic Head and Neck Cancer.
- Author
-
Kondratyev M, Pesic A, Ketela T, Stickle N, Beswick C, Shalev Z, Marastoni S, Samadian S, Dvorkin-Gheva A, Sayad A, Bashkurov M, Boasquevisque P, Datti A, Pugh TJ, Virtanen C, Moffat J, Grénman RA, Koritzinsky M, and Wouters BG
- Subjects
- Animals, Mice, Signal Transduction, Squamous Cell Carcinoma of Head and Neck, Humans, Carcinoma, Squamous Cell, Head and Neck Neoplasms
- Abstract
During cancer development, tumor cells acquire changes that enable them to invade surrounding tissues and seed metastasis at distant sites. These changes contribute to the aggressiveness of metastatic cancer and interfere with success of therapy. Our comprehensive analysis of "matched" pairs of HNSCC lines derived from primary tumors and corresponding metastatic sites identified several components of Notch3 signaling that are differentially expressed and/or altered in metastatic lines and confer a dependency on this pathway. These components were also shown to be differentially expressed between early and late stages of tumors in a TMA constructed from over 200 HNSCC patients. Finally, we show that suppression of Notch3 improves survival in mice in both subcutaneous and orthotopic models of metastatic HNSCC. Novel treatments targeting components of this pathway may prove effective in targeting metastatic HNSCC cells alone or in combination with conventional therapies., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
42. A Phase II Randomized Trial of Chemoradiation with or without Metformin in Locally Advanced Cervical Cancer.
- Author
-
Han K, Fyles A, Shek T, Croke J, Dhani N, D'Souza D, Lee TY, Chaudary N, Bruce J, Pintilie M, Cairns R, Vines D, Pakbaz S, Jaffray D, Metser U, Rouzbahman M, Milosevic M, and Koritzinsky M
- Subjects
- Female, Humans, Positron Emission Tomography Computed Tomography, Pandemics, Positron-Emission Tomography methods, Hypoxia, Radiopharmaceuticals, Uterine Cervical Neoplasms diagnostic imaging, Uterine Cervical Neoplasms drug therapy, Metformin therapeutic use, COVID-19, Nitroimidazoles
- Abstract
Purpose: Tumor hypoxia is associated with poor response to radiation (RT). We previously discovered a novel mechanism of metformin: enhancing tumor RT response by decreasing tumor hypoxia. We hypothesized that metformin would decrease tumor hypoxia and improve cervical cancer response to RT., Patients and Methods: A window-of-opportunity, phase II randomized trial was performed in stage IB-IVA cervical cancer. Patients underwent screening positron emission tomography (PET) imaging with hypoxia tracer fluoroazomycin arabinoside (FAZA). Only patients with FAZA uptake (hypoxic tumor) were included and randomized 2:1 to receive metformin in combination with chemoRT or chemoRT alone. A second FAZA-PET/CT scan was performed after 1 week of metformin or no intervention (control). The primary endpoint was a change in fractional hypoxic volume (FHV) between FAZA-PET scans, compared using the Wilcoxon signed-rank test. The study was closed early due to FAZA availability and the COVID-19 pandemic., Results: Of the 20 consented patients, 6 were excluded due to no FAZA uptake and 1 withdrew. FHV of 10 patients in the metformin arm decreased by an average of 10.2% (44.4%-34.2%) ± SD 16.9% after 1 week of metformin, compared with an average increase of 4.7% (29.1%-33.8%) ± 11.5% for the 3 controls (P = 0.027). Those with FHV reduction after metformin had significantly lower MATE2 expression. With a median follow-up of 2.8 years, the 2-year disease-free survival was 67% for the metformin arm versus 33% for controls (P = 0.09)., Conclusions: Metformin decreased cervical tumor hypoxia in this trial that selected for patients with hypoxic tumor. See related commentary by Lyng et al., p. 5233., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
43. Translational Control by 4E-BP1/2 Suppressor Proteins Regulates Mitochondrial Biosynthesis and Function during CD8 + T Cell Proliferation.
- Author
-
Dimitriou ID, Meiri D, Jitkova Y, Elford AR, Koritzinsky M, Schimmer AD, Ohashi PS, Sonenberg N, and Rottapel R
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Cell Cycle Proteins metabolism, Cell Proliferation, Mammals genetics, Mice, Organelle Biogenesis, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Protein Biosynthesis, Eukaryotic Initiation Factors genetics, Eukaryotic Initiation Factors metabolism, Phosphoproteins metabolism
- Abstract
CD8
+ T cell proliferation and differentiation into effector and memory states are high-energy processes associated with changes in cellular metabolism. CD28-mediated costimulation of T cells activates the PI3K/AKT/mammalian target of rapamycin signaling pathway and induces eukaryotic translation initiation factor 4E-dependent translation through the derepression by 4E-BP1 and 4E-BP2. In this study, we demonstrate that 4E-BP1/2 proteins are required for optimum proliferation of mouse CD8+ T cells and the development of an antiviral effector function. We show that translation of genes encoding mitochondrial biogenesis is impaired in T cells derived from 4E-BP1/2-deficient mice. Our findings demonstrate an unanticipated role for 4E-BPs in regulating a metabolic program that is required for cell growth and biosynthesis during the early stages of CD8+ T cell expansion., (Copyright © 2022 by The American Association of Immunologists, Inc.)- Published
- 2022
- Full Text
- View/download PDF
44. Repurposing Itraconazole and Hydroxychloroquine to Target Lysosomal Homeostasis in Epithelial Ovarian Cancer.
- Author
-
Marastoni S, Madariaga A, Pesic A, Nair SN, Li ZJ, Shalev Z, Ketela T, Colombo I, Mandilaras V, Cabanero M, Bruce JP, Li X, Garg S, Wang L, Chen EX, Gill S, Dhani NC, Zhang W, Pintilie M, Bowering V, Koritzinsky M, Rottapel R, Wouters BG, Oza AM, Joshua AM, and Lheureux S
- Subjects
- Humans, Female, Itraconazole pharmacology, Hydroxychloroquine pharmacology, Antifungal Agents metabolism, Carcinoma, Ovarian Epithelial drug therapy, Drug Repositioning, Chloroquine metabolism, Lysosomes, Homeostasis, Antineoplastic Agents pharmacology, Ovarian Neoplasms drug therapy
- Abstract
Drug repurposing is an attractive option for oncology drug development. Itraconazole is an antifungal ergosterol synthesis inhibitor that has pleiotropic actions including cholesterol antagonism, inhibition of Hedgehog and mTOR pathways. We tested a panel of 28 epithelial ovarian cancer (EOC) cell lines with itraconazole to define its spectrum of activity. To identify synthetic lethality in combination with itraconazole, a whole-genome drop-out genome-scale clustered regularly interspaced short palindromic repeats sensitivity screen in two cell lines (TOV1946 and OVCAR5) was performed. On this basis, we conducted a phase I dose-escalation study assessing the combination of itraconazole and hydroxychloroquine in patients with platinum refractory EOC (NCT03081702). We identified a wide spectrum of sensitivity to itraconazole across the EOC cell lines. Pathway analysis showed significant involvement of lysosomal compartments, the trans-golgi network and late endosomes/lysosomes; similar pathways are phenocopied by the autophagy inhibitor, chloroquine. We then demonstrated that the combination of itraconazole and chloroquine displayed Bliss defined synergy in EOC cancer cell lines. Furthermore, there was an association of cytotoxic synergy with the ability to induce functional lysosome dysfunction, by chloroquine. Within the clinical trial, 11 patients received at least one cycle of itraconazole and hydroxychloroquine. Treatment was safe and feasible with the recommended phase II dose of 300 and 600 mg twice daily, respectively. No objective responses were detected. Pharmacodynamic measurements on serial biopsies demonstrated limited pharmacodynamic impact. In vitro , itraconazole and chloroquine have synergistic activity and exert a potent antitumor effect by affecting lysosomal function. The drug combination had no clinical antitumor activity in dose escalation., Significance: The combination of the antifungal drug itraconazole with antimalarial drug hydroxychloroquine leads to a cytotoxic lysosomal dysfunction, supporting the rational for further research on lysosomal targeting in ovarian cancer., Competing Interests: S. Marastoni reports a Canadian patent number 3,116,081 pending. A. Madariaga reports personal fees from Clovis and AstraZeneca outside the submitted work. Z.J. Li reports a Canadian patent number to CA3,116,081 pending. I. Colombo reports personal fees from MSD; other from GSK, AstraZeneca, MSD, Bayer, and Oasmia outside the submitted work. J.P. Bruce reports other from Bowhead Health outside the submitted work. M. Koritzinsky reports other from The Princess Margaret Cancer Foundation during the conduct of the study; in addition, M. Koritzinsky has a Canadian patent number number 3,116,081 pending. B.G. Wouters reports grants from Ontario Institute for Cancer Research, Canadian Institutes for Cancer Research, and Princess Margaret Cancer Foundation during the conduct of the study; other from Northern Biologics outside the submitted work; in addition, B.G. Wouters has a Canadian patent number 3,116,081 issued. A.M. Oza is PI and on clinical trial steer- ing committees for trials with Clovis, GSK, AstraZeneca—all uncompensated. A.M. Oza is uncompensated CEO of Ozmosis Research, a Not For Profit Clin- ical Trials Management company associated with UHN. A.M. Joshua reports non-financial support from Mayne Pharma during the conduct of the study; other from Pricilium outside the submitted work; in addition, A.M. Joshua has a Canadian patent number 3,116,081 pending. S. Lheureux reports grants from OICR - TRI Ovarian Cancer during the conduct of the study; grants and personal fees from AstraZeneca, GSK; personal fees from Eisai, Merck, Shattuck Labs; grants from Roche, and outside the submitted work. No other disclosures were reported., (© 2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
45. Topographic mapping of the glioblastoma proteome reveals a triple-axis model of intra-tumoral heterogeneity.
- Author
-
Lam KHB, Leon AJ, Hui W, Lee SC, Batruch I, Faust K, Klekner A, Hutóczki G, Koritzinsky M, Richer M, Djuric U, and Diamandis P
- Subjects
- Antineoplastic Agents therapeutic use, Brain Neoplasms diagnosis, Brain Neoplasms drug therapy, Brain Neoplasms mortality, Cell Line, Tumor, Cohort Studies, Disease Progression, Drug Resistance, Neoplasm genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Glioblastoma diagnosis, Glioblastoma drug therapy, Glioblastoma mortality, Humans, Hypoxia diagnosis, Hypoxia drug therapy, Hypoxia mortality, Laser Capture Microdissection, Machine Learning, Models, Genetic, Neoplasm Proteins classification, Neoplasm Proteins metabolism, Proteomics methods, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Survival Analysis, Transcriptome, Brain Neoplasms genetics, Genetic Heterogeneity, Glioblastoma genetics, Hypoxia genetics, Neoplasm Proteins genetics, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Glioblastoma is an aggressive form of brain cancer with well-established patterns of intra-tumoral heterogeneity implicated in treatment resistance and progression. While regional and single cell transcriptomic variations of glioblastoma have been recently resolved, downstream phenotype-level proteomic programs have yet to be assigned across glioblastoma's hallmark histomorphologic niches. Here, we leverage mass spectrometry to spatially align abundance levels of 4,794 proteins to distinct histologic patterns across 20 patients and propose diverse molecular programs operational within these regional tumor compartments. Using machine learning, we overlay concordant transcriptional information, and define two distinct proteogenomic programs, MYC- and KRAS-axis hereon, that cooperate with hypoxia to produce a tri-dimensional model of intra-tumoral heterogeneity. Moreover, we highlight differential drug sensitivities and relative chemoresistance in glioblastoma cell lines with enhanced KRAS programs. Importantly, these pharmacological differences are less pronounced in transcriptional glioblastoma subgroups suggesting that this model may provide insights for targeting heterogeneity and overcoming therapy resistance., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
46. Strategic Training in Transdisciplinary Radiation Science for the 21st Century (STARS21): 15-Year Evaluation of an Innovative Research Training Program.
- Author
-
Patel PS, Vyravanathan S, Milne E, Bezjak A, Wouters BG, Bennewith K, Seuntjens J, Liu ZA, Harding SM, Liu FF, Koritzinsky M, and Koch CA
- Subjects
- Humans, Radiation Oncology education, Interdisciplinary Research, Female, Male, Surveys and Questionnaires, Mentors, Program Evaluation
- Abstract
Purpose: To evaluate the 15-year impact of a transdisciplinary research training program for graduate students, postdoctoral fellows, and clinical trainees focused on radiation science, entitled Strategic Training in Transdisciplinary Radiation Science for the 21st Century (STARS21) with a primary objective to build capacity in radiation research., Methods and Materials: Alumni (n = 128) and mentors (n = 41) who participated in STARS21 between 2003 and 2018 were sent an anonymized online survey designed to evaluate the program. Twelve alumni and 7 mentors also volunteered to participate in semistructured interviews. The transcribed interviews were coded and analyzed using NVivo12-Pro software. Alumni employment and publications were assessed from program records and by web-based search queries., Results: Alumni are located in 11 countries, and nearly 90% are employed in a research-oriented career and continue to publish in radiation medicine- or cancer-related fields. Of those invited, 46 alumni (36%) and 12 mentors (29%) completed the online survey. Approximately 87% of alumni valued interdisciplinary collaboration, and 80% indicated that STARS21 had encouraged them to pursue such collaborations. Alumni emphasized that STARS21 assisted their career development, and the majority of alumni and mentors would recommend STARS21 to other trainees (4.48 and 4.58, respectively; 5 = strongly agree). The time invested in the program was perceived by mentors as worthwhile for the knowledge and skills gained by trainees (4.67; 5 = strongly agree), and 64% of mentors indicated that these benefits were associated with improved trainee research productivity. From the alumni and mentor perspectives, the valuable skills acquired from STARS21 included scientific communication (85% and 83%, respectively) and networking (83% and 92%, respectively)., Conclusions: STARS21 is an innovative research training program that promotes interdisciplinary collaboration in radiation medicine research, which is valued by alumni and mentor respondents. Alumni can acquire important skill sets for career development, with a large proportion of alumni currently engaged in radiation research around the world., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
47. NOX4 links metabolic regulation in pancreatic cancer to endoplasmic reticulum redox vulnerability and dependence on PRDX4.
- Author
-
Jain P, Dvorkin-Gheva A, Mollen E, Malbeteau L, Xie M, Jessa F, Dhavarasa P, Chung S, Brown KR, Jang GH, Vora P, Notta F, Moffat J, Hedley D, Boutros PC, Wouters BG, and Koritzinsky M
- Subjects
- Endoplasmic Reticulum metabolism, Humans, NADP metabolism, NADPH Oxidase 4 genetics, NADPH Oxidase 4 metabolism, Oxidation-Reduction, Peroxiredoxins genetics, Peroxiredoxins metabolism, Reactive Oxygen Species metabolism, Hydrogen Peroxide metabolism, Hydrogen Peroxide pharmacology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism
- Abstract
There is an urgent need to identify vulnerabilities in pancreatic ductal adenocarcinoma (PDAC). PDAC cells acquire metabolic changes that augment NADPH production and cytosolic redox homeostasis. Here, we show that high NADPH levels drive activity of NADPH oxidase 4 (NOX4) expressed in the endoplasmic reticulum (ER) membrane. NOX4 produces H
2 O2 metabolized by peroxiredoxin 4 (PRDX4) in the ER lumen. Using functional genomics and subsequent in vitro and in vivo validations, we find that PDAC cell lines with high NADPH levels are dependent on PRDX4 for their growth and survival. PRDX4 addiction is associated with increased reactive oxygen species, a DNA-PKcs-governed DNA damage response and radiosensitivity, which can be rescued by depletion of NOX4 or NADPH. Hence, this study has identified NOX4 as a protein that paradoxically converts the reducing power of the cytosol to an ER-specific oxidative stress vulnerability in PDAC that may be therapeutically exploited by targeting PRDX4., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)- Published
- 2021
- Full Text
- View/download PDF
48. Mammary epithelial cells have lineage-rooted metabolic identities.
- Author
-
Mahendralingam MJ, Kim H, McCloskey CW, Aliar K, Casey AE, Tharmapalan P, Pellacani D, Ignatchenko V, Garcia-Valero M, Palomero L, Sinha A, Cruickshank J, Shetty R, Vellanki RN, Koritzinsky M, Stambolic V, Alam M, Schimmer AD, Berman HK, Eaves CJ, Pujana MA, Kislinger T, and Khokha R
- Subjects
- Animals, Biomarkers, Computational Biology methods, Female, Flow Cytometry methods, Gene Expression Profiling, High-Throughput Nucleotide Sequencing, Humans, Mammary Glands, Animal cytology, Mammary Glands, Animal metabolism, Mammary Glands, Human cytology, Metabolic Networks and Pathways, Mitochondria genetics, Mitochondria metabolism, Proteome, Proteomics methods, Cell Lineage, Energy Metabolism, Epithelial Cells metabolism, Mammary Glands, Human metabolism
- Abstract
Cancer metabolism adapts the metabolic network of its tissue of origin. However, breast cancer is not a disease of a single origin. Multiple epithelial populations serve as the culprit cell of origin for specific breast cancer subtypes, yet our knowledge of the metabolic network of normal mammary epithelial cells is limited. Using a multi-omic approach, here we identify the diverse metabolic programmes operating in normal mammary populations. The proteomes of basal, luminal progenitor and mature luminal cell populations revealed enrichment of glycolysis in basal cells and of oxidative phosphorylation in luminal progenitors. Single-cell transcriptomes corroborated lineage-specific metabolic identities and additional intra-lineage heterogeneity. Mitochondrial form and function differed across lineages, with clonogenicity correlating with mitochondrial activity. Targeting oxidative phosphorylation and glycolysis with inhibitors exposed lineage-rooted metabolic vulnerabilities of mammary progenitors. Bioinformatics indicated breast cancer subtypes retain metabolic features of their putative cell of origin. Thus, lineage-rooted metabolic identities of normal mammary cells may underlie breast cancer metabolic heterogeneity and targeting these vulnerabilities could advance breast cancer therapy.
- Published
- 2021
- Full Text
- View/download PDF
49. Emergence of Enzalutamide Resistance in Prostate Cancer is Associated with BCL-2 and IKKB Dependencies.
- Author
-
Liang Y, Jeganathan S, Marastoni S, Sharp A, Figueiredo I, Marcellus R, Mawson A, Shalev Z, Pesic A, Sweet J, Guo H, Uehling D, Gurel B, Neeb A, He HH, Montgomery B, Koritzinsky M, Oakes S, de Bono JS, Gleave M, Zoubeidi A, Wouters BG, and Joshua AM
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Benzamides therapeutic use, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Gene Knockdown Techniques, Humans, I-kappa B Kinase antagonists & inhibitors, I-kappa B Kinase genetics, Male, Nitriles therapeutic use, Phenylthiohydantoin therapeutic use, Prostate pathology, Prostate surgery, Prostatectomy, Prostatic Neoplasms, Castration-Resistant pathology, Prostatic Neoplasms, Castration-Resistant surgery, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 genetics, Sulfonamides pharmacology, Sulfonamides therapeutic use, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Benzamides pharmacology, I-kappa B Kinase metabolism, Nitriles pharmacology, Phenylthiohydantoin pharmacology, Prostatic Neoplasms, Castration-Resistant therapy, Proto-Oncogene Proteins c-bcl-2 metabolism
- Abstract
Purpose: Although enzalutamide (ENZ) has been widely used to treat de novo or castration-resistant metastatic prostate cancer, resistance develops and disease progression is ultimately inevitable. There are currently no approved targeted drugs to specifically delay or overcome ENZ resistance., Experimental Design: We selected several ENZ-resistant cell lines that replicated clinical characteristics of the majority of patients with ENZ-resistant disease. A high-throughput pharmacologic screen was utilized to identify compounds with greater cytotoxic effect for ENZ-resistant cell lines, compared with parental ENZ-sensitive cells. We validated the potential hits in vitro and in vivo , and used knockdown and overexpression assays to study the dependencies in ENZ-resistant prostate cancer., Results: ABT199 (BCL-2 inhibitor) and IMD0354 (IKKB inhibitor) were identified as potent and selective inhibitors of cell viability in ENZ-resistant cell lines in vitro and in vivo which were further validated using loss-of-function assays of BCL-2 and IKKB. Notably, we observed that overexpression of BCL-2 and IKKB in ENZ-sensitive cell lines was sufficient for the emergence of ENZ resistance. In addition, we confirmed that BCL-2 or IKKB inhibitors suppressed the development of ENZ resistance in xenografts. However, validation of both BCL-2 and IKKB in matched castration-sensitive/resistant clinical samples showed that, concurrent with the development of ENZ/abiraterone resistance in patients, only the protein levels of IKKB were increased., Conclusions: Our findings identify BCL-2 and IKKB dependencies in clinically relevant ENZ-resistant prostate cancer cells in vitro and in vivo , but indicate that IKKB upregulation appears to have greater relevance to the progression of human castrate-resistant prostate cancer., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
50. p38 MAPK Inhibition Mitigates Hypoxia-Induced AR Signaling in Castration-Resistant Prostate Cancer.
- Author
-
Cheung S, Jain P, So J, Shahidi S, Chung S, and Koritzinsky M
- Abstract
Background: Aberrant androgen receptor (AR) signaling is a major driver of castration-resistant prostate cancer (CRPC). Tumor hypoxia increases AR signaling and is associated with treatment resistance in prostate cancer. Heat shock protein 27 (Hsp27) is a molecular chaperone that is activated in response to heat shock and hypoxia. Hsp27 has previously been reported to facilitate AR nuclear translocation in a p38 mitogen-activated protein kinase (MAPK) dependent manner in castration-sensitive prostate cancer cell lines. Here, we evaluated the potential for inhibiting p38 MAPK/Hsp27 mediated AR signaling under normoxia and hypoxia in experimental models of CRPC., Methods: We inhibited p38 MAPK with SB203580 in prostate cancer cell lines and measured Hsp27 phosphorylation, AR activity, cell proliferation, and clonogenicity under normoxia and hypoxia. AR activity was measured using an androgen response element driven reporter assay and qPCR to measure expression of AR target genes. Xenograft-bearing mice were treated with SB203580 to measure tumor growth and serum prostate specific antigen (PSA)., Results: Our results indicate that p38 MAPK and Hsp27 are activated under normoxia and hypoxia in response to androgens in CRPC cells. p38 MAPK inhibition diminished Hsp27 activation and the hypoxia-mediated increase in AR activity. Additionally, inhibition of p38 MAPK activity decreased proliferation and survival of CRPC cells in vitro and prolonged the survival of tumor-bearing mice., Conclusions: These results suggest that p38 MAPK inhibition may represent a therapeutic strategy to disrupt AR signaling in the heterogeneous CRPC tumor microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.