124 results on '"Kunz-Schughart LA"'
Search Results
2. Increasing radiosensitivity by alleviating tumour hypoxia: a drug screen reveals atovaquone as a clinical candidate
- Author
-
Ashton, T, Fokas, E, Kunz-Schughart, LA, Folkes, LK, Anbalagan, S, Huether, M, Kelly, CJ, Pirovano, GM, Buffa, FM, Hammond, EM, Stratford, M, Muschel, RJ, Higgins, GS, and McKenna, WG
- Abstract
Tumour hypoxia renders cancer cells resistant to cancer therapy, resulting in markedly worse clinical outcomes. To find clinical candidate compounds that reduce hypoxia in tumours, we conducted a high throughput screen for oxygen consumption rate (OCR) reduction and identified a number of drugs with this property. For this study we focused on the antimalarial, atovaquone. Atovaquone rapidly decreases the OCR by more than 80% in a wide range of cancer cell lines at pharmacological concentrations. In addition, atovaquone eradicates hypoxia in FaDu, HCT116 and H1299 spheroids. Similarly, it virtually eliminates hypoxia in FaDu and HCT116 xenografts in nude mice, and causes a significant tumour growth delay when combined with radiation. Atovaquone is a ubiquinone analogue, and decreases the OCR by inhibiting mitochondrial complex III. We are now undertaking clinical studies to assess whether atovaquone reduces tumour hypoxia in patients, thereby increasing the efficacy of radiotherapy.
- Published
- 2016
3. Der Einfluss stromaler Fibroblasten auf die Tumorangiogenese im Kolonkarzinom
- Author
-
Schmid, SA, primary, Dietrich, A, additional, Gaumann, A, additional, and Kunz-Schughart, LA, additional
- Published
- 2009
- Full Text
- View/download PDF
4. Generalized, sublethal damage-based mathematical approach for improved modeling of clonogenic survival curve flattening upon hyperthermia, radiotherapy, and beyond.
- Author
-
De Mendoza AM, Michlíková S, Castro PS, Muñoz AG, Eckhardt L, Lange S, and Kunz-Schughart LA
- Subjects
- Humans, Radiotherapy methods, Combined Modality Therapy, Cell Survival radiation effects, Hyperthermia, Induced methods, Models, Biological
- Abstract
Objective . Mathematical modeling can offer valuable insights into the behavior of biological systems upon treatment. Different mathematical models (empirical, semi-empirical, and mechanistic) have been designed to predict the efficacy of either hyperthermia (HT), radiotherapy (RT), or their combination. However, mathematical approaches capable of modeling cell survival from shared general principles for both mono-treatments alone and their co-application are rare. Moreover, some cell cultures show dose-dependent saturation in response to HT or RT, manifesting in survival curve flattenings. An advanced survival model must, therefore, appropriately reflect such behavior. Approach . We propose a mathematical approach to model the effect of both treatments based on the general principle of sublethal damage (SLD) accumulation for the induction of cell death and irreversible proliferation arrest. Our approach extends Jung's model on heat-induced cellular inactivation by incorporating dose-dependent recovery rates that delineate changes in SLD restoration. Main results . The resulting unified model (Umodel) accurately describes HT and RT survival outcomes, applies to simultaneous thermoradiotherapy modeling, and is particularly suited to reproduce survival curve flattening phenomena. We demonstrate the Umodel's robust performance (R2 0.95) based on numerous clonogenic cell survival data sets from the literature and our experimental studies. Significance . The proposed Umodel allows using a single unified mathematical function based on generalized principles of accumulation of SLD with implemented radiosensitization, regardless of the type of energy deposited and the mechanism of action. It can reproduce various patterns of clonogenic survival curves, including any flattening, thus encompassing the variability of cell reactions to therapy, thereby potentially better reflecting overall tumor responses. Our approach opens a range of options for further model developments and strategic therapy outcome predictions of sequential treatments applied in different orders and varying recovery intervals between them., (Creative Commons Attribution license.)
- Published
- 2025
- Full Text
- View/download PDF
5. A concept of dual-responsive prodrugs based on oligomerization-controlled reactivity of ester groups: an improvement of cancer cells versus neutrophils selectivity of camptothecin.
- Author
-
Klemt I, Reshetnikov V, Dutta S, Bila G, Bilyy R, Cuartero IC, Hidalgo A, Wünsche A, Böhm M, Wondrak M, Kunz-Schughart LA, Tietze R, Beierlein F, Imhof P, Gensberger-Reigl S, Pischetsrieder M, Körber M, Jost T, and Mokhir A
- Abstract
Many known chemotherapeutic anticancer agents exhibit neutropenia as a dose-limiting side effect. In this paper we suggest a prodrug concept solving this problem for camptothecin (HO-cpt). The prodrug is programmed according to Boolean "AND" logic. In the absence of H
2 O2 (trigger T1), e.g. in the majority of normal cells, it exists as an inactive oligomer. In cancer cells and in primed neutrophils (high H2 O2 ), the oligomer is disrupted forming intermediate (inactive) lipophilic cationic species. These are accumulated in mitochondria (Mit) of cancer cells, where they are activated by hydrolysis at mitochondrial pH 8 (trigger T2) with formation of camptothecin. In contrast, the intermediates remain stable in neutrophils lacking Mit and therefore a source of T2. In this paper we demonstrated a proof-of-concept. Our prodrug exhibits antitumor activity both in vitro and in vivo , but is not toxic to normal cell and neutrophils in contrast to known single trigger prodrugs and the parent drug HO-cpt., Competing Interests: There are no conflicts to declare., (This journal is © The Royal Society of Chemistry.)- Published
- 2024
- Full Text
- View/download PDF
6. Efficient Radial-Shell Model for 3D Tumor Spheroid Dynamics with Radiotherapy.
- Author
-
Franke F, Michlíková S, Aland S, Kunz-Schughart LA, Voss-Böhme A, and Lange S
- Abstract
Understanding the complex dynamics of tumor growth to develop more efficient therapeutic strategies is one of the most challenging problems in biomedicine. Three-dimensional (3D) tumor spheroids, reflecting avascular microregions within a tumor, are an advanced in vitro model system to assess the curative effect of combinatorial radio(chemo)therapy. Tumor spheroids exhibit particular crucial pathophysiological characteristics such as a radial oxygen gradient that critically affect the sensitivity of the malignant cell population to treatment. However, spheroid experiments remain laborious, and determining long-term radio(chemo)therapy outcomes is challenging. Mathematical models of spheroid dynamics have the potential to enhance the informative value of experimental data, and can support study design; however, they typically face one of two limitations: while non-spatial models are computationally cheap, they lack the spatial resolution to predict oxygen-dependent radioresponse, whereas models that describe spatial cell dynamics are computationally expensive and often heavily parameterized, impeding the required calibration to experimental data. Here, we present an effectively one-dimensional mathematical model based on the cell dynamics within and across radial spheres which fully incorporates the 3D dynamics of tumor spheroids by exploiting their approximate rotational symmetry. We demonstrate that this radial-shell (RS) model reproduces experimental spheroid growth curves of several cell lines with and without radiotherapy, showing equal or better performance than published models such as 3D agent-based models. Notably, the RS model is sufficiently efficient to enable multi-parametric optimization within previously reported and/or physiologically reasonable ranges based on experimental data. Analysis of the model reveals that the characteristic change of dynamics observed in experiments at small spheroid volume originates from the spatial scale of cell interactions. Based on the calibrated parameters, we predict the spheroid volumes at which this behavior should be observable. Finally, we demonstrate how the generic parameterization of the model allows direct parameter transfer to 3D agent-based models.
- Published
- 2023
- Full Text
- View/download PDF
7. Targeting of p21-Activated Kinase 4 Radiosensitizes Glioblastoma Cells via Impaired DNA Repair.
- Author
-
Blankenstein LJ, Cordes N, Kunz-Schughart LA, and Vehlow A
- Subjects
- DNA Breaks, Double-Stranded radiation effects, DNA Repair radiation effects, Humans, RNA, Small Interfering, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma radiotherapy, p21-Activated Kinases genetics, p21-Activated Kinases metabolism
- Abstract
Glioblastoma is a devastating malignant disease with poor patient overall survival. Strong invasiveness and resistance to radiochemotherapy have challenged the identification of molecular targets that can finally improve treatment outcomes. This study evaluates the influence of all six known p21-activated kinase (PAK) protein family members on the invasion capacity and radio-response of glioblastoma cells by employing a siRNA-based screen. In a panel of human glioblastoma cell models, we identified PAK4 as the main PAK isoform regulating invasion and clonogenic survival upon irradiation and demonstrated the radiosensitizing potential of PAK4 inhibition. Mechanistically, we show that PAK4 depletion and pharmacological inhibition enhanced the number of irradiation-induced DNA double-strand breaks and reduced the expression levels of various DNA repair proteins. In conclusion, our data suggest PAK4 as a putative target for radiosensitization and impairing DNA repair in glioblastoma, deserving further scrutiny in extended combinatorial treatment testing.
- Published
- 2022
- Full Text
- View/download PDF
8. Validation of CD98hc as a Therapeutic Target for a Combination of Radiation and Immunotherapies in Head and Neck Squamous Cell Carcinoma.
- Author
-
Köseer AS, Loureiro LR, Jureczek J, Mitwasi N, González Soto KE, Aepler J, Bartsch T, Feldmann A, Kunz-Schughart LA, Linge A, Krause M, Bachmann M, Arndt C, and Dubrovska A
- Abstract
Most patients with head and neck squamous cell carcinomas (HNSCC) are diagnosed at a locally advanced stage and show heterogeneous treatment responses. Low SLC3A2 (solute carrier family 3 member 2) mRNA and protein (CD98hc) expression levels are associated with higher locoregional control in HNSCC patients treated with primary radiochemotherapy or postoperative radiochemotherapy, suggesting that CD98hc could be a target for HNSCC radiosensitization. One of the targeted strategies for tumor radiosensitization is precision immunotherapy, e.g., the use of chimeric antigen receptor (CAR) T cells. This study aimed to define the potential clinical value of new treatment approaches combining conventional radiotherapy with CD98hc-targeted immunotherapy. To address this question, we analyzed the antitumor activity of the combination of fractionated irradiation and switchable universal CAR (UniCAR) system against radioresistant HNSCC cells in 3D culture. CD98hc-redirected UniCAR T cells showed the ability to destroy radioresistant HNSCC spheroids. Also, the infiltration rate of the UniCAR T cells was enhanced in the presence of the CD98hc target module. Furthermore, sequential treatment with fractionated irradiation followed by CD98hc-redirected UniCAR T treatment showed a synergistic effect. Taken together, our obtained data underline the improved antitumor effect of the combination of radiotherapy with CD98hc-targeted immunotherapy. Such a combination presents an attractive approach for the treatment of high-risk HNSCC patients.
- Published
- 2022
- Full Text
- View/download PDF
9. Intracellular Amplifiers of Reactive Oxygen Species Affecting Mitochondria as Radiosensitizers.
- Author
-
Xu HG, Reshetnikov V, Wondrak M, Eckhardt L, Kunz-Schughart LA, Janko C, Tietze R, Alexiou C, Borchardt H, Aigner A, Gong W, Schmitt M, Sellner L, Daum S, Özkan HG, and Mokhir A
- Abstract
Radiotherapy (RT) efficacy can be improved by using radiosensitizers, i.e., drugs enhancing the effect of ionizing radiation (IR). One of the side effects of RT includes damage of normal tissue in close proximity to the treated tumor. This problem can be solved by applying cancer specific radiosensitizers. N -Alkylaminoferrocene-based (NAAF) prodrugs produce reactive oxygen species (ROS) in cancer cells, but not in normal cells. Therefore, they can potentially act as cancer specific radiosensitizers. However, early NAAF prodrugs did not exhibit this property. Since functional mitochondria are important for RT resistance, we assumed that NAAF prodrugs affecting mitochondria in parallel with increasing intracellular ROS can potentially exhibit synergy with RT. We applied sequential Cu
+ -catalyzed alkyne-azide cycloadditions (CuAAC) to obtain a series of NAAF derivatives with the goal of improving anticancer efficacies over already existing compounds. One of the obtained prodrugs (2c) exhibited high anticancer activity with IC50 values in the range of 5-7.1 µM in human ovarian carcinoma, Burkitt's lymphoma, pancreatic carcinoma and T-cell leukemia cells retained moderate water solubility and showed cancer specificity. 2c strongly affects mitochondria of cancer cells, leading to the amplification of mitochondrial and total ROS production and thus causing cell death via necrosis and apoptosis. We observed that 2c acts as a radiosensitizer in human head and neck squamous carcinoma cells. This is the first demonstration of a synergy between the radiotherapy and NAAF-based ROS amplifiers.- Published
- 2021
- Full Text
- View/download PDF
10. MISpheroID: a knowledgebase and transparency tool for minimum information in spheroid identity.
- Author
-
Peirsman A, Blondeel E, Ahmed T, Anckaert J, Audenaert D, Boterberg T, Buzas K, Carragher N, Castellani G, Castro F, Dangles-Marie V, Dawson J, De Tullio P, De Vlieghere E, Dedeyne S, Depypere H, Diosdi A, Dmitriev RI, Dolznig H, Fischer S, Gespach C, Goossens V, Heino J, Hendrix A, Horvath P, Kunz-Schughart LA, Maes S, Mangodt C, Mestdagh P, Michlíková S, Oliveira MJ, Pampaloni F, Piccinini F, Pinheiro C, Rahn J, Robbins SM, Siljamäki E, Steigemann P, Sys G, Takayama S, Tesei A, Tulkens J, Van Waeyenberge M, Vandesompele J, Wagemans G, Weindorfer C, Yigit N, Zablowsky N, Zanoni M, Blondeel P, and De Wever O
- Subjects
- Cell Culture Techniques methods, Gene Expression Regulation, Neoplastic, Humans, Neoplasms classification, Neoplasms metabolism, RNA-Seq, Reproducibility of Results, Spheroids, Cellular immunology, Spheroids, Cellular metabolism, Tumor Cells, Cultured, Biomarkers, Tumor genetics, Cell Proliferation, Knowledge Bases, Neoplasms pathology, Software, Spheroids, Cellular pathology, Tumor Microenvironment
- Abstract
Spheroids are three-dimensional cellular models with widespread basic and translational application across academia and industry. However, methodological transparency and guidelines for spheroid research have not yet been established. The MISpheroID Consortium developed a crowdsourcing knowledgebase that assembles the experimental parameters of 3,058 published spheroid-related experiments. Interrogation of this knowledgebase identified heterogeneity in the methodological setup of spheroids. Empirical evaluation and interlaboratory validation of selected variations in spheroid methodology revealed diverse impacts on spheroid metrics. To facilitate interpretation, stimulate transparency and increase awareness, the Consortium defines the MISpheroID string, a minimum set of experimental parameters required to report spheroid research. Thus, MISpheroID combines a valuable resource and a tool for three-dimensional cellular models to mine experimental parameters and to improve reproducibility., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
11. Models for Translational Proton Radiobiology-From Bench to Bedside and Back.
- Author
-
Suckert T, Nexhipi S, Dietrich A, Koch R, Kunz-Schughart LA, Bahn E, and Beyreuther E
- Abstract
The number of proton therapy centers worldwide are increasing steadily, with more than two million cancer patients treated so far. Despite this development, pending questions on proton radiobiology still call for basic and translational preclinical research. Open issues are the on-going discussion on an energy-dependent varying proton RBE (relative biological effectiveness), a better characterization of normal tissue side effects and combination treatments with drugs originally developed for photon therapy. At the same time, novel possibilities arise, such as radioimmunotherapy, and new proton therapy schemata, such as FLASH irradiation and proton mini-beams. The study of those aspects demands for radiobiological models at different stages along the translational chain, allowing the investigation of mechanisms from the molecular level to whole organisms. Focusing on the challenges and specifics of proton research, this review summarizes the different available models, ranging from in vitro systems to animal studies of increasing complexity as well as complementing in silico approaches.
- Published
- 2021
- Full Text
- View/download PDF
12. GLS-driven glutamine catabolism contributes to prostate cancer radiosensitivity by regulating the redox state, stemness and ATG5-mediated autophagy.
- Author
-
Mukha A, Kahya U, Linge A, Chen O, Löck S, Lukiyanchuk V, Richter S, Alves TC, Peitzsch M, Telychko V, Skvortsov S, Negro G, Aschenbrenner B, Skvortsova II, Mirtschink P, Lohaus F, Hölscher T, Neubauer H, Rivandi M, Labitzky V, Lange T, Franken A, Behrens B, Stoecklein NH, Toma M, Sommer U, Zschaeck S, Rehm M, Eisenhofer G, Schwager C, Abdollahi A, Groeben C, Kunz-Schughart LA, Baretton GB, Baumann M, Krause M, Peitzsch C, and Dubrovska A
- Subjects
- Animals, Autophagy, Autophagy-Related Protein 5 metabolism, Biomarkers, Pharmacological, Cell Line, Tumor, Glutaminase antagonists & inhibitors, Glutaminase genetics, Glutaminase metabolism, Humans, Male, Mice, Nude, Neoplastic Stem Cells metabolism, Oxidation-Reduction, Proto-Oncogene Proteins c-myc metabolism, Reactive Oxygen Species metabolism, Xenograft Model Antitumor Assays, Mice, Glutamine metabolism, Prostatic Neoplasms metabolism, Radiation Tolerance genetics
- Abstract
Radiotherapy is one of the curative treatment options for localized prostate cancer (PCa). The curative potential of radiotherapy is mediated by irradiation-induced oxidative stress and DNA damage in tumor cells. However, PCa radiocurability can be impeded by tumor resistance mechanisms and normal tissue toxicity. Metabolic reprogramming is one of the major hallmarks of tumor progression and therapy resistance. Specific metabolic features of PCa might serve as therapeutic targets for tumor radiosensitization and as biomarkers for identifying the patients most likely to respond to radiotherapy. The study aimed to characterize a potential role of glutaminase (GLS)-driven glutamine catabolism as a prognostic biomarker and a therapeutic target for PCa radiosensitization. Methods: We analyzed primary cell cultures and radioresistant (RR) derivatives of the conventional PCa cell lines by gene expression and metabolic assays to identify the molecular traits associated with radiation resistance. Relative radiosensitivity of the cell lines and primary cell cultures were analyzed by 2-D and 3-D clonogenic analyses. Targeting of glutamine (Gln) metabolism was achieved by Gln starvation, gene knockdown, and chemical inhibition. Activation of the DNA damage response (DDR) and autophagy was assessed by gene expression, western blotting, and fluorescence microscopy. Reactive oxygen species (ROS) and the ratio of reduced glutathione (GSH) to oxidized glutathione (GSSG) were analyzed by fluorescence and luminescence probes, respectively. Cancer stem cell (CSC) properties were investigated by sphere-forming assay, CSC marker analysis, and in vivo limiting dilution assays. Single circulating tumor cells (CTCs) isolated from the blood of PCa patients were analyzed by array comparative genome hybridization. Expression levels of the GLS1 and MYC gene in tumor tissues and amino acid concentrations in blood plasma were correlated to a progression-free survival in PCa patients. Results: Here, we found that radioresistant PCa cells and prostate CSCs have a high glutamine demand. GLS-driven catabolism of glutamine serves not only for energy production but also for the maintenance of the redox state. Consequently, glutamine depletion or inhibition of critical regulators of glutamine utilization, such as GLS and the transcription factor MYC results in PCa radiosensitization. On the contrary, we found that a combination of glutamine metabolism inhibitors with irradiation does not cause toxic effects on nonmalignant prostate cells. Glutamine catabolism contributes to the maintenance of CSCs through regulation of the alpha-ketoglutarate (α-KG)-dependent chromatin-modifying dioxygenase. The lack of glutamine results in the inhibition of CSCs with a high aldehyde dehydrogenase (ALDH) activity, decreases the frequency of the CSC populations in vivo and reduces tumor formation in xenograft mouse models. Moreover, this study shows that activation of the ATG5-mediated autophagy in response to a lack of glutamine is a tumor survival strategy to withstand radiation-mediated cell damage. In combination with autophagy inhibition, the blockade of glutamine metabolism might be a promising strategy for PCa radiosensitization. High blood levels of glutamine in PCa patients significantly correlate with a shorter prostate-specific antigen (PSA) doubling time. Furthermore, high expression of critical regulators of glutamine metabolism, GLS1 and MYC, is significantly associated with a decreased progression-free survival in PCa patients treated with radiotherapy. Conclusions: Our findings demonstrate that GLS-driven glutaminolysis is a prognostic biomarker and therapeutic target for PCa radiosensitization., Competing Interests: Competing Interests: In the past 5 years, Dr. Mechthild Krause received funding for her research projects by IBA (2016), Merck KGaA (2014-2018 for preclinical study; 2018-2020 for clinical study), Medipan GmbH (2014-2018). In the past 5 years, Dr. Krause, Dr. Linge and Dr. Löck have been involved in an ongoing publicly funded (German Federal Ministry of Education and Research) project with the companies Medipan, Attomol GmbH, GA Generic Assays GmbH, Gesellschaft für medizinische und wissenschaftliche genetische Analysen, Lipotype GmbH and PolyAn GmbH (2019-2021). For the present manuscript, none of the above mentioned funding sources were involved. In the past 5 years, Dr. Michael Baumann received funding for his research projects and for educational grants to the University of Dresden by Bayer AG (2016-2018), Merck KGaA (2014-open) and Medipan GmbH (2014-2018). He is on the supervisory board of HI-STEM gGmbH (Heidelberg) for the German Cancer Research Center (DKFZ, Heidelberg) and also member of the supervisory body of the Charité University Hospital, Berlin. As former chair of OncoRay (Dresden) and present CEO and Scientific Chair of the German Cancer Research Center (DKFZ, Heidelberg), he has been or is responsible for collaborations with a multitude of companies and institutions, worldwide. In this capacity, he has discussed potential projects and signed contracts for research funding and/or collaborations with industry and academia for his institute(s) and staff, including but not limited to pharmaceutical companies such as Bayer, Boehringer Ingelheim, Bosch, Roche and other companies such as Siemens, IBA, Varian, Elekta, Bruker, etc. In this role, he was/is also responsible for the commercial technology transfer activities of his institute(s), including the creation of start-ups and licensing. This includes the DKFZ-PSMA617 related patent portfolio [WO2015055318 (A1), ANTIGEN (PSMA)] and similar IP portfolios. Dr. Baumann confirms that, to the best of his knowledge, none of the above funding sources were involved in the preparation of this paper. Other co-authors declare that they have no conflict of interest., (© The author(s).)
- Published
- 2021
- Full Text
- View/download PDF
13. Efficient Heat Shock Response Affects Hyperthermia-Induced Radiosensitization in a Tumor Spheroid Control Probability Assay.
- Author
-
Chen O, Michlíková S, Eckhardt L, Wondrak M, De Mendoza AM, Krause M, McLeod DD, and Kunz-Schughart LA
- Abstract
Hyperthermia (HT) combined with irradiation is a well-known concept to improve the curative potential of radiotherapy. Technological progress has opened new avenues for thermoradiotherapy, even for recurrent head and neck squamous cell carcinomas (HNSCC). Preclinical evaluation of the curative radiosensitizing potential of various HT regimens remains ethically, economically, and technically challenging. One key objective of our study was to refine an advanced 3-D assay setup for HT + RT research and treatment testing. For the first time, HT-induced radiosensitization was systematically examined in two differently radioresponsive HNSCC spheroid models using the unique in vitro "curative" analytical endpoint of spheroid control probability. We further investigated the cellular stress response mechanisms underlying the HT-related radiosensitization process with the aim to unravel the impact of HT-induced proteotoxic stress on the overall radioresponse. HT disrupted the proteome's thermal stability, causing severe proteotoxic stress. It strongly enhanced radiation efficacy and affected paramount survival and stress response signaling networks. Transcriptomics, q-PCR, and western blotting data revealed that HT + RT co-treatment critically triggers the heat shock response (HSR). Pre-treatment with chemical chaperones intensified the radiosensitizing effect, thereby suppressing HT-induced Hsp27 expression. Our data suggest that HT-induced radiosensitization is adversely affected by the proteotoxic stress response. Hence, we propose the inhibition of particular heat shock proteins as a targeting strategy to improve the outcome of combinatorial HT + RT.
- Published
- 2021
- Full Text
- View/download PDF
14. Mathematical model for the thermal enhancement of radiation response: thermodynamic approach.
- Author
-
De Mendoza AM, Michlíková S, Berger J, Karschau J, Kunz-Schughart LA, and McLeod DD
- Abstract
Radiotherapy can effectively kill malignant cells, but the doses required to cure cancer patients may inflict severe collateral damage to adjacent healthy tissues. Recent technological advances in the clinical application has revitalized hyperthermia treatment (HT) as an option to improve radiotherapy (RT) outcomes. Understanding the synergistic effect of simultaneous thermoradiotherapy via mathematical modelling is essential for treatment planning. We here propose a theoretical model in which the thermal enhancement ratio (TER) relates to the cell fraction being radiosensitised by the infliction of sublethal damage through HT. Further damage finally kills the cell or abrogates its proliferative capacity in a non-reversible process. We suggest the TER to be proportional to the energy invested in the sensitisation, which is modelled as a simple rate process. Assuming protein denaturation as the main driver of HT-induced sublethal damage and considering the temperature dependence of the heat capacity of cellular proteins, the sensitisation rates were found to depend exponentially on temperature; in agreement with previous empirical observations. Our findings point towards an improved definition of thermal dose in concordance with the thermodynamics of protein denaturation. Our predictions well reproduce experimental in vitro and in vivo data, explaining the thermal modulation of cellular radioresponse for simultaneous thermoradiotherapy.
- Published
- 2021
- Full Text
- View/download PDF
15. Dual role of ER stress in response to metabolic co-targeting and radiosensitivity in head and neck cancer cells.
- Author
-
Chen O, Manig F, Lehmann L, Sorour N, Löck S, Yu Z, Dubrovska A, Baumann M, Kessler BM, Stasyk O, and Kunz-Schughart LA
- Subjects
- Activating Transcription Factor 4 antagonists & inhibitors, Activating Transcription Factor 4 genetics, Activating Transcription Factor 4 metabolism, Apoptosis drug effects, Arginine deficiency, Arginine metabolism, Cell Culture Techniques, Cell Line, Tumor, Cell Proliferation drug effects, Culture Media chemistry, Endoribonucleases antagonists & inhibitors, Endoribonucleases genetics, Endoribonucleases metabolism, Eukaryotic Initiation Factor-2 genetics, Eukaryotic Initiation Factor-2 metabolism, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms pathology, Humans, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA Interference, RNA, Small Interfering metabolism, Signal Transduction drug effects, Squamous Cell Carcinoma of Head and Neck metabolism, Squamous Cell Carcinoma of Head and Neck pathology, Transcription Factor CHOP antagonists & inhibitors, Transcription Factor CHOP genetics, Transcription Factor CHOP metabolism, Canavanine pharmacology, Endoplasmic Reticulum Stress drug effects, Radiation Tolerance drug effects, X-Rays
- Abstract
Arginine deprivation therapy (ADT) is a new metabolic targeting approach with high therapeutic potential for various solid cancers. Combination of ADT with low doses of the natural arginine analog canavanine effectively sensitizes malignant cells to irradiation. However, the molecular mechanisms determining the sensitivity of intrinsically non-auxotrophic cancers to arginine deficiency are still poorly understood. We here show for the first time that arginine deficiency is accompanied by global metabolic changes and protein/membrane breakdown, and results in the induction of specific, more or less pronounced (severe vs. mild) ER stress responses in head and neck squamous cell carcinoma (HNSCC) cells that differ in their intrinsic ADT sensitivity. Combination of ADT with canavanine triggered catastrophic ER stress via the eIF2α-ATF4(GADD34)-CHOP pathway, thereby inducing apoptosis; the same signaling arm was irrelevant in ADT-related radiosensitization. The particular strong supra-additive effect of ADT, canavanine and irradiation in both intrinsically more and less sensitive cancer cells supports the rational of ER stress pathways as novel target for improving multi-modal metabolic anti-cancer therapy.
- Published
- 2021
- Full Text
- View/download PDF
16. Indospicine combined with arginine deprivation triggers cancer cell death via caspase-dependent apoptosis.
- Author
-
Shuvayeva GY, Bobak YP, Vovk OI, Kunz-Schughart LA, Fletcher MT, and Stasyk OV
- Subjects
- Arginase metabolism, Arginine chemistry, Cell Line, Tumor, Cell Survival drug effects, Humans, Norleucine chemistry, Norleucine pharmacology, Protein Biosynthesis drug effects, Signal Transduction drug effects, Substrate Specificity drug effects, Apoptosis drug effects, Arginine deficiency, Neoplasms pathology, Norleucine analogs & derivatives
- Abstract
Arginine-deprivation therapy is a rapidly developing metabolic anticancer approach. To overcome the resistance of some cancer cells to this monotherapy, rationally designed combination modalities are needed. In this report, we evaluated for the first time indospicine, an arginine analogue of Indigofera plant genus origin, as potential enhancer compound for the metabolic therapy that utilizes recombinant human arginase I. We demonstrate that indospicine at low micromolar concentrations is selectively toxic for human colorectal cancer cells only in the absence of arginine. In arginine-deprived cancer cells indospicine deregulates some prosurvival pathways (PI3K-Akt and MAPK) and activates mammalian target of rapamycin, exacerbates endoplasmic reticulum stress and triggers caspase-dependent apoptosis, which is reversed by the exposure to translation inhibitors. Simultaneously, indospicine is not degraded by recombinant human arginase I and does not inhibit this arginine-degrading enzyme at its effective dose. The obtained results emphasize the potential of arginine structural analogues as efficient components for combinatorial metabolic targeting of malignant cells., (© 2020 International Federation for Cell Biology.)
- Published
- 2021
- Full Text
- View/download PDF
17. Reciprocal interactions between tumour cell populations enhance growth and reduce radiation sensitivity in prostate cancer.
- Author
-
Paczkowski M, Kretzschmar WW, Markelc B, Liu SK, Kunz-Schughart LA, Harris AL, Partridge M, Byrne HM, and Kannan P
- Subjects
- Cell Line, Tumor, Humans, Male, Spheroids, Cellular, Models, Biological, Neoplasm Recurrence, Local etiology, Prostatic Neoplasms, Radiation Tolerance
- Abstract
Intratumoural heterogeneity (ITH) contributes to local recurrence following radiotherapy in prostate cancer. Recent studies also show that ecological interactions between heterogeneous tumour cell populations can lead to resistance in chemotherapy. Here, we evaluated whether interactions between heterogenous populations could impact growth and response to radiotherapy in prostate cancer. Using mixed 3D cultures of parental and radioresistant populations from two prostate cancer cell lines and a predator-prey mathematical model to investigate various types of ecological interactions, we show that reciprocal interactions between heterogeneous populations enhance overall growth and reduce radiation sensitivity. The type of interaction influences the time of regrowth after radiation, and, at the population level, alters the survival and cell cycle of each population without eliminating either one. These interactions can arise from oxygen constraints and from cellular cross-talk that alter the tumour microenvironment. These findings suggest that ecological-type interactions are important in radiation response and could be targeted to reduce local recurrence.
- Published
- 2021
- Full Text
- View/download PDF
18. Publisher Correction: Spectral and spatial shaping of laser-driven proton beams using a pulsed high-field magnet beamline.
- Author
-
Brack FE, Kroll F, Gaus L, Bernert C, Beyreuther E, Cowan TE, Karsch L, Kraft S, Kunz-Schughart LA, Lessmann E, Metzkes-Ng J, Obst-Huebl L, Pawelke J, Rehwald M, Schlenvoigt HP, Schramm U, Sobiella M, Szabó ER, Ziegler T, and Zeil K
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
19. Microenvironmentally-driven Plasticity of CD44 isoform expression determines Engraftment and Stem-like Phenotype in CRC cell lines.
- Author
-
Dinger TF, Chen O, Dittfeld C, Hetze L, Hüther M, Wondrak M, Löck S, Eicheler W, Breier G, and Kunz-Schughart LA
- Subjects
- AC133 Antigen metabolism, Animals, Biomarkers, Tumor metabolism, Cancer-Associated Fibroblasts pathology, Cell Line, Tumor, Cell Plasticity, Cell Separation, Female, Flow Cytometry, Human Umbilical Vein Endothelial Cells, Humans, Mice, Protein Isoforms metabolism, Xenograft Model Antitumor Assays, Carcinogenesis pathology, Colorectal Neoplasms pathology, Hyaluronan Receptors metabolism, Neoplastic Stem Cells pathology, Tumor Microenvironment
- Abstract
Theranostic biomarkers for putative cancer stem-like cells (CSC) in colorectal cancer (CRC) are of particular interest in translational research to develop patient-individualized treatment strategies. Surface proteins still under debate are CD44 and CD133. The structural and functional diversity of these antigens, as well as their plasticity, has only just begun to be understood. Our study aimed to gain novel insight into the plasticity of CD133/CD44, thereby proving the hypothesis of marker-associated tumorigenic and non-tumorigenic phenotypes to be environmentally driven. Methods: CD133/CD44 profiles of 20 CRC cell lines were monitored; three models with distinct surface patterns in vitro were systematically examined. CD133/CD44 subpopulations were isolated by FACS and analyzed upon in vitro growth and/or in limiting dilution engraftment studies. The experimental setup included biomarker analyses on the protein (flow cytometry, Western blotting, immunofluorescence) and mRNA levels (RT-/qPCR) as well as CD44 gene sequencing. Results: In general, we found that (i) the in vitro CD133/CD44 pattern never determined engraftment and (ii) the CD133/CD44 population distributions harmonized under in vivo conditions. The LS1034 cell line appeared as a unique model due to its de novo in vivo presentation of CD44. CD44v8-10 was identified as main transcript, which was stronger expressed in primary human CRC than in normal colon tissues. Biomarker pattern of LS1034 cells in vivo reflected secondary engraftment: the tumorigenic potential was highest in CD133
+ /CD44+ , intermediate in CD133+ /CD44- and entirely lost in CD133- /CD44- subfractions. Both CD44+ and CD44- LS1034 cells gave rise to tumorigenic and non-tumorigenic progeny and were convertible - but only as long as they expressed CD133 in vivo . The highly tumorigenic CD133+ /CD44(v8-10)+ LS1034 cells were localized in well-oxygenated perivascular but not hypoxic regions. From a multitude of putative modulators, only the direct interaction with stromal fibroblasts triggered an essential, in vivo -like enhancement of CD44v8-10 presentation in vitro . Conclusion: Environmental conditions modulate CD133/CD44 phenotypes and tumorigenic potential of CRC subpopulations. The identification of fibroblasts as drivers of cancer-specific CD44 expression profile and plasticity sheds light on the limitation of per se dynamic surface antigens as biomarkers. It can also explain the location of putative CD133/CD44-positive CRC CSC in the perivascular niche, which is likely to comprise cancer-associated fibroblasts. The LS1034 in vitro/in vivo model is a valuable tool to unravel the mechanism of stromal-induced CD44v8-10 expression and identify further therapeutically relevant, mutual interrelations between microenvironment and tumorigenic phenotype., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)- Published
- 2020
- Full Text
- View/download PDF
20. Spectral and spatial shaping of laser-driven proton beams using a pulsed high-field magnet beamline.
- Author
-
Brack FE, Kroll F, Gaus L, Bernert C, Beyreuther E, Cowan TE, Karsch L, Kraft S, Kunz-Schughart LA, Lessmann E, Metzkes-Ng J, Obst-Huebl L, Pawelke J, Rehwald M, Schlenvoigt HP, Schramm U, Sobiella M, Szabó ER, Ziegler T, and Zeil K
- Abstract
Intense laser-driven proton pulses, inherently broadband and highly divergent, pose a challenge to established beamline concepts on the path to application-adapted irradiation field formation, particularly for 3D. Here we experimentally show the successful implementation of a highly efficient (50% transmission) and tuneable dual pulsed solenoid setup to generate a homogeneous (laterally and in depth) volumetric dose distribution (cylindrical volume of 5 mm diameter and depth) at a single pulse dose of 0.7 Gy via multi-energy slice selection from the broad input spectrum. The experiments were conducted at the Petawatt beam of the Dresden Laser Acceleration Source Draco and were aided by a predictive simulation model verified by proton transport studies. With the characterised beamline we investigated manipulation and matching of lateral and depth dose profiles to various desired applications and targets. Using an adapted dose profile, we performed a first proof-of-technical-concept laser-driven proton irradiation of volumetric in-vitro tumour tissue (SAS spheroids) to demonstrate concurrent operation of laser accelerator, beam shaping, dosimetry and irradiation procedure of volumetric biological samples.
- Published
- 2020
- Full Text
- View/download PDF
21. SATB1 as oncogenic driver and potential therapeutic target in head & neck squamous cell carcinoma (HNSCC).
- Author
-
Panchal O, Wichmann G, Grenman R, Eckhardt L, Kunz-Schughart LA, Franke H, Dietz A, and Aigner A
- Subjects
- Animals, Apoptosis, Cell Cycle Checkpoints, Cell Line, Tumor, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms metabolism, Humans, Matrix Attachment Region Binding Proteins antagonists & inhibitors, Matrix Attachment Region Binding Proteins genetics, Mice, Mice, Inbred NOD, Mice, SCID, Nanoparticles chemistry, Neoplasm Staging, Neuregulin-1 genetics, Neuregulin-1 metabolism, RNA Interference, RNA, Small Interfering chemistry, RNA, Small Interfering metabolism, RNA, Small Interfering therapeutic use, Receptor, ErbB-3 genetics, Receptor, ErbB-3 metabolism, Squamous Cell Carcinoma of Head and Neck drug therapy, Squamous Cell Carcinoma of Head and Neck metabolism, Transplantation, Heterologous, Up-Regulation, Head and Neck Neoplasms pathology, Matrix Attachment Region Binding Proteins metabolism, Squamous Cell Carcinoma of Head and Neck pathology
- Abstract
The Special AT-rich sequence binding protein 1 (SATB1) is a genome organizer protein that controls gene expression of numerous genes by regulating chromatin architecture and targeting chromatin-remodeling/-modifying enzymes onto specific chromatin regions. SATB1 is overexpressed in various tumors. In head and neck squamous cell carcinoma (HNSCC), SATB1 upregulation is correlated with TNM classification, metastasis, poor prognosis and reduced overall survival. In this paper, we comprehensively analyze cellular and molecular effects of SATB1 in a large set of primary cell lines from primary HNSCC or metastases, using RNAi-mediated knockdown in vitro and, therapeutically, in tumor xenograft mouse models in vivo. In a series of 15 cell lines, major differences in SATB1 levels are observed. In various 2-D and 3-D assays, growth inhibition upon efficient siRNA-mediated SATB1 knockdown depends on the cell line rather than initial SATB1 levels. Inhibitory effects are found to be based on cell cycle deceleration, apoptosis induction, decreased HER3 and Heregulin A&B expression, and effects on EMT genes. In vivo, systemic treatment of tumor xenograft-bearing mice with siRNAs formulated in polymeric nanoparticles inhibits tumor growth of two HNSCC xenograft models, resulting from therapeutic SATB1 reduction and concomitant decrease of proliferation and induction of apoptosis. In conclusion, SATB1 represents a promising target in HNSCC, affecting crucial cellular processes and molecular pathways.
- Published
- 2020
- Full Text
- View/download PDF
22. An ovarian spheroid based tumor model that represents vascularized tumors and enables the investigation of nanomedicine therapeutics.
- Author
-
Singh MS, Goldsmith M, Thakur K, Chatterjee S, Landesman-Milo D, Levy T, Kunz-Schughart LA, Barenholz Y, and Peer D
- Subjects
- Animals, Cell Line, Tumor, Doxorubicin pharmacology, Female, Heterografts, Humans, Mice, Neoplasm Transplantation, Polyethylene Glycols pharmacology, Xenograft Model Antitumor Assays, Bevacizumab pharmacology, Doxorubicin analogs & derivatives, Neoplasms, Experimental blood supply, Neoplasms, Experimental drug therapy, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Ovarian Neoplasms blood supply, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Spheroids, Cellular metabolism, Spheroids, Cellular pathology
- Abstract
The failure of cancer therapies in clinical settings is often attributed to the lack of a relevant tumor model and pathological heterogeneity across tumor types in the clinic. The objective of this study was to develop a robust in vivo tumor model that better represents clinical tumors for the evaluation of anti-cancer therapies. We successfully developed a simple mouse tumor model based on 3D cell culture by injecting a single spheroid and compared it to a tumor model routinely used by injecting cell suspension from 2D monolayer cell culture. We further characterized both tumors with cellular markers for the presence of myofibroblasts, pericytes, endothelial cells and extracellular matrix to understand the role of the tumor microenvironment. We further investigated the effect of chemotherapy (doxorubicin), nanomedicine (Doxil®), biological therapy (Avastin®) and their combination. Our results showed that the substantial blood vasculature in the 3D spheroid model enhances the delivery of Doxil® by 2.5-fold as compared to the 2D model. Taken together, our data suggest that the 3D tumors created by simple subcutaneous spheroid injection represents a robust and more vascular murine tumor model which is a clinically relevant platform to test anti-cancer therapy in solid tumors.
- Published
- 2020
- Full Text
- View/download PDF
23. Mutant IDH1 Differently Affects Redox State and Metabolism in Glial Cells of Normal and Tumor Origin.
- Author
-
Biedermann J, Preussler M, Conde M, Peitzsch M, Richter S, Wiedemuth R, Abou-El-Ardat K, Krüger A, Meinhardt M, Schackert G, Leenders WP, Herold-Mende C, Niclou SP, Bjerkvig R, Eisenhofer G, Temme A, Seifert M, Kunz-Schughart LA, Schröck E, and Klink B
- Abstract
IDH1
R132H (isocitrate dehydrogenase 1) mutations play a key role in the development of low-grade gliomas. IDH1wt converts isocitrate to α-ketoglutarate while reducing nicotinamide adenine dinucleotide phosphate (NADP+ ), whereas IDH1R132H uses α-ketoglutarate and NADPH to generate the oncometabolite 2-hydroxyglutarate (2-HG). While the effects of 2-HG have been the subject of intense research, the 2-HG independent effects of IDH1R132H are still ambiguous. The present study demonstrates that IDH1R132H expression but not 2-HG alone leads to significantly decreased tricarboxylic acid (TCA) cycle metabolites, reduced proliferation, and enhanced sensitivity to irradiation in both glioblastoma cells and astrocytes in vitro. Glioblastoma cells, but not astrocytes, showed decreased NADPH and NAD+ levels upon IDH1R132H transduction. However, in astrocytes IDH1R132H led to elevated expression of the NAD-synthesizing enzyme nicotinamide phosphoribosyltransferase (NAMPT). These effects were not 2-HG mediated. This suggests that IDH1R132H cells utilize NAD+ to restore NADP pools, which only astrocytes could compensate via induction of NAMPT. We found that the expression of NAMPT is lower in patient-derived IDH1-mutant glioma cells and xenografts compared to IDH1-wildtype models. The Cancer Genome Atlas (TCGA) data analysis confirmed lower NAMPT expression in IDH1-mutant versus IDH1-wildtype gliomas. We show that the IDH1 mutation directly affects the energy homeostasis and redox state in a cell-type dependent manner. Targeting the impairments in metabolism and redox state might open up new avenues for treating IDH1-mutant gliomas.- Published
- 2019
- Full Text
- View/download PDF
24. The CD98 Heavy Chain Is a Marker and Regulator of Head and Neck Squamous Cell Carcinoma Radiosensitivity.
- Author
-
Digomann D, Kurth I, Tyutyunnykova A, Chen O, Löck S, Gorodetska I, Peitzsch C, Skvortsova II, Negro G, Aschenbrenner B, Eisenhofer G, Richter S, Heiden S, Porrmann J, Klink B, Schwager C, Dowle AA, Hein L, Kunz-Schughart LA, Abdollahi A, Lohaus F, Krause M, Baumann M, Linge A, and Dubrovska A
- Subjects
- Amino Acids metabolism, Biological Transport, Biomarkers, Tumor, Cell Line, Tumor, Chemoradiotherapy, Citric Acid Cycle, Fusion Regulatory Protein 1, Heavy Chain metabolism, Gene Expression, Gene Knockdown Techniques, Humans, Immunohistochemistry, Large Neutral Amino Acid-Transporter 1 genetics, Large Neutral Amino Acid-Transporter 1 metabolism, Oxidative Stress genetics, Squamous Cell Carcinoma of Head and Neck mortality, Squamous Cell Carcinoma of Head and Neck pathology, Squamous Cell Carcinoma of Head and Neck radiotherapy, Fusion Regulatory Protein 1, Heavy Chain genetics, Radiation Tolerance genetics, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Purpose: The heavy chain of the CD98 protein (CD98hc) is encoded by the SLC3A2 gene. Together with the light subunit LAT1, CD98hc constitutes a heterodimeric transmembrane amino acid transporter. High SLC3A2 mRNA expression levels are associated with poor prognosis in patients with head and neck squamous cell carcinoma (HNSCC) treated with radiochemotherapy. Little is known regarding the CD98hc protein-mediated molecular mechanisms of tumor radioresistance., Experimental Design: CD98hc protein expression levels were correlated with corresponding tumor control dose 50 (TCD
50 ) in HNSCC xenograft models. Expression levels of CD98hc and LAT1 in HNSCC cells were modulated by siRNA or CRISPR/Cas9 gene editing. HNSCC cell phenotypes were characterized by transcription profiling, plasma membrane proteomics, metabolic analysis, and signaling pathway activation. Expression levels of CD98hc and LAT1 proteins were examined by IHC analysis of tumor tissues from patients with locally advanced HNSCC treated with primary radiochemotherapy (RCTx). Primary endpoint was locoregional tumor control (LRC)., Results: High expression levels of CD98hc resulted in an increase in mTOR pathway activation, amino acid metabolism, and DNA repair as well as downregulation of oxidative stress and autophagy. High expression levels of CD98hc and LAT1 proteins were significantly correlated and associated with an increase in radioresistance in HNSCC in vitro and in vivo models. High expression of both proteins identified a poor prognosis subgroup in patients with locally advanced HNSCC after RCTx., Conclusions: We found that CD98hc-associated signaling mechanisms play a central role in the regulation of HNSCC radioresistance and may be a promising target for tumor radiosensitization., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
25. Oxidative Phosphorylation as an Emerging Target in Cancer Therapy.
- Author
-
Ashton TM, McKenna WG, Kunz-Schughart LA, and Higgins GS
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Biomarkers, Energy Metabolism drug effects, Humans, Metabolic Networks and Pathways, Mitochondria drug effects, Mitochondria genetics, Mitochondria metabolism, Neoplasms etiology, Molecular Targeted Therapy methods, Neoplasms metabolism, Neoplasms therapy, Oxidative Phosphorylation drug effects
- Abstract
Cancer cells have upregulated glycolysis compared with normal cells, which has led many to the assumption that oxidative phosphorylation (OXPHOS) is downregulated in all cancers. However, recent studies have shown that OXPHOS can be also upregulated in certain cancers, including leukemias, lymphomas, pancreatic ductal adenocarcinoma, high OXPHOS subtype melanoma, and endometrial carcinoma, and that this can occur even in the face of active glycolysis. OXPHOS inhibitors could therefore be used to target cancer subtypes in which OXPHOS is upregulated and to alleviate therapeutically adverse tumor hypoxia. Several drugs including metformin, atovaquone, and arsenic trioxide are used clinically for non-oncologic indications, but emerging data demonstrate their potential use as OXPHOS inhibitors. We highlight novel applications of OXPHOS inhibitors with a suitable therapeutic index to target cancer cell metabolism. Clin Cancer Res; 24(11); 2482-90. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
26. Arginine Deprivation Therapy: Putative Strategy to Eradicate Glioblastoma Cells by Radiosensitization.
- Author
-
Hinrichs CN, Ingargiola M, Käubler T, Löck S, Temme A, Köhn-Luque A, Deutsch A, Vovk O, Stasyk O, and Kunz-Schughart LA
- Subjects
- Glioblastoma pathology, Humans, Radiation-Sensitizing Agents pharmacology, Arginine metabolism, Glioblastoma drug therapy, Glioblastoma radiotherapy, Radiation-Sensitizing Agents therapeutic use
- Abstract
Tumor cells-even if nonauxotrophic-are often highly sensitive to arginine deficiency. We hypothesized that arginine deprivation therapy (ADT) if combined with irradiation could be a new treatment strategy for glioblastoma (GBM) patients because systemic ADT is independent of local penetration and diffusion limitations. A proof-of-principle in vitro study was performed with ADT being mimicked by application of recombinant human arginase or arginine-free diets. ADT inhibited two-dimensional (2-D) growth and cell-cycle progression, and reduced growth recovery after completion of treatment in four different GBM cell line models. Cells were less susceptible to ADT alone in the presence of citrulline and in a three-dimensional (3-D) environment. Migration and 3-D invasion were not unfavorably affected. However, ADT caused a significant radiosensitization that was more pronounced in a GBM cell model with p53 loss of function as compared with its p53-wildtype counterpart. The synergistic effect was independent of basic and induced argininosuccinate synthase or argininosuccinate lyase protein expression and not abrogated by the presence of citrulline. The radiosensitizing potential was maintained or even more distinguishable in a 3-D environment as verified in p53-knockdown and p53-wildtype U87-MG cells via a 60-day spheroid control probability assay. Although the underlying mechanism is still ambiguous, the observation of ADT-induced radiosensitization is of great clinical interest, in particular for patients with GBM showing high radioresistance and/or p53 loss of function. Mol Cancer Ther; 17(2); 393-406. ©2017 AACR See all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology.", (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
27. A Complex Scenario and Underestimated Challenge: The Tumor Microenvironment, ER Stress, and Cancer Treatment.
- Author
-
Chen OI, Bobak YP, Stasyk OV, and Kunz-Schughart LA
- Subjects
- Antineoplastic Agents chemistry, Humans, Neoplasms metabolism, Signal Transduction drug effects, Antineoplastic Agents pharmacology, Endoplasmic Reticulum Stress drug effects, Neoplasms drug therapy, Tumor Microenvironment drug effects
- Abstract
The paradoxical role of ER stress in malignant diseases is only just being unraveled and remains incompletely understood. A particular challenge is the complex interplay between spaciotemporal and locoregional microenvironmental constraints in solid tumors and stress responses upon treatment; thus, the potential for new combinatorial therapeutic options to foster the coincidence of ER stress-related deadly events is likely to be underestimated. Without claiming this review to be complete, we present a comprehensive overview of the signaling mechanisms associated with the unfolded protein response (UPR) and the molecular link to cell survival and death mechanisms. We (i) delineate the mechanistic scenario and outcome of the UPR; (ii) discuss the role of ER stress in cancer development and progression; (iii) highlight the impact of various environmental conditions and stress stimuli, such as nutrient limitation and tumor hypoxia, in this context; and (iv) attempt to shed some light on the putative link between DNA damage, irradiation, and ER stress to emphasize the potential of therapeutic targeting of ER stress pathways for combined cancer treatments., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.)
- Published
- 2018
- Full Text
- View/download PDF
28. Nanoparticles for radiooncology: Mission, vision, challenges.
- Author
-
Kunz-Schughart LA, Dubrovska A, Peitzsch C, Ewe A, Aigner A, Schellenburg S, Muders MH, Hampel S, Cirillo G, Iemma F, Tietze R, Alexiou C, Stephan H, Zarschler K, Vittorio O, Kavallaris M, Parak WJ, Mädler L, and Pokhrel S
- Subjects
- Animals, Contrast Media chemical synthesis, Evidence-Based Medicine, Forecasting, Humans, Treatment Outcome, Diagnostic Imaging methods, Nanoparticles therapeutic use, Neoplasms diagnostic imaging, Neoplasms radiotherapy, Radiation Oncology trends, Theranostic Nanomedicine trends
- Abstract
Cancer is one of the leading non-communicable diseases with highest mortality rates worldwide. About half of all cancer patients receive radiation treatment in the course of their disease. However, treatment outcome and curative potential of radiotherapy is often impeded by genetically and/or environmentally driven mechanisms of tumor radioresistance and normal tissue radiotoxicity. While nanomedicine-based tools for imaging, dosimetry and treatment are potential keys to the improvement of therapeutic efficacy and reducing side effects, radiotherapy is an established technique to eradicate the tumor cells. In order to progress the introduction of nanoparticles in radiooncology, due to the highly interdisciplinary nature, expertise in chemistry, radiobiology and translational research is needed. In this report recent insights and promising policies to design nanotechnology-based therapeutics for tumor radiosensitization will be discussed. An attempt is made to cover the entire field from preclinical development to clinical studies. Hence, this report illustrates (1) the radio- and tumor-biological rationales for combining nanostructures with radiotherapy, (2) tumor-site targeting strategies and mechanisms of cellular uptake, (3) biological response hypotheses for new nanomaterials of interest, and (4) challenges to translate the research findings into clinical trials., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
29. The why and how of amino acid analytics in cancer diagnostics and therapy.
- Author
-
Manig F, Kuhne K, von Neubeck C, Schwarzenbolz U, Yu Z, Kessler BM, Pietzsch J, and Kunz-Schughart LA
- Subjects
- Amino Acids metabolism, Animals, Biomarkers, Tumor analysis, Biomarkers, Tumor metabolism, Chromatography, High Pressure Liquid methods, Chromatography, Liquid, Electrophoresis methods, Gas Chromatography-Mass Spectrometry methods, Humans, Mice, Neoplasms chemistry, Neoplasms metabolism, Amino Acids analysis, Neoplasms diagnosis, Neoplasms therapy
- Abstract
Pathological alterations in cell functions are frequently accompanied by metabolic reprogramming including modifications in amino acid metabolism. Amino acid detection is thus integral to the diagnosis of many hereditary metabolic diseases. The development of malignant diseases as metabolic disorders comes along with a complex dysregulation of genetic and epigenetic factors affecting metabolic enzymes. Cancer cells might transiently or permanently become auxotrophic for non-essential or semi-essential amino acids such as asparagine or arginine. Also, transformed cells are often more susceptible to local shortage of essential amino acids such as methionine than normal tissues. This offers new points of attacking unique metabolic features in cancer cells. To better understand these processes, highly sensitive methods for amino acid detection and quantification are required. Our review summarizes the main methodologies for amino acid detection with a particular focus on applications in biomedicine and cancer, provides a historical overview of the methodological pre-requisites in amino acid analytics. We compare classical and modern approaches such as the combination of gas chromatography and liquid chromatography with mass spectrometry (GC-MS/LC-MS). The latter is increasingly applied in clinical routine. We therefore illustrate an LC-MS workflow for analyzing arginine and methionine as well as their precursors and analogs in biological material. Pitfalls during protocol development are discussed, but LC-MS emerges as a reliable and sensitive tool for the detection of amino acids in biological matrices. Quantification is challenging, but of particular interest in cancer research as targeting arginine and methionine turnover in cancer cells represent novel treatment strategies., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. Co-application of canavanine and irradiation uncouples anticancer potential of arginine deprivation from citrulline availability.
- Author
-
Kurlishchuk Y, Vynnytska-Myronovska B, Grosse-Gehling P, Bobak Y, Manig F, Chen O, Merker SR, Henle T, Löck S, Stange DE, Stasyk O, and Kunz-Schughart LA
- Subjects
- Apoptosis drug effects, Apoptosis radiation effects, Argininosuccinate Synthase genetics, Argininosuccinate Synthase metabolism, Cell Cycle drug effects, Cell Cycle radiation effects, Cell Line, Tumor, Colorectal Neoplasms metabolism, DNA Methylation drug effects, DNA Methylation radiation effects, Gene Expression, Humans, Intestinal Mucosa metabolism, Intestinal Mucosa radiation effects, Radiation Tolerance, Spheroids, Cellular, Tumor Cells, Cultured, Arginine metabolism, Canavanine administration & dosage, Citrulline metabolism, Radiation, Ionizing
- Abstract
The moderate anticancer effect of arginine deprivation in clinical trials has been linked to an induced argininosuccinate synthetase (ASS1) expression in initially ASS1-negative tumors, and ASS1-positive cancers are anticipated as non-responders. Our previous studies indicated that arginine deprivation and low doses of the natural arginine analog canavanine can enhance radioresponse. However, the efficacy of the proposed combination in the presence of extracellular citrulline, the substrate for arginine synthesis by ASS1, remains to be elucidated, in particular for malignant cells with positive and/or inducible ASS1 as in colorectal cancer (CRC). Here, the physiological citrulline concentration of 0.05 mM was insufficient to overcome cell cycle arrest and radiosensitization triggered by arginine deficiency. Hyperphysiological citrulline (0.4 mM) did not entirely compensate for the absence of arginine and significantly decelerated cell cycling. Similar levels of canavanine-induced apoptosis were detected in the absence of arginine regardless of citrulline supplementation both in 2-D and advanced 3-D assays, while normal colon epithelial cells in organoid/colonosphere culture were unaffected. Notably, canavanine tremendously enhanced radiosensitivity of arginine-starved 3-D CRC spheroids even in the presence of hyperphysiological citrulline. We conclude that the novel combinatorial targeting strategy of metabolic-chemo-radiotherapy has great potential for the treatment of malignancies with inducible ASS1 expression.
- Published
- 2016
- Full Text
- View/download PDF
31. The anti-malarial atovaquone increases radiosensitivity by alleviating tumour hypoxia.
- Author
-
Ashton TM, Fokas E, Kunz-Schughart LA, Folkes LK, Anbalagan S, Huether M, Kelly CJ, Pirovano G, Buffa FM, Hammond EM, Stratford M, Muschel RJ, Higgins GS, and McKenna WG
- Subjects
- Animals, Biguanides pharmacology, Electron Transport Complex III metabolism, High-Throughput Screening Assays, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mice, Inbred BALB C, Mice, Nude, Oxygen Consumption drug effects, Pyrimidines biosynthesis, Spheroids, Cellular drug effects, Spheroids, Cellular metabolism, Spheroids, Cellular pathology, Tumor Cells, Cultured, Antimalarials pharmacology, Atovaquone pharmacology, Radiation Tolerance drug effects, Tumor Hypoxia drug effects
- Abstract
Tumour hypoxia renders cancer cells resistant to cancer therapy, resulting in markedly worse clinical outcomes. To find clinical candidate compounds that reduce hypoxia in tumours, we conduct a high-throughput screen for oxygen consumption rate (OCR) reduction and identify a number of drugs with this property. For this study we focus on the anti-malarial, atovaquone. Atovaquone rapidly decreases the OCR by more than 80% in a wide range of cancer cell lines at pharmacological concentrations. In addition, atovaquone eradicates hypoxia in FaDu, HCT116 and H1299 spheroids. Similarly, it reduces hypoxia in FaDu and HCT116 xenografts in nude mice, and causes a significant tumour growth delay when combined with radiation. Atovaquone is a ubiquinone analogue, and decreases the OCR by inhibiting mitochondrial complex III. We are now undertaking clinical studies to assess whether atovaquone reduces tumour hypoxia in patients, thereby increasing the efficacy of radiotherapy.
- Published
- 2016
- Full Text
- View/download PDF
32. Efficacy of Beta1 Integrin and EGFR Targeting in Sphere-Forming Human Head and Neck Cancer Cells.
- Author
-
Zscheppang K, Kurth I, Wachtel N, Dubrovska A, Kunz-Schughart LA, and Cordes N
- Abstract
Background: Resistance to radiotherapy continues to be a limiting factor in the treatment of cancer including head and neck squamous cell carcinoma (HNSCC). Simultaneous targeting of β1 integrin and EGFR was shown to have a higher radiosensitizing potential than mono-targeting in the majority of tested HNSCC cancer models. As tumor-initiating cells (TIC) are thought to play a key role for therapy resistance and recurrence and can be enriched in sphere forming conditions, this study investigated the efficacy of β1 integrin/EGFR targeting without and in combination with X-ray irradiation on the behavior of sphere-forming cells (SFC)., Methods: HNSCC cell lines (UTSCC15, UTSCC5, Cal33, SAS) were injected subcutaneously into nude mice for tumor up-take and plated for primary and secondary sphere formation under non-adhesive conditions which is thought to reflect the enrichment of SFC and their self-renewal capacity, respectively. Treatment was accomplished by inhibitory antibodies for β1 integrin (AIIB2) and EGFR (Cetuximab) as well as X-ray irradiation (2 - 6 Gy single doses). Further, flow cytometry for TIC marker expression and cell cycling as well as Western blotting for DNA repair protein expression and phosphorylation were employed., Results: We found higher primary and secondary sphere forming capacity of SAS cells relative to other HNSCC cell lines, which was in line with the tumor up-take rates of SAS versus UTSCC15 cells. AIIB2 and Cetuximab administration had minor cytotoxic and no radiosensitizing effects on SFC. Intriguingly, secondary SAS spheres, representing the fraction of surviving SFC upon passaging, showed greatly enhanced radiosensitivity compared to primary spheres. Intriguingly, neither AIIB2 nor Cetuximab significantly altered basal sphere forming capacity and radiosensitivity. While an increased accumulation of G0/G1 phase cells was observable in secondary SAS spheres, DNA double strand break repair indicated no difference on the basis of significantly enhanced ATM and Chk2 dephosphorylation upon irradiation., Conclusions: In the HNSCC model, sphere-forming conditions select for cells, which are unsusceptible to both anti-β1 integrin and anti-EGFR inhibitory antibodies. With regard to primary and secondary sphere formation, our data suggest that both of these SFC fractions express distinct survival strategies independent from β1 integrin and EGFR and that future work is warranted to better understand SFC survival and enrichment before and after treatment to untangle the underlying mechanisms for identifying novel, druggable cancer targets in SFC.
- Published
- 2016
- Full Text
- View/download PDF
33. Arginine starvation in colorectal carcinoma cells: Sensing, impact on translation control and cell cycle distribution.
- Author
-
Vynnytska-Myronovska BO, Kurlishchuk Y, Chen O, Bobak Y, Dittfeld C, Hüther M, Kunz-Schughart LA, and Stasyk OV
- Subjects
- HCT116 Cells, HT29 Cells, Humans, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Tumor Cells, Cultured, Tumor Suppressor Protein p53 metabolism, Arginine metabolism, Cell Cycle, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Protein Biosynthesis
- Abstract
Tumor cells rely on a continued exogenous nutrient supply in order to maintain a high proliferative activity. Although a strong dependence of some tumor types on exogenous arginine sources has been reported, the mechanisms of arginine sensing by tumor cells and the impact of changes in arginine availability on translation and cell cycle regulation are not fully understood. The results presented herein state that human colorectal carcinoma cells rapidly exhaust the internal arginine sources in the absence of exogenous arginine and repress global translation by activation of the GCN2-mediated pathway and inhibition of mTOR signaling. Tumor suppressor protein p53 activation and G1/G0 cell cycle arrest support cell survival upon prolonged arginine starvation. Cells with the mutant or deleted TP53 fail to stop cell cycle progression at defined cell cycle checkpoints which appears to be associated with reduced recovery after durable metabolic stress triggered by arginine withdrawal., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
34. Arginine deprivation induces endoplasmic reticulum stress in human solid cancer cells.
- Author
-
Bobak Y, Kurlishchuk Y, Vynnytska-Myronovska B, Grydzuk O, Shuvayeva G, Redowicz MJ, Kunz-Schughart LA, and Stasyk O
- Subjects
- Canavanine pharmacology, Cell Line, Tumor, Cycloheximide pharmacology, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Dimethyl Sulfoxide pharmacology, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum Chaperone BiP, Endoplasmic Reticulum Stress genetics, HCT116 Cells, HT29 Cells, Heat-Shock Proteins genetics, Heat-Shock Proteins metabolism, Humans, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Organ Specificity, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Regulatory Factor X Transcription Factors, Signal Transduction, Transcription Factor CHOP genetics, Transcription Factor CHOP metabolism, Transcription Factors genetics, Transcription Factors metabolism, Tunicamycin pharmacology, Unfolded Protein Response drug effects, Arginine deficiency, Culture Media pharmacology, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum Stress drug effects, Gene Expression Regulation, Neoplastic
- Abstract
Deprivation for the single amino acid arginine is a rapidly developing metabolic anticancer therapy, which allows growth control in a number of highly malignant tumors. Here we report that one of the responses of human solid cancer cells to arginine starvation is the induction of prolonged endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR). Systematic study of two colorectal carcinoma HCT-116 and HT29, glioblastoma U251 MG and ovarian carcinoma SKOV3 cell lines revealed, however, that the ER stress triggered by the absence of arginine does not result in massive apoptosis despite a profound upregulation of the proapoptotic gene CHOP. Instead, Akt- and MAPK-dependent pathways were activated which may counteract proapoptotic signaling. Treatment with DMSO as a disaggregating agent or with cycloheximide to block protein synthesis reduced ER stress evoked by arginine deprivation. On the other hand, ER stress and apoptosis induction in arginine-starved cells could be critically augmented by the arginine analog of plant origin canavanine, but not by the classic ER stress inducer tunicamycin. Our data suggest that canavanine treatment applied under the lack of arginine may enhance the efficacy of arginine deprivation-based anticancer therapy., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Aldehyde Dehydrogenase Is Regulated by β-Catenin/TCF and Promotes Radioresistance in Prostate Cancer Progenitor Cells.
- Author
-
Cojoc M, Peitzsch C, Kurth I, Trautmann F, Kunz-Schughart LA, Telegeev GD, Stakhovsky EA, Walker JR, Simin K, Lyle S, Fuessel S, Erdmann K, Wirth MP, Krause M, Baumann M, and Dubrovska A
- Subjects
- Aldehyde Dehydrogenase genetics, Aldehyde Dehydrogenase 1 Family, Animals, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Nude, Neoplasm Transplantation, Neoplastic Stem Cells radiation effects, Prostatic Neoplasms pathology, Radiation Tolerance, Retinal Dehydrogenase, Transcriptome, Wnt Signaling Pathway, Aldehyde Dehydrogenase metabolism, Neoplastic Stem Cells enzymology, Prostatic Neoplasms enzymology, beta Catenin physiology
- Abstract
Radiotherapy is a curative treatment option in prostate cancer. Nevertheless, patients with high-risk prostate cancer are prone to relapse. Identification of the predictive biomarkers and molecular mechanisms of radioresistance bears promise to improve cancer therapies. In this study, we show that aldehyde dehydrogenase (ALDH) activity is indicative of radioresistant prostate progenitor cells with an enhanced DNA repair capacity and activation of epithelial-mesenchymal transition (EMT). Gene expression profiling of prostate cancer cells, their radioresistant derivatives, ALDH(+) and ALDH(-) cell populations revealed the mechanisms, which link tumor progenitors to radioresistance, including activation of the WNT/β-catenin signaling pathway. We found that expression of the ALDH1A1 gene is regulated by the WNT signaling pathway and co-occurs with expression of β-catenin in prostate tumor specimens. Inhibition of the WNT pathway led to a decrease in ALDH(+) tumor progenitor population and to radiosensitization of cancer cells. Taken together, our results indicate that ALDH(+) cells contribute to tumor radioresistance and their molecular targeting may enhance the effectiveness of radiotherapy., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
36. Potential of a Cetuximab-based radioimmunotherapy combined with external irradiation manifests in a 3-D cell assay.
- Author
-
Ingargiola M, Runge R, Heldt JM, Freudenberg R, Steinbach J, Cordes N, Baumann M, Kotzerke J, Brockhoff G, and Kunz-Schughart LA
- Subjects
- Antineoplastic Agents therapeutic use, Carcinoma, Squamous Cell diagnostic imaging, Cell Survival, Cetuximab, Dose-Response Relationship, Radiation, Drug Carriers, ErbB Receptors metabolism, Head and Neck Neoplasms diagnostic imaging, Humans, Ligands, Monte Carlo Method, Probability, Radiation Tolerance drug effects, Radionuclide Imaging, Radiotherapy methods, Spheroids, Cellular cytology, Tumor Cells, Cultured cytology, X-Rays, Yttrium Radioisotopes chemistry, Antibodies, Monoclonal, Humanized therapeutic use, Carcinoma, Squamous Cell radiotherapy, Head and Neck Neoplasms radiotherapy, Radioimmunotherapy methods
- Abstract
Targeting epidermal growth factor receptor (EGFR)-overexpressing tumors with radiolabeled anti-EGFR antibodies is a promising strategy for combination with external radiotherapy. In this study, we evaluated the potential of external plus internal irradiation by [(90) Y]Y-CHX-A″-DTPA-C225 (Y-90-C225) in a 3-D environment using FaDu and SAS head and neck squamous cell carcinoma (HNSCC) spheroid models and clinically relevant endpoints such as spheroid control probability (SCP) and spheroid control dose 50% (SCD50 , external irradiation dose inducing 50% loss of spheroid regrowth). Spheroids were cultured using a standardized platform. Therapy response after treatment with C225, CHX-A"-DTPA-C225 (DTPA-C225), [(90) Y]Y-CHX-A"-DTPA (Y-90-DTPA) and Y-90-C225 alone or in combination with X-ray was evaluated by long-term monitoring (60 days) of spheroid integrity and volume growth. Penetration kinetics into spheroids and EGFR binding capacities on spheroid cells were identical for unconjugated C225 and Y-90-C225. Spheroid-associated radioactivity upon exposure to the antibody-free control conjugate Y-90-DTPA was negligible. Determination of the SCD50 demonstrated higher intrinsic radiosensitivity of FaDu as compared with SAS spheroids. Treatment with unconjugated C225 alone did not affect spheroid growth and cell viability. Also, C225 treatment after external irradiation showed no additive effect. However, the combination of external irradiation with Y-90-C225 (1 µg/ml, 24 hr) resulted in a considerable benefit as reflected by a pronounced reduction of the SCD50 from 16 Gy to 9 Gy for SAS spheroids and a complete loss of regrowth for FaDu spheroids due to the pronounced accumulation of internal dose caused by the continuous exposure to cell-bound radionuclide upon Y-90-C225-EGFR interaction., (© 2014 UICC.)
- Published
- 2014
- Full Text
- View/download PDF
37. Cytotoxic properties of radionuclide-conjugated Cetuximab without and in combination with external irradiation in head and neck cancer cells in vitro.
- Author
-
Eke I, Ingargiola M, Förster C, Kunz-Schughart LA, Baumann M, Runge R, Freudenberg R, Kotzerke J, Heldt JM, Pietzsch HJ, Steinbach J, and Cordes N
- Subjects
- Cell Count, Cell Line, Tumor, Cell Survival drug effects, Cell Survival radiation effects, Cetuximab, Combined Modality Therapy, DNA Breaks, Double-Stranded drug effects, DNA Breaks, Double-Stranded radiation effects, ErbB Receptors metabolism, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic radiation effects, Humans, Intracellular Space drug effects, Intracellular Space metabolism, Intracellular Space radiation effects, Molecular Targeted Therapy, Pentetic Acid chemistry, Phosphorylation drug effects, Phosphorylation radiation effects, Protein Transport drug effects, Protein Transport radiation effects, Signal Transduction drug effects, Signal Transduction radiation effects, Yttrium Radioisotopes chemistry, Yttrium Radioisotopes therapeutic use, Antibodies, Monoclonal, Humanized chemistry, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Head and Neck Neoplasms pathology
- Abstract
Purpose: Epidermal growth factor receptor (EGFR) is critically involved in progression and therapy resistance of squamous cell carcinoma (SCC). Albeit EGFR targeting could improve the effect of radiotherapy on patients' outcome, the clinical results failed to meet expectations from preclinical studies. In this work, we evaluated the potential of the radionuclide Yttrium-90 ((90)Y) bound to Cetuximab ((90)Y-Cetuximab) as novel targeting approach for SCC cells in vitro., Materials and Methods: FaDu and A431 cell lines were used. EGFR subcellular localization, clonogenic survival, radiation-induced γH2AX foci and EGFR signaling were examined. Cells were treated with DTPA, DTPA-Cetuximab, (90)Y and (90)Y-Cetuximab alone or in combination with external X-ray irradiation., Results: Dose- and cell line-dependently, (90)Y-Cetuximab mediated a significant reduction in clonogenicity relative to unbound (90)Y. Combined 2-Gy external radiation plus 2-Gy equivalent dose of (90)Y-Cetuximab was more effective than equivalent doses of (90)Y and X-ray radiation. Analogous effects were observed in the number of residual radiation-induced foci. Additionally, EGFR, ERK1/2 and AKT phosphorylation showed alterations upon different treatments., Conclusions: Our findings show that Cetuximab-conjugated (90)Y has a significant potential to eradicate human SCC cells. A combination of radioimmunotherapeutic compounds and external radiotherapy might be a promising treatment strategy for clinical application.
- Published
- 2014
- Full Text
- View/download PDF
38. Macromolecule extravasation-xenograft size matters: a systematic study using probe-based confocal laser endomicroscopy (pCLE).
- Author
-
Dietrich A, Stewart J, Huether M, Helm M, Schuetze C, Schnittler HJ, Jaffray DA, and Kunz-Schughart LA
- Subjects
- Animals, Female, HCT116 Cells, Humans, Mice, Mice, Inbred C57BL, Muscles blood supply, Muscles pathology, Neoplasms, Experimental, Tongue blood supply, Tongue pathology, Xenograft Model Antitumor Assays, Capillary Permeability, Extravasation of Diagnostic and Therapeutic Materials pathology, Microscopy, Confocal methods, Neovascularization, Pathologic pathology
- Abstract
Purpose: Profound changes of the vasculature in tumors critically impact drug delivery and therapy response. We aimed at developing a procedure to monitor morphological and functional parameters of the vasculature in subcutaneous xenograft models commonly applied for therapy testing by using probe-based confocal laser endomicroscopy., Procedures: By monitoring various normal and diseased tissues, we established an experimental and analytical set-up to systematically analyze tracer extravasation from the microvasculature. Application of the approach in two xenograft models (HCT-116 and SW620) was realized consecutively throughout tumor growth., Results: The incidence of dilated vessels increased with xenograft size in both models while macromolecule extravasation and tracer accumulation in the tumor tissue, respectively, was significantly reduced throughout growth. The development of dilated/ultradilated vessels correlated with tracer extravasation only in the HCT-116 but not the SW620 model. The underlying mechanisms are still ambiguous and discussed., Conclusions: Our findings clearly indicate that both xenograft type and size matter for drug delivery and therapy testing.
- Published
- 2013
- Full Text
- View/download PDF
39. Three-dimensional environment renders cancer cells profoundly less susceptible to a single amino acid starvation.
- Author
-
Vynnytska-Myronovska B, Kurlishchuk Y, Bobak Y, Dittfeld C, Kunz-Schughart LA, and Stasyk O
- Subjects
- Apoptosis, Cell Line, Tumor, Humans, Neoplasms physiopathology, Amino Acids metabolism, Cell Culture Techniques methods, Neoplasms metabolism
- Abstract
Increased amino acid requirement of malignant cells is exploited in metabolic antitumor therapy, e.g., enzymotherapies based on arginine or methionine deprivation. However, studies on animal models and clinical trials revealed that solid tumors are much less susceptible to single amino acid starvation than could be expected from the in vitro data. We conducted a comparative analysis of the response of several tumor cell lines to single amino acid starvation in 2-D monolayer versus 3-D spheroid culture. We revealed for the first time that in comparison with monolayer culture tumor cells, spheroids are much less susceptible to the deprivation of individual amino acids (i.e., arginine, leucine, lysine or methionine). Accordingly, even after prolonged (up to 10 days) starvation, spheroid cells could readily resume proliferation when appropriate amino acid was resupplemented. In the case of arginine deprivation, similar apoptosis induction was detected both in 2-D and 3-D culture, suggesting that this process does not determine the level of tumor cell sensitivity to this kind of treatment. It was also observed that spheroids much better mimic the in vivo ability of tumor cells to utilize citrulline as arginine precursor for growth in amino acid deficient environment. We conclude that 3-D spheroid culture better reflects in vivo tumor cell response to single amino acid starvation than 2-D monolayer culture and should be used as an integral model in the studies of this type of antitumor metabolic targeting.
- Published
- 2013
- Full Text
- View/download PDF
40. Simultaneous PLK1 inhibition improves local tumour control after fractionated irradiation.
- Author
-
Krause M, Kummer B, Deparade A, Eicheler W, Pfitzmann D, Yaromina A, Kunz-Schughart LA, and Baumann M
- Subjects
- Animals, Cell Cycle drug effects, Cell Line, Tumor, Female, Humans, Male, Mice, Neoplasms pathology, Polo-Like Kinase 1, Cell Cycle Proteins antagonists & inhibitors, Neoplasms radiotherapy, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors, Pteridines pharmacology, Radiation-Sensitizing Agents pharmacology
- Abstract
Purpose: Polo-like kinase 1 (PLK1) plays an important role in mitotic progression, is frequently overexpressed and associated with a poor prognosis of cancer patients, thus providing a promising target in anticancer treatment. Aim of the current project was to evaluate the effect of the novel PLK1 inhibitor BI 6727 in combination with irradiation., Material and Methods: In vitro proliferation and radiation cell survival assays as well as in vivo local tumour control assays after single treatment and combined radiation and drug application were carried out using the squamous cell carcinoma models A431 and FaDu. In addition, cell cycle phases were monitored in vitro and in vivo., Results: BI 6727 showed a dose-dependent antiproliferative effect and an increase in the mitotic fraction. BI 6727 alone reduced clonogenic cell survival, while radiosensitivity in vitro (SF2) and in vivo (single-dose TCD(50) under clamped hypoxia) was not affected. In contrast, local tumour control was significantly improved after application of BI 6727 simultaneously to fractionated irradiation (A431: TCD(50) = 60.5 Gy [95% C.I. 57; 63] after IR alone and <30 Gy after combined treatment; FaDu: 49.5 Gy [43; 56 Gy] versus 32.9 Gy [26; 40])., Conclusions: Despite the lack of direct cellular radiosensitisation, PLK1 inhibition with BI 6727 during fractionated irradiation significantly improves local tumour control when compared to irradiation alone. This result is likely explained by a considerable effect on cell cycle and an independent cytotoxic potential of BI 6727., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
41. CD133 as a biomarker for putative cancer stem cells in solid tumours: limitations, problems and challenges.
- Author
-
Grosse-Gehling P, Fargeas CA, Dittfeld C, Garbe Y, Alison MR, Corbeil D, and Kunz-Schughart LA
- Subjects
- AC133 Antigen, Antigens, CD genetics, Biomarkers, Tumor genetics, Gene Expression Regulation, Neoplastic, Glycoproteins genetics, Humans, Neoplasms diagnosis, Neoplasms genetics, Neoplastic Stem Cells pathology, Peptides genetics, Prognosis, Protein Processing, Post-Translational, Tumor Microenvironment physiology, Antigens, CD metabolism, Biomarkers, Tumor metabolism, Glycoproteins metabolism, Neoplasms metabolism, Neoplastic Stem Cells metabolism, Peptides metabolism
- Abstract
The cancer stem cell (CSC) hypothesis, despite the limitations of the currently available models and assays, has ushered in a new era of excitement in cancer research. The development of novel strategies for anti-tumour therapy relies on the use of biomarkers to identify, enrich, and/or isolate the cell population(s) of interest. In this context, various cell characteristics and antigen expression profiles are discussed as surrogate markers. The cell surface expression of the human prominin-1 (CD133) antigen, in particular of the AC133 epitope, is among those that have been most frequently studied in solid cancers, although no mechanism has yet been proposed to link CD133 expression with the CSC phenotype. Some inconsistencies between published data can be ascribed to different analytical tools as well as methodological limitations and pitfalls, highlighted in the present review. Therefore, a comprehensive overview on the current state of knowledge in this growing and exciting field with an emphasis on the most recent studies is presented. We highlight the link between the tumour microenvironment, tumour cell plasticity, and CD133 expression, and evaluate the utility of CD133 expression as a prognostic marker., (Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
42. EGFR/JIP-4/JNK2 signaling attenuates cetuximab-mediated radiosensitization of squamous cell carcinoma cells.
- Author
-
Eke I, Schneider L, Förster C, Zips D, Kunz-Schughart LA, and Cordes N
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Blotting, Western, Cell Culture Techniques methods, Cell Line, Tumor, Cetuximab, Drug Resistance, Neoplasm physiology, Humans, Immunoprecipitation, Mice, Mice, Nude, Protein Array Analysis, RNA, Small Interfering, Signal Transduction physiology, Transfection, Adaptor Proteins, Signal Transducing metabolism, Antibodies, Monoclonal pharmacology, Carcinoma, Squamous Cell metabolism, ErbB Receptors metabolism, Mitogen-Activated Protein Kinase 9 metabolism, Radiation-Sensitizing Agents pharmacology
- Abstract
EGF receptor (EGFR) promotes tumor growth as well as radio- and chemoresistance in various human malignancies including squamous cell carcinomas (SCC). In addition to deactivation of prosurvival signaling, cetuximab-mediated EGFR targeting might concomitantly induce self-attenuating signaling bypasses. Identification of such bypass mechanisms is key to improve the efficacy of targeted approaches. Here, we show great similarity of EGFR signaling and radiation survival in cetuximab-treated SCC cells grown in a more physiologic three-dimensional extracellular matrix and as tumor xenografts in contrast to conventional monolayer cell cultures. Using phosphoproteome arrays, we observed strong induction of JNK2 phosphorylation potentially resulting from cetuximab-inhibited EGFR through c-jun-NH(2)-kinase (JNK)-interacting protein-4 (JIP-4), which was identified using an immunoprecipitation-mass spectrometric approach. Inhibition of this signaling bypass by JIP-4 or JNK2 knockdown or pharmacologic JNK2 inhibition enhanced cetuximab efficacy and tumor cell radiosensitivity. Our findings add new facets to EGFR signaling and indicate signaling bypass possibilities of cancer cells to improve their survival on cetuximab treatment. By deactivation of cetuximab-self-attenuating JNK2-dependent signaling, the cytotoxicity, and radiosensitizing potential of cetuximab can be augmented.
- Published
- 2013
- Full Text
- View/download PDF
43. [Tumor pathophysiology].
- Author
-
Kunz-Schughart LA, Mueller-Klieser W, and Vaupel P
- Subjects
- Animals, Blood Glucose metabolism, Cell Hypoxia radiation effects, Energy Metabolism radiation effects, Humans, Lactic Acid metabolism, Neoplasm Invasiveness, Neoplasm Transplantation, Neoplasms pathology, Phenotype, Pyruvic Acid metabolism, Transplantation, Heterologous, Neoplasms physiopathology, Neoplasms radiotherapy
- Published
- 2012
- Full Text
- View/download PDF
44. Rapid re-expression of CD133 protein in colorectal cancer cell lines in vitro and in vivo.
- Author
-
Peickert S, Waurig J, Dittfeld C, Dietrich A, Garbe Y, Kabus L, Baumann M, Grade M, Ried T, and Kunz-Schughart LA
- Subjects
- AC133 Antigen, Animals, Female, HCT116 Cells, HT29 Cells, Humans, Mice, Neoplasms, Experimental metabolism, Antigens, CD metabolism, Biomarkers, Tumor metabolism, Colorectal Neoplasms metabolism, Glycoproteins metabolism, Peptides metabolism
- Abstract
Studies related to the cancer stem cell hypothesis are challenging because of the imperfect tools to identify cell populations of interest and controversy on the usefulness of established cancer cell lines. We previously found CD133 to not be selective for a tumor-propagating or radioresistant population in a near-diploid, microsatellite-instable colorectal carcinoma (CRC) cell line. Because of discrepant literature data, we herein systematically analyzed the behavior of microsatellite-stable cell line subpopulations reflecting the more frequent carcinogenesis pathway in spontaneous CRC. CD133⁺ and CD133(-/low) populations were isolated by fluorescence-activated cell sorting and further processed. HT29 and SW620 cells were studied in detail in monolayer and/or spheroid culture assays and upon subcutaneous injection in NMRI (nu/nu) mice using a limiting dilution approach. CD133(-/low) HT29 cells showed a significantly lower clonogenic survival and reduced spheroid formation capacity than their CD133⁺ counterparts. However, the cell populations neither differed in growth kinetics and response to treatment in vitro nor in tumor formation capacity when injecting as low as 10 cells. CD133(-/low) HT29 cells rapidly re-expressed CD133 protein in vitro and in vivo as shown by flow cytometry and/or western blot analyses, and they also showed a particular survival benefit under tissue normoxic conditions. In contrast, CD133 protein in the CD133⁺ population was quite stable throughout culturing. The observation of CD133 re-expression and lack of difference in tumor take rate of subpopulations was confirmed in SW620 cells. Here, we found cell density to affect CD133 re-expression in the CD133(-)-sorted population. And even SW480 cells, classified as a CD133⁻ cell line, presented some CD133 protein on their surface upon in vivo engraftment. We conclude that (i) CD133 protein expression shows high plasticity in CRC cell lines, and (ii) in vitro CD133 status on the cell surface neither determines tumorigenic potential nor CD133 profile in vivo.
- Published
- 2012
- Full Text
- View/download PDF
45. Flow cytometric cell-based assay to preselect antibody constructs for radionuclide conjugation.
- Author
-
Ingargiola M, Dittfeld C, Runge R, Zenker M, Heldt JM, Steinbach J, Cordes N, Baumann M, Kotzerke J, and Kunz-Schughart LA
- Subjects
- Antibodies, Monoclonal immunology, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal, Humanized, Binding, Competitive, Cell Line, Tumor, Cetuximab, Chelating Agents, Flow Cytometry methods, Fluorescence, Humans, Immunoconjugates immunology, Immunoconjugates metabolism, Isothiocyanates chemistry, Pentetic Acid analogs & derivatives, Pentetic Acid chemistry, Protein Binding, Radiopharmaceuticals immunology, Radiopharmaceuticals metabolism, Yttrium Radioisotopes, Antibodies, Monoclonal chemistry, Drug Carriers chemistry, ErbB Receptors metabolism, Immunoassay, Immunoconjugates chemistry, Radiopharmaceuticals chemistry
- Abstract
Radiolabeled antibodies (Abs) are an attractive tool for targeting and delivering particle emitters for therapy or imaging applications. The labeling of Abs with metal radionuclides requires chelating agents and can cause loss of binding to their ligands. The aim of the present approach was to design an easy-handling flow cytometric cell-based assay to evaluate Ab-binding capacity of conjugates of the therapeutic Ab Cetuximab and to verify the most promising candidate in a competitive radioactive binding experiment. The final setup for flow cytometric assessment of cellular binding capacities of epidermal growth factor receptor (EGFR)/ErbB1-directed Ab conjugates is based on (a) the selection of a robust cell line model (b) the definition of nonsaturated staining concentrations for the unconjugated reference Ab Cetuximab plus implementation of a reasonable isotype control, and (c) the calculation of relative Ab affinities based on the flow cytometric data. Two (FaDu, SAS) out of the three cell lines with different total and cell surface expression levels of EGFR turned out to be adequate models but the application of one cell line was sufficient to estimate reduced binding capacities of conjugates relative to Cetuximab. Only 1/11 conjugate Abs exhibited a fluorescence signal comparable to unconjugated Cetuximab and was applied for radiolabeling with Yttrium-90. Unaltered binding affinity of this conjugate was proven in a competitive radioactive Ab-binding study. We conclude that the flow cytometric assay is reliable and that the relative binding capacity of Cetuximab is neither affected by covalent modification with CHX-A"-DTPA (N-[(R)-2-Amino-3-(p-isothiocyanato-phenyl) propyl]-trans-(S,S)-cyclohexane-1,2-diamine-N,N,N',N",N"-pentaacetic acid) with a final chelator-to-Ab ratio of 5 nor by subsequent radiolabeling. [(90)Y]Y-CHX-A"-DTPA-Cetuximab thus qualifies for preclinical treatment testing as a prerequisite for therapeutic application., (Copyright © 2012 International Society for Advancement of Cytometry.)
- Published
- 2012
- Full Text
- View/download PDF
46. Single amino acid arginine starvation efficiently sensitizes cancer cells to canavanine treatment and irradiation.
- Author
-
Vynnytska-Myronovska B, Bobak Y, Garbe Y, Dittfeld C, Stasyk O, and Kunz-Schughart LA
- Subjects
- Apoptosis drug effects, Arginine genetics, Canavanine metabolism, Cell Line, Tumor, Enzyme Inhibitors therapeutic use, HCT116 Cells, HT29 Cells, Humans, Spheroids, Cellular metabolism, Spheroids, Cellular pathology, Arginine metabolism, Canavanine pharmacology, Cytoprotection drug effects, Neoplasms, Glandular and Epithelial drug therapy, Neoplasms, Glandular and Epithelial metabolism, Neoplasms, Glandular and Epithelial radiotherapy, Radiation Tolerance drug effects
- Abstract
Single amino acid arginine deprivation is a promising strategy in modern metabolic anticancer therapy. Its potency to inhibit tumor growth warrants the search for rational chemo- and radio-therapeutic approaches to be co-applied. In this report, we evaluated, for the first time, the efficacy of arginine deprivation as anticancer therapy in three-dimensional (3D) cultures of human tumor cells, and propose a new combinatorial metabolic-chemo-radio-treatment regime based on arginine starvation, low doses of arginine natural analog canavanine and irradiation. A sophisticated experimental setup was designed to evaluate the impact of arginine starvation on four human epithelial cancer cell lines in 2D monolayer and 3D spheroid culture. Radioresponse was assessed in colony formation assays and by monitoring spheroid regrowth probability following single dose irradiation using a standardized spheroid-based test platform. Surviving fraction at 2 Gy (SF(2Gy)) and spheroid control dose(50) (SCD(50) ) were calculated as analytical endpoints. Cancer cells in spheroids are much more resistant to arginine starvation than in 2D culture. Spheroid volume stagnated during arginine deprivation, but even after 10 days of starvation, 100% of the spheroids regrew. Combination treatment, however, was remarkably efficient. In particular, pretreatment of cancer cells with the arginine-degrading enzyme arginase combined with or without low concentration of canavanine substantially enhanced cell radioresponse reflected by a loss in spheroid regrowth probability and SCD(50) values reduced by a factor of 1.5-3. Our data strongly suggest that arginine withdrawal alone or in combination with canavanine is a promising antitumor strategy with potential to enhance cancer cure by irradiation., (Copyright © 2011 UICC.)
- Published
- 2012
- Full Text
- View/download PDF
47. Effects of three modifiers of glycolysis on ATP, lactate, hypoxia, and growth in human tumor cell lines in vivo.
- Author
-
Yaromina A, Meyer S, Fabian C, Zaleska K, Sattler UG, Kunz-Schughart LA, Mueller-Klieser W, Zips D, and Baumann M
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Coumaric Acids pharmacology, Dichloroacetic Acid pharmacology, Enzyme Inhibitors pharmacology, Female, Glycolysis drug effects, Humans, Hydrogen-Ion Concentration, Lactic Acid metabolism, Mice, Oxamic Acid pharmacology, Transplantation, Heterologous, Adenosine Triphosphate metabolism, Gene Expression Regulation, Neoplastic drug effects, Oxygen Consumption drug effects
- Abstract
Background: High pretreatment tumor lactate content is associated with poor outcome after fractionated irradiation in human squamous cell carcinoma (hSCC) xenografts. Therefore, decreasing lactate content might be a promising approach for increasing tumor radiosensitivity. As the basis for such experiments, the effects of the biochemical inhibitors pyruvate dehydrogenase kinase dichloroacetate (DCA), lactate dehydrogenase oxamate, and monocarboxylic acid transporter-1 α-cyano-4-hydroxycinnamate (CHC) on tumor micromilieu and growth were investigated., Materials and Methods: Oxygen consumption (OCR) and extracellular acidification rates (ECAR) were measured in FaDu and UT-SCC-5 hSCC in response to DCA in vitro. Mice bearing FaDu, UT-SCC-5, and WiDr colorectal adenocarcinoma received either DCA in drinking water or DCA injected twice a day, or CHC injected daily. WiDr was also treated daily with oxamate. FaDu and UT-SCC-5 were either excised 8 days after treatment for histology or tumor growth was monitored. WiDr tumors were excised at 8 mm. Effect of inhibitors on ATP, lactate, hypoxia, and Ki67 labeling index (LI) was evaluated., Results: DCA increased OCR and decreased ECAR in vitro. None of the treatments with inhibitors significantly changed lactate content, hypoxia levels, and Ki67 LI in the three tumor lines in vivo. ATP concentration significantly decreased after only daily twice injections of DCA in FaDu accompanied by a significant increase in necrotic fraction. Tumor growth was not affected by any of the treatments., Conclusion: Overall, tumor micromilieu and tumor growth could not be changed by glycolysis modifiers in the three tumor cell lines in vivo. Further studies are necessary to explore the impact of metabolic targets on radiation response.
- Published
- 2012
- Full Text
- View/download PDF
48. Cancer stem cells as a predictive factor in radiotherapy.
- Author
-
Brunner TB, Kunz-Schughart LA, Grosse-Gehling P, and Baumann M
- Subjects
- Humans, Biomarkers, Tumor physiology, Neoplasm Recurrence, Local, Neoplasms radiotherapy, Neoplastic Stem Cells physiology, Tumor Microenvironment physiology
- Abstract
Cancer stem cell research is one of the most thriving and competitive areas in oncology research because it has the potential to dramatically affect clinical outcomes. Led by progress in hematology, cancer stem cell research has now provided evidence to play an important role for solid cancers as well. Because radiotherapy is only second to surgery in terms of its curative potency, it is very important for radiation oncologists to learn whether progress in cancer stem cell biology can enable them to exploit this knowledge to help cure more patients suffering from cancer. The present article gives an overview about the challenges of the cancer stem cell concept and highlights some important phenomena that are under intense investigation, such as phenotypic plasticity of stemness and impact and dynamics of microenvironmental niches. We discuss the potential and limitations of current experimental and theragnostic tools and end up with an agenda for future research as outlook for translational possibilities in the clinic., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
49. Sodium-iodide symporter positive cells after intracellular uptake of (99m)Tc versus α-emitter 211At. Reduction of clonogenic survival and characterization of DNA damage.
- Author
-
Kotzerke J, Wendisch M, Freudenberg R, Runge R, Oehme L, Meyer GJ, Kunz-Schughart LA, and Wunderlich G
- Subjects
- Animals, Cell Line, Cell Survival drug effects, Cell Survival radiation effects, Colony-Forming Units Assay, Dose-Response Relationship, Radiation, Electrons, Radiation Dosage, Rats, Thyroid Gland cytology, Thyroid Gland radiation effects, Astatine pharmacokinetics, Astatine pharmacology, DNA Damage physiology, Symporters metabolism, Technetium pharmacokinetics, Technetium pharmacology, Thyroid Gland physiology
- Abstract
Purpose: We evaluated the DNA damaging potential of Auger electrons emitted in the decay of (99m)Tc compared to α-particles of 211At., Material and Methods: The impact of (99m)Tc and 211At was monitored in a NIS-expressing rat thyroid cell model PCCl3 with varying, yet defined intra- and extracellular radionuclide distribution (using ± perchlorate). The radiotoxicity of (99m)Tc and 211At was studied by the comet assay under neutral and alkaline conditions and colony formation., Results: In the presence of perchlorate, the radioactivity yielding 37% cellular survival, A37, was estimated to be (0.27 ± 0.02) MBq/ml and (450 ± 30) MBq/ml for 211At and (99m)Tc, respectively. In absence of perchlorate, cellular radiotracer uptake was similar for both radionuclides (2.2%, 2.7%), yet the A37 was reduced by 82% for the α-emitter and by 95% for (99m)Tc. Cellular dose increased by a factor of 5 (211At) and 38 (99mTc). Comet assays revealed an increased DNA damage after intracellular uptake of both radiotracers., Conclusions: The data indicate damage to the cell to occur from absorbed dose without recognizable contribution from intracellular heterogeneity of radionuclide distribution. Comet assay under alkaline and neutral conditions did not reveal any shift to more complex DNA damage after radionuclide uptake. Cellular uptake of (99m)Tc and 211At increased cellular dose and reduced clonogenic survival.
- Published
- 2012
- Full Text
- View/download PDF
50. Characterization and modulation of fibroblast/endothelial cell co-cultures for the in vitro preformation of three-dimensional tubular networks.
- Author
-
Eckermann CW, Lehle K, Schmid SA, Wheatley DN, and Kunz-Schughart LA
- Subjects
- Apoptosis, Cell Count, Cell Survival, Cells, Cultured, Coculture Techniques methods, Culture Media chemistry, DNA Fragmentation, Fibroblast Growth Factors chemistry, Fibroblasts physiology, Human Umbilical Vein Endothelial Cells physiology, Humans, Neovascularization, Physiologic physiology, Serum chemistry, Spheroids, Cellular metabolism, Time Factors, Vascular Endothelial Growth Factor A chemistry, Fibroblasts cytology, Human Umbilical Vein Endothelial Cells cytology, Spheroids, Cellular physiology, Tissue Engineering
- Abstract
Various assays of different complexity are used in research on angiogenesis in health and disease. The results of these assays increasingly impact the field of tissue engineering because preformed microvascular networks may connect and conduct to the vascular system of the host, thereby helping us to support the survival of implanted cells and tissue constructs. An interesting model that supports the formation of EC (endothelial cells) tubular structures in vitro is based on co-culturing them with fibroblasts. Our initial multilayer approach was recently transferred into a three-dimensional spheroid model using HUVEC (human umbilical vein endothelial cells) as model cells. The aim of the present study is to further characterize, extend and validate this fibroblast/EC spheroid co-culture system. We have evaluated the model with a maximum size of 600-650 μm attained on day 3 from inoculation of 4×104 fibroblasts with 1×104 EC. Cell count and spheroid diameter significantly decreased as a function of time, but the EC network that developed over a period of 14 days in culture was clearly visible and viable, and central cell death was excluded. We successfully included HMVEC (human microvascular endothelial cells) of dermal origin in the system and replaced FBS (fetal bovine serum) with human AB serum, which positively impacted the EC network formation at optimized concentrations. The need for exogenous growth factors [VEGF (vascular endothelial growth factor), EGF (epithelial growth factor), bFGF (basic fibroblast growth factor) and IGF-1 (insulin-like growth factor-1)] routinely added to classical EC media was also assessed. The behaviour of both fibroblasts and EC in response to a combination of these exogenous growth factors differed critically in fibroblast/EC spheroid co-cultures compared with the same cells in the multilayer approach. VEGF was the most relevant exogenous factor for EC network formation in fibroblast/EC multilayers, but was ineffective in the spheroid system. IGF-1 was found, in general, to be dispensable; however, while it had a negative impact on EC networking in the presence of bFGF and EGF in the multilayer, it did not in the spheroid approach. We conclude that the critical determinants of EC network formation and cell survival are not universal, but have to be specifically optimized for each culture model.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.