69 results on '"Medová M"'
Search Results
2. Supplementary materials, methods, and figures 1-4 from Impact of p53 Status on Radiosensitization of Tumor Cells by MET Inhibition–Associated Checkpoint Abrogation
- Author
-
Mikami, K., primary, Medová, M., primary, Nisa, L., primary, Francica, P., primary, Glück, A.A., primary, Tschan, M.P., primary, Blaukat, A., primary, Bladt, F., primary, Aebersold, D.M., primary, and Zimmer, Y., primary
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Impact of p53 Status on Radiosensitization of Tumor Cells by MET Inhibition–Associated Checkpoint Abrogation
- Author
-
Mikami, K., primary, Medová, M., primary, Nisa, L., primary, Francica, P., primary, Glück, A.A., primary, Tschan, M.P., primary, Blaukat, A., primary, Bladt, F., primary, Aebersold, D.M., primary, and Zimmer, Y., primary
- Published
- 2023
- Full Text
- View/download PDF
4. Separation and detection of oxidation products of fluorodeoxyglucose and glucose by high-performance liquid chromatography–electrospray ionisation mass spectrometry
- Author
-
Búriová, E, Medová, M, Macášek, F, and Brúder, P
- Published
- 2004
- Full Text
- View/download PDF
5. Investigating a novel potential ATM/ATR/DNA-PK phosphorylation site on the MET RTK as a link between MET addiction and radioresistance
- Author
-
Koch, J.P., primary, Roth, S.M., additional, Quintin, A., additional, Gavini, J., additional, Stroka, D.M., additional, Aebersold, D.M., additional, Zimmer, Y., additional, and Medová, M., additional
- Published
- 2016
- Full Text
- View/download PDF
6. Depletion of FOXM1 via MET targeting underlies establishment of a DNA damage-induced senescence program in gastric cancer
- Author
-
Francica, P., primary, Lluís, N., additional, Aebersold, D.M., additional, Langer, R., additional, Bladt, F., additional, Blaukat, A., additional, Stroka, D.M., additional, Martínez, M. Rodríguez, additional, Zimmer, Y., additional, and Medová, M., additional
- Published
- 2016
- Full Text
- View/download PDF
7. Tepotinib. Selective MET receptor tyrosine kinase inhibitor, Treatment of lung and liver cancers
- Author
-
Medová, M., primary
- Published
- 2016
- Full Text
- View/download PDF
8. THU-039 - Direct and indirect hepatoprotective mechanism of CBLB502 a TLR5 agonist
- Author
-
Melin, N., Fahrner, R., Keogh, A., Medova, M., Zimmer, Y., Aebersold, D., Candinas, D., Beldi, G., and Stroka, D.
- Published
- 2018
- Full Text
- View/download PDF
9. Impact of p53 Status on Radiosensitization of Tumor Cells by MET Inhibition–Associated Checkpoint Abrogation
- Author
-
Mikami, K., primary, Medová, M., additional, Nisa, L., additional, Francica, P., additional, Glück, A.A., additional, Tschan, M.P., additional, Blaukat, A., additional, Bladt, F., additional, Aebersold, D.M., additional, and Zimmer, Y., additional
- Published
- 2015
- Full Text
- View/download PDF
10. 720 - Investigating a novel potential ATM/ATR/DNA-PK phosphorylation site on the MET RTK as a link between MET addiction and radioresistance
- Author
-
Koch, J.P., Roth, S.M., Quintin, A., Gavini, J., Stroka, D.M., Aebersold, D.M., Zimmer, Y., and Medová, M.
- Published
- 2016
- Full Text
- View/download PDF
11. 719 - Depletion of FOXM1 via MET targeting underlies establishment of a DNA damage-induced senescence program in gastric cancer
- Author
-
Francica, P., Lluís, N., Aebersold, D.M., Langer, R., Bladt, F., Blaukat, A., Stroka, D.M., Martínez, M. Rodríguez, Zimmer, Y., and Medová, M.
- Published
- 2016
- Full Text
- View/download PDF
12. 702 - The role of MET in tumor resistance to radiation therapy: a phosphoproteomic approach
- Author
-
Orlando, E., Bensimon, A., Medovà, M., Aebersold, D.M., Aebersold, R., and Zimmer, Y.
- Published
- 2016
- Full Text
- View/download PDF
13. HPV and p53 status as precision determinants of head and neck cancer response to DNA-PKcs inhibition in combination with irradiation.
- Author
-
Hayrapetyan L, Roth SM, Quintin A, Hovhannisyan L, Medo M, Riedo R, Ott JG, Albers J, Aebersold DM, Zimmer Y, and Medová M
- Abstract
Major risk factors of head and neck squamous cell carcinoma (HNSCC) are tobacco use and human papillomavirus (HPV). HPV E6 oncoprotein leads to p53 degradation, whereas HPV-negative cancers are frequently associated with TP53 mutations. Peposertib is a potent and selective, orally administered small-molecule inhibitor of the catalytic subunit of the DNA-dependent kinase (DNA-PKcs), a key regulator of non-homologous end joining (NHEJ). NHEJ inhibition along with irradiation (IR)-induced DNA double-strand breaks has the potential to increase antitumor treatment efficacy. Here, we investigated the responses of a panel of HNSCC models with distinct HPV and p53 status to treatments with IR, DNA-PKcs inhibition, and their combination in-vitro and in-vivo. IR-induced DNA damage combined with peposertib administration shortly before IR results in decreased cell viability and proliferation and causes DNA repair delay in all studied HNSCC cell lines. However, our data confirm that the actual cell fate upon this treatment is determined by cellular p53 and/or HPV status. Cells lacking functional p53 due to its degradation by HPV or due to a loss-of-function mutation are arrested in the G2/M phase of the cell cycle and eliminated by apoptosis whereas p53-proficient HNSCC cell lines preferentially undergo senescence. This is also recapitulated in-vivo, where HPV+ UD-SCC-2 xenografts display stronger and more durable responses to the combined treatment as compared to p53 wild-type UM-SCC-74A tumors. In conclusion, DNA-PKcs inhibitor peposertib should be further studied as a potential radiosensitizer for HNSCCs, taking into consideration the genetic background and the HPV status of a particular tumor.
- Published
- 2024
- Full Text
- View/download PDF
14. A comprehensive comparison of tools for fitting mutational signatures.
- Author
-
Medo M, Ng CKY, and Medová M
- Subjects
- Humans, Computational Biology methods, Software, Algorithms, DNA Mutational Analysis methods, Mutation, Neoplasms genetics
- Abstract
Mutational signatures connect characteristic mutational patterns in the genome with biological or chemical processes that take place in cancers. Analysis of mutational signatures can help elucidate tumor evolution, prognosis, and therapeutic strategies. Although tools for extracting mutational signatures de novo have been extensively benchmarked, a similar effort is lacking for tools that fit known mutational signatures to a given catalog of mutations. We fill this gap by comprehensively evaluating twelve signature fitting tools on synthetic mutational catalogs with empirically driven signature weights corresponding to eight cancer types. On average, SigProfilerSingleSample and SigProfilerAssignment/MuSiCal perform best for small and large numbers of mutations per sample, respectively. We further show that ad hoc constraining the list of reference signatures is likely to produce inferior results. Evaluation of real mutational catalogs suggests that the activity of signatures that are absent in the reference catalog poses considerable problems to all evaluated tools., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. E2F1-Associated Purine Synthesis Pathway Is a Major Component of the MET-DNA Damage Response Network.
- Author
-
Poliaková Turan M, Riedo R, Medo M, Pozzato C, Friese-Hamim M, Koch JP, Coggins SA, Li Q, Kim B, Albers J, Aebersold DM, Zamboni N, Zimmer Y, and Medová M
- Subjects
- Animals, Mice, Humans, DNA Repair drug effects, Cell Line, Tumor, Xenograft Model Antitumor Assays, Signal Transduction drug effects, DNA Damage drug effects, Purines biosynthesis, Purines metabolism, E2F1 Transcription Factor metabolism, E2F1 Transcription Factor genetics, Proto-Oncogene Proteins c-met metabolism, Proto-Oncogene Proteins c-met genetics
- Abstract
Various lines of investigation support a signaling interphase shared by receptor tyrosine kinases and the DNA damage response. However, the underlying network nodes and their contribution to the maintenance of DNA integrity remain unknown. We explored MET-related metabolic pathways in which interruption compromises proper resolution of DNA damage. Discovery metabolomics combined with transcriptomics identified changes in pathways relevant to DNA repair following MET inhibition (METi). METi by tepotinib was associated with the formation of γH2AX foci and with significant alterations in major metabolic circuits such as glycolysis, gluconeogenesis, and purine, pyrimidine, amino acid, and lipid metabolism. 5'-Phosphoribosyl-N-formylglycinamide, a de novo purine synthesis pathway metabolite, was consistently decreased in in vitro and in vivo MET-dependent models, and METi-related depletion of dNTPs was observed. METi instigated the downregulation of critical purine synthesis enzymes including phosphoribosylglycinamide formyltransferase, which catalyzes 5'-phosphoribosyl-N-formylglycinamide synthesis. Genes encoding these enzymes are regulated through E2F1, whose levels decrease upon METi in MET-driven cells and xenografts. Transient E2F1 overexpression prevented dNTP depletion and the concomitant METi-associated DNA damage in MET-driven cells. We conclude that DNA damage following METi results from dNTP reduction via downregulation of E2F1 and a consequent decline of de novo purine synthesis., Significance: Maintenance of genome stability prevents disease and affiliates with growth factor receptor tyrosine kinases. We identified de novo purine synthesis as a pathway in which key enzymatic players are regulated through MET receptor and whose depletion via MET targeting explains MET inhibition-associated formation of DNA double-strand breaks. The mechanistic importance of MET inhibition-dependent E2F1 downregulation for interference with DNA integrity has translational implications for MET-targeting-based treatment of malignancies., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
16. An optimized protocol for the generation and monitoring of conditional orthotopic lung cancer in the KP mouse model using an adeno-associated virus vector compatible with biosafety level 1.
- Author
-
Deng H, Ge H, Dubey C, Losmanova T, Medová M, Konstantinidou G, Mutlu SM, Birrer FE, Brodie TM, Stroka D, Wang W, Peng RW, Dorn P, and Marti TM
- Subjects
- Mice, Animals, Humans, Proto-Oncogene Proteins p21(ras) genetics, Dependovirus genetics, X-Ray Microtomography, Tumor Suppressor Protein p53, Containment of Biohazards, Disease Models, Animal, Genetic Vectors genetics, Lung Neoplasms genetics, Lung Neoplasms pathology, Adenocarcinoma of Lung
- Abstract
Background: The inducible Kras/p53 lung adenocarcinoma mouse model, which faithfully recapitulates human disease, is routinely initiated by the intratracheal instillation of a virus-based Cre recombinase delivery system. Handling virus-based delivery systems requires elevated biosafety levels, e.g., biosafety level 2 (BSL-2). However, in experimental animal research facilities, following exposure to viral vectors in a BSL-2 environment, rodents may not be reclassified to BSL-1 according to standard practice, preventing access to small animal micro-computed tomography (micro-CT) scanners that are typically housed in general access areas such as BSL-1 rooms. Therefore, our goal was to adapt the protocol so that the Cre-induced KP mouse model could be handled under BSL-1 conditions during the entire procedure., Results: The Kras-Lox-STOP-Lox-G12D/p53 flox/flox (KP)-based lung adenocarcinoma mouse model was activated by intratracheal instillation of either an adenoviral-based or a gutless, adeno-associated viral-based Cre delivery system. Tumor growth was monitored over time by micro-CT. We have successfully substituted the virus-based Cre delivery system with a commercially available, gutless, adeno-associated, Cre-expressing vector that allows the KP mouse model to be handled and imaged in a BSL-1 facility. By optimizing the anesthesia protocol and switching to a microscope-guided vector instillation procedure, productivity was increased and procedure-related complications were significantly reduced. In addition, repeated micro-CT analysis of individual animals allowed us to monitor tumor growth longitudinally, dramatically reducing the number of animals required per experiment. Finally, we documented the evolution of tumor volume for different doses, which revealed that individual tumor nodules induced by low-titer AAV-Cre transductions can be monitored over time by micro-CT., Conclusion: Modifications to the anesthesia and instillation protocols increased the productivity of the original KP protocol. In addition, the switch to a gutless, adeno-associated, Cre-expressing vector allowed longitudinal monitoring of tumor growth under BSL-1 conditions, significantly reducing the number of animals required for an experiment, in line with the 3R principles., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
17. Author Correction: Tumour mutations in long noncoding RNAs enhance cell fitness.
- Author
-
Esposito R, Lanzós A, Uroda T, Ramnarayanan S, Büchi I, Polidori T, Guillen-Ramirez H, Mihaljevic A, Merlin BM, Mela L, Zoni E, Hovhannisyan L, McCluggage F, Medo M, Basile G, Meise DF, Zwyssig S, Wenger C, Schwarz K, Vancura A, Bosch-Guiteras N, Andrades Á, Tham AM, Roemmele M, Medina PP, Ochsenbein AF, Riether C, Kruithof-de Julio M, Zimmer Y, Medová M, Stroka D, Fox A, and Johnson R
- Published
- 2023
- Full Text
- View/download PDF
18. Tumour mutations in long noncoding RNAs enhance cell fitness.
- Author
-
Esposito R, Lanzós A, Uroda T, Ramnarayanan S, Büchi I, Polidori T, Guillen-Ramirez H, Mihaljevic A, Merlin BM, Mela L, Zoni E, Hovhannisyan L, McCluggage F, Medo M, Basile G, Meise DF, Zwyssig S, Wenger C, Schwarz K, Vancura A, Bosch-Guiteras N, Andrades Á, Tham AM, Roemmele M, Medina PP, Ochsenbein AF, Riether C, Kruithof-de Julio M, Zimmer Y, Medová M, Stroka D, Fox A, and Johnson R
- Subjects
- Animals, Mice, Mutation, Oncogenes, Genomics, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Neoplasms genetics
- Abstract
Long noncoding RNAs (lncRNAs) are linked to cancer via pathogenic changes in their expression levels. Yet, it remains unclear whether lncRNAs can also impact tumour cell fitness via function-altering somatic "driver" mutations. To search for such driver-lncRNAs, we here perform a genome-wide analysis of fitness-altering single nucleotide variants (SNVs) across a cohort of 2583 primary and 3527 metastatic tumours. The resulting 54 mutated and positively-selected lncRNAs are significantly enriched for previously-reported cancer genes and a range of clinical and genomic features. A number of these lncRNAs promote tumour cell proliferation when overexpressed in in vitro models. Our results also highlight a dense SNV hotspot in the widely-studied NEAT1 oncogene. To directly evaluate the functional significance of NEAT1 SNVs, we use in cellulo mutagenesis to introduce tumour-like mutations in the gene and observe a significant and reproducible increase in cell fitness, both in vitro and in a mouse model. Mechanistic studies reveal that SNVs remodel the NEAT1 ribonucleoprotein and boost subnuclear paraspeckles. In summary, this work demonstrates the utility of driver analysis for mapping cancer-promoting lncRNAs, and provides experimental evidence that somatic mutations can act through lncRNAs to enhance pathological cancer cell fitness., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. A DNA-PK phosphorylation site on MET regulates its signaling interface with the DNA damage response.
- Author
-
Koch JP, Roth SM, Quintin A, Gavini J, Orlando E, Riedo R, Pozzato C, Hayrapetyan L, Aebersold R, Stroka DM, Aebersold DM, Medo M, Zimmer Y, and Medová M
- Subjects
- Humans, Cell Cycle Proteins genetics, DNA metabolism, DNA Damage, Mitosis genetics, Phosphorylation, DNA-Activated Protein Kinase genetics, DNA-Activated Protein Kinase metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
The DNA damage response (DDR) is intertwined with signaling pathways downstream of oncogenic receptor tyrosine kinases (RTKs). To drive research into the application of targeted therapies as radiosensitizers, a better understanding of this molecular crosstalk is necessary. We present here the characterization of a previously unreported MET RTK phosphosite, Serine 1016 (S1016) that represents a potential DDR-MET interface. MET S1016 phosphorylation increases in response to irradiation and is mainly targeted by DNA-dependent protein kinase (DNA-PK). Phosphoproteomics unveils an impact of the S1016A substitution on the overall long-term cell cycle regulation following DNA damage. Accordingly, the abrogation of this phosphosite strongly perturbs the phosphorylation of proteins involved in the cell cycle and formation of the mitotic spindle, enabling cells to bypass a G2 arrest upon irradiation and leading to the entry into mitosis despite compromised genome integrity. This results in the formation of abnormal mitotic spindles and a lower proliferation rate. Altogether, the current data uncover a novel signaling mechanism through which the DDR uses a growth factor receptor system for regulating and maintaining genome stability., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
20. CAR T cell-based immunotherapy and radiation therapy: potential, promises and risks.
- Author
-
Hovhannisyan L, Riether C, Aebersold DM, Medová M, and Zimmer Y
- Subjects
- Humans, Receptors, Antigen, T-Cell, Immunotherapy, Immunotherapy, Adoptive adverse effects, T-Lymphocytes, Neoplasms radiotherapy, Neoplasms etiology, Hematologic Neoplasms etiology
- Abstract
CAR T cell-based therapies have revolutionized the treatment of hematological malignancies such as leukemia and lymphoma within the last years. In contrast to the success in hematological cancers, the treatment of solid tumors with CAR T cells is still a major challenge in the field and attempts to overcome these hurdles have not been successful yet. Radiation therapy is used for management of various malignancies for decades and its therapeutic role ranges from local therapy to a priming agent in cancer immunotherapy. Combinations of radiation with immune checkpoint inhibitors have already proven successful in clinical trials. Therefore, a combination of radiation therapy may have the potential to overcome the current limitations of CAR T cell therapy in solid tumor entities. So far, only limited research was conducted in the area of CAR T cells and radiation. In this review we will discuss the potential and risks of such a combination in the treatment of cancer patients., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
21. In Reply to Raaijmakers et al.
- Author
-
Dennstädt F, Glatzer M, Medová M, and Putora PM
- Published
- 2023
- Full Text
- View/download PDF
22. Correction: An oncogene addiction phosphorylation signature and its derived scores inform tumor responsiveness to targeted therapies.
- Author
-
Orlando E, Medo M, Bensimon A, Quintin A, Riedo R, Roth SM, Riether C, Marti TM, Aebersold DM, Medová M, Aebersold R, and Zimmer Y
- Published
- 2023
- Full Text
- View/download PDF
23. Parameters of the Lyman Model for Calculation of Normal-Tissue Complication Probability: A Systematic Literature Review.
- Author
-
Dennstädt F, Medová M, Putora PM, and Glatzer M
- Subjects
- Humans, Probability, Radiobiology, Radiotherapy Planning, Computer-Assisted, Rectum radiation effects
- Abstract
Purpose: The Lyman model is one of the most used radiobiological models for calculation of normal-tissue complication probability (NTCP). Since its introduction in 1985, many authors have published parameter values for the model based on clinical data of different radiotherapeutic situations. This study attempted to collect the entirety of radiobiological parameter sets published to date and provide an overview of the data basis for different variations of the model. Furthermore, it sought to compare the parameter values and calculated NTCPs for selected endpoints with sufficient data available., Methods and Materials: A systematic literature analysis was performed, searching for publications that provided parameters for the different variations of the Lyman model in the Medline database using PubMed. Parameter sets were grouped into 13 toxicity-related endpoint groups. For 3 selected endpoint groups (≤25% reduction of saliva 12 months after irradiation of the parotid, symptomatic pneumonitis after irradiation of the lung, and bleeding of grade 2 or less after irradiation of the rectum), parameter values were compared and differences in calculated NTCP values were analyzed., Results: A total of 509 parameter sets from 130 publications were identified. Considerable heterogeneities were detected regarding the number of parameters available for different radio-oncological situations. Furthermore, for the 3 selected endpoints, large differences in published parameter values were found. These translated into great variations of calculated NTCPs, with maximum ranges of 35.2% to 93.4% for the saliva endpoint, of 39.4% to 90.4% for the pneumonitis endpoint, and of 5.4% to 99.3% for the rectal bleeding endpoint., Conclusions: The detected heterogeneity of the data as well as the large variations of published radiobiological parameters underline the necessity for careful interpretation when using such parameters for NTCP calculations. Appropriate selection of parameters and validation of values are essential when using the Lyman model., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. Beyond keratinocyte differentiation: emerging new biology of small proline-rich proteins.
- Author
-
Zabini A, Zimmer Y, and Medová M
- Subjects
- Humans, Proteins metabolism, Keratinocytes, Biology, Proline metabolism, Cornified Envelope Proline-Rich Proteins metabolism
- Abstract
Small proline-rich proteins (SPRRPs) are traditionally known for their function in keratinocyte homeostasis. Recent evidence demonstrates their involvement in additional diverse physiological processes ranging from p53 signaling and direct prevention of DNA damage to bactericidal activities. We highlight these novel, intriguing roles of SPRRPs and discuss them in the context of relevant pathological conditions., Competing Interests: Declaration of interests There are no conflicts of interest to disclose., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. Prognostic and Predictive Biomarkers in Head and Neck Squamous Cell Carcinoma Treated with Radiotherapy-A Systematic Review.
- Author
-
Schanne DH, Koch A, Elicin O, Giger R, Medová M, Zimmer Y, and Aebersold DM
- Abstract
Background : Radiotherapy is a mainstay in head and neck squamous cell carcinoma (HNSCC) treatment but is mostly applied without stratification by molecular diagnostics. Development of reliable biomarkers may have the potential to improve radiotherapy (RT) efficacy and reduce toxicity. We conducted a systematic review to summarize the field of biomarkers in HNSCC treated by RT. Methods : Pubmed and EMBASE were searched independently by two researchers following pre-defined inclusion and exclusion criteria. Z curves were generated to investigate publication bias. OncoKB was used for identification of druggable targets. Results : 134 manuscripts remained for data extraction. 12% of tumors were AJCC/UICC stage I-II and 82% were stage III-IV. The most common biomarkers were proteins (39%), DNA (14%) and mRNA (9%). Limiting analysis to prospective data and statistically significant results, we found three potentially druggable targets: ERCC2, PTCH1 and EGFR. Regarding data quality, AJCC/UICC stage was missing in 32% of manuscripts. 73% of studies were retrospective and only 7% were based on prospective randomized trials. Z-curves indicated the presence of publication bias. Conclusion : An abundance of potential biomarkers in HNSCC is available but data quality is limited by retrospective collection, lack of validation and publication bias. Improved study design and reporting quality might accelerate successful development of personalized treatments in HNSCC.
- Published
- 2022
- Full Text
- View/download PDF
26. An oncogene addiction phosphorylation signature and its derived scores inform tumor responsiveness to targeted therapies.
- Author
-
Orlando E, Medo M, Bensimon A, Quintin A, Riedo R, Roth SM, Riether C, Marti TM, Aebersold DM, Medová M, Aebersold R, and Zimmer Y
- Subjects
- Humans, Oncogene Addiction, Precision Medicine, Phosphorylation, Cell Line, Tumor, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Mutation, Cytoskeletal Proteins, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology
- Abstract
Purpose: Oncogene addiction provides important therapeutic opportunities for precision oncology treatment strategies. To date the cellular circuitries associated with driving oncoproteins, which eventually establish the phenotypic manifestation of oncogene addiction, remain largely unexplored. Data suggest the DNA damage response (DDR) as a central signaling network that intersects with pathways associated with deregulated addicting oncoproteins with kinase activity in cancer cells., Experimental: DESIGN: We employed a targeted mass spectrometry approach to systematically explore alterations in 116 phosphosites related to oncogene signaling and its intersection with the DDR following inhibition of the addicting oncogene alone or in combination with irradiation in MET-, EGFR-, ALK- or BRAF (V600)-positive cancer models. An NSCLC tissue pipeline combining patient-derived xenografts (PDXs) and ex vivo patient organotypic cultures has been established for treatment responsiveness assessment., Results: We identified an 'oncogene addiction phosphorylation signature' (OAPS) consisting of 8 protein phosphorylations (ACLY S455, IF4B S422, IF4G1 S1231, LIMA1 S490, MYCN S62, NCBP1 S22, P3C2A S259 and TERF2 S365) that are significantly suppressed upon targeted oncogene inhibition solely in addicted cell line models and patient tissues. We show that the OAPS is present in patient tissues and the OAPS-derived score strongly correlates with the ex vivo responses to targeted treatments., Conclusions: We propose a score derived from OAPS as a quantitative measure to evaluate oncogene addiction of cancer cell samples. This work underlines the importance of protein phosphorylation assessment for patient stratification in precision oncology and corresponding identification of tumor subtypes sensitive to inhibition of a particular oncogene., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. Synergistic effect of the TLR5 agonist CBLB502 and its downstream effector IL-22 against liver injury.
- Author
-
Melin N, Sánchez-Taltavull D, Fahrner R, Keogh A, Dosch M, Büchi I, Zimmer Y, Medová M, Beldi G, Aebersold DM, Candinas D, and Stroka D
- Subjects
- Animals, Cell Line, Tumor, Hepatocytes metabolism, Liver drug effects, Liver metabolism, Mice, Inbred C57BL, Radiation-Protective Agents pharmacology, Signal Transduction drug effects, Interleukin-22, Mice, Hepatocytes drug effects, Interleukins metabolism, Liver injuries, Peptides pharmacology, Toll-Like Receptor 5 drug effects
- Abstract
The toll-like receptor 5 (TLR5) agonist, CBLB502/Entolimod, is a peptide derived from bacterial flagellin and has been shown to protect against radiation-induced tissue damage in animal models. Here we investigated the protective mechanism of CBLB502 in the liver using models of ischemia-reperfusion injury and concanavalin A (ConA) induced immuno-hepatitis. We report that pretreatment of mice with CBLB502 provoked a concomitant activation of NF-κB and STAT3 signaling in the liver and reduced hepatic damage in both models. To understand the underlying mechanism, we screened for cytokines in the serum of CBLB502 treated animals and detected high levels of IL-22. There was no transcriptional upregulation of IL-22 in the liver, rather it was found in extrahepatic tissues, mainly the colon, mesenteric lymph nodes (MLN), and spleen. RNA-seq analysis on isolated hepatocytes demonstrated that the concomitant activation of NF-κB signaling by CBLB502 and STAT3 signaling by IL-22 produced a synergistic cytoprotective transcriptional signature. In IL-22 knockout mice, the loss of IL-22 resulted in a decrease of hepatic STAT3 activation, a reduction in the cytoprotective signature, and a loss of hepatoprotection following ischemia-reperfusion-induced liver injury. Taken together, these findings suggest that CBLB502 protects the liver by increasing hepatocyte resistance to acute liver injury through the cooperation of TLR5-NF-κB and IL-22-STAT3 signaling pathways.
- Published
- 2021
- Full Text
- View/download PDF
28. Enhancing CRISPR deletion via pharmacological delay of DNA-PKcs.
- Author
-
Bosch-Guiteras N, Uroda T, Guillen-Ramirez HA, Riedo R, Gazdhar A, Esposito R, Pulido-Quetglas C, Zimmer Y, Medová M, and Johnson R
- Subjects
- Animals, DNA genetics, DNA metabolism, DNA End-Joining Repair, DNA-Activated Protein Kinase metabolism, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins metabolism, CRISPR-Cas Systems genetics, DNA Breaks, Double-Stranded, DNA-Activated Protein Kinase antagonists & inhibitors, Gene Editing, Sequence Deletion
- Abstract
CRISPR-Cas9 deletion (CRISPR-del) is the leading approach for eliminating DNA from mammalian cells and underpins a variety of genome-editing applications. Target DNA, defined by a pair of double-strand breaks (DSBs), is removed during nonhomologous end-joining (NHEJ). However, the low efficiency of CRISPR-del results in laborious experiments and false-negative results. By using an endogenous reporter system, we show that repression of the DNA-dependent protein kinase catalytic subunit (DNA-PKcs)-an early step in NHEJ-yields substantial increases in DNA deletion. This is observed across diverse cell lines, gene delivery methods, commercial inhibitors, and guide RNAs, including those that otherwise display negligible activity. We further show that DNA-PKcs inhibition can be used to boost the sensitivity of pooled functional screens and detect true-positive hits that would otherwise be overlooked. Thus, delaying the kinetics of NHEJ relative to DSB formation is a simple and effective means of enhancing CRISPR-deletion., (© 2021 Bosch-Guiteras et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2021
- Full Text
- View/download PDF
29. DNA-PK in human malignant disorders: Mechanisms and implications for pharmacological interventions.
- Author
-
Medová M, Medo M, Hovhannisyan L, Muñoz-Maldonado C, Aebersold DM, and Zimmer Y
- Subjects
- Animals, DNA Breaks, Double-Stranded, DNA-Activated Protein Kinase metabolism, Disease Progression, Epithelial-Mesenchymal Transition physiology, Humans, Neoplasms genetics, Signal Transduction, Antineoplastic Agents pharmacology, DNA-Activated Protein Kinase antagonists & inhibitors, Neoplasms drug therapy
- Abstract
The DNA-PK holoenzyme is a fundamental element of the DNA damage response machinery (DDR), which is responsible for cellular genomic stability. Consequently, and predictably, over the last decades since its identification and characterization, numerous pre-clinical and clinical studies reported observations correlating aberrant DNA-PK status and activity with cancer onset, progression and responses to therapeutic modalities. Notably, various studies have established in recent years the role of DNA-PK outside the DDR network, corroborating its role as a pleiotropic complex involved in transcriptional programs that operate biologic processes as epithelial to mesenchymal transition (EMT), hypoxia, metabolism, nuclear receptors signaling and inflammatory responses. In particular tumor entities as prostate cancer, immense research efforts assisted mapping and describing the overall signaling networks regulated by DNA-PK that control metastasis and tumor progression. Correspondingly, DNA-PK emerges as an obvious therapeutic target in cancer and data pertaining to various pharmacological approaches have been published, largely in context of combination with DNA-damaging agents (DDAs) that act by inflicting DNA double strand breaks (DSBs). Currently, new generation inhibitors are tested in clinical trials. Several excellent reviews have been published in recent years covering the biology of DNA-PK and its role in cancer. In the current article we are aiming to systematically describe the main findings on DNA-PK signaling in major cancer types, focusing on both preclinical and clinical reports and present a detailed current status of the DNA-PK inhibitors repertoire., Competing Interests: Declaration of Competing Interest The authors declare that there are no conflicts of interest., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
30. Editorial: Exploiting DNA Damage Response in the Era of Precision Oncology.
- Author
-
Zimmer Y, Reinhardt HC, and Medová M
- Published
- 2020
- Full Text
- View/download PDF
31. The LIM Protein Ajuba Augments Tumor Metastasis in Colon Cancer.
- Author
-
Dommann N, Sánchez-Taltavull D, Eggs L, Birrer F, Brodie T, Salm L, Baier FA, Medová M, Humbert M, Tschan MP, Beldi G, Candinas D, and Stroka D
- Abstract
Colorectal cancer, along with its high potential for recurrence and metastasis, is a major health burden. Uncovering proteins and pathways required for tumor cell growth is necessary for the development of novel targeted therapies. Ajuba is a member of the LIM domain family of proteins whose expression is positively associated with numerous cancers. Our data shows that Ajuba is highly expressed in human colon cancer tissue and cell lines. Publicly available data from The Cancer Genome Atlas shows a negative correlation between survival and Ajuba expression in patients with colon cancer. To investigate its function, we transduced SW480 human colon cancer cells, with lentiviral constructs to knockdown or overexpress Ajuba protein. The transcriptome of the modified cell lines was analyzed by RNA sequencing. Among the pathways enriched in the differentially expressed genes, were cell proliferation, migration and differentiation. We confirmed our sequencing data with biological assays; cells depleted of Ajuba were less proliferative, more sensitive to irradiation, migrated less and were less efficient in colony formation. In addition, loss of Ajuba expression decreased the tumor burden in a murine model of colorectal metastasis to the liver. Taken together, our data supports that Ajuba promotes colon cancer growth, migration and metastasis and therefore is a potential candidate for targeted therapy.
- Published
- 2020
- Full Text
- View/download PDF
32. Deciphering MET-dependent modulation of global cellular responses to DNA damage by quantitative phosphoproteomics.
- Author
-
Bensimon A, Koch JP, Francica P, Roth SM, Riedo R, Glück AA, Orlando E, Blaukat A, Aebersold DM, Zimmer Y, Aebersold R, and Medová M
- Subjects
- Animals, Cell Line, Tumor, DNA Repair radiation effects, Down-Regulation radiation effects, Epithelium radiation effects, Female, Humans, Mesoderm radiation effects, Mice, Neoplasm Proteins metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation radiation effects, Radiation, Ionizing, Reproducibility of Results, Substrate Specificity radiation effects, Xenograft Model Antitumor Assays, DNA Damage, Epithelium pathology, Mesoderm pathology, Phosphoproteins metabolism, Proteomics
- Abstract
Increasing evidence suggests that interference with growth factor receptor tyrosine kinase (RTK) signaling can affect DNA damage response (DDR) networks, with a consequent impact on cellular responses to DNA-damaging agents widely used in cancer treatment. In that respect, the MET RTK is deregulated in abundance and/or activity in a variety of human tumors. Using two proteomic techniques, we explored how disrupting MET signaling modulates global cellular phosphorylation response to ionizing radiation (IR). Following an immunoaffinity-based phosphoproteomic discovery survey, we selected candidate phosphorylation sites for extensive characterization by targeted proteomics focusing on phosphorylation sites in both signaling networks. Several substrates of the DDR were confirmed to be modulated by sequential MET inhibition and IR, or MET inhibition alone. Upon combined treatment, for two substrates, NUMA1 S395 and CHEK1 S345, the gain and loss of phosphorylation, respectively, were recapitulated using invivo tumor models by immunohistochemistry, with possible utility in future translational research. Overall, we have corroborated phosphorylation sites at the intersection between MET and the DDR signaling networks, and suggest that these represent a class of proteins at the interface between oncogene-driven proliferation and genomic stability., (© 2020 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
33. MET targeting: time for a rematch.
- Author
-
Koch JP, Aebersold DM, Zimmer Y, and Medová M
- Subjects
- Animals, Carcinogenesis metabolism, Hepatocyte Growth Factor metabolism, Humans, Neoplasms metabolism, Proto-Oncogene Mas, Signal Transduction physiology, Proto-Oncogene Proteins c-met metabolism
- Abstract
MET, the receptor tyrosine kinase (RTK) for hepatocyte growth factor, is a proto-oncogene involved in embryonic development and throughout life in homeostasis and tissue regeneration. Deregulation of MET signaling has been reported in numerous malignancies, prompting great interest in MET targeting for cancer therapy. The present review offers a summary of the biology of MET and its known functions in normal physiology and carcinogenesis, followed by an overview of the most relevant MET-targeting strategies and corresponding clinical trials, highlighting both past setbacks and promising future prospects. By placing their efforts on a more precise stratification strategy through the genetic analysis of tumors, modern trials such as the NCI-MATCH trial could revive the past enthusiasm for MET-targeted therapy.
- Published
- 2020
- Full Text
- View/download PDF
34. Targeting the MET Receptor Tyrosine Kinase as a Strategy for Radiosensitization in Locoregionally Advanced Head and Neck Squamous Cell Carcinoma.
- Author
-
Nisa L, Francica P, Giger R, Medo M, Elicin O, Friese-Hamim M, Wilm C, Stroh C, Bojaxhiu B, Quintin A, Caversaccio MD, Dettmer MS, Buchwalder M, Brodie TM, Aebersold DM, Zimmer Y, Carey TE, and Medová M
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Xenograft Model Antitumor Assays, Protein Kinase Inhibitors therapeutic use, Radiation-Sensitizing Agents therapeutic use, Squamous Cell Carcinoma of Head and Neck drug therapy
- Abstract
Radiotherapy (RT) along with surgery is the mainstay of treatment in head and neck squamous cell carcinoma (HNSCC). Radioresistance represents a major source of treatment failure, underlining the urgent necessity to explore and implement effective radiosensitization strategies. The MET receptor widely participates in the acquisition and maintenance of an aggressive phenotype in HNSCC and modulates the DNA damage response following ionizing radiation (IR). Here, we assessed MET expression and mutation status in primary and metastatic lesions within a cohort of patients with advanced HNSCC. Moreover, we investigated the radiosensitization potential of the MET inhibitor tepotinib in a panel of cell lines, in vitro and in vivo , as well as in ex vivo patient-derived organotypic tissue cultures (OTC). MET was highly expressed in 62.4% of primary tumors and in 53.6% of lymph node metastases (LNM), and in 6 of 9 evaluated cell lines. MET expression in primaries and LNMs was significantly associated with decreased disease control in univariate survival analyses. Tepotinib abrogated MET phosphorylation and to distinct extent MET downstream signaling. Pretreatment with tepotinib resulted in variable radiosensitization, enhanced DNA damage, cell death, and G
2 -M-phase arrest. Combination of tepotinib with IR led to significant radiosensitization in one of two tested in vivo models. OTCs revealed differential patterns of response toward tepotinib, irradiation, and combination of both modalities. The molecular basis of tepotinib-mediated radiosensitization was studied by a CyTOF-based single-cell mass cytometry approach, which uncovered that MET inhibition modulated PI3K activity in cells radiosensitized by tepotinib but not in the resistant ones., (©2019 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
35. ProtRank: bypassing the imputation of missing values in differential expression analysis of proteomic data.
- Author
-
Medo M, Aebersold DM, and Medová M
- Subjects
- Humans, Models, Statistical, Phosphopeptides metabolism, Phosphoproteins metabolism, ROC Curve, Research Design, Algorithms, Gene Expression Profiling, Proteomics
- Abstract
Background: Data from discovery proteomic and phosphoproteomic experiments typically include missing values that correspond to proteins that have not been identified in the analyzed sample. Replacing the missing values with random numbers, a process known as "imputation", avoids apparent infinite fold-change values. However, the procedure comes at a cost: Imputing a large number of missing values has the potential to significantly impact the results of the subsequent differential expression analysis., Results: We propose a method that identifies differentially expressed proteins by ranking their observed changes with respect to the changes observed for other proteins. Missing values are taken into account by this method directly, without the need to impute them. We illustrate the performance of the new method on two distinct datasets and show that it is robust to missing values and, at the same time, provides results that are otherwise similar to those obtained with edgeR which is a state-of-art differential expression analysis method., Conclusions: The new method for the differential expression analysis of proteomic data is available as an easy to use Python package.
- Published
- 2019
- Full Text
- View/download PDF
36. A Comparative Analysis of Individual RAS Mutations in Cancer Biology.
- Author
-
Muñoz-Maldonado C, Zimmer Y, and Medová M
- Abstract
In human cells, three closely related RAS genes, termed HRAS, KRAS , and NRAS , encode four highly homologous proteins. RAS proteins are small GTPases involved in a broad spectrum of key molecular and cellular activities, including proliferation and survival among others. Gain-of-function missense mutations, mostly located at codons 12, 13, and 61, constitutively activate RAS proteins and can be detected in various types of human cancers. KRAS is the most frequently mutated, followed by NRAS and HRAS . However, each isoform exhibits distinctive mutation frequency at each codon, supporting the hypothesis that different RAS mutants may lead to distinct biologic manifestations. This review is focused on the differences in signaling and phenotype, as well as on transcriptomics, proteomics, and metabolomics profiles related to individual RAS-mutated variants. Additionally, association of these mutants with particular targeted outcomes and rare mutations at additional RAS codons are discussed., (Copyright © 2019 Muñoz-Maldonado, Zimmer and Medová.)
- Published
- 2019
- Full Text
- View/download PDF
37. Metabolomics reveals tepotinib-related mitochondrial dysfunction in MET-activating mutations-driven models.
- Author
-
Poliaková M, Felser A, Pierzchala K, Nuoffer JM, Aebersold DM, Zimmer Y, Zamboni N, and Medová M
- Subjects
- Animals, Citric Acid Cycle, Mice, Mitochondria physiology, Mutation, NIH 3T3 Cells, Proto-Oncogene Proteins c-met physiology, Reactive Oxygen Species metabolism, Metabolomics, Mitochondria drug effects, Piperidines pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met antagonists & inhibitors, Pyridazines pharmacology, Pyrimidines pharmacology
- Abstract
Genetic aberrations in the hepatocyte growth factor receptor tyrosine kinase MET induce oncogenic addiction in various types of human cancers, advocating MET as a viable anticancer target. Here, we report that MET signaling plays an important role in conferring a unique metabolic phenotype to cellular models expressing MET-activating mutated variants that are either sensitive or resistant toward MET small molecule inhibitors. MET phosphorylation downregulated by the specific MET inhibitor tepotinib resulted in markedly decreased viability and increased apoptosis in tepotinib-sensitive cells. Moreover, prior to the induction of MET inhibition-dependent cell death, tepotinib also led to an altered metabolic signature, characterized by a prominent reduction of metabolite ions related to amino sugar metabolism, gluconeogenesis, glycine and serine metabolism, and of numerous TCA cycle-related metabolites such as succinate, malate, and citrate. Functionally, a decrease in oxygen consumption rate, a reduced citrate synthase activity, a drop in membrane potential, and an associated misbalanced mitochondrial function were observed exclusively in MET inhibitor-sensitive cells. These data imply that interference with metabolic state can be considered an early indicator of efficient MET inhibition and particular changes reported here could be explored in the future as markers of efficacy of anti-MET therapies., (© 2019 Federation of European Biochemical Societies.)
- Published
- 2019
- Full Text
- View/download PDF
38. Oncogene addiction as a foundation of targeted cancer therapy: The paradigm of the MET receptor tyrosine kinase.
- Author
-
Orlando E, Aebersold DM, Medová M, and Zimmer Y
- Subjects
- Clinical Trials as Topic, Drug Design, Humans, Molecular Targeted Therapy, Neoplasms genetics, Neoplasms metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met chemistry, Proto-Oncogene Proteins c-met genetics, Signal Transduction drug effects, Neoplasms drug therapy, Oncogene Addiction genetics, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-met antagonists & inhibitors
- Abstract
Following nearly two decades of its conception, the phenomenon of oncogene addiction still represents a key concept of how progresses in basic research can translate to unprecedented translational breakthroughs. Coined by Bernard Weinstein, this term refers to the phenomenon by which cancer cells can exhibit dependence on a single oncogenic protein or signaling pathway for sustaining proliferation and survival, despite the wide burden of genetic lesions characterizing their genomic background, revealing thus a promising Achilles' heel of cancer. Importantly, this concept aided the design and clinical implementation of molecularly targeted anticancer therapies, further supporting the paradigm shift witnessed in clinical oncology towards an individual-based, personalized era. In this review, we outline the path of discovery concerning the oncogene addiction concept and focus on the MET receptor tyrosine kinase as a model addicting oncoprotein to further explore potential and pitfalls stemming from the implementation of anticancer strategies targeting tumor dependencies beyond their blending with other therapeutic opportunities., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
39. Comprehensive Genomic Profiling of Patient-matched Head and Neck Cancer Cells: A Preclinical Pipeline for Metastatic and Recurrent Disease.
- Author
-
Nisa L, Barras D, Medová M, Aebersold DM, Medo M, Poliaková M, Koch J, Bojaxhiu B, Eliçin O, Dettmer MS, Angelino P, Giger R, Borner U, Caversaccio MD, Carey TE, Ho L, McKee TA, Delorenzi M, and Zimmer Y
- Subjects
- Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases chemistry, Down-Regulation, Drug Resistance, Neoplasm, Female, Gene Dosage, Gene Expression Regulation, Neoplastic, Gene Regulatory Networks, Head and Neck Neoplasms drug therapy, Humans, Male, Mutation, Neoplasm Recurrence, Local drug therapy, Protein Domains, Sequence Analysis, RNA, Squamous Cell Carcinoma of Head and Neck drug therapy, Exome Sequencing, Class I Phosphatidylinositol 3-Kinases genetics, Cornified Envelope Proline-Rich Proteins genetics, Gene Expression Profiling methods, Genomics methods, Head and Neck Neoplasms genetics, Neoplasm Recurrence, Local genetics, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Metastases and tumor recurrence have a major prognostic impact in head and neck squamous cell carcinoma (HNSCC); however, cellular models that comprehensively characterize metastatic and recurrent HNSCC are lacking. To this end, we obtained genomic, transcriptomic, and copy number profiles of the UM-SCC cell line panel, encompassing patient-matched metastatic and recurrent cells. UM-SCC cells recapitulate the most prevalent genomic alterations described in HNSCC, featuring common TP53, PI3K, NOTCH, and Hippo pathway mutations. This analysis identified a novel F977Y kinase domain PIK3CA mutation exclusively present in a recurrent cell line (UM-SCC14B), potentially conferring resistance to PI3K inhibitors. Small proline-rich protein 2A (SPRR2A), a protein involved in epithelial homeostasis and invasion, was one of the most consistently downregulated transcripts in metastatic and recurrent UM-SCC cells. Assessment of SPRR2A protein expression in a clinical cohort of patients with HNSCC confirmed common SPRR2A downregulation in primary tumors (61.9% of cases) and lymph node metastases (31.3%), but not in normal tissue. High expression of SPRR2A in lymph node metastases was, along with nonoropharyngeal location of the primary tumor, an independent prognostic factor for regional disease recurrence after surgery and radiotherapy (HR 2.81; 95% CI, 1.16-6.79; P = 0.02). These results suggest that SPRR2A plays a dual role in invasion and therapeutic resistance in HNSCC, respectively through its downregulation and overexpression. IMPLICATIONS: The current study reveals translationally relevant mechanisms underlying metastasis and recurrence in HNSCC and represents an adjuvant tool for preclinical research in this disease setting. Underlining its discovery potential this approach identified a PIK3CA-resistant mutation as well as SPRR2A as possible theragnostic markers., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
40. Identification of a MET-eIF4G1 translational regulation axis that controls HIF-1α levels under hypoxia.
- Author
-
Glück AA, Orlando E, Leiser D, Poliaková M, Nisa L, Quintin A, Gavini J, Stroka DM, Berezowska S, Bubendorf L, Blaukat A, Aebersold DM, Medová M, and Zimmer Y
- Subjects
- Animals, Cell Line, Tumor, Disease-Free Survival, Gene Expression Regulation, Neoplastic genetics, Humans, Mice, Mitogen-Activated Protein Kinases genetics, Phosphorylation genetics, Signal Transduction genetics, Carcinoma, Non-Small-Cell Lung genetics, Eukaryotic Initiation Factor-4G genetics, Hypoxia genetics, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Lung Neoplasms genetics, Proto-Oncogene Proteins c-met genetics
- Abstract
Poor oxygenation is a common hallmark of solid cancers that strongly associates with aggressive tumor progression and treatment resistance. While a hypoxia-inducible factor 1α (HIF-1α)-associated transcriptional overexpression of the hepatocyte growth factor (HGF) receptor tyrosine kinase (RTK) MET has been previously documented, any regulation of the HIF-1α system through MET downstream signaling in hypoxic tumors has not been yet described. By using MET-driven in vitro as well as ex vivo tumor organotypic fresh tissue models we report that MET targeting results in depletion of HIF-1α and its various downstream targets. Mechanistically, we provide evidence that MET regulates HIF-1α levels through a protein translation mechanism that relies on phosphorylation modulation of the eukaryotic initiation factor 4G1 (eIF4G1) on serine 1232 (Ser-1232). Targeted phosphoproteomics data demonstrate a significant drop in eIF4G1 Ser-1232 phosphorylation following MET targeting, which is linked to an increased affinity between eIF4G1 and eIF4E. Since phosphorylation of eIF4G1 on Ser-1232 is largely mediated through mitogen-activated protein kinase (MAPK), we show that expression of a constitutively active K-RAS variant is sufficient to abrogate the inhibitory effect of MET targeting on the HIF-1α pathway with subsequent resistance of tumor cells to MET targeting under hypoxic conditions. Analysis of The Cancer Genome Atlas data demonstrates frequent co-expression of MET, HIF-1α and eIF4G1 in various solid tumors and its impact on disease-free survival of non-small cell lung cancer patients. Clinical relevance of the MET-eIF4G1-HIF-1α pathway is further supported by a co-occurrence of their expression in common tumor regions of individual lung cancer patients.
- Published
- 2018
- Full Text
- View/download PDF
41. OMIP-045: Characterizing human head and neck tumors and cancer cell lines with mass cytometry.
- Author
-
Brodie TM, Tosevski V, and Medová M
- Subjects
- Cell Line, Tumor, Flow Cytometry methods, Humans, Signal Transduction physiology, Head and Neck Neoplasms pathology
- Published
- 2018
- Full Text
- View/download PDF
42. The relevance of tyrosine kinase inhibitors for global metabolic pathways in cancer.
- Author
-
Poliaková M, Aebersold DM, Zimmer Y, and Medová M
- Subjects
- Animals, Energy Metabolism drug effects, Glucose metabolism, Glycolysis drug effects, Humans, Lipid Metabolism drug effects, Signal Transduction drug effects, Enzyme Inhibitors therapeutic use, Neoplasms metabolism, Protein-Tyrosine Kinases metabolism
- Abstract
Tumor metabolism is a thrilling discipline that focuses on mechanisms used by cancer cells to earn crucial building blocks and energy to preserve growth and overcome resistance to various treatment modalities. At the same time, therapies directed specifically against aberrant signalling pathways driven by protein tyrosine kinases (TKs) involved in proliferation, metastasis and growth count for several years to promising anti-cancer approaches. In this respect, small molecule inhibitors are the most widely used clinically relevant means for targeted therapy, with a rising number of approvals for TKs inhibitors. In this review, we discuss recent observations related to TKs-associated metabolism and to metabolic feedback that is initialized as cellular response to particular TK-targeted therapies. These observations provide collective evidence that therapeutic responses are primarily linked to such pathways as regulation of lipid and amino acid metabolism, TCA cycle and glycolysis, advocating therefore the development of further effective targeted therapies against a broader spectrum of TKs to treat patients whose tumors display deregulated signalling driven by these proteins.
- Published
- 2018
- Full Text
- View/download PDF
43. PIK3CA hotspot mutations differentially impact responses to MET targeting in MET-driven and non-driven preclinical cancer models.
- Author
-
Nisa L, Häfliger P, Poliaková M, Giger R, Francica P, Aebersold DM, Charles RP, Zimmer Y, and Medová M
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Class I Phosphatidylinositol 3-Kinases, Disease Models, Animal, Drug Resistance, Neoplasm genetics, Mice, NIH 3T3 Cells, Neoplasms metabolism, Neoplasms pathology, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met metabolism, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Mutation, Neoplasms genetics, Phosphatidylinositol 3-Kinases genetics, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met genetics
- Abstract
Background: The MET receptor tyrosine kinase represents a promising target in cancer. PIK3CA activating mutations are common in several tumor types and can potentially confer resistance to anti-receptor tyrosine kinase therapy., Methods: MET and/or PI3K pathway inhibition was assessed in NIH3T3 cells harboring MET-activating point mutation with or without ectopic expression of PIK3CA
E545K and PIK3CAH1047R , as well as in MET-expressing head and neck cancer cells with endogenous PIK3CA mutations. Endpoints included PI3K pathway activation, cell proliferation, colony-forming ability, cell death, wound-healing, and an in vivo model., Results: PIK3CAE545K and PIK3CAH1047R confer resistance to MET inhibition in MET-driven models. PIK3CAH1047R was more potent than PIK3CAE545K at inducing resistance in PI3K pathway activation, cell proliferation, colony-forming ability, induction of cell death and wound-healing upon MET inhibition. Resistance to MET inhibition could be synergistically overcome by co-targeting PI3K. Furthermore, combined MET/PI3K inhibition led to enhanced anti-tumor activity in vivo in tumors harboring PIK3CAH1047R . In head and neck cancer cells the combination of MET/PI3K inhibitors led to more-than-additive effects., Conclusions: PIK3CA mutations can lead to resistance to MET inhibition, supporting future clinical evaluation of combinations of PI3K and MET inhibitors in common scenarios of malignant neoplasms featuring aberrant MET expression and PIK3CA mutations.- Published
- 2017
- Full Text
- View/download PDF
44. Senescence as biologic endpoint following pharmacological targeting of receptor tyrosine kinases in cancer.
- Author
-
Francica P, Aebersold DM, and Medová M
- Subjects
- Biomarkers metabolism, DNA Damage, Humans, Molecular Targeted Therapy, Neoplasms enzymology, Neoplasms pathology, Oncogenes, Oxidative Stress, Radiotherapy, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Antineoplastic Agents therapeutic use, Cellular Senescence drug effects, Neoplasms therapy, Receptor Protein-Tyrosine Kinases metabolism
- Abstract
Cellular senescence was first described in 1961 in a seminal study by Hayflick and Moorhead as a limit to the replicative lifespan of somatic cells after serial cultivation. Since then, major advances in our understanding of senescence have been achieved suggesting that this mechanism is activated also by oncogenic stimuli, oxidative stress and DNA damage, giving rise to the concept of premature senescence. Regardless of the initial trigger, numerous experimental observations have been provided to support the notion that both replicative and premature senescence play pivotal roles in early stages of tumorigenesis and in response of tumor cells to anticancer treatments. Moreover, various studies have suggested that the induction of senescence by both chemo- and radiotherapy in a variety of cancer types correlates with treatment outcome. As it is widely accepted that cellular senescence may function as a fundamental barrier of tumor progression, the significance of senescence for clinical interventions that make use of novel molecular targeting-based modalities needs to be well defined. Interestingly, despite numerous studies evaluating efficacies of receptor tyrosine kinases (RTKs) targeting strategies in both preclinical and clinical settings, the relevance of RTKs inhibition-associated senescence in tumors remains less characterized. Here we review the available literature that describes premature senescence as a major mechanism following targeting of RTKs in preclinical as well as in clinical settings. Additionally, we discuss the possible role of diverse RTKs in regulating the induction of senescence following cellular stress and possible implications of this crosstalk in identification of biomarkers of inhibitor-mediated chemo- and radiosensitization approaches., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
45. Hepatocyte growth factor secreted by bone marrow stem cell reduce ER stress and improves repair in alveolar epithelial II cells.
- Author
-
Nita I, Hostettler K, Tamo L, Medová M, Bombaci G, Zhong J, Allam R, Zimmer Y, Roth M, Geiser T, and Gazdhar A
- Subjects
- Alveolar Epithelial Cells drug effects, Cell Line, Tumor, Cells, Cultured, Culture Media, Conditioned pharmacology, Endoplasmic Reticulum Chaperone BiP, Hepatocyte Growth Factor metabolism, Humans, Thapsigargin toxicity, Tunicamycin toxicity, Alveolar Epithelial Cells metabolism, Bone Marrow Cells metabolism, Endoplasmic Reticulum Stress, Hepatocyte Growth Factor pharmacology
- Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a progressive, irreversible lung disease with complex pathophysiology. Evidence of endoplasmic reticulum (ER) stress has been reported in alveolar epithelial cells (AEC) in IPF patients. Secreted mediators from bone marrow stem cells (BMSC-cm) have regenerative properties. In this study we investigate the beneficial effects of BMSC-cm on ER stress response in primary AEC and ER stressed A549 cells. We hypothesize that BMSC-cm reduces ER stress. Primary AEC isolated from IPF patients were treated with BMSC-cm. To induce ER stress A549 cells were incubated with Tunicamycin or Thapsigargin and treated with BMSC-cm, or control media. Primary IPF-AEC had high Grp78 and CHOP gene expression, which was lowered after BMSC-cm treatment. Similar results were observed in ER stressed A549 cells. Alveolar epithelial repair increased in presence of BMSC-cm in ER stressed A549 cells. Hepatocyte growth factor (HGF) was detected in biologically relevant levels in BMSC-cm. Neutralization of HGF in BMSC-cm attenuated the beneficial effects of BMSC-cm including synthesis of surfactant protein C (SP-C) in primary AEC, indicating a crucial role of HGF in ER homeostasis and alveolar epithelial repair. Our data suggest that BMSC-cm may be a potential therapeutic option for treating pulmonary fibrosis., Competing Interests: The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
46. Depletion of FOXM1 via MET Targeting Underlies Establishment of a DNA Damage-Induced Senescence Program in Gastric Cancer.
- Author
-
Francica P, Nisa L, Aebersold DM, Langer R, Bladt F, Blaukat A, Stroka D, Martínez MR, Zimmer Y, and Medová M
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Cellular Senescence genetics, DNA Damage genetics, Drug Resistance, Neoplasm drug effects, Gene Amplification drug effects, Gene Amplification genetics, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic genetics, Humans, Signal Transduction drug effects, Signal Transduction genetics, Stomach Neoplasms genetics, Cellular Senescence drug effects, DNA Damage drug effects, Forkhead Box Protein M1 genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met genetics, Stomach Neoplasms drug therapy
- Abstract
Purpose: Deregulated signaling via the MET receptor tyrosine kinase is abundant in gastric tumors, with up to 80% of cases displaying aberrant MET expression. A growing body of evidence suggests MET as a potential target for tumor radiosensitization., Experimental Design: Cellular proliferation and DNA damage-induced senescence were studied in a panel of MET-overexpressing human gastric cancer cell lines as well as in xenograft models after MET inhibition and/or ionizing radiation. Pathways activation and protein expression were assessed by immunoblotting and immunohistochemistry. Tumor tissue microarrays (91 gastric cancer patients) were generated and copy number alteration (178 patients) and gene expression (373 patients) data available at The Cancer Genome Atlas were analyzed to assess the coalterations of MET and FOXM1., Results: MET targeting administered before ionizing radiation instigates DNA damage-induced senescence (∼80%, P < 0.001) rather than cell death. MET inhibition-associated senescence is linked to the blockade of MAPK pathway, correlates with downregulation of FOXM1, and can be abrogated (11.8% vs. 95.3%, P < 0.001) by ectopic expression of FOXM1 in the corresponding gastric tumor cells. Cells with ectopic FOXM1 expression demonstrate considerable (∼20%, P < 0.001) growth advantage despite MET targeting, suggesting a novel clinically relevant resistance mechanism to MET inhibition as the copresence of both MET and FOXM1 protein (33%) and mRNA (30%) overexpression as well as gene amplification (24,7%) are common in patients with gastric cancer., Conclusions: FOXM1, a negative regulator of senescence, has been identified as a key downstream effector and potential clinical biomarker that mediates MET signaling following infliction of DNA damage in gastric tumors. Clin Cancer Res; 22(21); 5322-36. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
47. KRAS and HRAS mutations confer resistance to MET targeting in preclinical models of MET-expressing tumor cells.
- Author
-
Leiser D, Medová M, Mikami K, Nisa L, Stroka D, Blaukat A, Bladt F, Aebersold DM, and Zimmer Y
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Humans, MAP Kinase Signaling System, Mice, NIH 3T3 Cells, Neoplasms pathology, Proto-Oncogene Proteins c-met antagonists & inhibitors, Pyridazines pharmacology, Pyrimidines pharmacology, Genes, ras, Mutation, Neoplasms enzymology, Proto-Oncogene Proteins c-met metabolism
- Abstract
The MET receptor tyrosine kinase is often deregulated in human cancers and several MET inhibitors are evaluated in clinical trials. Similarly to EGFR, MET signals through the RAS-RAF-ERK/MAPK pathway which plays key roles in cell proliferation and survival. Mutations of genes encoding for RAS proteins, particularly in KRAS, are commonly found in various tumors and are associated with constitutive activation of the MAPK pathway. It was shown for EGFR, that KRAS mutations render upstream EGFR inhibition ineffective in EGFR-positive colorectal cancers. Currently, there are no clinical studies evaluating MET inhibition impairment due to RAS mutations. To test the impact of RAS mutations on MET targeting, we generated tumor cells responsive to the MET inhibitor EMD1214063 that express KRAS G12V, G12D, G13D and HRAS G12V variants. We demonstrate that these MAPK-activating RAS mutations differentially interfere with MET-mediated biological effects of MET inhibition. We report increased residual ERK1/2 phosphorylation indicating that the downstream pathway remains active in presence of MET inhibition. Consequently, RAS variants counteracted MET inhibition-induced morphological changes as well as anti-proliferative and anchorage-independent growth effects. The effect of RAS mutants was reversed when MET inhibition was combined with MEK inhibitors AZD6244 and UO126. In an in vivo mouse xenograft model, MET-driven tumors harboring mutated RAS displayed resistance to MET inhibition. Taken together, our results demonstrate for the first time in details the role of KRAS and HRAS mutations in resistance to MET inhibition and suggest targeting both MET and MEK as an effective strategy when both oncogenic drivers are expressed., (Copyright © 2015 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
48. Impact of MET targeting on tumor-associated angiogenesis and growth of MET mutations-driven models of liver cancer.
- Author
-
Piguet AC, Medová M, Keogh A, Glück AA, Aebersold DM, Dufour JF, and Zimmer Y
- Abstract
Deregulated expression of the MET receptor tyrosine kinase has been reported in up to 50% of patients with hepatocellular carcinoma, the most abundant form of liver cancers, and is associated with decreased survival. Consequently, MET is considered as a molecular target in this malignancy, whose progression is highly dependent on extensive angiogenesis. Here we studied the impact of MET small molecule inhibitors on angiogenesis-associated parameters and growth of xenograft liver models consisting of cells expressing MET-mutated variants M1268T and Y1248H, which exhibit constitutive kinase activity. We demonstrate that MET mutations expression is associated with significantly increased production of vascular endothelial growth factor, which is blocked by MET targeting only in cells expressing the M1268T inhibitor-sensitive but not in the Y1248H inhibitor-resistant variant. Decrease in vascular endothelial growth factor production is also associated with reduction of tyrosine phopshorylation of the vascular endothelial growth factor receptor 2 expressed on primary liver sinusoidal endothelial cells and with inhibition of vessel formation. Furthermore, MET inhibition demonstrated an efficient anti-tumor activity and considerable reduction in microvessel density only against the M1268T-derived intrahepatic tumors. Collectively, our data support the role of targeting MET-associated angiogenesis as a major biological determinant for liver tumor growth control.
- Published
- 2015
- Full Text
- View/download PDF
49. Interplay between receptor tyrosine kinases and hypoxia signaling in cancer.
- Author
-
Glück AA, Aebersold DM, Zimmer Y, and Medová M
- Subjects
- Animals, Cell Hypoxia physiology, Humans, Hypoxia genetics, Hypoxia-Inducible Factor 1 physiology, Receptor Protein-Tyrosine Kinases genetics, Signal Transduction, Hypoxia metabolism, Neoplasms genetics, Neoplasms metabolism, Receptor Protein-Tyrosine Kinases physiology
- Abstract
Deregulated signaling via receptor tyrosine kinase (RTK) pathways is prevalent in numerous types of human cancers and is commonly correlated with worst prognosis, resistance to various treatment modalities and increased mortality. Likewise, hypoxic tumors are often manifested by aggressive mode of growth and progression following an adaptive genetic reprogramming with consequent transcriptional activation of genes encoding proteins, which support tumor survival under low oxygen-related conditions. Consequently, both the hypoxia-inducible factor (HIF) system, which is the major mediator of hypoxia-related signaling, and numerous RTK systems are considered critical molecular targets in current cancer therapy. It is now evident that there is an intricate molecular crosstalk between RTKs and hypoxia-related signaling in the sense that hypoxia can activate expression of particular RTKs and/or their corresponding ligands, while some RTK systems have been shown to trigger activation of the HIF machinery. Moreover, signaling regulation of some RTK systems under hypoxic conditions has also been documented to take place in a HIF-independent manner. With this review we aim at overviewing the most current observations on that topic and highlight the importance of the potential co-drugging the HIF system along with particular relevant RTKs for better tumor growth control., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
50. Profiling invasiveness in head and neck cancer: recent contributions of genomic and transcriptomic approaches.
- Author
-
Nisa L, Aebersold DM, Giger R, Caversaccio MD, Borner U, Medová M, and Zimmer Y
- Abstract
High-throughput molecular profiling approaches have emerged as precious research tools in the field of head and neck translational oncology. Such approaches have identified and/or confirmed the role of several genes or pathways in the acquisition/maintenance of an invasive phenotype and the execution of cellular programs related to cell invasion. Recently published new-generation sequencing studies in head and neck squamous cell carcinoma (HNSCC) have unveiled prominent roles in carcinogenesis and cell invasion of mutations involving NOTCH1 and PI3K-patwhay components. Gene-expression profiling studies combined with systems biology approaches have allowed identifying and gaining further mechanistic understanding into pathways commonly enriched in invasive HNSCC. These pathways include antigen-presenting and leucocyte adhesion molecules, as well as genes involved in cell-extracellular matrix interactions. Here we review the major insights into invasiveness in head and neck cancer provided by high-throughput molecular profiling approaches.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.