90 results on '"Radimerski T"'
Search Results
2. Both IKKβ/NFκB and PKA/HSL pathways are involved in lipopolysaccharides-induced lipolysis in human adipocytes: T2:OS5.3
- Author
-
Grisouard, J, Bouillet, E, Timper, K, Radimerski, T, Dembinski, K, Frey, D M, Peterli, R, Zulewski, H, Keller, U, Müller, B, and Christ-Crain, M
- Published
- 2010
3. Metformin induces glucose uptake in human preadipocyte-derived adipocytes from various fat depots
- Author
-
Fischer, M., Timper, K., Radimerski, T., Dembinski, K., Frey, D. M., Zulewski, H., Keller, U., Müller, B., Christ-Crain, M., and Grisouard, J.
- Published
- 2010
- Full Text
- View/download PDF
4. Insulin signaling and growth control in Drosophila
- Author
-
Stocker, H., Oldham, S., Brogiolo, W., Rintelen, F., Radimerski, T., Thomas, G., and Hafen, Ernst
- Published
- 2001
5. Effects of Jak2 Type 1 Inhibitors NVP-BSK805 and NVP-BVB808 on Jak2 Mutation-Positive and Bcr-Abl-Positive Cell Lines
- Author
-
Radimerski T, Michal Schwarz, Manley Pw, Peggy Grille, Frauke Ringel, Marque F, le Coutre P, Bernd Dörken, Jaspal Kaeda, and Christian Oberender
- Subjects
Fusion Proteins, bcr-abl ,Apoptosis ,Pharmacology ,Biology ,Piperazines ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Quinoxalines ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,medicine ,Humans ,Phosphorylation ,Protein Kinase Inhibitors ,Cell Proliferation ,Myeloproliferative Disorders ,Kinase ,Tumor Suppressor Proteins ,Imatinib ,Hematology ,General Medicine ,Janus Kinase 2 ,medicine.disease ,Haematopoiesis ,Pyrimidines ,Nilotinib ,Benzamides ,Mutation ,Imatinib Mesylate ,Cancer research ,Signal transduction ,K562 Cells ,Janus kinase ,Tyrosine kinase ,medicine.drug ,Chronic myelogenous leukemia - Abstract
Janus kinases are critical components of signaling pathways that regulate hematopoiesis. Mutations of the non-receptor tyrosine kinase JAK2 are found in many BCR-ABL-negative myeloproliferative neoplasms. Preclinical results support that JAK2 inhibitors could show efficacy in treating chronic myeloproliferative neoplasms. JAK2 has also been postulated to play a role in BCR-ABL signal transduction. Therefore, inhibitors of JAK2 kinases are turning into therapeutic strategies for treatment of chronic myelogenous leukemia (CML). In this study, the effects of two novel JAK2 inhibitors, NVP-BSK805 and NVP-BVB808, have been investigated in cell lines expressing either BCR-ABL or mutant JAK2. Possible synergies between NVP-BSK805/NVP-BVB808 and the kinase inhibitors imatinib and nilotinib were assessed. Proliferation and apoptosis tests with both substances showed response in the following cell lines: CHRF-288-11, SET-2 and UKE-1. All BCR-ABL-positive cell lines showed some reduction in proliferation, but with half-maximal growth-inhibitory values >1 µM. Combination of the JAK2 inhibitors with imatinib and nilotinib showed no significant additive or synergistic effects, although all BCR-ABL-positive cell lines responded well to both CML therapeutic agents. Interestingly, it seemed that the combination of imatinib with NVP-BSK805 had a protective effect on the cells. Combination treatment with nilotinib did not show this effect.
- Published
- 2014
6. HSP90 inhibition leads to degradation of the TYK2 kinase and apoptotic cell death in T-cell acute lymphoblastic leukemia
- Author
-
Akahane, K, primary, Sanda, T, additional, Mansour, M R, additional, Radimerski, T, additional, DeAngelo, D J, additional, Weinstock, D M, additional, and Look, A T, additional
- Published
- 2015
- Full Text
- View/download PDF
7. Differential effects of 5AMP-activated protein kinase (AMPK) activation by metformin and AICAR on glucose uptake and lipolysis in human adipocytes
- Author
-
Radimerski, T, Seboek, D, Kola, B, Linscheid, P, Zulewski, H, Grossman, A, Keller, U, Mueller, B, Korbonits, M, and Christ-Crain, M
- Published
- 2007
8. 24: Proffered Paper: JAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: A rationale for co-targeting these pathways in metastatic breast cancer
- Author
-
Britschgi, A., primary, Andraos, R., additional, Brinkhaus, H., additional, Klebba, I., additional, Romanet, V., additional, Mueller, U., additional, Murakami, M., additional, Radimerski, T., additional, and Bentires-Alj, M., additional
- Published
- 2014
- Full Text
- View/download PDF
9. The Drosophila forkhead transcription factor FOXO mediates the reduction in cell number associated with reduced insulin signaling
- Author
-
Jünger, M. A., Rintelen, F., Stocker, H., Jonathan Wasserman, Végh, M., Radimerski, T., Greenberg, M. E., Hafen, E., University of Zurich, and Hafen, E
- Subjects
Male ,10127alt Institute of Zoology (former) ,Transcription, Genetic ,Molecular Sequence Data ,Genes, Insect ,1100 General Agricultural and Biological Sciences ,Protein Serine-Threonine Kinases ,3-Phosphoinositide-Dependent Protein Kinases ,1300 General Biochemistry, Genetics and Molecular Biology ,Animals ,Drosophila Proteins ,Insulin ,Amino Acid Sequence ,Caenorhabditis elegans Proteins ,lcsh:QH301-705.5 ,Cells, Cultured ,Cell Death ,Sequence Homology, Amino Acid ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Forkhead Transcription Factors ,Organ Size ,Up-Regulation ,Oxidative Stress ,Phenotype ,lcsh:Biology (General) ,Protein Biosynthesis ,Mutation ,Insulin Receptor Substrate Proteins ,570 Life sciences ,biology ,590 Animals (Zoology) ,Drosophila ,Female ,Energy Intake ,Research Article ,Signal Transduction ,Transcription Factors - Abstract
Background Forkhead transcription factors belonging to the FOXO subfamily are negatively regulated by protein kinase B (PKB) in response to signaling by insulin and insulin-like growth factor in Caenorhabditis elegans and mammals. In Drosophila, the insulin-signaling pathway regulates the size of cells, organs, and the entire body in response to nutrient availability, by controlling both cell size and cell number. In this study, we present a genetic characterization of dFOXO, the only Drosophila FOXO ortholog. Results Ectopic expression of dFOXO and human FOXO3a induced organ-size reduction and cell death in a manner dependent on phosphoinositide (PI) 3-kinase and nutrient levels. Surprisingly, flies homozygous for dFOXO null alleles are viable and of normal size. They are, however, more sensitive to oxidative stress. Furthermore, dFOXO function is required for growth inhibition associated with reduced insulin signaling. Loss of dFOXO suppresses the reduction in cell number but not the cell-size reduction elicited by mutations in the insulin-signaling pathway. By microarray analysis and subsequent genetic validation, we have identified d4E-BP, which encodes a translation inhibitor, as a relevant dFOXO target gene. Conclusion Our results show that dFOXO is a crucial mediator of insulin signaling in Drosophila, mediating the reduction in cell number in insulin-signaling mutants. We propose that in response to cellular stresses, such as nutrient deprivation or increased levels of reactive oxygen species, dFOXO is activated and inhibits growth through the action of target genes such as d4E-BP.
- Published
- 2003
10. The Drosophila forkhead transcription factor FOXO mediates the reduction in cell number associated with reduced insulin signaling.
- Author
-
Jünger, M A, Rintelen, F, Stocker, H, Wasserman, J D, Végh, M, Radimerski, T, Greenberg, M E, Hafen, E, Jünger, M A, Rintelen, F, Stocker, H, Wasserman, J D, Végh, M, Radimerski, T, Greenberg, M E, and Hafen, E
- Abstract
BACKGROUND: Forkhead transcription factors belonging to the FOXO subfamily are negatively regulated by protein kinase B (PKB) in response to signaling by insulin and insulin-like growth factor in Caenorhabditis elegans and mammals. In Drosophila, the insulin-signaling pathway regulates the size of cells, organs, and the entire body in response to nutrient availability, by controlling both cell size and cell number. In this study, we present a genetic characterization of dFOXO, the only Drosophila FOXO ortholog. RESULTS: Ectopic expression of dFOXO and human FOXO3a induced organ-size reduction and cell death in a manner dependent on phosphoinositide (PI) 3-kinase and nutrient levels. Surprisingly, flies homozygous for dFOXO null alleles are viable and of normal size. They are, however, more sensitive to oxidative stress. Furthermore, dFOXO function is required for growth inhibition associated with reduced insulin signaling. Loss of dFOXO suppresses the reduction in cell number but not the cell-size reduction elicited by mutations in the insulin-signaling pathway. By microarray analysis and subsequent genetic validation, we have identified d4E-BP, which encodes a translation inhibitor, as a relevant dFOXO target gene. CONCLUSION: Our results show that dFOXO is a crucial mediator of insulin signaling in Drosophila, mediating the reduction in cell number in insulin-signaling mutants. We propose that in response to cellular stresses, such as nutrient deprivation or increased levels of reactive oxygen species, dFOXO is activated and inhibits growth through the action of target genes such as d4E-BP.
- Published
- 2003
11. Rheb is an essential regulator of S6K in controlling cell growth in Drosophila.
- Author
-
Stocker, H, Radimerski, T, Schindelholz, B, Wittwer, F, Belawat, P, Daram, P, Breuer, S, Thomas, G, Hafen, E, Stocker, H, Radimerski, T, Schindelholz, B, Wittwer, F, Belawat, P, Daram, P, Breuer, S, Thomas, G, and Hafen, E
- Abstract
Understanding the mechanisms through which multicellular organisms regulate cell, organ and body growth is of relevance to developmental biology and to research on growth-related diseases such as cancer. Here we describe a new effector in growth control, the small GTPase Rheb (Ras homologue enriched in brain). Mutations in the Drosophila melanogaster Rheb gene were isolated as growth-inhibitors, whereas overexpression of Rheb promoted cell growth. Our genetic and biochemical analyses suggest that Rheb functions downstream of the tumour suppressors Tsc1 (tuberous sclerosis 1)-Tsc2 in the TOR (target of rapamycin) signalling pathway to control growth, and that a major effector of Rheb function is ribosomal S6 kinase (S6K).
- Published
- 2003
12. Insulin Resistance and Increased Lipolysis in Bone Marrow Derived Adipocytes Stimulated with Agonists of Toll-like Receptors
- Author
-
Franchini, M., primary, Monnais, E., additional, Seboek, D., additional, Radimerski, T., additional, Zini, E., additional, Kaufmann, K., additional, Lutz, T., additional, Reusch, C., additional, Ackermann, M., additional, Muller, B., additional, and Linscheid, P., additional
- Published
- 2010
- Full Text
- View/download PDF
13. dS6K-regulated cell growth is dPKB/dPI(3)K-independent, but requires dPDK1.
- Author
-
Radimerski, T, Montagne, J, Rintelen, F, Stocker, H, van der Kaay, J, Downes, C P, Hafen, E, Thomas, G, Radimerski, T, Montagne, J, Rintelen, F, Stocker, H, van der Kaay, J, Downes, C P, Hafen, E, and Thomas, G
- Abstract
Genetic studies in Drosophila melanogaster underscore the importance of the insulin-signalling pathway in controlling cell, organ and animal size. Effectors of this pathway include Chico (the insulin receptor substrate homologue), dPI(3)K, dPKB, dPTEN, and dS6K. Mutations in any of these components have a striking effect on cell size and number, with the exception of dS6K. Mutants in dS6K affect cell size but not cell number, seemingly consistent with arguments that dS6K is a distal effector in the signalling pathway, directly controlled by dTOR, a downstream effector of dPI(3)K and dPKB. Unexpectedly, recent studies showed that dS6K activity is unimpaired in chico-deficient larvae, suggesting that dS6K activation may be mediated through the dPI(3)K docking sites of the Drosophila insulin receptor. Here, we show genetically, pharmacologically and biochemically that dS6K resides on an insulin signalling pathway distinct from that of dPKB, and surprisingly also from that of dPI(3)K. More striking, despite dPKB-dPI(3)K-independence, dS6K activity is dependent on the Drosophila homologue of the phosphoinositide-dependent protein kinase 1, dPDK1, demonstrating that both dPDK1, as well as dTOR, mediated dS6K activation is phosphatidylinositide-3,4,5-trisphosphate (PIP3)-independent.
- Published
- 2002
14. Orally Active 2-Amino Thienopyrimidine Inhibitors of the Hsp90 Chaperone
- Author
-
Brough, P.A., primary, Barril, X., additional, Borgognoni, J., additional, Chene, P., additional, Davies, N.G.M., additional, Davis, B., additional, Drysdale, M.J., additional, Dymock, B., additional, Eccles, S.A., additional, Garcia-Echeverria, C., additional, Fromont, C., additional, Hayes, A., additional, Hubbard, R.E., additional, Jordan, A.M., additional, Rugaard-Jensen, M., additional, Massey, A., additional, Merret, A., additional, Padfield, A., additional, Parsons, R., additional, Radimerski, T., additional, Raynaud, F.I., additional, Robertson, A., additional, Roughley, S.D., additional, Schoepfer, J., additional, Simmonite, H., additional, Surgenor, A., additional, Valenti, M., additional, Walls, S., additional, Webb, P., additional, Wood, M., additional, Workman, P., additional, and Wright, L.M., additional
- Published
- 2009
- Full Text
- View/download PDF
15. 155 POSTER Pharmakokinetic/phamacodynamic relationship in human xenograft models and PBMC's treated with the Hsp90 inhibitor NVP-AUY922
- Author
-
Jensen, M.R., primary, Ide, S., additional, Brueggen, J., additional, Schoepfer, J., additional, Motwani, M., additional, Wang, X., additional, Radimerski, T., additional, Quadt, C., additional, Garcia-Echeverria, C., additional, and Chene, P., additional
- Published
- 2008
- Full Text
- View/download PDF
16. Genetic and biochemical characterization of dTOR, the Drosophila homolog of the target of rapamycin.
- Author
-
Oldham, S, Montagne, J, Radimerski, T, Thomas, G, Hafen, E, Oldham, S, Montagne, J, Radimerski, T, Thomas, G, and Hafen, E
- Abstract
The adaptation of growth in response to nutritional changes is essential for the proper development of all organisms. Here we describe the identification of the Drosophila homolog of the target of rapamycin (TOR), a candidate effector for nutritional sensing. Genetic and biochemical analyses indicate that dTOR impinges on the insulin signaling pathway by autonomously affecting growth through modulating the activity of dS6K. However, in contrast to other components in the insulin signaling pathway, partial loss of dTOR function preferentially reduces growth of the endoreplicating tissues. These results are consistent with dTOR residing on a parallel amino acid sensing pathway.
- Published
- 2000
17. Insulin Signaling: Lessons from the Drosophila Tuberous Sclerosis Complex, a Tumor Suppressor
- Author
-
Montagne, J., primary, Radimerski, T., additional, and Thomas, G., additional
- Published
- 2001
- Full Text
- View/download PDF
18. Bim and Mcl-1 exert key roles in regulating JAK2V617F cell survival
- Author
-
Guthy Daniel A, Andraos Rita, Qian Zhiyan, Rubert Joëlle, and Radimerski Thomas
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The JAK2V617F mutation plays a major role in the pathogenesis of myeloproliferative neoplasms and is found in the vast majority of patients suffering from polycythemia vera and in roughly every second patient suffering from essential thrombocythemia or from primary myelofibrosis. The V617F mutation is thought to provide hematopoietic stem cells and myeloid progenitors with a survival and proliferation advantage. It has previously been shown that activated JAK2 promotes cell survival by upregulating the anti-apoptotic STAT5 target gene Bcl-xL. In this study, we have investigated the role of additional apoptotic players, the pro-apoptotic protein Bim as well as the anti-apoptotic protein Mcl-1. Methods Pharmacological inhibition of JAK2/STAT5 signaling in JAK2V617F mutant SET-2 and MB-02 cells was used to study effects on signaling, cell proliferation and apoptosis by Western blot analysis, WST-1 proliferation assays and flow cytometry. Cells were transfected with siRNA oligos to deplete candidate pro- and anti-apoptotic proteins. Co-immunoprecipitation assays were performed to assess the impact of JAK2 inhibition on complexes of pro- and anti-apoptotic proteins. Results Treatment of JAK2V617F mutant cell lines with a JAK2 inhibitor was found to trigger Bim activation. Furthermore, Bim depletion by RNAi suppressed JAK2 inhibitor-induced cell death. Bim activation following JAK2 inhibition led to enhanced sequestration of Mcl-1, besides Bcl-xL. Importantly, Mcl-1 depletion by RNAi was sufficient to compromise JAK2V617F mutant cell viability and sensitized the cells to JAK2 inhibition. Conclusions We conclude that Bim and Mcl-1 have key opposing roles in regulating JAK2V617F cell survival and propose that inactivation of aberrant JAK2 signaling leads to changes in Bim complexes that trigger cell death. Thus, further preclinical evaluation of combinations of JAK2 inhibitors with Bcl-2 family antagonists that also tackle Mcl-1, besides Bcl-xL, is warranted to assess the therapeutic potential for the treatment of chronic myeloproliferative neoplasms.
- Published
- 2011
- Full Text
- View/download PDF
19. The Drosophila Forkhead transcription factor FOXO mediates the reduction in cell number associated with reduced insulin signaling
- Author
-
Végh Mátyás, Stocker Hugo, Wasserman Jonathan D, Rintelen Felix, Jünger Martin A, Radimerski Thomas, Greenberg Michael E, and Hafen Ernst
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Background Forkhead transcription factors belonging to the FOXO subfamily are negatively regulated by protein kinase B (PKB) in response to signaling by insulin and insulin-like growth factor in Caenorhabditis elegans and mammals. In Drosophila, the insulin-signaling pathway regulates the size of cells, organs, and the entire body in response to nutrient availability, by controlling both cell size and cell number. In this study, we present a genetic characterization of dFOXO, the only Drosophila FOXO ortholog. Results Ectopic expression of dFOXO and human FOXO3a induced organ-size reduction and cell death in a manner dependent on phosphoinositide (PI) 3-kinase and nutrient levels. Surprisingly, flies homozygous for dFOXO null alleles are viable and of normal size. They are, however, more sensitive to oxidative stress. Furthermore, dFOXO function is required for growth inhibition associated with reduced insulin signaling. Loss of dFOXO suppresses the reduction in cell number but not the cell-size reduction elicited by mutations in the insulin-signaling pathway. By microarray analysis and subsequent genetic validation, we have identified d4E-BP, which encodes a translation inhibitor, as a relevant dFOXO target gene. Conclusion Our results show that dFOXO is a crucial mediator of insulin signaling in Drosophila, mediating the reduction in cell number in insulin-signaling mutants. We propose that in response to cellular stresses, such as nutrient deprivation or increased levels of reactive oxygen species, dFOXO is activated and inhibits growth through the action of target genes such as d4E-BP.
- Published
- 2003
- Full Text
- View/download PDF
20. Author Correction: DCAF1-based PROTACs with activity against clinically validated targets overcoming intrinsic- and acquired-degrader resistance.
- Author
-
Schröder M, Renatus M, Liang X, Meili F, Zoller T, Ferrand S, Gauter F, Li X, Sigoillot F, Gleim S, Stachyra TM, Thomas JR, Begue D, Khoshouei M, Lefeuvre P, Andraos-Rey R, Chung B, Ma R, Pinch B, Hofmann A, Schirle M, Schmiedeberg N, Imbach P, Gorses D, Calkins K, Bauer-Probst B, Maschlej M, Niederst M, Maher R, Henault M, Alford J, Ahrne E, Tordella L, Hollingworth G, Thomä NH, Vulpetti A, Radimerski T, Holzer P, Carbonneau S, and Thoma CR
- Published
- 2024
- Full Text
- View/download PDF
21. Design, Synthesis, and In Vitro and In Vivo Evaluation of Cereblon Binding Bruton's Tyrosine Kinase (BTK) Degrader CD79b Targeted Antibody-Drug Conjugates.
- Author
-
Zhang A, Seiss K, Laborde L, Palacio-Ramirez S, Guthy D, Lanter M, Lorber J, Vulpetti A, Arista L, Zoller T, Radimerski T, Thoma C, Hebach C, Tschantz WR, Karpov A, Hollingworth GJ, D'Alessio JA, Ferretti S, and Burger MT
- Subjects
- Agammaglobulinaemia Tyrosine Kinase, Antibodies, Monoclonal chemistry, Immunoconjugates chemistry
- Abstract
Antibody-drug conjugates (ADCs) are an established modality that allow for targeted delivery of a potent molecule, or payload, to a desired site of action. ADCs, wherein the payload is a targeted protein degrader, are an emerging area in the field. Herein we describe our efforts of delivering a Bruton's tyrosine kinase (BTK) bifunctional degrader 1 via a CD79b mAb (monoclonal antibody) where the degrader is linked at the ligase binding portion of the payload via a cleavable linker to the mAb. The resulting CD79b ADCs, 3 and 4 , exhibit in vitro degradation and cytotoxicity comparable with that of 1 , and ADC 3 can achieve more sustained in vivo degradation than intravenously administered 1 with markedly reduced systemic exposure of the payload.
- Published
- 2024
- Full Text
- View/download PDF
22. DCAF1-based PROTACs with activity against clinically validated targets overcoming intrinsic- and acquired-degrader resistance.
- Author
-
Schröder M, Renatus M, Liang X, Meili F, Zoller T, Ferrand S, Gauter F, Li X, Sigoillot F, Gleim S, Stachyra TM, Thomas JR, Begue D, Khoshouei M, Lefeuvre P, Andraos-Rey R, Chung B, Ma R, Pinch B, Hofmann A, Schirle M, Schmiedeberg N, Imbach P, Gorses D, Calkins K, Bauer-Probst B, Maschlej M, Niederst M, Maher R, Henault M, Alford J, Ahrne E, Tordella L, Hollingworth G, Thomä NH, Vulpetti A, Radimerski T, Holzer P, Carbonneau S, and Thoma CR
- Subjects
- Proteolysis, Ubiquitin metabolism, Carrier Proteins metabolism, Ubiquitin-Protein Ligases metabolism, Proteolysis Targeting Chimera
- Abstract
Targeted protein degradation (TPD) mediates protein level through small molecule induced redirection of E3 ligases to ubiquitinate neo-substrates and mark them for proteasomal degradation. TPD has recently emerged as a key modality in drug discovery. So far only a few ligases have been utilized for TPD. Interestingly, the workhorse ligase CRBN has been observed to be downregulated in settings of resistance to immunomodulatory inhibitory drugs (IMiDs). Here we show that the essential E3 ligase receptor DCAF1 can be harnessed for TPD utilizing a selective, non-covalent DCAF1 binder. We confirm that this binder can be functionalized into an efficient DCAF1-BRD9 PROTAC. Chemical and genetic rescue experiments validate specific degradation via the CRL4
DCAF1 E3 ligase. Additionally, a dasatinib-based DCAF1 PROTAC successfully degrades cytosolic and membrane-bound tyrosine kinases. A potent and selective DCAF1-BTK-PROTAC (DBt-10) degrades BTK in cells with acquired resistance to CRBN-BTK-PROTACs while the DCAF1-BRD9 PROTAC (DBr-1) provides an alternative strategy to tackle intrinsic resistance to VHL-degrader, highlighting DCAF1-PROTACS as a promising strategy to overcome ligase mediated resistance in clinical settings., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
23. Discovery of New Binders for DCAF1, an Emerging Ligase Target in the Targeted Protein Degradation Field.
- Author
-
Vulpetti A, Holzer P, Schmiedeberg N, Imbach-Weese P, Pissot-Soldermann C, Hollingworth GJ, Radimerski T, Thoma CR, Stachyra TM, Wojtynek M, Maschlej M, Chau S, Schuffenhauer A, Fernández C, Schröder M, and Renatus M
- Abstract
In this study, we describe the rapid identification of potent binders for the WD40 repeat domain (WDR) of DCAF1. This was achieved by two rounds of iterative focused screening of a small set of compounds selected on the basis of internal WDR domain knowledge followed by hit expansion. Subsequent structure-based design led to nanomolar potency binders with a clear exit vector enabling DCAF1-based bifunctional degrader exploration., Competing Interests: The authors declare no competing financial interest., (© 2023 American Chemical Society.)
- Published
- 2023
- Full Text
- View/download PDF
24. A high-throughput drug screen reveals means to differentiate triple-negative breast cancer.
- Author
-
Vulin M, Jehanno C, Sethi A, Correia AL, Obradović MMS, Couto JP, Coissieux MM, Diepenbruck M, Preca BT, Volkmann K, der Maur PA, Schmidt A, Münst S, Sauteur L, Kloc M, Palafox M, Britschgi A, Unterreiner V, Galuba O, Claerr I, Lopez-Romero S, Galli GG, Baeschlin D, Okamoto R, Soysal SD, Mechera R, Weber WP, Radimerski T, and Bentires-Alj M
- Subjects
- Breast pathology, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation, Estrogen Receptor alpha, Humans, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- Abstract
Plasticity delineates cancer subtypes with more or less favourable outcomes. In breast cancer, the subtype triple-negative lacks expression of major differentiation markers, e.g., estrogen receptor α (ERα), and its high cellular plasticity results in greater aggressiveness and poorer prognosis than other subtypes. Whether plasticity itself represents a potential vulnerability of cancer cells is not clear. However, we show here that cancer cell plasticity can be exploited to differentiate triple-negative breast cancer (TNBC). Using a high-throughput imaging-based reporter drug screen with 9 501 compounds, we have identified three polo-like kinase 1 (PLK1) inhibitors as major inducers of ERα protein expression and downstream activity in TNBC cells. PLK1 inhibition upregulates a cell differentiation program characterized by increased DNA damage, mitotic arrest, and ultimately cell death. Furthermore, cells surviving PLK1 inhibition have decreased tumorigenic potential, and targeting PLK1 in already established tumours reduces tumour growth both in cell line- and patient-derived xenograft models. In addition, the upregulation of genes upon PLK1 inhibition correlates with their expression in normal breast tissue and with better overall survival in breast cancer patients. Our results indicate that differentiation therapy based on PLK1 inhibition is a potential alternative strategy to treat TNBC., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
25. Transient Inhibition of the JAK/STAT Pathway Prevents B-ALL Development in Genetically Predisposed Mice.
- Author
-
Casado-García A, Isidro-Hernández M, Oak N, Mayado A, Mann-Ran C, Raboso-Gallego J, Alemán-Arteaga S, Buhles A, Sterker D, Sánchez EG, Martínez-Cano J, Blanco O, Orfao A, Alonso-López D, De Las Rivas J, Riesco S, Prieto-Matos P, González-Murillo Á, García Criado FJ, García Cenador MB, Radimerski T, Ramírez-Orellana M, Cobaleda C, Yang JJ, Vicente-Dueñas C, Weiss A, Nichols KE, and Sánchez-García I
- Subjects
- Animals, Humans, Mice, PAX5 Transcription Factor genetics, STAT Transcription Factors, Signal Transduction genetics, Janus Kinases genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma
- Abstract
Preventing development of childhood B-cell acute lymphoblastic leukemia (B-ALL), a disease with devastating effects, is a longstanding and unsolved challenge. Heterozygous germline alterations in the PAX5 gene can lead to B-ALL upon accumulation of secondary mutations affecting the JAK/STAT signaling pathway. Preclinical studies have shown that this malignant transformation occurs only under immune stress such as exposure to infectious pathogens. Here we show in Pax5+/- mice that transient, early-life administration of clinically relevant doses of ruxolitinib, a JAK1/2 inhibitor, significantly mitigates the risk of B-ALL following exposure to infection; 1 of 29 animals treated with ruxolitinib developed B-ALL versus 8 of 34 untreated mice. Ruxolitinib treatment preferentially targeted Pax5+/- versus wild-type B-cell progenitors and exerted unique effects on the Pax5+/- B-cell progenitor transcriptional program. These findings provide the first in vivo evidence for a potential strategy to prevent B-ALL development., Significance: JAK/STAT inhibition suppresses tumorigenesis in a B-ALL-susceptible mouse model, presenting a novel approach to prevent B-ALL onset., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
26. Optimization of the In Vivo Potency of Pyrazolopyrimidine MALT1 Protease Inhibitors by Reducing Metabolism and Increasing Potency in Whole Blood.
- Author
-
Quancard J, Simic O, Pissot Soldermann C, Aichholz R, Blatter M, Renatus M, Erbel P, Melkko S, Endres R, Sorge M, Kieffer L, Wagner T, Beltz K, Mcsheehy P, Wartmann M, Régnier CH, Calzascia T, Radimerski T, Bigaud M, Weiss A, Bornancin F, and Schlapbach A
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Caspase Inhibitors chemical synthesis, Caspase Inhibitors metabolism, Caspase Inhibitors pharmacokinetics, Cell Line, Tumor, Female, Half-Life, Humans, Mice, Inbred BALB C, Mice, SCID, Microsomes, Liver metabolism, Neoplasms drug therapy, Pyrazoles chemical synthesis, Pyrazoles metabolism, Pyrazoles pharmacokinetics, Pyrimidines chemical synthesis, Pyrimidines metabolism, Pyrimidines pharmacokinetics, Rats, Sprague-Dawley, Sheep, Urea chemical synthesis, Urea metabolism, Urea pharmacokinetics, Xenograft Model Antitumor Assays, Blood metabolism, Caspase Inhibitors therapeutic use, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Pyrazoles therapeutic use, Pyrimidines therapeutic use, Urea therapeutic use
- Abstract
The paracaspase MALT1 has gained increasing interest as a target for the treatment of subsets of lymphomas as well as autoimmune diseases, and there is a need for suitable compounds to explore the therapeutic potential of this target. Here, we report the optimization of the in vivo potency of pyrazolopyrimidines, a class of highly selective allosteric MALT1 inhibitors. High doses of the initial lead compound led to tumor stasis in an activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) xenograft model, but this compound suffered from a short in vivo half-life and suboptimal potency in whole blood. Guided by metabolism studies, we identified compounds with reduced metabolic clearance and increased in vivo half-life. In the second optimization step, masking one of the hydrogen-bond donors of the central urea moiety through an intramolecular interaction led to improved potency in whole blood. This was associated with improved in vivo potency in a mechanistic model of B cell activation. The optimized compound led to tumor regression in a CARD11 mutant ABC-DLBCL lymphoma xenograft model.
- Published
- 2020
- Full Text
- View/download PDF
27. Discovery of Potent, Highly Selective, and In Vivo Efficacious, Allosteric MALT1 Inhibitors by Iterative Scaffold Morphing.
- Author
-
Pissot Soldermann C, Simic O, Renatus M, Erbel P, Melkko S, Wartmann M, Bigaud M, Weiss A, McSheehy P, Endres R, Santos P, Blank J, Schuffenhauer A, Bold G, Buschmann N, Zoller T, Altmann E, Manley PW, Dix I, Buchdunger E, Scesa J, Quancard J, Schlapbach A, Bornancin F, Radimerski T, and Régnier CH
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Caspase Inhibitors chemical synthesis, Caspase Inhibitors pharmacology, Drug Discovery, Female, Humans, Immunity, Humoral drug effects, Male, Mice, Inbred BALB C, Molecular Structure, Pyrazoles chemical synthesis, Pyrazoles pharmacology, Pyrazoles therapeutic use, Pyrimidines chemical synthesis, Pyrimidines pharmacology, Pyrimidines therapeutic use, Rats, Sprague-Dawley, Structure-Activity Relationship, T-Lymphocytes drug effects, Urea pharmacology, Xenograft Model Antitumor Assays, Caspase Inhibitors therapeutic use, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Neoplasms drug therapy, Urea analogs & derivatives, Urea therapeutic use
- Abstract
MALT1 plays a central role in immune cell activation by transducing NF-κB signaling, and its proteolytic activity represents a key node for therapeutic intervention. Two cycles of scaffold morphing of a high-throughput biochemical screening hit resulted in the discovery of MLT-231, which enabled the successful pharmacological validation of MALT1 allosteric inhibition in preclinical models of humoral immune responses and B-cell lymphomas. Herein, we report the structural activity relationships (SARs) and analysis of the physicochemical properties of a pyrazolopyrimidine-derived compound series. In human T-cells and B-cell lymphoma lines, MLT-231 potently and selectively inhibits the proteolytic activity of MALT1 in NF-κB-dependent assays. Both in vitro and in vivo profiling of MLT-231 support further optimization of this in vivo tool compound toward preclinical characterization.
- Published
- 2020
- Full Text
- View/download PDF
28. A proteomic study of myeloproliferative neoplasms using reverse-phase protein arrays.
- Author
-
Tognon R, Almeida-E-Silva DC, Andraos-Rey R, Ristov M, Ambrósio L, de Almeida FC, de Souza Nunes N, Xisto Souto E, de Lourdes Perobelli L, Simões BP, Alexander Guthy D, Radimerski T, and Attié de Castro F
- Subjects
- Humans, Janus Kinase 2 genetics, Mutation, Phosphatidylinositol 3-Kinases, Protein Array Analysis, Proteomics, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders genetics, Neoplasms
- Abstract
Myeloproliferative neoplasms polycythemia vera (PV), essential thrombocythaemia (ET) and primary myelofibrosis constitute a group of haematological diseases. The comprehensive assessment of signaling pathway activation in blood cells may aid the understanding of MPN pathophysiology. Thus, levels of post-translational protein modifications and total protein expression were determined in MPN patients and control leukocytes by using reverse-phase protein arrays (RPPA). Compared to control samples, p-SRC, p-CTNNB1, c-MYC, MCL-1, p-MDM2, BAX and CCNB1 showed higher expression in PV samples than controls. P-JAK2/JAK2 and pro-apoptotic BIM showed differential expression between JAK2 V617F-positive and -negative ET patients. Apoptosis, cancer and PI3K/AKT pathways proteins showed differential expression among the studied groups. For most of the proteins analyzed using Western-Blot and RPPA, RPPA showed higher sensitivity to detect subtle differences. Taken together, our data indicate deregulated protein expression in MPN patients compared to controls. Thus, RPPA may be a useful method for broad proteome analysis in MPN patients´ leukocytes.
- Published
- 2020
- Full Text
- View/download PDF
29. Identification of the HECT E3 ligase UBR5 as a regulator of MYC degradation using a CRISPR/Cas9 screen.
- Author
-
Schukur L, Zimmermann T, Niewoehner O, Kerr G, Gleim S, Bauer-Probst B, Knapp B, Galli GG, Liang X, Mendiola A, Reece-Hoyes J, Rapti M, Barbosa I, Reschke M, Radimerski T, and Thoma CR
- Subjects
- Apoptosis, Humans, Neoplasms genetics, Neoplasms metabolism, Protein Binding, Proto-Oncogene Proteins c-myc genetics, Tumor Cells, Cultured, Ubiquitin-Protein Ligases genetics, CRISPR-Cas Systems, Neoplasms pathology, Proteolysis, Proto-Oncogene Proteins c-myc metabolism, Ubiquitin-Protein Ligases metabolism, Ubiquitination
- Abstract
MYC oncoprotein is a multifunctional transcription factor that regulates the expression of a large number of genes involved in cellular growth, proliferation and metabolism. Altered MYC protein level lead to cellular transformation and tumorigenesis. MYC is deregulated in > 50% of human cancers, rendering it an attractive drug target. However, direct inhibition of this class of proteins using conventional small molecules is challenging due to their intrinsically disordered state. To discover novel posttranslational regulators of MYC protein stability and turnover, we established a genetic screen in mammalian cells by combining a fluorescent protein-based MYC abundance sensor, CRISPR/Cas9-based gene knockouts and next-generation sequencing. Our screen identifies UBR5, an E3 ligase of the HECT-type family, as a novel regulator of MYC degradation. Even in the presence of the well-described and functional MYC ligase, FBXW7, UBR5 depletion leads to accumulation of MYC in cells. We demonstrate interaction of UBR5 with MYC and reduced K48-linked ubiquitination of MYC upon loss of UBR5 in cells. Interestingly, in cancer cell lines with amplified MYC expression, depletion of UBR5 resulted in reduced cell survival, as a consequence of MYC stabilization. Finally, we show that MYC and UBR5 are co-amplified in more than 40% of cancer cells and that MYC copy number amplification correlates with enhanced transcriptional output of UBR5. This suggests that UBR5 acts as a buffer in MYC amplified settings and protects these cells from apoptosis.
- Published
- 2020
- Full Text
- View/download PDF
30. Benzimidazolyl-pyrazolo[3,4- b ]pyridinones, Selective Inhibitors of MOLT-4 Leukemia Cell Growth and Sea Urchin Embryo Spiculogenesis: Target Quest.
- Author
-
Lichitsky BV, Komogortsev AN, Dudinov AA, Krayushkin MM, Khodot EN, Samet AV, Silyanova EA, Konyushkin LD, Karpov AS, Gorses D, Radimerski T, Semenova MN, Kiselyov AS, and Semenov VV
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Benzimidazoles chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Combinatorial Chemistry Techniques, Drug Screening Assays, Antitumor, Humans, Molecular Structure, Pyrazoles chemistry, Pyridones chemistry, Sea Urchins embryology, Antineoplastic Agents pharmacology, Benzimidazoles pharmacology, Embryo, Nonmammalian drug effects, Pyrazoles pharmacology, Pyridones pharmacology
- Abstract
1,3-Substituted pyrazolo[3,4- b ]pyridinones 11 - 18 were synthesized by a three-component condensation of Meldrum's acid with aryl aldehydes and 1,3-substituted 5-aminopyrazoles. Their biological activity was evaluated using the in vivo phenotypic sea urchin embryo assay and the in vitro cytotoxicity screen against human cancer cell lines. In the sea urchin embryo model, 1-benzimidazolyl-pyrazolo[3,4- b ]pyridinones 11 caused inhibition of hatching and spiculogenesis at sub-micromolar concentrations. These compounds also selectively and potently inhibited growth of the MOLT-4 leukemia cell line. Subsequent structure-activity relationship studies determined the benzimidazolyl fragment as an essential pharmacophore for both effects. We applied numerous techniques for target identification. A preliminary QSAR target identification search did not result in tangible leads. Attempts to prepare a relevant photoaffinity probe that retained potency in both assays were not successful. Compounds 11 were further characterized for their activity in a wild-type versus Notch-mutant leukemia cell lines, and in in vitro panels of kinases and matrix metalloproteinases. Using a series of diverse modulators of spiculogenesis as standards, we excluded multiple signaling networks including Notch, Wnt/β-catenin, receptor tyrosine kinases (VEGF/VEGFR, FGF/FGFR), PI3K, and Raf-MEK-ERK as possible targets of 11 . On the other hand, matrix metalloproteinase-9/hatching enzyme was identified as one potential target.
- Published
- 2019
- Full Text
- View/download PDF
31. JAK2-mutant hematopoietic cells display metabolic alterations that can be targeted to treat myeloproliferative neoplasms.
- Author
-
Rao TN, Hansen N, Hilfiker J, Rai S, Majewska JM, Leković D, Gezer D, Andina N, Galli S, Cassel T, Geier F, Delezie J, Nienhold R, Hao-Shen H, Beisel C, Di Palma S, Dimeloe S, Trebicka J, Wolf D, Gassmann M, Fan TW, Lane AN, Handschin C, Dirnhofer S, Kröger N, Hess C, Radimerski T, Koschmieder S, Čokić VP, and Skoda RC
- Subjects
- Animals, Humans, Mice, Mutation, Hematopoietic Stem Cells metabolism, Janus Kinase 2 genetics, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism
- Abstract
Increased energy requirement and metabolic reprogramming are hallmarks of cancer cells. We show that metabolic alterations in hematopoietic cells are fundamental to the pathogenesis of mutant JAK2-driven myeloproliferative neoplasms (MPNs). We found that expression of mutant JAK2 augmented and subverted metabolic activity of MPN cells, resulting in systemic metabolic changes in vivo, including hypoglycemia, adipose tissue atrophy, and early mortality. Hypoglycemia in MPN mouse models correlated with hyperactive erythropoiesis and was due to a combination of elevated glycolysis and increased oxidative phosphorylation. Modulating nutrient supply through high-fat diet improved survival, whereas high-glucose diet augmented the MPN phenotype. Transcriptomic and metabolomic analyses identified numerous metabolic nodes in JAK2-mutant hematopoietic stem and progenitor cells that were altered in comparison with wild-type controls. We studied the consequences of elevated levels of Pfkfb3, a key regulatory enzyme of glycolysis, and found that pharmacological inhibition of Pfkfb3 with the small molecule 3PO reversed hypoglycemia and reduced hematopoietic manifestations of MPNs. These effects were additive with the JAK1/2 inhibitor ruxolitinib in vivo and in vitro. Inhibition of glycolysis by 3PO altered the redox homeostasis, leading to accumulation of reactive oxygen species and augmented apoptosis rate. Our findings reveal the contribution of metabolic alterations to the pathogenesis of MPNs and suggest that metabolic dependencies of mutant cells represent vulnerabilities that can be targeted for treating MPNs., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
32. A conditional inducible JAK2V617F transgenic mouse model reveals myeloproliferative disease that is reversible upon switching off transgene expression.
- Author
-
Chapeau EA, Mandon E, Gill J, Romanet V, Ebel N, Powajbo V, Andraos-Rey R, Qian Z, Kininis M, Zumstein-Mecker S, Ito M, Hynes NE, Tiedt R, Hofmann F, Eshkind L, Bockamp E, Kinzel B, Mueller M, Murakami M, Baffert F, and Radimerski T
- Subjects
- Amino Acid Substitution, Animals, Disease Models, Animal, Humans, Mice, Mice, Transgenic, Mutation, Missense, Gene Expression Regulation, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, Janus Kinase 2 biosynthesis, Janus Kinase 2 genetics, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology, Transgenes
- Abstract
Aberrant activation of the JAK/STAT pathway is thought to be the critical event in the pathogenesis of the chronic myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia and primary myelofibrosis. The most frequent genetic alteration in these pathologies is the activating JAK2V617F mutation, and expression of the mutant gene in mouse models was shown to cause a phenotype resembling the human diseases. Given the body of genetic evidence, it has come as a sobering finding that JAK inhibitor therapy only modestly suppresses the JAK2V617F allele burden, despite showing clear benefits in terms of reducing splenomegaly and constitutional symptoms in patients. To gain a better understanding if JAK2V617F is required for maintenance of myeloproliferative disease once it has evolved, we generated a conditional inducible transgenic JAK2V617F mouse model using the SCL-tTA-2S tet-off system. Our model corroborates that expression of JAK2V617F in hematopoietic stem and progenitor cells recapitulates key hallmarks of human myeloproliferative neoplasms, and exhibits gender differences in disease manifestation. The disease was found to be transplantable, and importantly, reversible when transgenic JAK2V617F expression was switched off. Our results indicate that mutant JAK2V617F-specific inhibitors should result in profound disease modification by disabling the myeloproliferative clone bearing mutant JAK2., Competing Interests: Some of the authors are full-time employees of Novartis Pharma AG (Emilie A. Chapeau, Emeline Mandon, Vincent Romanet, Nicolas Ebel, Rita Andraos-Rey, Zhiyan Qian, Miltos Kininis, Sabine Zumstein-Mecker, Ralph Tiedt, Francesco Hofmann, Matthias Mueller, Fabienne Baffert), or have been full-time employees of Novartis Pharma AG (Violetta Powajbo, Moriko Ito, Bernd Kinzel, Masato Murakami, Thomas Radimerski). Novartis Pharma AG had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Ruxolitinib is a marketed product of Incyte, for which Novartis Pharma AG received exclusive development and commercialization rights outside of the United States. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2019
- Full Text
- View/download PDF
33. Discovery and Structural Characterization of ATP-Site Ligands for the Wild-Type and V617F Mutant JAK2 Pseudokinase Domain.
- Author
-
McNally R, Li Q, Li K, Dekker C, Vangrevelinghe E, Jones M, Chène P, Machauer R, Radimerski T, and Eck MJ
- Subjects
- Cell Line, Crystallography, X-Ray, Humans, Janus Kinase 2 chemistry, Janus Kinase 2 genetics, Ligands, Phosphorylation, Protein Conformation, Adenosine Triphosphate metabolism, Janus Kinase 2 metabolism, Mutation
- Abstract
The oncogenic V617F mutation lies in the pseudokinase domain of JAK2, marking it as a potential target for development of compounds that might inhibit the pathogenic activity of the mutant protein. We used differential scanning fluorimetry to identify compounds that bind the JAK2 pseudokinase domain. Crystal structures of five candidate compounds with the wild-type domain reveal their modes of binding. Exploration of analogs of screening hit BI-D1870 led to the identification of compound 2, a 123 nM ligand for the pseudokinase domain. Interestingly, crystal structures of the V617F domain in complex with two unrelated compounds reveal a conformation that is characteristic of the wild-type domain, rather than that previously observed for the V617F mutant. These structures suggest that certain ATP-site ligands can modulate the V617F allosteric site, thereby providing a mechanistic rationale for targeting the pseudokinase domain and a structural foundation for development of more potent and pseudokinase-selective compounds.
- Published
- 2019
- Full Text
- View/download PDF
34. Targeting compensatory MEK/ERK activation increases JAK inhibitor efficacy in myeloproliferative neoplasms.
- Author
-
Stivala S, Codilupi T, Brkic S, Baerenwaldt A, Ghosh N, Hao-Shen H, Dirnhofer S, Dettmer MS, Simillion C, Kaufmann BA, Chiu S, Keller M, Kleppe M, Hilpert M, Buser AS, Passweg JR, Radimerski T, Skoda RC, Levine RL, and Meyer SC
- Subjects
- Amino Acid Substitution, Animals, Becaplermin genetics, Becaplermin metabolism, Cell Line, Tumor, Drug Delivery Systems, Hematologic Neoplasms enzymology, Hematologic Neoplasms genetics, Hematologic Neoplasms pathology, Humans, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, MAP Kinase Signaling System genetics, Mice, Myeloproliferative Disorders enzymology, Myeloproliferative Disorders genetics, Myeloproliferative Disorders pathology, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Platelet-Derived Growth Factor genetics, Platelet-Derived Growth Factor metabolism, Receptor, Platelet-Derived Growth Factor alpha genetics, Receptor, Platelet-Derived Growth Factor alpha metabolism, Receptor, Platelet-Derived Growth Factor beta genetics, Receptor, Platelet-Derived Growth Factor beta metabolism, Receptors, Thrombopoietin genetics, Receptors, Thrombopoietin metabolism, Hematologic Neoplasms drug therapy, Janus Kinase 2 antagonists & inhibitors, MAP Kinase Signaling System drug effects, Mutation, Missense, Myeloproliferative Disorders drug therapy, Neoplasm Proteins antagonists & inhibitors, Protein Kinase Inhibitors pharmacology
- Abstract
Constitutive JAK2 signaling is central to myeloproliferative neoplasm (MPN) pathogenesis and results in activation of STAT, PI3K/AKT, and MEK/ERK signaling. However, the therapeutic efficacy of current JAK2 inhibitors is limited. We investigated the role of MEK/ERK signaling in MPN cell survival in the setting of JAK inhibition. Type I and II JAK2 inhibition suppressed MEK/ERK activation in MPN cell lines in vitro, but not in Jak2V617F and MPLW515L mouse models in vivo. JAK2 inhibition ex vivo inhibited MEK/ERK signaling, suggesting that cell-extrinsic factors maintain ERK activation in vivo. We identified PDGFRα as an activated kinase that remains activated upon JAK2 inhibition in vivo, and PDGF-AA/PDGF-BB production persisted in the setting of JAK inhibition. PDGF-BB maintained ERK activation in the presence of ruxolitinib, consistent with its function as a ligand-induced bypass for ERK activation. Combined JAK/MEK inhibition suppressed MEK/ERK activation in Jak2V617F and MPLW515L mice with increased efficacy and reversal of fibrosis to an extent not seen with JAK inhibitors. This demonstrates that compensatory ERK activation limits the efficacy of JAK2 inhibition and dual JAK/MEK inhibition provides an opportunity for improved therapeutic efficacy in MPNs and in other malignancies driven by aberrant JAK-STAT signaling.
- Published
- 2019
- Full Text
- View/download PDF
35. N-aryl-piperidine-4-carboxamides as a novel class of potent inhibitors of MALT1 proteolytic activity.
- Author
-
Schlapbach A, Revesz L, Pissot Soldermann C, Zoller T, Régnier CH, Bornancin F, Radimerski T, Blank J, Schuffenhauer A, Renatus M, Erbel P, Melkko S, Heng R, Simic O, Endres R, Wartmann M, and Quancard J
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Hepatocytes drug effects, Humans, Jurkat Cells, Microsomes drug effects, Molecular Structure, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism, Piperidines chemical synthesis, Piperidines chemistry, Proteolysis drug effects, Rats, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Piperidines pharmacology
- Abstract
Starting from a weak screening hit, potent and selective inhibitors of the MALT1 protease function were elaborated. Advanced compounds displayed high potency in biochemical and cellular assays. Compounds showed activity in a mechanistic Jurkat T cell activation assay as well as in the B-cell lymphoma line OCI-Ly3, which suggests potential use of MALT1 inhibitors in the treatment of autoimmune diseases as well as B-cell lymphomas with a dysregulated NF-κB pathway. Initially, rat pharmacokinetic properties of this compound series were dominated by very high clearance which could be linked to amide cleavage. Using a rat hepatocyte assay a good in vitro-in vivo correlation could be established which led to the identification of compounds with improved PK properties., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
36. Ex vivo drug response profiling detects recurrent sensitivity patterns in drug-resistant acute lymphoblastic leukemia.
- Author
-
Frismantas V, Dobay MP, Rinaldi A, Tchinda J, Dunn SH, Kunz J, Richter-Pechanska P, Marovca B, Pail O, Jenni S, Diaz-Flores E, Chang BH, Brown TJ, Collins RH, Uhrig S, Balasubramanian GP, Bandapalli OR, Higi S, Eugster S, Voegeli P, Delorenzi M, Cario G, Loh ML, Schrappe M, Stanulla M, Kulozik AE, Muckenthaler MU, Saha V, Irving JA, Meisel R, Radimerski T, Von Stackelberg A, Eckert C, Tyner JW, Horvath P, Bornhauser BC, and Bourquin JP
- Subjects
- Antineoplastic Combined Chemotherapy Protocols pharmacology, Cells, Cultured, Coculture Techniques, Heterografts, Humans, Mesenchymal Stem Cells pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Drug sensitivity and resistance testing on diagnostic leukemia samples should provide important functional information to guide actionable target and biomarker discovery. We provide proof of concept data by profiling 60 drugs on 68 acute lymphoblastic leukemia (ALL) samples mostly from resistant disease in cocultures of bone marrow stromal cells. Patient-derived xenografts retained the original pattern of mutations found in the matched patient material. Stromal coculture did not prevent leukemia cell cycle activity, but a specific sensitivity profile to cell cycle-related drugs identified samples with higher cell proliferation both in vitro and in vivo as leukemia xenografts. In patients with refractory relapses, individual patterns of marked drug resistance and exceptional responses to new agents of immediate clinical relevance were detected. The BCL2-inhibitor venetoclax was highly active below 10 nM in B-cell precursor ALL (BCP-ALL) subsets, including MLL -AF4 ALL, and in some T-cell ALLs (T-ALLs), predicting in vivo activity as a single agent and in combination with dexamethasone and vincristine. Unexpected sensitivity to dasatinib with half maximal inhibitory concentration values below 20 nM was detected in 2 independent T-ALL cohorts, which correlated with similar cytotoxic activity of the SRC inhibitor KX2-391 and inhibition of SRC phosphorylation. A patient with refractory T-ALL was treated with dasatinib on the basis of drug profiling information and achieved a 5-month remission. Thus, drug profiling captures disease-relevant features and unexpected sensitivity to relevant drugs, which warrants further exploration of this functional assay in the context of clinical trials to develop drug repurposing strategies for patients with urgent medical needs.TCF3-HLF ALL, and in some T-cell ALLs (T-ALLs), predicting in vivo activity as a single agent and in combination with dexamethasone and vincristine. Unexpected sensitivity to dasatinib with half maximal inhibitory concentration values below 20 nM was detected in 2 independent T-ALL cohorts, which correlated with similar cytotoxic activity of the SRC inhibitor KX2-391 and inhibition of SRC phosphorylation. A patient with refractory T-ALL was treated with dasatinib on the basis of drug profiling information and achieved a 5-month remission. Thus, drug profiling captures disease-relevant features and unexpected sensitivity to relevant drugs, which warrants further exploration of this functional assay in the context of clinical trials to develop drug repurposing strategies for patients with urgent medical needs., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
37. JAK2 exon 12 mutant mice display isolated erythrocytosis and changes in iron metabolism favoring increased erythropoiesis.
- Author
-
Grisouard J, Li S, Kubovcakova L, Rao TN, Meyer SC, Lundberg P, Hao-Shen H, Romanet V, Murakami M, Radimerski T, Dirnhofer S, and Skoda RC
- Subjects
- Animals, Base Sequence, Erythrocytes pathology, Exons, Iron metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Polycythemia metabolism, Polycythemia physiopathology, Erythropoiesis, Janus Kinase 2 genetics, Mutation, Polycythemia genetics
- Abstract
Mutations in JAK2 exon 12 are frequently found in patients with polycythemia vera (PV) that do not carry a JAK2-V617F mutation. The majority of these patients display isolated erythrocytosis. We generated a mouse model that expresses JAK2-N542-E543del, the most frequent JAK2 exon 12 mutation found in PV patients. Mice expressing the human JAK2-N542-E543del (Ex12) showed a strong increase in red blood cell parameters but normal neutrophil and platelet counts, and reduced overall survival. Erythropoiesis was increased in the bone marrow and spleen, with normal megakaryopoiesis and absence of myelofibrosis in histopathology. Erythroid progenitors and precursors were increased in hematopoietic tissues, but the numbers of megakaryocytic precursors were unchanged. Phosphorylation Stat3 and Erk1/2 proteins were increased, and a trend toward increased phospho-Stat5 and phospho-Stat1 was noted. However, Stat1 knock out in Ex12 mice induced no changes in platelet or red cell parameters, indicating that Stat1 does not play a central role in mediating the effects of Ex12 signaling on megakaryopoiesis or erythropoiesis. Ex12 mice showed decreased expression of hepcidin and increased expression of transferrin receptor-1 and erythroferrone, suggesting that the strong erythroid phenotype in Ex12 mutant mice is favored by changes in iron metabolism that optimize iron availability to allow maximal production of red cells., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
38. Activity of the Type II JAK2 Inhibitor CHZ868 in B Cell Acute Lymphoblastic Leukemia.
- Author
-
Wu SC, Li LS, Kopp N, Montero J, Chapuy B, Yoda A, Christie AL, Liu H, Christodoulou A, van Bodegom D, van der Zwet J, Layer JV, Tivey T, Lane AA, Ryan JA, Ng SY, DeAngelo DJ, Stone RM, Steensma D, Wadleigh M, Harris M, Mandon E, Ebel N, Andraos R, Romanet V, Dölemeyer A, Sterker D, Zender M, Rodig SJ, Murakami M, Hofmann F, Kuo F, Eck MJ, Silverman LB, Sallan SE, Letai A, Baffert F, Vangrevelinghe E, Radimerski T, Gaul C, and Weinstock DM
- Subjects
- Aminopyridines pharmacology, Animals, Antineoplastic Agents pharmacology, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Apoptosis, Benzimidazoles pharmacology, Cell Line, Tumor, Cytoprotection drug effects, Drug Synergism, Humans, Janus Kinase 2 chemistry, Janus Kinase 2 genetics, Mice, Mutation, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Protein Kinase Inhibitors pharmacology, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Aminopyridines administration & dosage, Antineoplastic Agents administration & dosage, Benzimidazoles administration & dosage, Dexamethasone administration & dosage, Drug Resistance, Neoplasm drug effects, Janus Kinase 2 antagonists & inhibitors, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Kinase Inhibitors administration & dosage
- Abstract
A variety of cancers depend on JAK2 signaling, including the high-risk subset of B cell acute lymphoblastic leukemias (B-ALLs) with CRLF2 rearrangements. Type I JAK2 inhibitors induce paradoxical JAK2 hyperphosphorylation in these leukemias and have limited activity. To improve the efficacy of JAK2 inhibition in B-ALL, we developed the type II inhibitor CHZ868, which stabilizes JAK2 in an inactive conformation. CHZ868 potently suppressed the growth of CRLF2-rearranged human B-ALL cells, abrogated JAK2 signaling, and improved survival in mice with human or murine B-ALL. CHZ868 and dexamethasone synergistically induced apoptosis in JAK2-dependent B-ALLs and further improved in vivo survival compared to CHZ868 alone. These data support the testing of type II JAK2 inhibition in patients with JAK2-dependent leukemias and other disorders., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
39. CHZ868, a Type II JAK2 Inhibitor, Reverses Type I JAK Inhibitor Persistence and Demonstrates Efficacy in Myeloproliferative Neoplasms.
- Author
-
Meyer SC, Keller MD, Chiu S, Koppikar P, Guryanova OA, Rapaport F, Xu K, Manova K, Pankov D, O'Reilly RJ, Kleppe M, McKenney AS, Shih AH, Shank K, Ahn J, Papalexi E, Spitzer B, Socci N, Viale A, Mandon E, Ebel N, Andraos R, Rubert J, Dammassa E, Romanet V, Dölemeyer A, Zender M, Heinlein M, Rampal R, Weinberg RS, Hoffman R, Sellers WR, Hofmann F, Murakami M, Baffert F, Gaul C, Radimerski T, and Levine RL
- Subjects
- Animals, Antineoplastic Agents pharmacology, Benzamides administration & dosage, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Mutation, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Protein Kinase Inhibitors pharmacology, Pyrimidines administration & dosage, Receptors, Thrombopoietin genetics, Receptors, Thrombopoietin metabolism, Sequence Analysis, RNA, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents administration & dosage, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 genetics, Myeloproliferative Disorders drug therapy, Protein Kinase Inhibitors administration & dosage
- Abstract
Although clinically tested JAK inhibitors reduce splenomegaly and systemic symptoms, molecular responses are not observed in most myeloproliferative neoplasm (MPN) patients. We previously demonstrated that MPN cells become persistent to type I JAK inhibitors that bind the active conformation of JAK2. We investigated whether CHZ868, a type II JAK inhibitor, would demonstrate activity in JAK inhibitor persistent cells, murine MPN models, and MPN patient samples. JAK2 and MPL mutant cell lines were sensitive to CHZ868, including type I JAK inhibitor persistent cells. CHZ868 showed significant activity in murine MPN models and induced reductions in mutant allele burden not observed with type I JAK inhibitors. These data demonstrate that type II JAK inhibition is a viable therapeutic approach for MPN patients., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
40. JAK-STAT pathway activation in malignant and nonmalignant cells contributes to MPN pathogenesis and therapeutic response.
- Author
-
Kleppe M, Kwak M, Koppikar P, Riester M, Keller M, Bastian L, Hricik T, Bhagwat N, McKenney AS, Papalexi E, Abdel-Wahab O, Rampal R, Marubayashi S, Chen JJ, Romanet V, Fridman JS, Bromberg J, Teruya-Feldstein J, Murakami M, Radimerski T, Michor F, Fan R, and Levine RL
- Subjects
- Animals, Antineoplastic Agents pharmacology, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Bone Marrow Cells pathology, Cytokines metabolism, Disease Models, Animal, Gene Deletion, Humans, Inflammation Mediators metabolism, Janus Kinase 1 antagonists & inhibitors, Janus Kinase 2 antagonists & inhibitors, Janus Kinases genetics, Leukocyte Common Antigens genetics, Leukocyte Common Antigens metabolism, Mice, Mice, Knockout, Mutation, Myeloid Cells drug effects, Myeloid Cells metabolism, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Myeloproliferative Disorders pathology, Primary Myelofibrosis genetics, Primary Myelofibrosis metabolism, Primary Myelofibrosis pathology, Protein Kinase Inhibitors pharmacology, STAT Transcription Factors genetics, Cell Transformation, Neoplastic metabolism, Janus Kinases metabolism, Myeloproliferative Disorders metabolism, STAT Transcription Factors metabolism, Signal Transduction drug effects
- Abstract
Unlabelled: The identification of JAK2/MPL mutations in patients with myeloproliferative neoplasms (MPN) has led to the clinical development of JAK kinase inhibitors, including ruxolitinib. Ruxolitinib reduces splenomegaly and systemic symptoms in myelofibrosis and improves overall survival; however, the mechanism by which JAK inhibitors achieve efficacy has not been delineated. Patients with MPN present with increased levels of circulating proinflammatory cytokines, which are mitigated by JAK inhibitor therapy. We sought to elucidate mechanisms by which JAK inhibitors attenuate cytokine-mediated pathophysiology. Single-cell profiling demonstrated that hematopoietic cells from myelofibrosis models and patient samples aberrantly secrete inflammatory cytokines. Pan-hematopoietic Stat3 deletion reduced disease severity and attenuated cytokine secretion, with similar efficacy as observed with ruxolitinib therapy. In contrast, Stat3 deletion restricted to MPN cells did not reduce disease severity or cytokine production. Consistent with these observations, we found that malignant and nonmalignant cells aberrantly secrete cytokines and JAK inhibition reduces cytokine production from both populations., Significance: Our results demonstrate that JAK-STAT3-mediated cytokine production from malignant and nonmalignant cells contributes to MPN pathogenesis and that JAK inhibition in both populations is required for therapeutic efficacy. These findings provide novel insight into the mechanisms by which JAK kinase inhibition achieves therapeutic efficacy in MPNs., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
41. The identification and characterization of a STAT5 gene signature in hematologic malignancies.
- Author
-
Sonkin D, Palmer M, Rong X, Horrigan K, Regnier CH, Fanton C, Holash J, Pinzon-Ortiz M, Squires M, Sirulnik A, Radimerski T, Schlegel R, Morrissey M, and Cao ZA
- Subjects
- Animals, Cell Line, Tumor, Female, Hematologic Neoplasms drug therapy, Heterografts, Humans, Janus Kinases metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Nitriles, Pyrazoles pharmacology, Pyrimidines, Signal Transduction drug effects, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, STAT5 Transcription Factor genetics, STAT5 Transcription Factor metabolism
- Abstract
Background: The JAK-STAT pathway is an important signaling pathway downstream of multiple cytokine and growth factor receptors. Dysregulated JAK-STAT signaling has been implicated in the pathogenesis of multiple human malignancies., Objective: Given this pivotal role of JAK-STAT dysregulation, it is important to identify patients with an overactive JAK-STAT pathway for possible treatment with JAK inhibitors., Methods: We developed a gene signature assay to detect overactive JAK-STAT signaling. The cancer cell line encyclopedia and associated gene-expression data were used to correlate the activation status of STAT5 with the induction of a set of STAT5 target genes., Results: Four target genes were identified (PIM1, CISH, SOCS2, and ID1), the expression of which correlated significantly with pSTAT5 status in 40 hematologic tumor cell lines. In pSTAT5-positive models, the expression of the gene signature genes decreased following ruxolitinib treatment, which corresponded to pSTAT5 downmodulation. In pSTAT5-negative cell lines, neither pSTAT5 modulation nor a change in signature gene expression was observed following ruxolitinib treatment., Conclusions: The gene signature can potentially be used to stratify or enrich for patient populations with activated JAK-STAT5 signaling that might benefit from treatments targeting JAK-STAT signaling. Furthermore, the 4-gene signature is a predictor of the pharmacodynamic effects of ruxolitinib.
- Published
- 2015
- Full Text
- View/download PDF
42. Effects of Jak2 type 1 inhibitors NVP-BSK805 and NVP-BVB808 on Jak2 mutation-positive and Bcr-Abl-positive cell lines.
- Author
-
Ringel F, Kaeda J, Schwarz M, Oberender C, Grille P, Dörken B, Marque F, Manley PW, Radimerski T, and le Coutre P
- Subjects
- Apoptosis drug effects, Benzamides administration & dosage, Cell Line, Tumor, Cell Proliferation drug effects, Fusion Proteins, bcr-abl metabolism, Humans, Imatinib Mesylate, K562 Cells, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive enzymology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders enzymology, Myeloproliferative Disorders genetics, Phosphorylation drug effects, Piperazines administration & dosage, Protein Kinase Inhibitors administration & dosage, Pyrimidines administration & dosage, Quinoxalines administration & dosage, STAT5 Transcription Factor metabolism, Tumor Suppressor Proteins metabolism, Fusion Proteins, bcr-abl genetics, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 genetics, Mutation, Protein Kinase Inhibitors pharmacology, Quinoxalines pharmacology
- Abstract
Janus kinases are critical components of signaling pathways that regulate hematopoiesis. Mutations of the non-receptor tyrosine kinase JAK2 are found in many BCR-ABL-negative myeloproliferative neoplasms. Preclinical results support that JAK2 inhibitors could show efficacy in treating chronic myeloproliferative neoplasms. JAK2 has also been postulated to play a role in BCR-ABL signal transduction. Therefore, inhibitors of JAK2 kinases are turning into therapeutic strategies for treatment of chronic myelogenous leukemia (CML). In this study, the effects of two novel JAK2 inhibitors, NVP-BSK805 and NVP-BVB808, have been investigated in cell lines expressing either BCR-ABL or mutant JAK2. Possible synergies between NVP-BSK805/NVP-BVB808 and the kinase inhibitors imatinib and nilotinib were assessed. Proliferation and apoptosis tests with both substances showed response in the following cell lines: CHRF-288-11, SET-2 and UKE-1. All BCR-ABL-positive cell lines showed some reduction in proliferation, but with half-maximal growth-inhibitory values >1 µM. Combination of the JAK2 inhibitors with imatinib and nilotinib showed no significant additive or synergistic effects, although all BCR-ABL-positive cell lines responded well to both CML therapeutic agents. Interestingly, it seemed that the combination of imatinib with NVP-BSK805 had a protective effect on the cells. Combination treatment with nilotinib did not show this effect., (© 2014 S. Karger AG, Basel.)
- Published
- 2014
- Full Text
- View/download PDF
43. JAK1/2 and Pan-deacetylase inhibitor combination therapy yields improved efficacy in preclinical mouse models of JAK2V617F-driven disease.
- Author
-
Evrot E, Ebel N, Romanet V, Roelli C, Andraos R, Qian Z, Dölemeyer A, Dammassa E, Sterker D, Cozens R, Hofmann F, Murakami M, Baffert F, and Radimerski T
- Subjects
- Acetylation, Animals, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Disease Models, Animal, Histone Deacetylase Inhibitors adverse effects, Histone Deacetylase Inhibitors therapeutic use, Histone Deacetylases drug effects, Histones metabolism, Hydroxamic Acids adverse effects, Indoles adverse effects, Janus Kinase 1 metabolism, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, Mice, Nitriles, Panobinostat, Polycythemia Vera drug therapy, Pyrazoles adverse effects, Pyrimidines, Reticulin analysis, STAT5 Transcription Factor drug effects, STAT5 Transcription Factor metabolism, Spleen cytology, Spleen metabolism, Splenomegaly drug therapy, Thrombocytosis drug therapy, Hydroxamic Acids therapeutic use, Indoles therapeutic use, Janus Kinase 1 antagonists & inhibitors, Janus Kinase 2 antagonists & inhibitors, Primary Myelofibrosis drug therapy, Pyrazoles therapeutic use
- Abstract
Purpose: The myeloproliferative neoplasm myelofibrosis is characterized by frequent deregulation of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling, and JAK inhibitors were shown to reduce splenomegaly and ameliorate disease-related symptoms. However, the mutant clone and bone marrow fibrosis persist in the majority of patients. Using preclinical models, we explored whether JAK and pan-deacetylase inhibitor combination yielded additional benefits., Experimental Design: The combination of the JAK1/2 inhibitor ruxolitinib and panobinostat was investigated using two different mouse models of JAK2(V617F)-driven disease. A Ba/F3 JAK2(V617F) cell-driven leukemic disease model was used to identify tolerated and efficacious doses. The drugs were then evaluated alone and in combination in a mouse model of myeloproliferative neoplasm-like disease based on transplantation of bone marrow transduced with a retrovirus expressing JAK2(V617F). Exposures were determined in blood and tissues, and phosphorylated STAT5 and acetylated histone H3 pharmacodynamic readouts were assessed in spleen and bone marrow. Histologic analysis was conducted on spleen and bone marrow, including staining of reticulin fibers in the latter organ., Results: The combination of ruxolitinib and panobinostat was found to have a more profound effect on splenomegaly, as well as on bone marrow and spleen histology, compared with either agent alone, and the analysis of pharmacodynamic readouts showed that ruxolitinib and panobinostat have nonoverlapping and complementary effects., Conclusion: Combining JAK1/2 and pan-deacetylase inhibitors was fairly well tolerated and resulted in improved efficacy in mouse models of JAK2(V617F)-driven disease compared with the single agents. Thus, the combination of ruxolitinib and panobinostat may represent a promising novel therapeutic modality for myeloproliferative neoplasms., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
44. Targeting PI3K, HER2 and the IL-8/JAK2 axis in metastatic breast cancer: Which combination makes the whole greater than the sum of its parts?
- Author
-
Britschgi A, Radimerski T, and Bentires-Alj M
- Subjects
- Animals, Breast drug effects, Breast immunology, Breast metabolism, Breast Neoplasms immunology, Breast Neoplasms metabolism, Drug Synergism, Female, Humans, Immunomodulation drug effects, Interleukin-8 antagonists & inhibitors, Interleukin-8 metabolism, Janus Kinase 2 metabolism, Molecular Targeted Therapy, Neoplasm Metastasis, Phosphatidylinositol 3-Kinase metabolism, Receptor, ErbB-2 metabolism, Signal Transduction drug effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm, Janus Kinase 2 antagonists & inhibitors, Models, Biological, Phosphoinositide-3 Kinase Inhibitors, Receptor, ErbB-2 antagonists & inhibitors
- Abstract
The widespread hyperactivation of the PI3K/mTOR pathway in human cancer has made it a prime target for the treatment of this disease. However, a variety of resistance mechanisms involving (re)activation of the targeted pathway or of parallel survival signaling cascades have limited the clinical success of inhibitors targeting PI3K and/or mTOR. Recent studies delineated new crosstalks between PI3K, HER2, JAK2 and IL-8 signaling, which can explain the limited efficacy of PI3K blockade when inhibitors of this pathway are used as single agents. In this review, we summarize molecular mechanisms of resistance to inhibitors of the PI3K/mTOR pathway, provide an outline of new connections between crucial oncogenic signaling pathways, and discuss the potential of new combination therapy approaches to overcome resistance., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
45. Differential effects of hydroxyurea and INC424 on mutant allele burden and myeloproliferative phenotype in a JAK2-V617F polycythemia vera mouse model.
- Author
-
Kubovcakova L, Lundberg P, Grisouard J, Hao-Shen H, Romanet V, Andraos R, Murakami M, Dirnhofer S, Wagner KU, Radimerski T, and Skoda RC
- Subjects
- Alleles, Amino Acid Substitution, Animals, Bone Marrow Transplantation, Disease Models, Animal, Female, Hematopoiesis drug effects, Hematopoiesis genetics, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 deficiency, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Mutation, Nitriles, Phenotype, Polycythemia Vera metabolism, Polycythemia Vera pathology, Pyrimidines, STAT5 Transcription Factor metabolism, Signal Transduction drug effects, Hydroxyurea pharmacology, Janus Kinase 2 genetics, Polycythemia Vera drug therapy, Polycythemia Vera genetics, Pyrazoles pharmacology
- Abstract
To establish a preclinical animal model for testing drugs with potential effects on myeloproliferative neoplasms (MPNs), we first performed a detailed phenotypic characterization of Cre-inducible transgenic JAK2-V617F mice. Deleting the conditional mouse Jak2-knockout alleles increased erythropoiesis and accentuated the polycythemia vera phenotype, but did not alter platelet or granulocyte levels. In a transplantation assay, JAK2-V617F(+) BM cells had an advantage over wild-type competitor cells. Using this competitive repopulation assay, we compared the effects of INC424 (ruxolitinib), a dual Jak1/Jak2 inhibitor, and hydroxyurea (HU). HU led to weight loss, but did not reduce spleen weight. The hematologic parameters were lowered and a slight decrease of the mutant allele burden was noted. INC424 had little effect on body weight, but strongly decreased spleen size and rapidly normalized RBC and neutrophil parameters. No significant decrease in the mutant allele burden was observed. INC424 reduced the phospho-Stat5 levels, whereas HU strongly increased phospho-Stat5, most likely because of the elevated erythropoietin levels in response to the HU-induced anemia. This compensatory increase in JAK/STAT signaling may counteract the beneficial effects of cytoreduction at higher doses of HU and represents an adverse effect that should be avoided.
- Published
- 2013
- Full Text
- View/download PDF
46. Glucose-dependent insulinotropic polypeptide induces cytokine expression, lipolysis, and insulin resistance in human adipocytes.
- Author
-
Timper K, Grisouard J, Sauter NS, Herzog-Radimerski T, Dembinski K, Peterli R, Frey DM, Zulewski H, Keller U, Müller B, and Christ-Crain M
- Subjects
- Adipocytes metabolism, Adult, Anti-Obesity Agents pharmacology, Cyclic AMP-Dependent Protein Kinases metabolism, Cytokines metabolism, Gastric Inhibitory Polypeptide physiology, Humans, Interleukin 1 Receptor Antagonist Protein genetics, Interleukin 1 Receptor Antagonist Protein metabolism, Interleukin-1beta genetics, Interleukin-1beta metabolism, Interleukin-6 genetics, Interleukin-6 metabolism, Lactones pharmacology, Lipolysis genetics, Middle Aged, NF-kappa B metabolism, Orlistat, Signal Transduction drug effects, Signal Transduction genetics, Up-Regulation drug effects, Up-Regulation genetics, Adipocytes drug effects, Cytokines genetics, Gastric Inhibitory Polypeptide pharmacology, Insulin Resistance genetics, Insulin Resistance physiology, Lipolysis drug effects
- Abstract
Obesity-related insulin resistance is linked to a chronic state of systemic and adipose tissue-derived inflammation. Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone also acting on adipocytes. We investigated whether GIP affects inflammation, lipolysis, and insulin resistance in human adipocytes. Human subcutaneous preadipocyte-derived adipocytes, differentiated in vitro, were treated with human GIP to analyze mRNA expression and protein secretion of cytokines, glycerol, and free fatty acid release and insulin-induced glucose uptake. GIP induced mRNA expression of IL-6, IL-1β, and the IL-1 receptor antagonist IL-1Ra, whereas TNFα, IL-8, and monocyte chemotactic protein (MCP)-1 remained unchanged. Cytokine induction involved PKA and the NF-κB pathway as well as an autocrine IL-1 effect. Furthermore, GIP potentiated IL-6 and IL-1Ra secretion in the presence of LPS, IL-1β, and TNFα. GIP induced lipolysis via activation of hormone-sensitive lipase and was linked to NF-κB activation. Finally, chronic GIP treatment impaired insulin-induced glucose uptake possibly due to the observed impaired translocation of glucose transporter GLUT4. In conclusion, GIP induces an inflammatory and prolipolytic response via the PKA -NF-κB-IL-1 pathway and impairs insulin sensitivity of glucose uptake in human adipocytes.
- Published
- 2013
- Full Text
- View/download PDF
47. JAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: a rationale for cotargeting these pathways in metastatic breast cancer.
- Author
-
Britschgi A, Andraos R, Brinkhaus H, Klebba I, Romanet V, Müller U, Murakami M, Radimerski T, and Bentires-Alj M
- Subjects
- Animals, Breast Neoplasms drug therapy, Cell Death drug effects, Cell Line, Tumor, Female, Humans, Insulin Receptor Substrate Proteins metabolism, Interleukin-8 metabolism, Janus Kinase 2 metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, SCID, Phosphatidylinositol 3-Kinases metabolism, Receptors, Interleukin-8A metabolism, STAT5 Transcription Factor metabolism, TOR Serine-Threonine Kinases metabolism, Breast Neoplasms metabolism, Janus Kinase 2 antagonists & inhibitors, Neoplasm Metastasis drug therapy, Phosphoinositide-3 Kinase Inhibitors, STAT5 Transcription Factor antagonists & inhibitors, Signal Transduction drug effects, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Hyperactive PI3K/mTOR signaling is prevalent in human malignancies and its inhibition has potent antitumor consequences. Unfortunately, single-agent targeted cancer therapy is usually short-lived. We have discovered a JAK2/STAT5-evoked positive feedback loop that dampens the efficacy of PI3K/mTOR inhibition. Mechanistically, PI3K/mTOR inhibition increased IRS1-dependent activation of JAK2/STAT5 and secretion of IL-8 in several cell lines and primary breast tumors. Genetic or pharmacological inhibition of JAK2 abrogated this feedback loop and combined PI3K/mTOR and JAK2 inhibition synergistically reduced cancer cell number and tumor growth, decreased tumor seeding and metastasis, and also increased overall survival of the animals. Our results provide a rationale for combined targeting of the PI3K/mTOR and JAK2/STAT5 pathways in triple-negative breast cancer, a particularly aggressive and currently incurable disease., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
48. Modulation of activation-loop phosphorylation by JAK inhibitors is binding mode dependent.
- Author
-
Andraos R, Qian Z, Bonenfant D, Rubert J, Vangrevelinghe E, Scheufler C, Marque F, Régnier CH, De Pover A, Ryckelynck H, Bhagwat N, Koppikar P, Goel A, Wyder L, Tavares G, Baffert F, Pissot-Soldermann C, Manley PW, Gaul C, Voshol H, Levine RL, Sellers WR, Hofmann F, and Radimerski T
- Subjects
- Animals, Binding Sites, Cell Line, Tumor, Humans, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 chemistry, Mice, Phosphorylation drug effects, Protein Binding, Protein Structure, Tertiary, STAT5 Transcription Factor metabolism, Janus Kinases antagonists & inhibitors, Janus Kinases chemistry, Protein Kinase Inhibitors pharmacology
- Abstract
Janus kinase (JAK) inhibitors are being developed for the treatment of rheumatoid arthritis, psoriasis, myeloproliferative neoplasms, and leukemias. Most of these drugs target the ATP-binding pocket and stabilize the active conformation of the JAK kinases. This type I binding mode can lead to an increase in JAK activation loop phosphorylation, despite blockade of kinase function. Here we report that stabilizing the inactive state via type II inhibition acts in the opposite manner, leading to a loss of activation loop phosphorylation. We used X-ray crystallography to corroborate the binding mode and report for the first time the crystal structure of the JAK2 kinase domain in an inactive conformation. Importantly, JAK inhibitor-induced activation loop phosphorylation requires receptor interaction, as well as intact kinase and pseudokinase domains. Hence, depending on the respective conformation stabilized by a JAK inhibitor, hyperphosphorylation of the activation loop may or may not be elicited.
- Published
- 2012
- Full Text
- View/download PDF
49. Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition.
- Author
-
Weigert O, Lane AA, Bird L, Kopp N, Chapuy B, van Bodegom D, Toms AV, Marubayashi S, Christie AL, McKeown M, Paranal RM, Bradner JE, Yoda A, Gaul C, Vangrevelinghe E, Romanet V, Murakami M, Tiedt R, Ebel N, Evrot E, De Pover A, Régnier CH, Erdmann D, Hofmann F, Eck MJ, Sallan SE, Levine RL, Kung AL, Baffert F, Radimerski T, and Weinstock DM
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, DNA Primers genetics, Female, Flow Cytometry, Gene Expression Profiling, HSP90 Heat-Shock Proteins metabolism, Humans, Immunoblotting, Immunohistochemistry, Isoxazoles therapeutic use, Janus Kinase 2 metabolism, Leukemia, B-Cell drug therapy, Leukemia, B-Cell genetics, Luciferases, Mice, Mice, Inbred BALB C, Mutagenesis, Mutation, Missense genetics, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Phosphorylation, RNA, Small Interfering genetics, Receptors, Cytokine genetics, Receptors, Cytokine metabolism, Resorcinols therapeutic use, X-Ray Microtomography, HSP90 Heat-Shock Proteins antagonists & inhibitors, Isoxazoles pharmacology, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 genetics, Leukemia, B-Cell enzymology, Myeloproliferative Disorders enzymology, Resorcinols pharmacology, Signal Transduction physiology
- Abstract
Enzymatic inhibitors of Janus kinase 2 (JAK2) are in clinical development for the treatment of myeloproliferative neoplasms (MPNs), B cell acute lymphoblastic leukemia (B-ALL) with rearrangements of the cytokine receptor subunit cytokine receptor-like factor 2 (CRLF2), and other tumors with constitutive JAK2 signaling. In this study, we identify G935R, Y931C, and E864K mutations within the JAK2 kinase domain that confer resistance across a panel of JAK inhibitors, whether present in cis with JAK2 V617F (observed in MPNs) or JAK2 R683G (observed in B-ALL). G935R, Y931C, and E864K do not reduce the sensitivity of JAK2-dependent cells to inhibitors of heat shock protein 90 (HSP90), which promote the degradation of both wild-type and mutant JAK2. HSP90 inhibitors were 100-1,000-fold more potent against CRLF2-rearranged B-ALL cells, which correlated with JAK2 degradation and more extensive blockade of JAK2/STAT5, MAP kinase, and AKT signaling. In addition, the HSP90 inhibitor AUY922 prolonged survival of mice xenografted with primary human CRLF2-rearranged B-ALL further than an enzymatic JAK2 inhibitor. Thus, HSP90 is a promising therapeutic target in JAK2-driven cancers, including those with genetic resistance to JAK enzymatic inhibitors.
- Published
- 2012
- Full Text
- View/download PDF
50. Both inflammatory and classical lipolytic pathways are involved in lipopolysaccharide-induced lipolysis in human adipocytes.
- Author
-
Grisouard J, Bouillet E, Timper K, Radimerski T, Dembinski K, Frey DM, Peterli R, Zulewski H, Keller U, Müller B, and Christ-Crain M
- Subjects
- Adipocytes drug effects, Adipocytes pathology, Cell Differentiation, Cells, Cultured, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Enzyme Inhibitors pharmacology, Gene Expression Regulation drug effects, Gene Expression Regulation immunology, Glycerol metabolism, Humans, I-kappa B Kinase antagonists & inhibitors, Interleukin-6 genetics, Interleukin-6 immunology, Interleukin-8 genetics, Interleukin-8 immunology, Isoproterenol pharmacology, Lipolysis drug effects, Adipocytes immunology, Inflammation Mediators metabolism, Interleukin-6 metabolism, Interleukin-8 metabolism, Lipolysis immunology, Lipopolysaccharides immunology
- Abstract
High fat diet-induced endotoxaemia triggers low-grade inflammation and lipid release from adipose tissue. This study aims to unravel the cellular mechanisms leading to the lipopolysaccharide (LPS) effects in human adipocytes. Subcutaneous pre-adipocytes surgically isolated from patients were differentiated into mature adipocytes in vitro. Lipolysis was assessed by measurement of glycerol release and mRNA expression of pro-inflammatory cytokines were evaluated by real-time PCR. Treatment with LPS for 24 h induced a dose-dependent increase in interleukin (IL)-6 and IL-8 mRNA expression. At 1 µg/ml LPS, IL-6 and IL-8 were induced to 19.5 ± 1.8-fold and 662.7 ± 91.5-fold (P < 0.01 vs basal), respectively. From 100 ng/ml to 1 µg/ml, LPS-induced lipolysis increased to a plateau of 3.1-fold above basal level (P < 0.001 vs basal). Co-treatment with inhibitors of inhibitory kappa B kinase kinase beta (IKKβ) or NF-κB inhibited LPS-induced glycerol release. Co-treatment with the protein kinase A (PKA) inhibitor H-89, the lipase inhibitor orlistat or the hormone-sensitive lipase (HSL) inhibitor CAY10499 abolished the lipolytic effects of LPS. Co-treatment with the MAPK inhibitor, U0126 also reduced LPS-induced glycerol release. Inhibition of lipolysis by orlistat or CAY10499 reduced LPS-induced IL-6 and IL-8 mRNA expression. Induction of lipolysis by the synthetic catecholamine isoproterenol or the phosphodiesterase type III inhibitor milrinone did not alter basal IL-6 and IL-8 mRNA expression after 24 treatments whereas these compounds enhanced LPS-induced IL-6 and IL-8 mRNA expression. Both the inflammatory IKKβ/NF-κB pathway and the lipolytic PKA/HSL pathways mediate LPS-induced lipolysis. In turn, LPS-induced lipolysis reinforces the expression of pro-inflammatory cytokines and, thereby, triggers its own lipolytic activity.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.