111 results on '"Christian Rommel"'
Search Results
2. Data from PI3Kδ Inhibitors in Cancer: Rationale and Serendipity Merge in the Clinic
- Author
-
Christian Rommel and David A. Fruman
- Abstract
Several phosphoinositide 3-kinase (PI3K) inhibitors are in the clinic and many more are in preclinical development. CAL-101, a selective inhibitor of the PI3Kδ isoform, has shown remarkable success in certain hematologic malignancies. Although PI3Kδ signaling plays a central role in lymphocyte biology, the degree of single-agent therapeutic activity of CAL-101 during early-phase development has been somewhat unexpected. CAL-101 works in part by blocking signals from the microenvironment that normally sustain leukemia and lymphoma cells in a protective niche. As PI3Ks enter the arena of molecular-targeted therapies, CAL-101 provides proof of principle that isoform-selective compounds can be effective in selected cancer types and patient populations.Significance: A key question is whether compounds targeting a single PI3K catalytic isoform can provide meaningful single agent efficacy in cancer cells that express multiple isoforms. Clinical studies of the drug CAL-101 have provided a significant advance by showing that selective targeting of PI3Kδ achieves efficacy in chronic lymphocytic leukemia, in part through targeting the tumor microenvironment. Cancer Discovery; 1(7); 562–72. ©2011 AACR.
- Published
- 2023
3. Supplementary Figure 1 from PI3Kδ Inhibitors in Cancer: Rationale and Serendipity Merge in the Clinic
- Author
-
Christian Rommel and David A. Fruman
- Abstract
PDF file - 122K
- Published
- 2023
4. Supplementary Figure 4 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 523KB, Cell cycle analysis of trastuzumab/lapatinib resistant cells.
- Published
- 2023
5. Supplementary Figure Legend from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 85KB.
- Published
- 2023
6. Supplementary Figure 6 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 233KB, In vitro cell death induced by lapatinib and INK-128 of trastuzumab/lapatinib resistant cell lines.
- Published
- 2023
7. Supplementary Figure 8 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 281KB, Trastuzumab treatment in trastuzumab-resistant patient derived-xenografts.
- Published
- 2023
8. Supplementary Figure 5 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 593KB, In vitro cell death induced by lapatinib and INK-128 of trastuzumab/lapatinib resistant cell lines.
- Published
- 2023
9. Data from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
Purpose: The PI3K/Akt/mTOR pathway is an attractive target in HER2-positive breast cancer that is refractory to anti-HER2 therapy. The hypothesis is that the suppression of this pathway results in sensitization to anti-HER2 agents. However, this combinatorial strategy has not been comprehensively tested in models of trastuzumab and lapatinib resistance.Experimental Design: We analyzed in vitro cell viability and induction of apoptosis in five different cell lines resistant to trastuzumab and lapatinib. Inhibition of HER2/HER3 phosphorylation, PI3K/Akt/mTOR, and extracellular signal-regulated kinase (ERK) signaling pathways was evaluated by Western blotting. Tumor growth inhibition after treatment with lapatinib, INK-128, or the combination of both agents was evaluated in three different animal models: two cell-based xenograft models refractory to both trastuzumab and lapatinib and a xenograft derived from a patient who relapsed on trastuzumab-based therapy.Results: The addition of lapatinib to INK-128 prevented both HER2 and HER3 phosphorylation induced by INK-128, resulting in inhibition of both PI3K/Akt/mTOR and ERK pathways. This dual blockade produced synergistic induction of cell death in five different HER2-positive cell lines resistant to trastuzumab and lapatinib. In vivo, both cell line–based and patient-derived xenografts showed exquisite sensitivity to the antitumor activity of the combination of lapatinib and INK-128, which resulted in durable tumor shrinkage and exhibited no signs of toxicity in these models.Conclusions: The simultaneous blockade of both PI3K/Akt/mTOR and ERK pathways obtained by combining lapatinib with INK-128 acts synergistically in inducing cell death and tumor regression in breast cancer models refractory to anti-HER2 therapy. Clin Cancer Res; 18(9); 2603–12. ©2012 AACR.
- Published
- 2023
10. Supplementary Figure 3 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 714KB, Combination index (CI) of the combination of lapatinib and INK-128 in resistant cell lines.
- Published
- 2023
11. Supplementary Figure 1 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 760KB, Biochemical analyses of trastuzumab/lapatinib sensitive cells treated with lapatinib.
- Published
- 2023
12. Supplementary Figure 2 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 792KB, Biochemical analyses of trastuzumab/lapatinib resistant cells treated with lapatinib.
- Published
- 2023
13. Supplementary Figure 7 from Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Maurizio Scaltriti, José Baselga, Josep Tabernero, Christian Rommel, Yi Liu, Katti Jessen, José Pérez, Claudia Aura, Judit Grueso, Marta Guzmán, Maria Teresa Calvo, Violeta Serra, Yasir H. Ibrahim, and Celina García-García
- Abstract
PDF file, 386KB, In vivo treatments of JIMT1 xenografts.
- Published
- 2023
14. Kennzahlenbasiertes F & E-Controlling im Kontext der Elektromobilität
- Author
-
Andreas Mayr, Christian Rommel, Veronika Schaller, and Jörg Franke
- Subjects
Strategy and Management ,General Engineering ,Management Science and Operations Research - Abstract
Die deutsche Automobilindustrie steht angesichts der Elektromobilität vor einem radikalen Wandel. Um ihre Technologieführerschaft beizubehalten, stehen die F & E-Abteilungen der etablierten Akteure unter hohem Druck. Im Rahmen dieses Beitrags wird gezeigt, wie sich ein passgenaues Kennzahlengerüst für das Controlling des operativen F & E-Prozesses erarbeiten lässt. Die Validierung der vorgestellten Methode erfolgt anhand eines Maschinen- und Anlagenbauers im Bereich der Elektromobilität, der GROB-WERKE GmbH & Co. KG in Mindelheim.
- Published
- 2022
15. Establishing standardized immune phenotyping of metastatic melanoma by digital pathology
- Author
-
Bettina Sobottka, Marta Nowak, Anja Laura Frei, Martina Haberecker, Samuel Merki, Mitchell P. Levesque, Reinhard Dummer, Holger Moch, Viktor Hendrik Koelzer, Rudolf Aebersold, Melike Ak, Faisal S. Al-Quaddoomi, Jonas Albinus, Ilaria Alborelli, Sonali Andani, Per-Olof Attinger, Marina Bacac, Daniel Baumhoer, Beatrice Beck-Schimmer, Niko Beerenwinkel, Christian Beisel, Lara Bernasconi, Anne Bertolini, Bernd Bodenmiller, Ximena Bonilla, Ruben Casanova, Stéphane Chevrier, Natalia Chicherova, Maya D'Costa, Esther Danenberg, Natalie Davidson, Monica-Andreea Drăganmoch, Stefanie Engler, Martin Erkens, Katja Eschbach, Cinzia Esposito, André Fedier, Pedro Ferreira, Joanna Ficek, Bruno Frey, Sandra Goetze, Linda Grob, Gabriele Gut, Detlef Günther, Pirmin Haeuptle, Viola Heinzelmann-Schwarz, Sylvia Herter, Rene Holtackers, Tamara Huesser, Anja Irmisch, Francis Jacob, Andrea Jacobs, Tim M. Jaeger, Katharina Jahn, Alva R. James, Philip M. Jermann, André Kahles, Abdullah Kahraman, Werner Kuebler, Jack Kuipers, Christian P. Kunze, Christian Kurzeder, Kjong-Van Lehmann, Sebastian Lugert, Gerd Maass, Markus G. Manz, Philipp Markolin, Julien Mena, Ulrike Menzel, Julian M. Metzler, Nicola Miglino, Emanuela S. Milani, Simone Muenst, Riccardo Murri, Charlotte K.Y. Ng, Stefan Nicolet, Patrick G.A. Pedrioli, Lucas Pelkmans, Salvatore Piscuoglio, Michael Prummer, Mathilde Ritter, Christian Rommel, María L. Rosano-González, Gunnar Rätsch, Natascha Santacroce, Jacobo Sarabia del Castillo, Ramona Schlenker, Petra C. Schwalie, Severin Schwan, Tobias Schär, Gabriela Senti, Franziska Singer, Sujana Sivapatham, Berend Snijder, Vipin T. Sreedharan, Stefan Stark, Daniel J. Stekhoven, Alexandre P.A. Theocharides, Tinu M. Thomas, Markus Tolnay, Vinko Tosevski, Nora C. Toussaint, Mustafa A. Tuncel, Marina Tusup, Audrey Van Drogen, Marcus Vetter, Tatjana Vlajnic, Sandra Weber, Walter P. Weber, Rebekka Wegmann, Michael Weller, Fabian Wendt, Norbert Wey, Andreas Wicki, Mattheus HE Wildschut, Bernd Wollscheid, Shuqing Yu, Johanna Ziegler, Marc Zimmermann, Martin Zoche, Gregor Zuend, University of Zurich, Sobottka-Brillout, Bettina, and Koelzer, Viktor Hendrik
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,610 Medicine & health ,Disease ,Predictive markers ,Article ,Pathology and Forensic Medicine ,1307 Cell Biology ,Immune system ,10049 Institute of Pathology and Molecular Pathology ,1312 Molecular Biology ,Medicine ,Compartment (pharmacokinetics) ,Molecular Biology ,Melanoma ,business.industry ,10177 Dermatology Clinic ,Digital pathology ,10060 Epidemiology, Biostatistics and Prevention Institute (EBPI) ,Cell Biology ,Biomarker (cell) ,2734 Pathology and Forensic Medicine ,10032 Clinic for Oncology and Hematology ,Cohort ,Imaging the immune system ,business ,CD8 - Abstract
CD8+ tumor-infiltrating T cells can be regarded as one of the most relevant predictive biomarkers in immune-oncology. Highly infiltrated tumors, referred to as inflamed (clinically “hot”), show the most favorable response to immune checkpoint inhibitors in contrast to tumors with a scarce immune infiltrate called immune desert or excluded (clinically “cold”). Nevertheless, quantitative and reproducible methods examining their prevalence within tumors are lacking. We therefore established a computational diagnostic algorithm to quantitatively measure spatial densities of tumor-infiltrating CD8+ T cells by digital pathology within the three known tumor compartments as recommended by the International Immuno-Oncology Biomarker Working Group in 116 prospective metastatic melanomas of the Swiss Tumor Profiler cohort. Workflow robustness was confirmed in 33 samples of an independent retrospective validation cohort. The introduction of the intratumoral tumor center compartment proved to be most relevant for establishing an immune diagnosis in metastatic disease, independent of metastatic site. Cut-off values for reproducible classification were defined and successfully assigned densities into the respective immune diagnostic category in the validation cohort with high sensitivity, specificity, and precision. We provide a robust diagnostic algorithm based on intratumoral and stromal CD8+ T-cell densities in the tumor center compartment that translates spatial densities of tumor-infiltrating CD8+ T cells into the clinically relevant immune diagnostic categories “inflamed”, “excluded”, and “desert”. The consideration of the intratumoral tumor center compartment allows immune phenotyping in the clinically highly relevant setting of metastatic lesions, even if the invasive margin compartment is not captured in biopsy material., The authors present a robust diagnostic algorithm based on digital pathology and image analysis that quantifies intratumoral and stromal CD8+ T-cell densities in the tumor center and invasive margin compartment in metastatic melanoma. Spatial CD8+ T-cell densities are translated into the clinically relevant immune diagnostic categories “inflamed”, “excluded”, and “desert”. Their approach also allows efficient immune phenotyping of metastatic lesions, on biopsy material or even in the absence of material from the invasive margin.
- Published
- 2021
- Full Text
- View/download PDF
16. The Tumor Profiler Study: integrated, multi-omic, functional tumor profiling for clinical decision support
- Author
-
René Holtackers, Simone Muenst, Martin Zoche, Rudolf Aebersold, Stéphane Chevrier, Tobias Schär, Bettina Sobottka, Marc Zimmermann, Abdullah Kahraman, Lucas Pelkmans, Faisal Alquaddoomi, Philip Jermann, Natascha Santacroce, Andreas Wicki, Norbert Wey, Nora C. Toussaint, Monica-Andreea Drăgan, Mattheus H.E. Wildschut, Ruben Casanova, Shuqing Yu, Markus Tolnay, Marcus Vetter, Mustafa Anil Tuncel, Ximena Bonilla, Stefan Nicolet, Gabriele Gut, Stefan G. Stark, Philipp Markolin, Bruno S. Frey, Ramona Schlenker, Rebekka Wegmann, Walter P. Weber, Lara Bernasconi, Emanuela S. Milani, Viktor H. Koelzer, Christian Rommel, Christian P. Kunze, Sylvia Herter, Cinzia Esposito, Gabriela Senti, Michael Prummer, Katja Eschbach, Bernd Wollscheid, Riccardo Murri, Salvatore Piscuoglio, Mathilde Ritter, Mitchell P. Levesque, Christian Beisel, Tim M. Jaeger, Viola Heinzelmann-Schwarz, Gunnar Rätsch, Severin Schwan, Marina Bacac, Reinhard Dummer, Joanna Ficek, Sandra Goetze, Tatjana Vlajnic, Martin Erkens, Ilaria Alborelli, Ulrike Menzel, Vinko Tosevski, Markus G. Manz, Werner Kuebler, Detlef Günther, Julian M. Metzler, Daniel J. Stekhoven, Christian Kurzeder, Anja Frei, Tamara Huesser, Marta Nowak, Melike Ak, Francis Jacob, Gregor Zuend, Berend Snijder, Martina Haberecker, Pirmin Haeuptle, Anja Irmisch, Maya D'Costa, Linda Grob, Natalia Chicherova, Bernd Bodenmiller, Johanna Ziegler, Per-Olof Attinger, Jack Kuipers, Katharina Jahn, Nicola Miglino, Natalie R. Davidson, Jacobo Sarabia del Castillo, André Fedier, Fabian Wendt, Esther Danenberg, Pedro Ferreira, María Lourdes Rosano-Gonzalez, Sebastian Lugert, Andrea Jacobs, Charlotte K.Y. Ng, Alva Rani James, Tinu M. Thomas, André Kahles, Gerd Maass, Julien Mena, Jonas B. Albinus, Daniel Baumhoer, Sonali Andani, Petra C. Schwalie, Anne Bertolini, Marina Tusup, Franziska Singer, Alexandre Theocharides, Sandra Weber, Beatrice Beck-Schimmer, Sujana Sivapatham, Kjong-Van Lehmann, Stefanie Engler, Holger Moch, Niko Beerenwinkel, Vipin T. Sreedharan, Michael Weller, Audrey Van Drogen, Patrick G. A. Pedrioli, University of Zurich, Rätsch, Gunnar, and Levesque, Mitchell Paul
- Subjects
0301 basic medicine ,Decision support system ,Cancer Research ,Computer science ,Observational Trial ,Clinical Decision-Making ,610 Medicine & health ,Computational biology ,Clinical decision support system ,1307 Cell Biology ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,10049 Institute of Pathology and Molecular Pathology ,Humans ,Profiling (information science) ,1306 Cancer Research ,Prospective Studies ,Precision Medicine ,business.industry ,Computational Biology ,10177 Dermatology Clinic ,10060 Epidemiology, Biostatistics and Prevention Institute (EBPI) ,Cell Biology ,Decision Support Systems, Clinical ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,10032 Clinic for Oncology and Hematology ,2730 Oncology ,Personalized medicine ,business ,11493 Department of Quantitative Biomedicine - Abstract
The application and integration of molecular profiling technologies create novel opportunities for personalized medicine. Here, we introduce the Tumor Profiler Study, an observational trial combining a prospective diagnostic approach to assess the relevance of in-depth tumor profiling to support clinical decision-making with an exploratory approach to improve the biological understanding of the disease.
- Published
- 2021
- Full Text
- View/download PDF
17. PI3K-δ and PI3K-γ Inhibition by IPI-145 Abrogates Immune Responses and Suppresses Activity in Autoimmune and Inflammatory Disease Models
- Author
-
Alice R. Lim, Pingda Ren, Yi Liu, Jonathan P. DiNitto, Christian C. Fritz, Katti Jessen, Erin Murphy, Vito J. Palombella, Jennifer Lussier, Janid A. Ali, Brian D. Thomas, Kerry White, Lian-Sheng Li, Paul S. Changelian, Melissa Pink, Joi Dunbar, Jennifer Hoyt, Erin Brophy, Bonnie Tillotson, James R. Porter, John Macdougall, David G. Winkler, Christian Rommel, Kerrie Faia, Christian M. Martin, Jennifer L. Proctor, and Jeffery L. Kutok
- Subjects
Ovalbumin ,Clinical Biochemistry ,Arthritis ,Biology ,Biochemistry ,Article ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,Immune system ,Drug Discovery ,medicine ,Animals ,Humans ,Lupus Erythematosus, Systemic ,Rats, Wistar ,Collagen Type II ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Pharmacology ,Innate immune system ,Lupus erythematosus ,Dose-Response Relationship, Drug ,Molecular Structure ,CCL18 ,General Medicine ,Isoquinolines ,medicine.disease ,Asthma ,Rats ,Blockade ,Disease Models, Animal ,Basophil activation ,Purines ,Rats, Inbred Lew ,Immunology ,Molecular Medicine ,Female - Abstract
SummaryPhosphoinositide-3 kinase (PI3K)-δ and PI3K-γ are preferentially expressed in immune cells, and inhibitors targeting these isoforms are hypothesized to have anti-inflammatory activity by affecting the adaptive and innate immune response. We report on a potent oral PI3K-δ and PI3K-γ inhibitor (IPI-145) and characterize this compound in biochemical, cellular, and in vivo assays. These studies demonstrate that IPI-145 exerts profound effects on adaptive and innate immunity by inhibiting B and T cell proliferation, blocking neutrophil migration, and inhibiting basophil activation. We explored the therapeutic value of combined PI3K-δ and PI3K-γ blockade, and IPI-145 showed potent activity in collagen-induced arthritis, ovalbumin-induced asthma, and systemic lupus erythematosus rodent models. These findings support the hypothesis that inhibition of immune function can be achieved through PI3K-δ and PI3K-γ blockade, potentially leading to significant therapeutic effects in multiple inflammatory, autoimmune, and hematologic diseases.
- Published
- 2013
18. Dual mTORC1/2 and HER2 Blockade Results in Antitumor Activity in Preclinical Models of Breast Cancer Resistant to Anti-HER2 Therapy
- Author
-
Josep Tabernero, José Baselga, Christian Rommel, Maria Teresa Calvo, Katti Jessen, Marta Guzman, Celina Garcia-Garcia, Maurizio Scaltriti, José Francisco Pérez, Yi Liu, Claudia Aura, Yasir H. Ibrahim, Judit Grueso, and Violeta Serra
- Subjects
MAPK/ERK pathway ,Cancer Research ,Programmed cell death ,Receptor, ErbB-2 ,Blotting, Western ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Breast Neoplasms ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Lapatinib ,Mice ,Phosphatidylinositol 3-Kinases ,Trastuzumab ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,skin and connective tissue diseases ,Protein Kinase Inhibitors ,neoplasms ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Benzoxazoles ,business.industry ,TOR Serine-Threonine Kinases ,Cell Cycle ,Proteins ,Drug Synergism ,Xenograft Model Antitumor Assays ,Pyrimidines ,Oncology ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,Quinazolines ,Female ,business ,Proto-Oncogene Proteins c-akt ,Transcription Factors ,medicine.drug - Abstract
Purpose: The PI3K/Akt/mTOR pathway is an attractive target in HER2-positive breast cancer that is refractory to anti-HER2 therapy. The hypothesis is that the suppression of this pathway results in sensitization to anti-HER2 agents. However, this combinatorial strategy has not been comprehensively tested in models of trastuzumab and lapatinib resistance. Experimental Design: We analyzed in vitro cell viability and induction of apoptosis in five different cell lines resistant to trastuzumab and lapatinib. Inhibition of HER2/HER3 phosphorylation, PI3K/Akt/mTOR, and extracellular signal-regulated kinase (ERK) signaling pathways was evaluated by Western blotting. Tumor growth inhibition after treatment with lapatinib, INK-128, or the combination of both agents was evaluated in three different animal models: two cell-based xenograft models refractory to both trastuzumab and lapatinib and a xenograft derived from a patient who relapsed on trastuzumab-based therapy. Results: The addition of lapatinib to INK-128 prevented both HER2 and HER3 phosphorylation induced by INK-128, resulting in inhibition of both PI3K/Akt/mTOR and ERK pathways. This dual blockade produced synergistic induction of cell death in five different HER2-positive cell lines resistant to trastuzumab and lapatinib. In vivo, both cell line–based and patient-derived xenografts showed exquisite sensitivity to the antitumor activity of the combination of lapatinib and INK-128, which resulted in durable tumor shrinkage and exhibited no signs of toxicity in these models. Conclusions: The simultaneous blockade of both PI3K/Akt/mTOR and ERK pathways obtained by combining lapatinib with INK-128 acts synergistically in inducing cell death and tumor regression in breast cancer models refractory to anti-HER2 therapy. Clin Cancer Res; 18(9); 2603–12. ©2012 AACR.
- Published
- 2012
19. The translational landscape of mTOR signalling steers cancer initiation and metastasis
- Author
-
Nicholas T. Ingolia, Matthew R. Janes, Andrew C. Hsieh, Craig R. Stumpf, Pingda Ren, Carly Christensen, Yi Liu, Kevan M. Shokat, Jonathan S. Weissman, Katti Jessen, Shunyou Wang, Michael Bonham, Christian Rommel, Michael C. Martin, Annie Sher, Evan Y. Shi, Davide Ruggero, Morris E. Feldman, and Merritt Edlind
- Subjects
Male ,Cell Cycle Proteins ,Biology ,Bioinformatics ,Article ,Metastasis ,Mice ,Prostate cancer ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,Ribosome profiling ,Eukaryotic Initiation Factors ,Neoplasm Metastasis ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Regulation of gene expression ,Benzoxazoles ,Genome ,Multidisciplinary ,Cell growth ,TOR Serine-Threonine Kinases ,RPTOR ,Prostatic Neoplasms ,Phosphoproteins ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Repressor Proteins ,Eukaryotic Initiation Factor-4E ,Pyrimidines ,Protein Biosynthesis ,Cancer cell ,Cancer research ,Signal Transduction - Abstract
The mammalian target of rapamycin (mTOR) kinase is a master regulator of protein synthesis that couples nutrient sensing to cell growth and cancer. However, the downstream translationally regulated nodes of gene expression that may direct cancer development are poorly characterized. Using ribosome profiling, we uncover specialized translation of the prostate cancer genome by oncogenic mTOR signalling, revealing a remarkably specific repertoire of genes involved in cell proliferation, metabolism and invasion. We extend these findings by functionally characterizing a class of translationally controlled pro-invasion messenger RNAs that we show direct prostate cancer invasion and metastasis downstream of oncogenic mTOR signalling. Furthermore, we develop a clinically relevant ATP site inhibitor of mTOR, INK128, which reprograms this gene expression signature with therapeutic benefit for prostate cancer metastasis, for which there is presently no cure. Together, these findings extend our understanding of how the ‘cancerous’ translation machinery steers specific cancer cell behaviours, including metastasis, and may be therapeutically targeted.
- Published
- 2012
20. Critical role for phosphoinositide 3-kinase gamma in parasite invasion and disease progression of cutaneous leishmaniasis
- Author
-
Sanjay Varikuti, Stephanie Seveau, Steve Oghumu, Caroline C. Whitacre, Hannah E. Cummings, Anasuya Sarkar, Mark D. Wewers, Patrick K. Reville, Danuta Radzioch, Abhay R. Satoskar, Nicholas Zorko, Thomas Rückle, Joseph Barbi, Claudio M. Lezama-Davila, Tracy L. Keiser, Bao Lu, and Christian Rommel
- Subjects
Neutrophils ,Sodium stibogluconate ,Phagocytosis ,Leishmania mexicana ,Leishmaniasis, Cutaneous ,Host-Parasite Interactions ,Mice ,Phosphatidylinositol 3-Kinases ,Cutaneous leishmaniasis ,Quinoxalines ,parasitic diseases ,medicine ,Animals ,Humans ,Disease Resistance ,Phosphoinositide-3 Kinase Inhibitors ,Phagocytes ,Multidisciplinary ,Phosphoinositide 3-kinase ,biology ,Macrophages ,Leishmaniasis ,Biological Sciences ,Flow Cytometry ,biology.organism_classification ,medicine.disease ,Leishmania ,Mice, Inbred C57BL ,Chronic infection ,Microscopy, Fluorescence ,Antimony Sodium Gluconate ,Immunology ,biology.protein ,Thiazolidinediones ,medicine.drug - Abstract
Obligate intracellular pathogens such as Leishmania specifically target host phagocytes for survival and replication. Phosphoinositide 3-kinase γ (PI3Kγ), a member of the class I PI3Ks that is highly expressed by leukocytes, controls cell migration by initiating actin polymerization and cytoskeletal reorganization, which are processes also critical for phagocytosis. In this study, we demonstrate that class IB PI3K, PI3Kγ, plays a critical role in pathogenesis of chronic cutaneous leishmaniasis caused by L. mexicana . Using the isoform-selective PI3Kγ inhibitor, AS-605240 and PI3Kγ gene-deficient mice, we show that selective blockade or deficiency of PI3Kγ significantly enhances resistance against L. mexicana that is associated with a significant suppression of parasite entry into phagocytes and reduction in recruitment of host phagocytes as well as regulatory T cells to the site of infection. Furthermore, we demonstrate that AS-605240 is as effective as the standard antileishmanial drug sodium stibogluconate in treatment of cutaneous leishmaniasis caused by L. mexicana . These findings reveal a unique role for PI3Kγ in Leishmania invasion and establishment of chronic infection, and demonstrate that therapeutic targeting of host pathways involved in establishment of infection may be a viable strategy for treating infections caused by obligate intracellular pathogens such as Leishmania .
- Published
- 2012
21. MLN0128, a novel mTOR kinase inhibitor, disrupts survival signaling and triggers apoptosis in AML and AML stem/ progenitor cells
- Author
-
Kevin R. Coombes, Hagop M. Kantarjian, Suk Young Yoo, Qi Zhang, Yi Liu, Marina Konopleva, David A. Fruman, Duncan H. Mak, Katti Jessen, Christian Rommel, Steven M. Kornblau, Michael Andreeff, Zhihong Zeng, Rui Yu Wang, and Yihua Qiu
- Subjects
0301 basic medicine ,Cell Survival ,Apoptosis ,Mice, SCID ,03 medical and health sciences ,0302 clinical medicine ,AML ,Mice, Inbred NOD ,stem cells ,hemic and lymphatic diseases ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Progenitor cell ,Phosphorylation ,Protein kinase B ,neoplasms ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Mice, Knockout ,Benzoxazoles ,therapy ,Cell growth ,business.industry ,TOR Serine-Threonine Kinases ,Myeloid leukemia ,U937 Cells ,Xenograft Model Antitumor Assays ,3. Good health ,030104 developmental biology ,Cell killing ,Pyrimidines ,Oncology ,Leukemia, Myeloid ,030220 oncology & carcinogenesis ,Immunology ,Acute Disease ,Cancer research ,Neoplastic Stem Cells ,mTOR ,CyTOF ,Signal transduction ,Stem cell ,business ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,Research Paper - Abstract
mTOR activation leads to enhanced survival signaling in acute myeloid leukemia (AML) cells. The active-site mTOR inhibitors (asTORi) represent a promising new approach to targeting mTOR in AKT/mTOR signaling. MLN0128 is an orally-administered, second-generation asTORi, currently in clinical development. We examined the anti-leukemic effects and the mechanisms of action of MLN0128 in AML cell lines and primary samples, with a particular focus on its effect in AML stem/progenitor cells. MLN0128 inhibited cell proliferation and induced apoptosis in AML by attenuating the activity of mTOR complex 1 and 2. Using time-of-flight mass cytometry, we demonstrated that MLN0128 selectively targeted and functionally inhibited AML stem/progenitor cells with high AKT/mTOR signaling activity. Using the reverse-phase protein array technique, we measured expression and phosphorylation changes in response to MLN0128 in 151 proteins from 24 primary AML samples and identified several pro-survival pathways that antagonize MLN0128-induced cellular stress. A combined blockade of AKT/mTOR signaling and these pro-survival pathways facilitated AML cell killing. Our findings provide a rationale for the clinical use of MLN0128 to target AML and AML stem/progenitor cells, and support the use of combinatorial multi-targeted approaches in AML therapy.
- Published
- 2015
22. Immunity to protozoan parasite infection (PP-002)
- Author
-
H. Watanabe, P. Salotra, A. Yano, Mohsen Abolhassani, Mohammad Hossein Alimohammadian, M. Abdul Hafeez, F. Afrin, H. M. Snider, Amir Mizbani, C. H. N. Costa, Ali Khamesipour, M. Razavi, H. Xia, A. Zavaran Hoseini, R. Burchmore, Christopher A. Hunter, M. Makino, R. Correa-Oliveira, C. Shimokawa, A. P. Barral, M. Narita, E. M. Silva, R. K. M. T. Bairam, A. M. Silva, H. Borges, K. Nagamune, M. Suzuki, P. Hagan, R. M. A. Carvalho, M. Nakaya, F. Jafari, Fabio T. M. Costa, M. A. H. Khan, N. Ohta, P. Guirnalda, L. S. Elias, K. Takeshima, K. Hirayama, R. S. Vaz, D. Dasgupta, Gerson Chadi, T. K. Khiong, L. Cui, I. Bechmann, V. Dara, A. Östlund Farrants, Tajie H. Harris, P. Giusti, R. H. Panatieri, S. Rebelo, J. Uzonna, L. I. A. Pereira, K. Suzue, E. N. Miller, K. Suzuki, J. S. Wiley, Arlene H. Sharpe, Y. Kitamura, S. M. B. Jeronimo, M. P. Lees, F. D. Pretel, C. D. Gowda, L. Renia, L. M. G. Bahia-Oliveira, M. M. Molaei, Joseph Barbi, J. Argueta, S. Kobayashi, Z. Mou, N. C. Smith, Kayhan Azadmanesh, L. C. Ndhlovu, Y. Beuzard, J. Chavatte, Caroline C. Whitacre, M. Tasleem, Stephanie Seveau, A. Dolo, P. Giri, S. I. Castillo-Mendéz, S. Ajdary, Farnaz Zahedifard, G. A. DosReis, Houri Rezvan, X. Olivares López, F. Aosai, S. G. Yasawardene, M. Akhtar, A. U. Haq, J. Tavares, Jude E. Uzonna, M. I. Hiyane, L. Gutiérrez-Kobeh, S. Hamano, R. L. Rocha, C. M. V. Vendrame, C. E. Rosas-Jorquera, B. Niang, M. Islamuddin, E. Vannier, M. Rasouli, Bahram Kazemi, N. Khansari, Samaneh Saberi, S. A. Kaba, S. Dias, B. Mbengue, M. Mauduit, Hiva Azizi, D. E. Lanar, N. R. Palha, A. Ferreira, H. Goto, B. Lu, A. Yoshida, Y. Hamzavi, Kazumi Norose, S. Soeng, A. Gorgin karaji, Y. Chinzei, G. K. Katara, A. Dieye, Luiz Roberto Sardinha, Nicholas Zorko, M. Troye-Blomberg, Karina R. Bortoluci, A. A. Oeij, O. Doumbo, Y. Yamaguchi, L. Castellucci, M. Takahashi, Tahere Taheri, A. Gruner, H. Yoshida, Yasaman Taslimi, B. S. Dwarakanath, S. Rojas Hernández, N. Ishii, K. Honma, A. A. A. Mohammady, A. Latifynia, A. Khodadadi, N. Vega Martínez, S. Koyasu, B. Malleret, Akitoshi Kikumura, M. F. Lopes, E. Houpt, Masoud Moradi, J. V. Weyenbergh, N. Uemura, R. M. Siegel, S. Magez, C. Brando, E. Salles, K. Sugamura, Y. Miyahira, M. Moroda, A. Shibuya, Q. Guo, M. M. Awais, Samar Kumar Guha, Tracy L. Keiser, D. Liu, S. Boström, B. Traoré, L. D. Souza, M. R. D'Império-Lima, W. F. Pereira, P. Burkhard, L. V. C. Guillermo, Carlos Penha-Gonçalves, M. Carrasco Yépez, C. Mittelholzer, C. M. Gomes, Abhay R. Satoskar, H. Daneshvar, H. Hara, A. Kanayama, M. Kayibanda, H. Maruyama, C. Arama, A. Asao, T. Tamura, M. Barral-Netto, N. T. Huy, H. Kamiabi, S. A. Pinto, C. Claser, José M. Alvarez, R. Ramasamy, T. Kanda-Taniguchi, M. Kikuchi, T. Susy, Lígia A. Gonçalves, F. Ginhoux, Abdolmajid Fata, Srijit Khan, H. Nekouie, M. L. Dorta, A. Lima-Neto, A. L. Peixoto-Rangel, M. R. D'Império Lima, V. Khase Shahgoli, H. Hisaeda, S. Black, A. Raz, V. Bockstal, K. Salgado, R. Campos Rodríguez, W. V. Parreira, S. Varani, T. Ono, C. A. Zago, M. Miyakoda, M. Nateghi-Rostami, L. C. Reis, M. Yamazaki, F. Montalvão, Sedigheh Zakeri, M. Doroudian, M. D. T. Carvalho, A. Henri, F. L. Ribeiro-Gomes, D. Kimura, A. Montes de Oca, V. Ramesh, E. M. Carvalho, B. Diatta, Danuta Radzioch, Patrick K. Reville, L. Roubaix, L. F. Batista, A. Cordeiro-da-Silva, Mojtaba Sankian, M. E. McCoy, G. Chouhan, J. M. Blackwell, M. Irfan Anwar, P. Teo, Rima McLeod, R. Udomsangpetch, M. P. Soares, M. Udagawa, Q. Gao, F. Ribeiro-Dias, B. L. Lima, K. Kimura, Y. Inamine, E. Belnoue, Â. Chora, N. D. Jadid, M. Yasunami, A. R. K. Goldberg, S. Hejazi, S. E. Jamieson, P. Bonilla-us, J. Kalil, Mahmoud Eshagh Hosseini, M. J. Gharagozlou, L. C. Oliveira, A. Alborzi, M. Mahmoudian Sani, N. S. Vellozo, A. Farooque, Mohammad Ali Nilforoushzadeh, S. R. Phillips, Q. Fang, T. Imai, T. Taniguchi, S. Phompida, I. Bujila, D. Iravani, P. Jia, I. Hussain, F. Ahmad, M. Senba, T. Yanagi, N. Hosseini, R. Perraut, S. J. Fuller, Hannah E. Cummings, S. Umemoto, M. K. Mannoor, K. Jangpatarapongsa, K. Yui, C. Li, A. Varasteh, T. A. P. F. Pimentel, D. L. Costa, F. A. Asteal, A. Barral, E. Ramos Sanchez, Kenji Shibuya, Sima Rafati, M. N. Shuaibu, Saqib Ali, Barbara Papadopoulou, G. K. Helegbe, Nastaran Ansari, J. Sattabongkot, S. Kiany, Christian Rommel, T. Wickramarachchi, H. H. Wortis, J. Yamada, M. Inahuku, F. Abrishami, P. V. Udagama-Randeniya, E. P. Amaral, F. Muhammad, Thomas Rückle, B. Li, M. Resende, A. Vigario, A. F. Frade, Y. Yang, André Luis Bombeiro, M. Yuda, P. Reville, G. Snounou, Craig Gerard, H. M. Niknam, M. A. P. Oliveira, T. Hoshino, R. G. Peixe, S. Zavosh, V. Thomaz-Soccol, S. Inam, and Graham H. Coombs
- Subjects
Immunity ,Immunology ,Immunology and Allergy ,General Medicine ,Biology ,Protozoan parasite ,Microbiology - Published
- 2010
23. Chemokine receptor CCR2 undergoes transportin1-dependent nuclear translocation
- Author
-
Nicolas Favre, Christian Arod, Christian Pasquali, Christian Chabert, Montserrat Camps, and Christian Rommel
- Subjects
Proteomics ,CCR1 ,CCR2 ,Receptors, CCR2 ,Chemokine receptor CCR5 ,Active Transport, Cell Nucleus ,C-C chemokine receptor type 7 ,C-C chemokine receptor type 6 ,Biochemistry ,Cell Line ,Epitopes ,Chemokine receptor ,Tandem Mass Spectrometry ,Protein Interaction Mapping ,parasitic diseases ,Humans ,Immunoprecipitation ,RNA, Small Interfering ,Molecular Biology ,Chemokine CCL2 ,Cell Nucleus ,biology ,Chemotaxis ,beta Karyopherins ,Cell biology ,Hemagglutinins ,biology.protein ,CC chemokine receptors ,Chromatography, Liquid ,Signal Transduction ,CCL21 - Abstract
Chemokines (CCs) are small chemoattractant cytokines involved in a wide variety of biological and pathological processes. Released by cells in the milieu, and extracellular matrix and activating signalling cascades upon binding to specific G protein-coupled receptors (GPCRs), they trigger many cellular events. In various pathologies, CCs are directly responsible for excessive recruitment of leukocytes to inflammatory sites and recent studies using chemokine receptor (CCR) antagonists permitted these molecules to reach the market for medical use. While interaction of CCs with their receptors has been extensively documented, downstream GPCR signalling cascades triggered by CC are less well understood. Given the pivotal role of chemokine receptor 2 (CCR2) in monocyte recruitment, activation and differentiation and its implication in several autoimmune-inflammatory pathologies, we searched for potential new CCR2-interacting proteins by engineering a modified CCR2 that we used as bait. Herein, we show the direct interaction of CCR2 with transportin1 (TRN1), which we demonstrate is followed by CCR2 receptor internalization. Further characterization of this novel interaction revealed that TRN1-binding to CCR2 increased upon time in agonist treated cells and promotes its nuclear translocation in a TRN1-dependent manner. Finally, we provide evidence that following translocation, the receptor localizes at the outer edge of the nuclear envelope where it is finally released from TRN1.
- Published
- 2008
24. Isoform-Specific Functions of Phosphoinositide 3-Kinases: p110δ but Not p110γ Promotes Optimal Allergic Responses In Vivo
- Author
-
Khaled Ali, Hong Ji, Christian Rommel, Christian Pasquali, Nicolas Kuehn, Montserrat Camps, Thomas Rückle, Christian Chabert, Wayne Pearce, and Bart Vanhaesebroeck
- Subjects
Gene isoform ,biology ,Cell Degranulation ,Proto-Oncogene Proteins c-akt ,Immunology ,Degranulation ,Immunoglobulin E ,Mast cell ,Cell biology ,medicine.anatomical_structure ,P110δ ,In vivo ,biology.protein ,medicine ,Immunology and Allergy - Abstract
The leukocyte-enriched p110γ and p110δ isoforms of PI3K have been shown to control in vitro degranulation of mast cells induced by cross-linking of the high affinity receptor of IgE (FcεRI). However, the relative contribution of these PI3K isoforms in IgE-dependent allergic responses in vivo is controversial. A side-by-side comparative analysis of the role of p110γ and p110δ in mast cell function, using genetic approaches and newly developed isoform-selective pharmacologic inhibitors, confirms that both PI3K isoforms play an important role in FcεRI-activated mast cell degranulation in vitro. In vivo, however, only p110δ was found to be required for optimal IgE/Ag-dependent hypersensitivity responses in mice. These observations identify p110δ as a key therapeutic target among PI3K isoforms for allergy- and mast cell-related diseases.
- Published
- 2008
25. Tissue- and Stimulus-Dependent Role of Phosphatidylinositol 3-Kinase Isoforms for Neutrophil Recruitment Induced by Chemoattractants In Vivo
- Author
-
Vanessa, Pinho, Remo Castro, Russo, Remo, de Castro Russo, Flávio A, Amaral, Lirlândia P, de Sousa, Michele M, Barsante, Danielle G, de Souza, José C, Alves-Filho, Denise C, Cara, Joel S, Hayflick, Christian, Rommel, Thomas, Ruckle, Adriano G, Rossi, and Mauro M, Teixeira
- Subjects
Male ,Neutrophils ,Chemokine CXCL1 ,Immunology ,Inflammation ,Endogeny ,Biology ,Wortmannin ,Mice ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,In vivo ,medicine ,Animals ,Class Ib Phosphatidylinositol 3-Kinase ,Immunology and Allergy ,Mice, Knockout ,Chemotactic Factors ,Chemotaxis ,Neutrophil extracellular traps ,respiratory system ,Molecular biology ,Recombinant Proteins ,Isoenzymes ,Mice, Inbred C57BL ,CXCL1 ,Disease Models, Animal ,Neutrophil Infiltration ,chemistry ,medicine.symptom ,Intravital microscopy - Abstract
PI3K plays a fundamental role in regulating neutrophil recruitment into sites of inflammation but the role of the different isoforms of PI3K remains unclear. In this study, we evaluated the role of PI3Kgamma and PI3Kdelta for neutrophil influx induced by the exogenous administration or the endogenous generation of the chemokine CXCL1. Administration of CXCL1 in PI3Kgamma(-/-) or wild-type (WT) mice induced similar increases in leukocyte rolling, adhesion, and emigration in the cremaster muscle when examined by intravital microscopy. The induction of neutrophil recruitment into the pleural cavity or the tibia-femoral joint induced by the injection of CXCL1 was not significantly different in PI3Kgamma(-/-) or WT mice. Neutrophil influx was not altered by treatment of WT mice with a specific PI3Kdelta inhibitor, IC87114, or a specific PI3Kgamma inhibitor, AS605240. The administration of IC87114 prevented CXCL1-induced neutrophil recruitment only in presence of the PI3Kgamma inhibitor or in PI3Kgamma(-/-) mice. Ag challenge of immunized mice induced CXCR2-dependent neutrophil recruitment that was inhibited by wortmannin or by blockade of and PI3Kdelta in PI3Kgamma(-/-) mice. Neutrophil recruitment to bronchoalveolar lavage induced by exogenously added or endogenous production of CXCL1 was prevented in PI3Kgamma(-/-) mice. The accumulation of the neutrophils in lung tissues was significantly inhibited only in PI3Kgamma(-/-) mice treated with IC87114. Neutrophil recruitment induced by exogenous administration of C5a or fMLP appeared to rely solely on PI3Kgamma. Altogether, our data demonstrate that there is a tissue- and stimulus-dependent role of PI3Kgamma and PI3Kdelta for neutrophil recruitment induced by different chemoattractants in vivo.
- Published
- 2007
26. A Chemical Proteomics Approach to Phosphatidylinositol 3-Kinase Signaling in Macrophages
- Author
-
Christian Chabert, Christian Arod, Karl Mechtler, Christian Rommel, Christian Pasquali, Montserrat Camps, Ioannis Xenarios, Dominique Bertschy-Meier, Randy Booth, Marie Laure Curchod, Francis Vilbois, Colin G. Ferguson, and Glenn D. Prestwich
- Subjects
Proteomics ,Lipid Chemistry ,Akt/PKB signaling pathway ,Macrophages ,Intracellular Signaling Peptides and Proteins ,Biology ,Phosphatidylinositols ,Lipids ,Biochemistry ,Chromatography, Affinity ,Analytical Chemistry ,Pleckstrin homology domain ,Mice ,Phosphatidylinositol 3-Kinases ,Proteome ,Animals ,Phosphatidylinositol 3-kinase signaling ,Signal transduction ,Protein kinase A ,Molecular Biology ,Cells, Cultured ,Signal Transduction - Abstract
Prior work using lipid-based affinity matrices has been done to investigate distinct sets of lipid-binding proteins, and one series of experiments has proven successful in mammalian cells for the proteome-wide identification of lipid-binding proteins. However, most lipid-based proteomics screens require scaled up sample preparation, are often composed of multiple cell types, and are not adapted for simultaneous signal transduction studies. Herein we provide a chemical proteomics strategy that uses cleavable lipid "baits" with broad applicability to diverse biological samples. The novel baits were designed to avoid preparative steps to allow functional proteomics studies when the biological source is a limiting factor. Validation of the chemical baits was first confirmed by the selective isolation of several known endogenous phosphatidylinositol 3-kinase signaling proteins using primary bone marrow-derived macrophages. The use of this technique for cellular proteomics and MS/MS analysis was then demonstrated by the identification of known and potential novel lipid-binding proteins that was confirmed in vitro for several proteins by direct lipid-protein interactions. Further to the identification, the method is also compatible with subsequent signal transduction studies, notably for protein kinase profiling of the isolated lipid-bound protein complexes. Taken together, this integration of minimal scale proteomics, lipid chemistry, and activity-based readouts provides a significant advancement in the ability to identify and study the lipid proteome of single, relevant cell types.
- Published
- 2007
27. Protein-tyrosine Phosphatase H1 Controls Growth Hormone Receptor Signaling and Systemic Growth
- Author
-
Marie-Laure Curchod, Paola Zaratin, David M. Valenzuela, Yingzi Xue, Ann M. Clark, Dominique Perrin, Claudia Patrignani, Maria Chiara Magnone, Rob Hooft van Huijsduijnen, R Pescini, Iwona Pilecka, Jason Yasenchak, and Christian Rommel
- Subjects
Male ,medicine.medical_specialty ,medicine.medical_treatment ,Phosphatase ,Growth hormone receptor ,Biology ,Models, Biological ,Biochemistry ,Mice ,Growth factor receptor ,In vivo ,Catalytic Domain ,Internal medicine ,medicine ,Animals ,Humans ,Growth factor receptor inhibitor ,RNA, Messenger ,Insulin-Like Growth Factor I ,Phosphorylation ,Molecular Biology ,Cell Proliferation ,Insulin-like growth factor 1 receptor ,Mice, Knockout ,Growth factor ,Protein Tyrosine Phosphatase, Non-Receptor Type 3 ,Wild type ,Receptors, Somatotropin ,Cell Biology ,Endocrinology ,Liver ,Mutation ,Female ,Signal Transduction - Abstract
Several protein-tyrosine phosphatases (PTPs) have been implicated in the control of growth hormone receptor (GHR) signaling, but none have been shown to affect growth in vivo. We have applied a battery of molecular and cellular approaches to test a family-wide panel of PTPs for interference with GHR signaling. Among the subset of PTPs that showed activity in multiple readouts, we selected PTP-H1/PTPN3 for further in vivo studies and found that mice lacking the PTP-H1 catalytic domain show significantly enhanced growth over their wild type littermates. In addition, PTP-H1 mutant animals had enhanced plasma and liver mRNA expression of insulin-like growth factor 1, as well as increased bone density and mineral content. These observations point to a controlling role for PTP-H1 in modulating GHR signaling and systemic growth through insulin-like growth factor 1 secretion.
- Published
- 2007
28. Targeting dual-specificity phosphatases: manipulating MAP kinase signalling and immune responses
- Author
-
Charles R. Mackay, Montserrat Camps, Christian Rommel, and Kate L. Jeffrey
- Subjects
Pharmacology ,MAPK/ERK pathway ,Kinase ,Immunity ,General Medicine ,Protein tyrosine phosphatase ,Biology ,Phosphoric Monoester Hydrolases ,Substrate Specificity ,Cell biology ,Immune system ,Mitogen-activated protein kinase ,Drug Discovery ,Dual-specificity phosphatase ,biology.protein ,Animals ,Humans ,Mitogen-Activated Protein Kinases ,Protein Tyrosine Phosphatases ,Tyrosine ,Signal transduction ,Signal Transduction - Abstract
Dual-specificity phosphatases (DUSPs) are a subset of protein tyrosine phosphatases, many of which dephosphorylate threonine and tyrosine residues on mitogen-activated protein kinases (MAPKs), and hence are also referred to as MAPK phosphatases (MKPs). The regulated expression and activity of DUSP family members in different cells and tissues controls MAPK intensity and duration to determine the type of physiological response. For immune cells, DUSPs regulate responses in both positive and negative ways, and DUSP-deficient mice have been used to identify individual DUSPs as key regulators of immune responses. From a drug discovery perspective, DUSP family members are promising drug targets for manipulating MAPK-dependent immune responses in a cell-type and disease-context-dependent manner, to either boost or subdue immune responses in cancers, infectious diseases or inflammatory disorders.
- Published
- 2007
29. PI3Kδ and PI3Kγ: partners in crime in inflammation in rheumatoid arthritis and beyond?
- Author
-
Hong Ji, Montserrat Camps, and Christian Rommel
- Subjects
Gene isoform ,History ,business.industry ,Inflammation ,medicine.disease ,Computer Science Applications ,Education ,Immune system ,Rheumatoid arthritis ,Immunology ,Second messenger system ,medicine ,medicine.symptom ,Signal transduction ,business ,Signalling pathways ,PI3K/AKT/mTOR pathway - Abstract
The phosphoinositide 3-kinase (PI3K) isoforms PI3Kδ and PI3Kγ generate lipid second messengers that control an array of signalling pathways for numerous immune-cell functions. Recent studies indicate that specific targeting of these PI3K isoforms could be beneficial for treating inflammatory diseases. Dysregulated signal transduction in innate and adaptive immune cells is known to be associated with the development of various autoimmune and inflammatory diseases. Consequently, targeting intracellular signalling of the pro-inflammatory cytokine network heralds hope for the next generation of anti-inflammatory drugs. Phosphoinositide 3-kinases (PI3Ks) generate lipid-based second messengers that control an array of intracellular signalling pathways that are known to have important roles in leukocytes. In light of the recent progress in the development of selective PI3K inhibitors, and the beneficial effects of these inhibitors in models of acute and chronic inflammatory disorders, we discuss the therapeutic potential of blocking PI3K isoforms for the treatment of rheumatoid arthritis and other immune-mediated diseases.
- Published
- 2007
30. T-cell function is partially maintained in the absence of class IA phosphoinositide 3-kinase signaling
- Author
-
Jonathan A. Deane, Jean S. Oak, Thomas E. Lane, Travis I. Moore, Linda N. Stiles, Lewis C. Cantley, Ji Luo, Michael G. Kharas, Hong Ji, David A. Fruman, and Christian Rommel
- Subjects
T-Lymphocytes ,medicine.medical_treatment ,T cell ,Lymphocyte Cooperation ,Immunology ,Mice, Transgenic ,In Vitro Techniques ,Biochemistry ,Mice ,Phosphatidylinositol 3-Kinases ,medicine ,Animals ,IL-2 receptor ,PI3K/AKT/mTOR pathway ,Immunobiology ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Phosphoinositide 3-kinase ,biology ,TOR Serine-Threonine Kinases ,T-cell receptor ,CD28 ,Cell Biology ,Hematology ,Mice, Mutant Strains ,Cell biology ,Mice, Inbred C57BL ,Protein Subunits ,Cytokine ,medicine.anatomical_structure ,biology.protein ,Signal transduction ,Protein Kinases ,Signal Transduction - Abstract
The class IA subgroup of phosphoinositide 3-kinase (PI3K) is activated downstream of antigen receptors, costimulatory molecules, and cytokine receptors on lymphocytes. Targeted deletion of individual genes for class IA regulatory subunits severely impairs the development and function of B cells but not T cells. Here we analyze conditional mutant mice in which thymocytes and T cells lack the major class IA regulatory subunits p85α, p55α, p50α, and p85β. These cells exhibit nearly complete loss of PI3K signaling downstream of the T-cell receptor (TCR) and CD28. Nevertheless, T-cell development is largely unperturbed, and peripheral T cells show only partial impairments in proliferation and cytokine production in vitro. Both genetic and pharmacologic experiments suggest that class IA PI3K signaling plays a limited role in T-cell proliferation driven by TCR/CD28 clustering. In vivo, class IA–deficient T cells provide reduced help to B cells but show normal ability to mediate antiviral immunity. Together these findings provide definitive evidence that class IA PI3K regulatory subunits are essential for a subset of T-cell functions while challenging the notion that this signaling mechanism is a critical mediator of costimulatory signals downstream of CD28.
- Published
- 2006
31. PI3Kγ inhibition: towards an 'aspirin of the 21st century'?
- Author
-
Christian Rommel, Matthias Schwarz, and Thomas Rückle
- Subjects
Pharmacology ,Aspirin ,business.industry ,Anti-Inflammatory Agents, Non-Steroidal ,Drug target ,General Medicine ,Disease ,Bioinformatics ,Hematopoiesis ,Mice ,Human disease ,Drug Discovery ,Animals ,Humans ,Medicine ,Receptors, Chemokine ,Chemokines ,Enzyme Inhibitors ,business ,Phosphoinositide-3 Kinase Inhibitors ,medicine.drug - Abstract
Class IB phosphatidylinositol 3-kinase p110gamma (PI3Kgamma) has gained increasing attention as a promising drug target for the treatment of inflammatory disease. Extensive target-validation data are available, which are derived from studies using both pharmacological and genetic tools. More recent findings have uncovered further therapeutic applications for PI3Kgamma inhibitors, opening up potentially huge opportunities for these drugs. Several companies have been pursuing small-molecule PI3Kgamma inhibitor projects, but none of them has progressed to the clinic yet. Here, we discuss the insights gained so far and the main challenges that are emerging on the path to developing PI3Kgamma inhibitors for the treatment of human disease.
- Published
- 2006
32. Key role of the p110δ isoform of PI3K in B-cell antigen and IL-4 receptor signaling: comparative analysis of genetic and pharmacologic interference with p110δ function in B cells
- Author
-
Juliet L. Emery, Klaus Okkenhaug, Thomas Rückle, Christian Rommel, Antonio Bilancio, Montserrat Camps, Bart Vanhaesebroeck, Bilancio, Antonio, Okkenhaug, K, Camps, M, Emery, Jl, Ruckle, T, Rommel, C, and Vanhaesebroeck, B.
- Subjects
Male ,Apoptosis ,Retinoblastoma Protein ,Biochemistry ,Glycogen Synthase Kinase 3 ,Mice ,Phosphatidylinositol 3-Kinases ,GSK-3 ,Cyclin D2 ,Protein Isoforms ,Enzyme Inhibitors ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Mice, Knockout ,B-Lymphocytes ,Cell Cycle ,Forkhead Box Protein O3 ,Ribosomal Protein S6 Kinases, 70-kDa ,Cell Differentiation ,Forkhead Transcription Factors ,Hematology ,Cell biology ,Female ,Signal transduction ,Signal Transduction ,medicine.medical_specialty ,Class I Phosphatidylinositol 3-Kinases ,Immunology ,B-cell receptor ,Receptors, Antigen, B-Cell ,P70-S6 Kinase 1 ,Cyclin A ,Biology ,Cyclins ,Internal medicine ,Cyclin E ,medicine ,Animals ,Protein kinase B ,Cell Proliferation ,Glycogen Synthase Kinase 3 beta ,Akt/PKB signaling pathway ,Cell Biology ,Receptors, Interleukin-4 ,Mice, Inbred C57BL ,Endocrinology ,P110δ ,Calcium ,Interleukin-4 ,Proto-Oncogene Proteins c-akt - Abstract
Mouse gene-targeting studies have documented a central role of the p110delta isoform of phosphoinositide 3-kinase (PI3K) in B-cell development and function. A defect in B-cell antigen receptor (BCR) signaling is key to this B-cell phenotype. Here we further characterize this signaling defect and report that a p110delta-selective small molecule inhibitor mirrors the effect of genetic inactivation of p110delta in BCR signaling. p110delta activity is indispensable for BCR-induced DNA synthesis and phosphorylation of Akt/protein kinase B (PKB), forkhead transcription factor/forkhead box O3a (FOXO3a), and p70 S6 kinase (p70 S6K), with modest effects on the phosphorylation of glycogen synthase kinase 3 alpha/beta (GSK3alpha/beta) and extracellular signal-regulated kinase (Erk). The PI3K-dependent component of intracellular calcium mobilization also completely relies on p110delta catalytic activity. Resting B cells with inactive p110delta fail to enter the cell cycle, correlating with an incapacity to up-regulate the expression of cyclins D2, A, and E, and to phosphorylate the retinoblastoma protein (Rb). p110delta is also critical for interleukin 4 (IL-4)-induced phosphorylation of Akt/PKB and FOXO3a, and protection from apoptosis. Taken together, these data show that defects observed in p110delta mutant mice are not merely a consequence of altered B-cell differentiation, and emphasize the potential utility of p110delta as a drug target in autoimmune diseases in which B cells play a crucial role.
- Published
- 2006
33. Sequential activation of class IB and class IA PI3K is important for the primed respiratory burst of human but not murine neutrophils
- Author
-
Matthias P. Wymann, Martin R Turner, Bart Vanhaesebroeck, Alison M. Condliffe, Phillip T. Hawkins, Shaun P. Jackson, Keith Davidson, Christian Rommel, Edwin R. Chilvers, Louise M. C. Webb, Karen E. Anderson, Thomas Rückle, Emilio Hirsch, Klaus Okkenhaug, Montserrat Camps, Len R. Stephens, Chris D. Ellson, and Tom Crabbe
- Subjects
Class I Phosphatidylinositol 3-Kinases ,respiratory burst ,neutrophils ,signal transduction ,PI3K ,Immunology ,Priming (immunology) ,Inflammation ,Biochemistry ,Proinflammatory cytokine ,Mice ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Species Specificity ,medicine ,Animals ,Class Ib Phosphatidylinositol 3-Kinase ,Humans ,Enzyme Inhibitors ,Cells, Cultured ,NADPH oxidase ,Phosphoinositide 3-kinase ,biology ,Tumor Necrosis Factor-alpha ,Cell Biology ,Hematology ,N-Formylmethionine leucyl-phenylalanine ,Respiratory burst ,Cell biology ,Enzyme Activation ,Isoenzymes ,N-Formylmethionine Leucyl-Phenylalanine ,chemistry ,biology.protein ,Tumor necrosis factor alpha ,medicine.symptom ,Reactive Oxygen Species - Abstract
It is well established that preexposure of human neutrophils to proinflammatory cytokines markedly augments the production of reactive oxygen species (ROS) to subsequent stimuli. This priming event is thought to be critical for localizing ROS to the vicinity of the inflammation, maximizing their role in the resolution of the inflammation, and minimizing the damage to surrounding tissue. We have used a new generation of isoform-selective phosphoinositide 3-kinase (PI3K) inhibitors to show that ROS production under these circumstances is regulated by temporal control of class I PI3K activity. Stimulation of tumor necrosis factor-alpha (TNF-alpha)-primed human neutrophils with N-formyl-methionyl-leucyl-phenylalanine (fMLP) results in biphasic activation of PI3K; the first phase is largely dependent on PI3Kgamma, and the second phase is largely dependent on PI3Kdelta. The second phase of PI3K activation requires the first phase; it is this second phase that is augmented by TNF-alpha priming and that regulates parallel activation of ROS production. Surprisingly, although TNF-alpha-primed mouse bone marrow-derived neutrophils exhibit superficially similar patterns of PI3K activation and ROS production in response to fMLP, these responses are substantially lower and largely dependent on PI3Kgamma alone. These results start to define which PI3K isoforms are responsible for modulating neutrophil responsiveness to infection and inflammation.
- Published
- 2005
34. Therapeutic potential of phosphoinositide 3-kinase inhibitors
- Author
-
Beth E. Drees, Glenn D. Prestwich, Christian Rommel, and Gordon B. Mills
- Subjects
Pharmacology ,chemistry.chemical_classification ,Phosphoinositide 3-kinase ,Kinase ,High-throughput screening ,Cancer ,General Medicine ,Biology ,medicine.disease ,Cell biology ,Enzyme ,Immune system ,chemistry ,Drug development ,Drug Discovery ,Cancer research ,biology.protein ,medicine ,Signal transduction - Abstract
Originally discovered as oncogene-associated lipid kinases more than 15 years ago, the family of phosphoinositide 3-kinase (PI 3-K) enzymes has recently emerged as an important therapeutic target in human pathophysiology. After more than a decade with only a few inhibitors with low activity, poor isoform or kinase selectivity or unacceptable pharmacological and toxicological profiles, a variety of PI 3-K inhibitors with pan-isoform activity and isoform-selective inhibition have recently been identified. A growing excitement surrounds the success of signal transduction modifiers in cancer therapy, as well as in the therapy of other diseases. In combination with additional genetic evidence implicating the PI 3-K pathway in human disease, the search for more selective and more drug-like PI 3-K inhibitors has been reinvigorated. As a consequence, many biotech and pharmaceutical companies have established PI 3-K pathway drug development programmes. With the help of important insights provided by genetic knocko...
- Published
- 2004
35. Mining for Protein Kinase Substrates: Integration of Biochemistry, Genetics and Proteomics
- Author
-
Mark Velleca, Christian Rommel, Christian Pasquali, and Benjamin K. Benton
- Subjects
Biochemistry ,Biology ,Proteomics ,Protein kinase A ,Molecular Biology - Published
- 2004
36. Strategies for chemokine antagonists as therapeutics
- Author
-
Christine A. Power, Christian Rommel, Amanda E. I. Proudfoot, and Timothy N. C. Wells
- Subjects
CCR1 ,Chemokine ,biology ,Chemokine receptor CCR5 ,Immunology ,Proteins ,CXCR3 ,Antibodies ,Protein Transport ,Chemokine receptor ,Drug Design ,biology.protein ,Animals ,Humans ,Immunology and Allergy ,Receptors, Chemokine ,CCR10 ,Chemokines ,Signal transduction ,Glycosaminoglycans ,Signal Transduction ,Homing (hematopoietic) - Abstract
Chemokines are responsible for specific recruitment of leukocytes that are involved both in homing as well as in inflammation. Dysregulation of the system results in excessive recruitment to inflammatory sites and thus prevention of this recruitment is an effective anti-inflammatory strategy. Chemokine receptors are not limited only to cellular recruitment but are also the essential co-factor along with CD4 that enable HIV-1 viruses to infect cells. In this review we discuss the various points of intervention that can be addressed to provide anti-inflammatory and anti-HIV infectivity therapeutics. These include prevention of the receptor-ligand interaction, prevention of the chemokine-glycosaminoglycan interaction, interfering with the signaling pathways that are induced upon receptor activation, and modification of receptor trafficking pathways. We summarize the status of the approaches that have been undertaken to produce therapeutics that block chemokine action.
- Published
- 2003
37. Next-generation flow cytometry
- Author
-
Matthew R. Janes and Christian Rommel
- Subjects
Flow injection analysis ,medicine.diagnostic_test ,Chemistry ,Cell ,Biomedical Engineering ,Bioengineering ,Applied Microbiology and Biotechnology ,Molecular biology ,Flow cytometry ,medicine.anatomical_structure ,Phenotypic analysis ,Cell culture ,medicine ,Molecular Medicine ,Mass cytometry ,Biotechnology - Abstract
Mass cytometry dramatically enhances the dimensionality of fluorescence-based flow cytometry for phenotypic analysis of heterogeneous cell populations.
- Published
- 2011
38. PI3K and cancer: Lessons, challenges and opportunities
- Author
-
Christian Rommel and David A. Fruman
- Subjects
Antineoplastic Agents ,Biology ,Bioinformatics ,Medical and Health Sciences ,Article ,Phosphatidylinositol 3-Kinases ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Pharmacology & Pharmacy ,Enzyme Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Cancer ,Pharmacology ,Clinical Trials as Topic ,Drug discovery ,Kinase ,TOR Serine-Threonine Kinases ,General Medicine ,Receptor Cross-Talk ,Biological Sciences ,medicine.disease ,Clinical trial ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The central role of phosphoinositide 3-kinase (PI3K) activation in tumour cell biology has prompted a sizeable effort to target PI3K and/or downstream kinases such as AKT and mammalian target of rapamycin (mTOR) in cancer. However, emerging clinical data show limited single-agent activity of inhibitors targeting PI3K, AKT or mTOR at tolerated doses. One exception is the response to PI3Kδ inhibitors in chronic lymphocytic leukaemia, where a combination of cell-intrinsic and -extrinsic activities drive efficacy. Here, we review key challenges and opportunities for the clinical development of inhibitors targeting the PI3K-AKT-mTOR pathway. Through a greater focus on patient selection, increased understanding of immune modulation and strategic application of rational combinations, it should be possible to realize the potential of this promising class of targeted anticancer agents..©2014 Macmillan Publishers Limited. All rights reserved.
- Published
- 2014
39. Retroviral gene transfer of dominant negative Raf-1 mutants suppresses Ha-ras-induced transformation and delays tumor formation
- Author
-
Gerald Radziwill, Jovan Pavlovic, Thomas Heinicke, Christian Rommel, Michael Nawrath, and Karin Moelling
- Subjects
Cancer Research ,DNA, Complementary ,Time Factors ,Immunoblotting ,Mutant ,Genes, myc ,Reversion ,Mice, Nude ,Mice ,Transduction, Genetic ,Animals ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Genes, Dominant ,Mitogen-Activated Protein Kinase 1 ,Mice, Inbred ICR ,Models, Genetic ,Chemistry ,Effector ,Gene Transfer Techniques ,Contact inhibition ,3T3 Cells ,Neoplasms, Experimental ,Flow Cytometry ,Phenotype ,Molecular biology ,In vitro ,Proto-Oncogene Proteins c-raf ,Kinetics ,Transformation (genetics) ,Cell Transformation, Neoplastic ,Genes, ras ,Retroviridae ,Molecular Medicine ,Cell Division ,Neoplasm Transplantation ,Signal Transduction - Abstract
Activating mutants of ras are among the most frequently found genetic alterations in human cancers. Therefore, Ras appears to be an attractive target for therapeutic intervention using gene transfer. The protein kinase Raf-1 acts as a direct downstream effector of Ras and is involved in Ras-induced cellular transformation. Using the NIH3T3 fibroblast-derived tumor cell line PEJ, which expresses oncogenic Ha-rasG12V, we analyzed whether dominant negative mutants of Raf-1 can inhibit Ras-mediated transformation. Retroviral gene transfer was used to stably transduce PEJ cells with three different dominant negative mutants of Raf-1. This resulted in reversion of the transformed phenotype in vitro as evidenced by an increase in contact inhibition and reduced anchorage-independent growth. However, tumor formation in nude mice was significantly delayed only by one of these mutants. Therefore, dominant negative mutants of the oncoprotein Myc, which is known to synergize with Raf-1 in tumor formation, were transduced into PEJ cells expressing a dominant negative Raf mutant. This leads to killing of the cells. These results indicate that although interference with Ras-induced transformation using dominant negative mutants of Raf is feasible and effective in vitro using retroviral vectors, an additional block (e.g., that of Myc) is necessary to kill PEJ cells. These results also indicate that interference with Ras-dependent signaling is not sufficient for inhibition of tumor formation of PEJ cells in vivo.
- Published
- 2000
40. 14-3-3 Is Phosphorylated by Casein Kinase I on Residue 233
- Author
-
Ulrike Steinhussen, Thierry Dubois, Karin Moelling, Nick Morrice, Yasmina Soneji, Steven Howell, Alastair Aitken, and Christian Rommel
- Subjects
Biochemistry ,Chemistry ,Casein Kinase I ,Casein kinase 2, alpha 1 ,Cell Biology ,Mitogen-activated protein kinase kinase ,Casein kinase 2 ,Signal transduction ,Protein kinase A ,Casein kinases ,Molecular Biology ,Casein Kinase Ialpha - Abstract
14-3-3 proteins mediate interactions between proteins involved in signal transduction and cell cycle regulation. Phosphorylation of target proteins as well as 14-3-3 are important for protein-protein interactions. Here, we describe the purification of a protein kinase from porcine brain that phosphorylates 14-3-3 zeta on Thr-233. This protein kinase has been identified as casein kinase Ialpha (CKIalpha) by peptide mapping analysis and sequencing. Among mammalian 14-3-3, only 14-3-3 tau possesses a phosphorylatable residue at the same position (Ser-233), and we show that this residue is also phosphorylated by CKI. In addition, we show that 14-3-3 zeta is exclusively phosphorylated on Thr-233 in human embryonic kidney 293 cells. The residue 233 is located within a region shown to be important for the association of 14-3-3 to target proteins. We showed previously that, in 293 cells, only the unphosphorylated form of 14-3-3 zeta associates with the regulatory domain of c-Raf. We have now shown that in vivo phosphorylation of 14-3-3 zeta at the CKIalpha site (Thr-233) negatively regulates its binding to c-Raf, and may be important in Raf-mediated signal transduction.
- Published
- 1997
41. Selective inhibition of phosphoinositide 3-kinase p110α preserves lymphocyte function
- Author
-
Katti Jessen, Pingda Ren, Jean S. Oak, Michael B. Martin, Lomon So, Jeff M. Kucharski, Linda V. Kessler, Qiao Han Ke, Arun Manmadhan, Lian-Sheng Li, David A. Fruman, Sung Su Yea, Yi Liu, Matthew R. Janes, Mengrou Lu, and Christian Rommel
- Subjects
T cell ,T-Lymphocytes ,B-cell receptor ,Immunology ,Enzyme-Linked Immunosorbent Assay ,Biology ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Mice ,Cell Line, Tumor ,Neoplasms ,Marginal zone B-cell ,medicine ,Animals ,Humans ,Protein Isoforms ,Lymphocytes ,Enzyme Inhibitors ,Molecular Biology ,Protein kinase B ,B cell ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Mice, Inbred BALB C ,Phosphoinositide 3-kinase ,Germinal center ,Cell Biology ,Molecular biology ,Class Ia Phosphatidylinositol 3-Kinase ,medicine.anatomical_structure ,P110δ ,Drug Design ,biology.protein ,Calcium ,Immunosuppressive Agents ,Spleen ,Signal Transduction - Abstract
Class IA phosphoinositide 3-kinase (PI3K) is essential for clonal expansion, differentiation, and effector function of B and T lymphocytes. The p110δ catalytic isoform of PI3K is highly expressed in lymphocytes and plays a prominent role in B and T cell responses. Another class IA PI3K catalytic isoform, p110α, is a promising drug target in cancer but little is known about its function in lymphocytes. Here we used highly selective inhibitors to probe the function of p110α in lymphocyte responses in vitro and in vivo. p110α inhibition partially reduced B cell receptor (BCR)-dependent AKT activation and proliferation, and diminished survival supported by the cytokines BAFF and IL-4. Selective p110δ inhibition suppressed B cell responses much more strongly, yet maximal suppression was achieved by targeting multiple PI3K isoforms. In mouse and human T cells, inhibition of single class IA isoforms had little effect on proliferation, whereas pan-class I inhibition did suppress T cell expansion. In mice, selective p110α inhibition using the investigational agent MLN1117 (previously known as INK1117) did not disrupt the marginal zone B cell compartment and did not block T cell-dependent germinal center formation. In contrast, the selective p110δ inhibitor IC87114 strongly suppressed germinal center formation and reduced marginal zone B cell numbers, similar to a pan-class I inhibitor. These findings show that although acute p110α inhibition partially diminishes AKT activation, selective p110α inhibitors are likely to be less immunosuppressive in vivo compared with p110δ or pan-class I inhibitors.
- Published
- 2013
42. Phosphoinositide 3-kinase in Health and Disease : Volume 2
- Author
-
Christian Rommel, Bart Vanhaesebroeck, Peter K. Vogt, Christian Rommel, Bart Vanhaesebroeck, and Peter K. Vogt
- Subjects
- Cellular signal transduction, Protein kinases, Phosphoinositides--Physiological effect
- Abstract
PI3K has become a very intense area of research, with over 2000 publications on PI3K in PubMed for 2009 alone. The expectations for a therapeutic impact of intervention with PI3K activity are high, and progress in the clinical arena is being monitored by many. However, targeted therapies almost invariably encounter roadblocks, often exposing unresolved questions in the basic understanding of the target
- Published
- 2011
43. Direct interaction and N-terminal phosphorylation of c-Jun by c-Mil/Raf
- Author
-
Christian Rommel, Monika Niehof, Karin Moelling, and Gerald Radziwill
- Subjects
Cytoplasm ,Macromolecular Substances ,Proto-Oncogene Proteins c-jun ,Recombinant Fusion Proteins ,Coturnix ,In Vitro Techniques ,Protein Serine-Threonine Kinases ,Biology ,Peptide Mapping ,Proto-Oncogene Proteins ,Animals ,c-Raf ,Phosphorylation ,Binding site ,Protein kinase A ,Multidisciplinary ,Protein-Serine-Threonine Kinases ,Kinase ,c-jun ,Precipitin Tests ,Molecular biology ,Cell Compartmentation ,Proto-Oncogene Proteins c-raf ,Protein Binding ,Research Article - Abstract
c-Mil is the avian homologue of the mammalian serine/threonine kinase c-Raf-1. c-Mil/Raf is a mediator of signal transduction leading to gene expression via the c-Jun DNA-binding site, AP-1. Here we show that c-Mil immunopurified from MC29-virus-transformed quail fibroblasts phosphorylates c-Jun in vitro near its N terminus (Ser-63 and -73). Furthermore, the viral oncogene product Gag-Mil of the avian wild-type retrovirus MH2 phosphorylates c-Jun in vitro. A contribution by other known kinases phosphorylating c-Jun, such as the mitogen-activated protein kinases (MAPKs) and the c-Jun N-terminal kinases, was excluded by control reactions. c-Raf-1 and c-Jun directly interact in vitro as shown by various immobilized glutathione S-transferase-Raf fusion proteins which specify the cysteine-rich region of c-Mil/Raf as the major N-terminal binding site. An additional minor binding site is located in the C-terminal region. The biological relevance of these results is demonstrated by coimmunoprecipitation of c-Jun and c-Mil from 32P-labeled MC29- and MH2-transformed fibroblasts as well as normal quail embryo fibroblasts, whereby c-Jun was identified by tryptic phosphopeptide analysis. The complexed c-Jun exhibits a decreased electrophoretic mobility corresponding to a more highly phosphorylated state. Cell fractionation analyses indicate that the c-Mil/c-Jun complex is located in the cytoplasm. The data demonstrate that c-Jun can be a direct target of the protein kinase c-Mil/Raf, suggesting an alternative pathway, which leads to c-Jun phosphorylation independent of the MAPKs and MAPK-related proteins.
- Published
- 1995
44. Efficacy of the investigational mTOR kinase inhibitor MLN0128 / INK128 in models of B-cell acute lymphoblastic leukemia
- Author
-
Sharmila Mallya, Matthew R. Janes, Lian-Sheng Li, Katti Jessen, Michael B. Lilly, Yi Liu, David A. Fruman, Collin Vu, Leonard S. Sender, Michael B. Martin, Jose J. Limon, Christian Rommel, Marie P. Shieh, and Pingda Ren
- Subjects
Adult ,Male ,Cancer Research ,Mice, SCID ,Biology ,Philadelphia chromosome ,Article ,Colony-Forming Units Assay ,Mice ,In vivo ,Bone Marrow ,Mice, Inbred NOD ,hemic and lymphatic diseases ,medicine ,Medicine and Health Sciences ,Tumor Cells, Cultured ,Animals ,Humans ,Philadelphia Chromosome ,Mechanistic target of rapamycin ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Benzoxazoles ,Mice, Inbred BALB C ,Cell growth ,TOR Serine-Threonine Kinases ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Flow Cytometry ,Dasatinib ,Leukemia ,Disease Models, Animal ,medicine.anatomical_structure ,Pyrimidines ,Treatment Outcome ,Oncology ,Immunology ,Cancer research ,biology.protein ,Female ,Bone marrow ,medicine.drug - Abstract
The mechanistic target of rapamycin (mTOR) is a serine/threonine kinase whose activity contributes to leukemia proliferation and survival. Compounds targeting the mTOR active site inhibit rapamycin-resistant functions and have enhanced anti-cancer activity in mouse models. MLN0128 (formerly known as INK128) is a novel, orally active mTOR kinase inhibitor currently in clinical development. Here we evaluated MLN0128 in preclinical models of B-cell acute lymphoblastic leukemia (B-ALL). MLN0128 suppressed proliferation of B-ALL cell lines in vitro and reduced colony formation by primary human leukemia cells from adult and pediatric B-ALL patients. MLN0128 also boosted the efficacy of dasatinib in Philadelphia Chromosome-positive (Ph+) specimens. In a syngeneic mouse model of lymphoid BCR-ABL+ disease, daily oral dosing of MLN0128 rapidly cleared leukemic outgrowth. In primary xenografts of Ph+ B-ALL specimens, MLN0128 significantly enhanced the efficacy of dasatinib. In non-Ph B-ALL xenografts, single agent MLN0128 had a cytostatic effect that was most pronounced in mice with low disease burden. In all in vivo models, MLN0128 was well tolerated and did not suppress endogenous bone marrow proliferation. These findings support the rationale for clinical testing of MLN0128 in both adult and pediatric B-ALL and provide insight towards optimizing therapeutic efficacy of mTOR kinase inhibitors.
- Published
- 2012
45. Targeting of mTORC1/2 by the mTOR kinase inhibitor PP242 induces apoptosis in AML cells under conditions mimicking the bone marrow microenvironment
- Author
-
Wenbin Liu, Marina Konopleva, Yihua Qiu, Keith A. Baggerly, Michael Andreeff, Zhihong Zeng, Yue Xi Shi, David A. Fruman, Steven M. Kornblau, Katti Jessen, Christian Rommel, Hagop M. Kantarjian, Yi Liu, and Twee Tsao
- Subjects
Receptors, CXCR4 ,Indoles ,Immunology ,Blotting, Western ,Protein Array Analysis ,P70-S6 Kinase 1 ,Apoptosis ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Mice, SCID ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Real-Time Polymerase Chain Reaction ,Biochemistry ,mTORC2 ,Mice ,Bone Marrow ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,RNA, Messenger ,Phosphorylation ,Protein kinase B ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sirolimus ,Antibiotics, Antineoplastic ,Leukemia, Experimental ,Myeloid Neoplasia ,Reverse Transcriptase Polymerase Chain Reaction ,TOR Serine-Threonine Kinases ,RPTOR ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Flow Cytometry ,Coculture Techniques ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Purines ,Multiprotein Complexes ,Cancer research ,Bone marrow ,medicine.drug ,Signal Transduction - Abstract
The interactions between the bone marrow (BM) microenvironment and acute myeloid leukemia (AML) is known to promote survival of AML cells. In this study, we used reverse phase-protein array (RPPA) technology to measure changes in multiple proteins induced by stroma in leukemic cells. We then investigated the potential of an mTOR kinase inhibitor, PP242, to disrupt leukemia/stroma interactions, and examined the effects of PP242 in vivo using a mouse model. Using RPPA, we confirmed that multiple survival signaling pathways, including the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), were up-regulated in primary AML cells cocultured with stroma. PP242 effectively induced apoptosis in primary samples cultured with or without stroma. Mechanistically, PP242 attenuated the activities of mTORC1 and mTORC2, sequentially inhibited phosphorylated AKT, S6K, and 4EBP1, and concurrently suppressed chemokine receptor CXCR4 expression in primary leukemic cells and in stromal cells cultured alone or cocultured with leukemic cells. In the in vivo leukemia mouse model, PP242 inhibited mTOR signaling in leukemic cells and demonstrated a greater antileukemia effect than rapamycin. Our findings indicate that disrupting mTOR/AKT signaling with a selective mTOR kinase inhibitor can effectively target leukemic cells within the BM microenvironment.
- Published
- 2012
46. Investigational drug MLN0128, a novel TORC1/2 inhibitor, demonstrates potent oral antitumor activity in human breast cancer xenograft models
- Author
-
Yesim Gökmen-Polar, Rachel A. Toroni, George W. Sledge, Christian Rommel, Rutika Mehta, Sunil Badve, Kerry L. Sanders, and Yi Liu
- Subjects
Proto-Oncogene Proteins B-raf ,Vascular Endothelial Growth Factor A ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Class I Phosphatidylinositol 3-Kinases ,Mice, Nude ,Antineoplastic Agents ,Breast Neoplasms ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,Metastasis ,Mice ,Phosphatidylinositol 3-Kinases ,Breast cancer ,Internal medicine ,Cell Line, Tumor ,medicine ,PTEN ,Animals ,Humans ,Phosphorylation ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Benzoxazoles ,biology ,Neovascularization, Pathologic ,business.industry ,TOR Serine-Threonine Kinases ,PTEN Phosphohydrolase ,Cancer ,Endothelial Cells ,Proteins ,medicine.disease ,Primary tumor ,Xenograft Model Antitumor Assays ,Endocrinology ,Pyrimidines ,Oncology ,Multiprotein Complexes ,Mutation ,biology.protein ,Cancer research ,Female ,Breast disease ,business ,Proto-Oncogene Proteins c-akt - Abstract
Aberrant activation of the mammalian target of rapamycin (mTOR) signaling plays an important role in breast cancer progression and represents a potential therapeutic target for breast cancer. In this study, we report the impact of the investigational drug MLN0128, a potent and selective small molecule active-site TORC1/2 kinase inhibitor, on tumor growth and metastasis using human breast cancer xenograft models. We assessed in vitro antiproliferative activity of MLN0128 in a panel of breast cancer cell lines. We next evaluated the impact of MLN0128 on tumor growth, angiogenesis and metastasis using mammary fat pad xenograft models of a non-VEGF (ML20) and a VEGF-driven (MV165) MCF-7 sublines harboring PIK3CA mutations. MLN0128 potently inhibited cell proliferation in various breast cancer cell lines harboring PIK3CA (IC(50): 1.5-53 nM), PTEN (IC(50): 1-149 nM), KRAS, and/or BRAF mutations (IC(50): 13-162 nM), and in human endothelial cells (IC(50): 33-40 nM) in vitro. In vivo, MLN0128 decreased primary tumor growth significantly in both non-VEGF (ML20; p = 0.05) and VEGF-driven MCF-7 (MV165; p = 0.014) xenograft models. MLN0128 decreased the phosphorylation of Akt, S6, 4E-BP1, and NDRG1 in both models. In contrast, rapamycin increased Akt activity and failed to reduce the phosphorylation of 4E-BP1, PRAS40, and NDRG1. VEGF-induced lung metastasis in MV165 is inhibited by MLN0128 and rapamycin. In conclusion, MLN0128 inhibits TORC1/2-dependent signaling in preclinical models of breast cancer. MLN0128 appears to be superior in blocking mTORC1/2 signaling in contrast to rapamycin. Our findings support the clinical research of MLN0128 in patients with breast cancer and metastasis.
- Published
- 2012
47. PI3Kδ inhibitors in cancer: rationale and serendipity merge in the clinic
- Author
-
David A. Fruman and Christian Rommel
- Subjects
Gene isoform ,Tumor microenvironment ,Clinical Trials, Phase I as Topic ,business.industry ,Chronic lymphocytic leukemia ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,medicine.disease ,Lymphoma ,Clinical trial ,Leukemia ,Phosphatidylinositol 3-Kinases ,Oncology ,Purines ,Neoplasms ,Cancer cell ,Immunology ,medicine ,Cancer research ,Animals ,Humans ,business ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Quinazolinones - Abstract
Several phosphoinositide 3-kinase (PI3K) inhibitors are in the clinic and many more are in preclinical development. CAL-101, a selective inhibitor of the PI3Kδ isoform, has shown remarkable success in certain hematologic malignancies. Although PI3Kδ signaling plays a central role in lymphocyte biology, the degree of single-agent therapeutic activity of CAL-101 during early-phase development has been somewhat unexpected. CAL-101 works in part by blocking signals from the microenvironment that normally sustain leukemia and lymphoma cells in a protective niche. As PI3Ks enter the arena of molecular-targeted therapies, CAL-101 provides proof of principle that isoform-selective compounds can be effective in selected cancer types and patient populations. Significance: A key question is whether compounds targeting a single PI3K catalytic isoform can provide meaningful single agent efficacy in cancer cells that express multiple isoforms. Clinical studies of the drug CAL-101 have provided a significant advance by showing that selective targeting of PI3Kδ achieves efficacy in chronic lymphocytic leukemia, in part through targeting the tumor microenvironment. Cancer Discovery; 1(7); 562–72. ©2011 AACR.
- Published
- 2012
48. Defining the role of TORC1/2 in multiple myeloma
- Author
-
Michel B. Martin, Abdel Kareem Azab, Pingda Ren, Brittany Morgan, Yang Liu, Christian Rommel, Scott J. Rodig, Yong Zhang, Yi Liu, Irene M. Ghobrial, Charles P. Lin, Aldo M. Roccaro, Hai Ngo, Patricia Maiso, Phong Quang, Feda Azab, and Antonio Sacco
- Subjects
Stromal cell ,Immunology ,Blotting, Western ,Apoptosis ,Cell Cycle Proteins ,mTORC1 ,Mice, SCID ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Biochemistry ,Cell Line ,Mice ,Phosphatidylinositol 3-Kinases ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein kinase B ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Sirolimus ,Lymphoid Neoplasia ,Antibiotics, Antineoplastic ,Cell growth ,TOR Serine-Threonine Kinases ,Proteins ,Cell Biology ,Hematology ,Regulatory-Associated Protein of mTOR ,Phosphoproteins ,Xenograft Model Antitumor Assays ,Cell biology ,Rapamycin-Insensitive Companion of mTOR Protein ,Multiprotein Complexes ,RNA Interference ,Carrier Proteins ,Multiple Myeloma ,Proto-Oncogene Proteins c-akt ,medicine.drug ,Signal Transduction - Abstract
Mammalian target of rapamycin (mTOR) is a downstream serine/threonine kinase of the PI3K/Akt pathway that integrates signals from the tumor microenvironment to regulate multiple cellular processes. Rapamycin and its analogs have not shown significant activity in multiple myeloma (MM), likely because of the lack of inhibition of TORC2. In the present study, we investigated the baseline activity of the PI3K/Akt/mTOR pathway TORC1/2 in MM cell lines with different genetic abnormalities. TORC1/2 knock-down led to significant inhibition of the proliferation of MM cells, even in the presence of BM stromal cells. We also tested INK128, a dual TORC1/2 inhibitor, as a new therapeutic agent against these MM cell lines. We showed that dual TORC1/2 inhibition is much more active than TORC1 inhibition alone (rapamycin), even in the presence of cytokines or stromal cells. In vitro and in vivo studies showed that p-4EBP1 and p-Akt inhibition could be predictive markers of TORC2 inhibition in MM cell lines. Dual TORC1/2 inhibition showed better inhibition of adhesion to BM microenvironmental cells and inhibition of homing in vivo. These studies form the basis for further clinical testing of TORC1/2 inhibitors in MM.
- Published
- 2011
49. PI3 kinase δ is a key regulator of synoviocyte function in rheumatoid arthritis
- Author
-
Gary S. Firestein, Christian Rommel, Pingda Ren, Beatrix Bartok, William D. Bugbee, Scott T. Ball, David L. Boyle, and Yi Liu
- Subjects
Cell Survival ,Class I Phosphatidylinositol 3-Kinases ,medicine.medical_treatment ,Arthritis ,Apoptosis ,Gene Expression Regulation, Enzymologic ,Pathology and Forensic Medicine ,Arthritis, Rheumatoid ,Phosphatidylinositol 3-Kinases ,Transforming Growth Factor beta ,Osteoarthritis ,medicine ,Humans ,RNA, Messenger ,Enzyme Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Phosphoinositide-3 Kinase Inhibitors ,Platelet-Derived Growth Factor ,Phosphoinositide 3-kinase ,biology ,business.industry ,Growth factor ,Synovial Membrane ,Fibroblasts ,medicine.disease ,Immunology ,biology.protein ,Cancer research ,Cytokines ,Tumor necrosis factor alpha ,Inflammation Mediators ,business ,Proto-Oncogene Proteins c-akt ,Platelet-derived growth factor receptor ,Cell Division ,Transforming growth factor - Abstract
Class I phosphoinositide 3 kinase (PI3K) δ is a promising therapeutic target for rheumatoid arthritis (RA) because of its contribution to leukocyte biology. However, its contribution in fibroblasts has not been studied as a mechanism that contributes to efficacy. We investigated the expression and function of PI3Kδ in synovium and cultured fibroblast-like synoviocytes (FLS). Immunohistochemistry demonstrated that PI3Kδ is highly expressed in RA synovium, especially in the synovial lining. Using quantitative PCR and Western blot analysis, we found that PI3Kδ mRNA and protein expression is higher in RA than in osteoarthritis (OA) synovium. PI3Kδ was also expressed in cultured FLS, along with PI3Kα and PI3Kβ, whereas PI3Kγ was not detectable. PI3Kδ mRNA expression was selectively induced by inflammatory cytokines tumor necrosis factor (TNF) and interleukin-1 (IL-1) but not by growth factors platelet-derived growth factor (PDGF) and transforming growth factor β (TGFβ). The use of inhibitors that block individual PI3K isoforms, including the novel selective PI3Kδ inhibitor INK007, showed that PI3Kδ is required for PDGF- and TNF-induced Akt activation. PI3Kδ inhibition also diminished PDGF-mediated synoviocyte growth and sensitized cells to H 2 O 2 -induced apoptosis. These data are the first documentation of increased PI3Kδ expression in both RA synovium and cultured synoviocytes. Furthermore, these are the first data demonstrating that PI3Kδ is a major regulator of PDGF-mediated fibroblast growth and survival via Akt. Thus, targeting PI3Kδ in RA could modulate synoviocyte function via anti-inflammatory and disease-altering mechanisms.
- Published
- 2011
50. Phosphoinositide 3-kinase in Health and Disease
- Author
-
Peter K. Vogt, Christian Rommel, and Bart Vanhaesebroeck
- Subjects
Phosphoinositide 3-kinase ,Mediator ,Protein kinase domain ,Kinase ,Integrin ,biology.protein ,PTEN ,Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell biology - Abstract
PI3K - From the bench to the clinic and back.- Oncogenetic mutations of PIK3CA in human cancers.- Structural effects of oncogenic PI3Kalpha mutations.- Comparing the roles of the p110 and p110 isoforms of PI3K in signaling and cancer.- Phophatidylinositol 3-kinase (PI3K): the oncoprotein.- AKT Signaling in Physiology and Disease.- Faithfull modeling of PTEN loss driven diseases in the mouse.- PI3K as a target for therapy in haematological malignancies.- Clinical Development of Phosphatidylinositol-3 Kinase Pathway Inhibitors.- From the bench to the bed side: PI3K pathway inhibitors in clinical development.- New Inhibitors of the PI3K-Akt-mTOR Pathway: Insights into mTOR Signaling From a New Generation of Tor Kinase Domain Inhibitors (TORKinibs).- Small molecule inhibitors of the PI3-kinase family.- Targeting the RTK-PI3K-mTOR axis in malignant glioma: overcoming resistance.- Subject index Contents of sister volume 1: PI3K Book Introduction.- PDK1: The major transducer of PI 3-kinase actions.- Protein Kinase B (PKB/Akt), a key mediator of the PI3K signaling pathway.- Regulatory subunity of class IA PI3K.- The Regulation of Class IA PI 3-kinases by Inter-Subunit Interactions.- Phosphoinositide signalling pathways in metabolic regulation.- Role of RAS in the regulation of PI 3-kinase.- More than just kinases: the scaffolding function of PI3K.- PI3K signalling in neutrophils.- PI 3-kinase p110 Regulation of Platelet Integrin IIb 3.- PI3Ks in lymphocyte signaling and development.- the neurodevelopmental implication of PI3K signaling.- PI3kinase regulation of skeletal muscle hypertrophy and atrophy.- Taking PI3Kdelta and PI3Kgamma one step ahead - Dual active PI3Kdelta/gamma inhibotrs for the treatment of immune -mediated inflammatory diseases.- Subject index
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.