107 results on '"G. Draetta"'
Search Results
2. Down-regulation of the Notch pathway mediated by a gamma-secretase inhibitor induces anti-tumour effects in mouse models of T-cell leukaemia
- Author
-
J, Tammam, C, Ware, C, Efferson, J, O'Neil, S, Rao, X, Qu, J, Gorenstein, M, Angagaw, H, Kim, C, Kenific, K, Kunii, K J, Leach, G, Nikov, J, Zhao, X, Dai, J, Hardwick, M, Scott, C, Winter, L, Bristow, C, Elbi, J F, Reilly, T, Look, G, Draetta, Lht, Van der Ploeg, N E, Kohl, P R, Strack, and P K, Majumder
- Subjects
Membrane Potential, Mitochondrial ,Amyloid beta-Peptides ,Colon ,Transplantation, Heterologous ,Down-Regulation ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Cell Differentiation ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Research Papers ,Drug Administration Schedule ,Peptide Fragments ,Cyclic S-Oxides ,Mitochondrial Proteins ,Mice ,Cell Line, Tumor ,Thiadiazoles ,Animals ,Humans ,Amyloid Precursor Protein Secretases ,Intestinal Mucosa ,Receptor, Notch1 ,Neoplasm Transplantation ,Signal Transduction - Abstract
gamma-Secretase inhibitors (GSIs) block NOTCH receptor cleavage and pathway activation and have been under clinical evaluation for the treatment of malignancies such as T-cell acute lymphoblastic leukaemia (T-ALL). The ability of GSIs to decrease T-ALL cell viability in vitro is a slow process requiring8 days, however, such treatment durations are not well tolerated in vivo. Here we study GSI's effect on tumour and normal cellular processes to optimize dosing regimens for anti-tumour efficacy.Inhibition of the Notch pathway in mouse intestinal epithelium was used to evaluate the effect of GSIs and guide the design of dosing regimens for xenograft models. Serum Abeta(40) and Notch target gene modulation in tumours were used to evaluate the degree and duration of target inhibition. Pharmacokinetic and pharmacodynamic correlations with biochemical, immunohistochemical and profiling data were used to demonstrate GSI mechanism of action in xenograft tumours.Three days of70% Notch pathway inhibition was sufficient to provide an anti-tumour effect and was well tolerated. GSI-induced conversion of mouse epithelial cells to a secretory lineage was time- and dose-dependent. Anti-tumour efficacy was associated with cell cycle arrest and apoptosis that was in part due to Notch-dependent regulation of mitochondrial homeostasis.Intermittent but potent inhibition of Notch signalling is sufficient for anti-tumour efficacy in these T-ALL models. These findings provide support for the use of GSI in Notch-dependent malignancies and that clinical benefits may be derived from transient but potent inhibition of Notch.
- Published
- 2009
3. Transforming Growth Factor β1 Inhibition of p34cdc2 Phosphorylation and Histone H1 Kinase Activity Is Associated with G1/S-Phase Growth Arrest
- Author
-
P H, Howe, G, Draetta, and E B, Leof
- Subjects
Cell Cycle ,Epithelial Cells ,Cell Biology ,Phosphoproteins ,Epithelium ,Growth Inhibitors ,Cell Line ,Histones ,Mink ,Transforming Growth Factor beta ,CDC2 Protein Kinase ,Animals ,RNA Polymerase II ,Phosphorylation ,Molecular Biology ,Dichlororibofuranosylbenzimidazole ,Research Article - Abstract
Transforming growth factor beta 1 (TGF beta 1) is a potent inhibitor of epithelial cell proliferation. We present data which indicate that epithelial cell proliferation is inhibited when TGF beta 1 is added throughout the prereplicative G1 phase. Cultures become reversibly blocked in late G1 at the G1/S-phase boundary. The inhibitory effects of TGF beta 1 on cell growth occur in the presence of the RNA synthesis inhibitor 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole. Associated with this inhibitory effect is a decrease in the phosphorylation and histone H1 kinase activity of the p34cdc2 protein kinase. These data suggest that TGF beta 1 growth inhibition in epithelial cells involves the regulation of p34cdc2 activity at the G1/S transition.
- Published
- 1991
- Full Text
- View/download PDF
4. The cell cycle inhibitor p27 is an independent prognostic marker in small (T1a,b) invasive breast carcinomas
- Author
-
P, Tan, B, Cady, M, Wanner, P, Worland, B, Cukor, C, Magi-Galluzzi, P, Lavin, G, Draetta, M, Pagano, and M, Loda
- Subjects
Receptor, ErbB-2 ,Tumor Suppressor Proteins ,Age Factors ,Breast Neoplasms ,Cell Cycle Proteins ,Middle Aged ,Prognosis ,Immunohistochemistry ,Cyclin-Dependent Kinases ,Ki-67 Antigen ,Receptors, Estrogen ,Lymphatic Metastasis ,Biomarkers, Tumor ,Phosphoprotein Phosphatases ,Humans ,cdc25 Phosphatases ,Female ,Genes, Tumor Suppressor ,Tumor Suppressor Protein p53 ,Receptors, Progesterone ,Microtubule-Associated Proteins ,Cyclin-Dependent Kinase Inhibitor p27 ,In Situ Hybridization ,Retrospective Studies - Abstract
Breast carcinomasor = 1 cm in size (T1a,b) are being detected more frequently as a result of screening. Because traditional prognostic parameters are either lacking (tumor size) or rare (nodal metastases), a marker(s) is needed to identify the subset of patients who could benefit from adjuvant therapy. A retrospective series of 202 patients with stage T1a,b invasive breast carcinomas was evaluated. The clinicopathological features (age, histological grade, extensive in situ carcinoma, hormone receptor status, and nodal metastasis) as well as microvessel density and the expression of c-erb-B2, p53, MIB-1/Ki-67, and cdc25B were assessed. In addition, expression of the cell cycle inhibitor p27 was evaluated. Nineteen patients (18% of patients who had axillary dissection) had locoregional lymph node metastases. Forty-two % of them died of disease (median survival, 112 months), whereas mortality was 11% in node-negative patients (median survival, 168 months; P = 0.0055). Patients with low p27 expression had a median survival of 139 months (17% mortality) versus 174 months (9% mortality) in the group with high p27 expression (P = 0.0233). Lack of p27 was associated with poor prognosis when node-positive patients were excluded (P = 0.0252). Nodal status and low p27 were found to be the only independent prognostic parameters by both univariate and multivariate analysis, with relative risks of dying of disease of 4.9 (P = 0.001) and 3.4 (P = 0.0306), respectively. Assessment of p27, which yields prognostic information in node-negative patients, could be useful to identify patients with small, invasive breast carcinomas who might benefit from adjuvant therapy.
- Published
- 1997
5. Molecular cloning of PISSLRE, a novel putative member of the cdk family of protein serine/threonine kinases
- Author
-
R, Brambilla and G, Draetta
- Subjects
DNA, Complementary ,Base Sequence ,Sequence Homology, Amino Acid ,Molecular Sequence Data ,Tumor Cells, Cultured ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Protein Kinases ,Cyclin-Dependent Kinases - Abstract
Several members of the cdk family of protein kinases are involved in the regulation of the eukaryotic cell cycle. Using a PCR-based strategy we have screened different human tumor cell lines for cdk-related cDNAs. One clone isolated from the bladder carcinoma cell line RT112 encodes a novel protein kinase named PISSLRE, based on its predicted sequence at the conserved PSTAIRE motif. PISSLRE showed 50% amino acid identity with the previously isolated p58KGTA. PISSLRE contained all the structural elements featured by cyclin dependent kinases, including a proline in the PSTAIRE motif, which might be important for cyclin binding. PISSLRE was found expressed as 2.0 kb messenger RNA in a variety of human cell lines. Its expression was not restricted to tumor cells as it was detectable also in normal fibroblasts. In adult tissues, PISSLRE mRNA showed the highest expression in lung, liver and kidney. The broad expression pattern in adult tissues might suggest that PISSLRE could be involved in processes distinct from cell proliferation.
- Published
- 1994
6. Cyclin D1 protein oscillates and is essential for cell cycle progression in human tumour cell lines
- Author
-
J, Lukas, M, Pagano, Z, Staskova, G, Draetta, and J, Bartek
- Subjects
Oncogene Proteins ,Cyclins ,Cell Cycle ,Tumor Cells, Cultured ,Antibodies, Monoclonal ,Humans ,Cyclin D1 ,Immunohistochemistry ,Proto-Oncogene Mas ,Cells, Cultured ,Subcellular Fractions - Abstract
Among the key cell cycle regulators, cyclin D1 has been implicated most strongly in oncogenesis. This G1 cyclin is a putative proto-oncogene whose clonal rearrangement and/or amplification and mRNA overexpression occurs in several types of human neoplasias. We have now raised a series of monoclonal antibodies to human cyclin D1 and analysed its regulation at the protein level in 40 human tumour cell lines. We found that 12 cell lines displayed low or undetectable cyclin D1 protein level, while the remaining lines accumulated the protein to a level comparable to, or moderately higher than, that of four normal diploid non-immortalized cell types. The cell cycle-dependent oscillation and subcellular localization of cyclin D1 were similar in both tumour and normal cells. The protein localized to the nucleus of G1 cells, and it was reduced to immunocytochemically undetectable level in DNA-replicating cells. At the functional level, microinjection and electroporation of anti-D1 antibodies revealed that in most tumour cell lines studied, including those with amplification at the cyclin D1 locus, this cyclin is essential for cell cycle progression in G1. Some tumours, however, seem to have evolved mechanism(s) that enable them to bypass the requirement for functional cyclin D1.
- Published
- 1994
7. Modifications of cell cycle controlling nuclear proteins by transforming growth factor beta in the HaCaT keratinocyte cell line
- Author
-
Y, Landesman, M, Pagano, G, Draetta, V, Rotter, N E, Fusenig, and A, Kimchi
- Subjects
Keratinocytes ,Proto-Oncogene Proteins c-myc ,Gene Expression Regulation ,Transforming Growth Factor beta ,Cyclins ,Cell Cycle ,Immunoblotting ,Nuclear Proteins ,Tumor Suppressor Protein p53 ,Blotting, Northern ,Phosphoproteins ,Protein Kinases ,Cell Line - Published
- 1992
8. cdc2 protein kinase: structure-function relationships
- Author
-
M J, Marcote, M, Pagano, and G, Draetta
- Subjects
Structure-Activity Relationship ,Cyclins ,CDC2 Protein Kinase ,Cell Cycle ,Humans - Abstract
Activation of the cdc2 kinase in the cell cycle occurs upon binding to a regulatory subunit called cyclin. Cyclin A associates with both Cdc2 and its homologue Cdk2. The two complexes appear in S phase but cyclin A/Cdk2 is activated earlier than cyclin A/Cdc2. Several regions in Cdc2 are involved in binding cyclins A and B. Phosphorylation of cyclin/Cdk complexes ensures that the kinase activity peaks at a specific time in the cell cycle. Phosphorylation of Thr161 in Cdc2 is required for strong cyclin binding and kinase activity in vitro; its dephosphorylation is necessary for cells to exit mitosis. We have identified a novel 'Activating factor' that stimulates binding between cyclin and Cdc2 by inducing phosphorylation of Cdc2 on Thr161. We propose that Thr161 is targeted by an additional cell cycle regulatory pathway.
- Published
- 1992
9. Effect of the ribosome-inactivating protein saporin 6 on gene expression and kinase activity in leukemic cells
- Author
-
G. Saglio, M. Pagano, A. Gasperi-Campani, L. Roncuzzi, and G. Draetta
- Subjects
NFATC2 ,Oncology ,Saporin ,Ribosome-inactivating protein ,RUNX1T1 ,biology.protein ,Biology ,Autophagy-related protein 13 ,Kinase activity ,MAP3K7 ,Molecular biology ,GPS2 ,Cell biology - Published
- 1991
- Full Text
- View/download PDF
10. [Experimental research on the toxicity of PVC administered orally]
- Author
-
G, Faa, E, Dessy, A, Bucarelli, G, Frau, G, Zucca, and G, Draetta
- Subjects
Hyperplasia ,Gastric Mucosa ,Administration, Oral ,Animals ,Polyvinyls ,Intestinal Mucosa ,Polyvinyl Chloride ,Rats - Abstract
The AA. report the results of two experiments carried out on 24 rats Wistar treated with PVC dust ingestion. A group received 1 g of PVC dust in distilled water for only 15 days; the second group received PVC dust mixed with standard food almost 5 months till to 15 months. Pathological findings consist in marked hyperplasia of gastro-enteric mucosa, with polipoidfigures, and in a marked activation of pulmonary linforeticular tissue with cellular atipias. In one case a subcutaneous histiocytic lymphoma was observed.
- Published
- 1981
11. [Tumors of the large intestine. Statistical research and histopathological findings]
- Author
-
G, Zucca, E, Dessy, G, Faa, and G, Draetta
- Subjects
Male ,Rectal Neoplasms ,Carcinoma ,Age Factors ,Carcinoid Tumor ,Cecal Neoplasms ,Adenocarcinoma ,Middle Aged ,Adenocarcinoma, Mucinous ,Sex Factors ,Appendiceal Neoplasms ,Italy ,Colonic Neoplasms ,Intestinal Neoplasms ,Humans ,Female ,Intestine, Large ,Aged - Published
- 1980
12. Purification of calmodulin-stimulated phosphodiesterase by affinity chromatography on calmodulin fragment 1-77 linked to sepharose
- Author
-
G, Draetta and C B, Klee
- Subjects
Male ,Chromatography ,Chromatography, Ion Exchange ,Chromatography, Affinity ,Peptide Fragments ,Kinetics ,Durapatite ,Calmodulin ,3',5'-Cyclic-AMP Phosphodiesterases ,Testis ,Animals ,Cattle ,Female ,Indicators and Reagents ,Trypsin ,Hydroxyapatites ,Chromatography, High Pressure Liquid - Published
- 1988
13. [Malignant mesenchymal tumors of the larynx]
- Author
-
G, Draetta, M L, Pintus, G, Santa Cruz, and G, Pinto
- Subjects
Male ,Histiocytoma, Benign Fibrous ,Fibrosarcoma ,Lymphoma, Non-Hodgkin ,Humans ,Mesenchymoma ,Lymphoma, Large B-Cell, Diffuse ,Middle Aged ,Laryngeal Neoplasms ,Aged - Published
- 1980
14. p34, a protein kinase involved in cell cycle regulation in eukaryotic cells
- Author
-
G, Draetta, L, Brizuela, and D, Beach
- Subjects
CDC2 Protein Kinase ,Cell Cycle ,Genes, Fungal ,Schizosaccharomyces ,Humans ,Saccharomyces cerevisiae ,Phosphoproteins ,Models, Biological ,Protein Kinases - Published
- 1988
15. [Hamartochondromas of the lung. Case reports]
- Author
-
M, Pisano, G, Draetta, E, Valdes, F A, Serra, R, Versace, S, Furcas, and A I, Desogus
- Subjects
Adult ,Male ,Lung Neoplasms ,Hamartoma ,Humans ,Female ,Middle Aged ,Aged - Published
- 1982
16. The stereomorphologic disposition of the muscular capillary network. Experimental research into the effect of training, age, arterial insufficiency and chronic treatment with EPL
- Author
-
G, Brotzu, M, Carta, R, Cherchi, G, D'Alia, I P, Deriu, G, Diaz, G, Draetta, R, Montisci, G, Savona, and F, Testa Riva
- Subjects
Cell Nucleus ,Male ,Physical Education and Training ,Muscles ,Age Factors ,Phosphatidylcholines ,Animals ,Cell Count ,Female ,Aortic Valve Stenosis ,Capillaries ,Rats - Published
- 1984
17. Synthesis of p34, the mammalian homolog of the yeast cdc2+/CDC28 protein kinase, is stimulated during adenovirus-induced proliferation of primary baby rat kidney cells
- Author
-
G, Draetta, D, Beach, and E, Moran
- Subjects
Adenoviridae Infections ,Cell Cycle ,Immunologic Techniques ,Animals ,Tissue Distribution ,RNA, Messenger ,Protein Kinases ,Cells, Cultured ,Adenoviridae ,Cell Line ,Rats - Abstract
The homolog of the cdc2+/CDC28+ encoded protein kinases has previously been identified in HeLa cells by immunological methods. Here we have studied the distribution and synthesis of this 34 kd protein in rat tissues and cell lines. p34 was found in a variety of organs, including some such as brain that are not highly active in cell division. Kidney has very low levels of p34. However, proliferative activation of baby rat kidney cells with adenovirus caused rapid induction of p34 synthesis. Induction was dependent on the E1A gene of the virus but not E1B, and was not prevented by inhibition of cellular DNA synthesis with hydroxyurea. Increased synthesis of p34 is due, at least in part, to an increase in abundance of translatable p34 mRNA. These data are consistent with the possibility that p34 plays a role in cell division in higher vertebrates.
- Published
- 1988
18. Csk inhibition of c-Src activity requires both the SH2 and SH3 domains of Src
- Author
-
Sara A. Courtneidge, Manfred Koegl, G. Draetta, Giulio Superti-Furga, and S. Fumagalli
- Subjects
animal structures ,Protein Conformation ,Molecular Sequence Data ,Proto-Oncogene Proteins pp60(c-src) ,Peptide binding ,macromolecular substances ,Biology ,SH2 domain ,environment and public health ,General Biochemistry, Genetics and Molecular Biology ,SH3 domain ,Substrate Specificity ,CSK Tyrosine-Protein Kinase ,Schizosaccharomyces ,Amino Acid Sequence ,Cloning, Molecular ,Phosphorylation ,Molecular Biology ,Tyrosine-protein kinase CSK ,General Immunology and Microbiology ,General Neuroscience ,Wild type ,Protein-Tyrosine Kinases ,Cell biology ,Phenotype ,src-Family Kinases ,Biochemistry ,Mutation ,Tyrosine ,Genes, Lethal ,Peptides ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src ,Research Article - Abstract
The protein tyrosine kinase c-Src is negatively regulated by phosphorylation of Tyr527 in its carboxy-terminal tail. A kinase that phosphorylates Tyr527, called Csk, has recently been identified. We expressed c-Src in yeast to test the role of the SH2 and SH3 domains of Src in the negative regulation exerted by Tyr527 phosphorylation. Inducible expression of c-Src in Schizosaccharomyces pombe caused cell death. Co-expression of Csk counteracted this effect. Src proteins mutated in either the SH2 or SH3 domain were as lethal as wild type c-Src, but were insensitive to Csk, even though they were substrates for Csk in vivo. Peptide binding experiments revealed that Src proteins with mutant SH3 domains adopted a conformation in which the SH2 domain was not interacting with the tail. These data support the model of an SH2 domain-phosphorylated tail interaction repressing c-Src activity, but expand it to include a role for the SH3 domain. We propose that the SH3 domain contributes to the maintenance of the folded, inactive configuration of the Src molecule by stabilizing the SH2 domain-phosphorylated tail interaction. Moreover, the system we describe here allows for further study of the regulation of tyrosine kinases in a neutral background and in an organism amenable to genetic analysis.
19. Dissection of clonal heterogeneity unmasks pre-existing chemoresistance and new metabolic vulnerabilities in pancreatic cancer
- Author
-
Seth, Sahil, Li, Chieh-Yuan, Loponte, Sara, Ho, I-Lin, Corti, Denise, Sapio, Luigi, Poggetto, Edoardo Del, Peoples, Michael, Karpinets, Tatiana, Robinson, Frederick S., Jiang, Shan, Dutta, Prasanta, Marszalek, Joseph, Francesco, Maria E. Di, Heffernan, Timothy P., Giuliani, Virginia, Bhattacharya, Pratip K., Genovese, Giannicola, Futreal, Andrew, Draetta, Giulio, Viale, Andrea, Carugo, Alessandro, S. Seth, C.Y. Li, S. Loponte, I.L. Ho, D. Corti, L. Sapio, E. Del Poggetto, M. Peoples, T. Karpinets, F.S. Robinson, S. Jiang, P. Dutta, J. Marszalek, M.E. Di Francesco, T.P. Heffernan, V. Giuliani, P.K. Bhattacharya, G. Genovese, A. Futreal, G. Draetta, A. Viale and A. Carugo, Seth, Sahil, Li, Chieh-Yuan, Loponte, Sara, Ho, I-Lin, Corti, Denise, Sapio, Luigi, Poggetto, Edoardo Del, Peoples, Michael, Karpinets, Tatiana, Robinson, Frederick S., Jiang, Shan, Dutta, Prasanta, Marszalek, Joseph, Francesco, Maria E. Di, Heffernan, Timothy P., Giuliani, Virginia, Bhattacharya, Pratip K., Genovese, Giannicola, Futreal, Andrew, Draetta, Giulio, Viale, Andrea, and Carugo, Alessandro
- Abstract
Adaptive drug-resistance mechanisms allow human tumors to evade treatment through selection and expansion of treatment-resistant clones. Modeling the functional heterogeneity of tumors can unmask critical contributions of distinct tumor cell sub-populations toward identifying rational drug combinations. Here, studying clonal evolution of tumor cells derived from human pancreatic tumors, we demonstrate that in vitro adherent cultures and in vivo tumors are maintained by a common set of long term self-renewing tumorigenic cells that can be used to establish clonal replica tumors (CRTs), large cohorts of animals bearing human tumors with identical clonal composition. Using CRTs to conduct quantitative assessments of clonal dynamics and adaptive responses to therapeutic challenge over time, we uncovered that the tumorigenic compartment of pancreatic tumors maintains a multitude of functionally heterogeneous subpopulations of cells with differential degrees of sensitivity to therapeutics. High-throughput isolation and deep characterization of unique clonal lineages showed genetic and transcriptomic diversity underlying the functionally diverse subpopulations, positioning the origins of tumor heterogeneity within the long-term self-renewing compartment. Molecular annotation of gemcitabine-naïve clonal lineages with distinct responses to treatment in the context of CRTs generated signatures that can predict the response to chemotherapy and exposed pre-existing functional mechanisms of clonal resistance, primarily associated to DNA damage tolerance and mitochondrial respiration (OXPHOS). Further transcriptomic and metabolic characterization of residual tumor cells in patient derived xenograft models as well as in patients after chemoradiation showed that resistant cells that contribute to tumor relapse are metabolically rewired to upregulate OXPHOS. Combining a novel inhibitor of oxidative phosphorylation (IACS-10759) developed at the MD Anderson Institute for Applied Cancer Science, and currently in phase I clinical trial in acute myeloid leukemia and solid tumors, with standard of care drugs drastically reduces tumor clonal complexity, underscoring the promise of inhibiting mitochondrial respiration as a new therapeutic strategy to prolong patient survival by eradicating resistant clones that survive chemoradiation. Our study, correlating genomic and transcriptomic traits with specific functional phenotypes, uncovered new mechanisms that underlie intra-tumor sub-clonal heterogeneity, influence treatment response to drugs and sustain tumor relapse.
- Published
- 2019
20. Clonal dominance defines metastatic dissemination in pancreatic cancer.
- Author
-
Ho IL, Li CY, Wang F, Zhao L, Liu J, Yen EY, Dyke CA, Shah R, Liu Z, Çetin AO, Chu Y, Citron F, Attanasio S, Corti D, Darbaniyan F, Del Poggetto E, Loponte S, Liu J, Soeung M, Chen Z, Jiang S, Jiang H, Inoue A, Gao S, Deem A, Feng N, Ying H, Kim M, Giuliani V, Genovese G, Zhang J, Futreal A, Maitra A, Heffernan T, Wang L, Do KA, Gargiulo G, Draetta G, Carugo A, Lin R, and Viale A
- Subjects
- Humans, Gene Expression Profiling, Transcriptome, Ecosystem, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology
- Abstract
Tumors represent ecosystems where subclones compete during tumor growth. While extensively investigated, a comprehensive picture of the interplay of clonal lineages during dissemination is still lacking. Using patient-derived pancreatic cancer cells, we created orthotopically implanted clonal replica tumors to trace clonal dynamics of unperturbed tumor expansion and dissemination. This model revealed the multifaceted nature of tumor growth, with rapid changes in clonal fitness leading to continuous reshuffling of tumor architecture and alternating clonal dominance as a distinct feature of cancer growth. Regarding dissemination, a large fraction of tumor lineages could be found at secondary sites each having distinctive organ growth patterns as well as numerous undescribed behaviors such as abortive colonization. Paired analysis of primary and secondary sites revealed fitness as major contributor to dissemination. From the analysis of pro- and nonmetastatic isogenic subclones, we identified a transcriptomic signature able to identify metastatic cells in human tumors and predict patients' survival.
- Published
- 2024
- Full Text
- View/download PDF
21. PGC1α/β Expression Predicts Therapeutic Response to Oxidative Phosphorylation Inhibition in Ovarian Cancer.
- Author
-
Ghilardi C, Moreira-Barbosa C, Brunelli L, Ostano P, Panini N, Lupi M, Anastasia A, Fiordaliso F, Salio M, Formenti L, Russo M, Arrigoni E, Chiaradonna F, Chiorino G, Draetta G, Marszalek JR, Vellano CP, Pastorelli R, Bani M, Decio A, and Giavazzi R
- Subjects
- Animals, Female, Humans, Mice, Mitochondria metabolism, Oxidation-Reduction, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Oxidative Phosphorylation, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, RNA-Binding Proteins metabolism
- Abstract
Ovarian cancer is the deadliest gynecologic cancer, and novel therapeutic options are crucial to improve overall survival. Here we provide evidence that impairment of oxidative phosphorylation (OXPHOS) can help control ovarian cancer progression, and this benefit correlates with expression of the two mitochondrial master regulators PGC1α and PGC1β. In orthotopic patient-derived ovarian cancer xenografts (OC-PDX), concomitant high expression of PGC1α and PGC1β (PGC1α/β) fostered a unique transcriptional signature, leading to increased mitochondrial abundance, enhanced tricarboxylic acid cycling, and elevated cellular respiration that ultimately conferred vulnerability to OXPHOS inhibition. Treatment with the respiratory chain complex I inhibitor IACS-010759 caused mitochondrial swelling and ATP depletion that consequently delayed malignant progression and prolonged the lifespan of high PGC1α/β-expressing OC-PDX-bearing mice. Conversely, low PGC1α/β OC-PDXs were not affected by IACS-010759, thus pinpointing a selective antitumor effect of OXPHOS inhibition. The clinical relevance of these findings was substantiated by analysis of ovarian cancer patient datasets, which showed that 25% of all cases displayed high PGC1α/β expression along with an activated mitochondrial gene program. This study endorses the use of OXPHOS inhibitors to manage ovarian cancer and identifies the high expression of both PGC1α and β as biomarkers to refine the selection of patients likely to benefit most from this therapy., Significance: OXPHOS inhibition in ovarian cancer can exploit the metabolic vulnerabilities conferred by high PGC1α/β expression and offers an effective approach to manage patients on the basis of PGC1α/β expression., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
22. Discovery of 6-[(3 S ,4 S )-4-Amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl]-3-(2,3-dichlorophenyl)-2-methyl-3,4-dihydropyrimidin-4-one (IACS-15414), a Potent and Orally Bioavailable SHP2 Inhibitor.
- Author
-
Czako B, Sun Y, McAfoos T, Cross JB, Leonard PG, Burke JP, Carroll CL, Feng N, Harris AL, Jiang Y, Kang Z, Kovacs JJ, Mandal P, Meyers BA, Mseeh F, Parker CA, Yu SS, Williams CC, Wu Q, Di Francesco ME, Draetta G, Heffernan T, Marszalek JR, Kohl NE, and Jones P
- Subjects
- Administration, Oral, Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Enzyme Inhibitors administration & dosage, Enzyme Inhibitors chemistry, Humans, Mice, Molecular Structure, Neoplasms, Experimental drug therapy, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Protein Tyrosine Phosphatase, Non-Receptor Type 11 metabolism, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Drug Discovery, Enzyme Inhibitors pharmacology, Protein Tyrosine Phosphatase, Non-Receptor Type 11 antagonists & inhibitors
- Abstract
Src homology 2 (SH2) domain-containing phosphatase 2 (SHP2) plays a role in receptor tyrosine kinase (RTK), neurofibromin-1 (NF-1), and Kirsten rat sarcoma virus (KRAS) mutant-driven cancers, as well as in RTK-mediated resistance, making the identification of small-molecule therapeutics that interfere with its function of high interest. Our quest to identify potent, orally bioavailable, and safe SHP2 inhibitors led to the discovery of a promising series of pyrazolopyrimidinones that displayed excellent potency but had a suboptimal in vivo pharmacokinetic (PK) profile. Hypothesis-driven scaffold optimization led us to a series of pyrazolopyrazines with excellent PK properties across species but a narrow human Ether-à-go-go-Related Gene (hERG) window. Subsequent optimization of properties led to the discovery of the pyrimidinone series, in which multiple members possessed excellent potency, optimal in vivo PK across species, and no off-target activities including no hERG liability up to 100 μM. Importantly, compound 30 (IACS-15414) potently suppressed the mitogen-activated protein kinase (MAPK) pathway signaling and tumor growth in RTK-activated and KRAS
mut xenograft models in vivo.- Published
- 2021
- Full Text
- View/download PDF
23. PRMT1-dependent regulation of RNA metabolism and DNA damage response sustains pancreatic ductal adenocarcinoma.
- Author
-
Giuliani V, Miller MA, Liu CY, Hartono SR, Class CA, Bristow CA, Suzuki E, Sanz LA, Gao G, Gay JP, Feng N, Rose JL, Tomihara H, Daniele JR, Peoples MD, Bardenhagen JP, Geck Do MK, Chang QE, Vangamudi B, Vellano C, Ying H, Deem AK, Do KA, Genovese G, Marszalek JR, Kovacs JJ, Kim M, Fleming JB, Guccione E, Viale A, Maitra A, Emilia Di Francesco M, Yap TA, Jones P, Draetta G, Carugo A, Chedin F, and Heffernan TP
- Subjects
- Animals, Biocatalysis drug effects, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal prevention & control, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Enzyme Inhibitors pharmacology, Female, Humans, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms prevention & control, Protein-Arginine N-Methyltransferases metabolism, RNA metabolism, RNA Interference, Repressor Proteins metabolism, Tumor Burden drug effects, Xenograft Model Antitumor Assays methods, Mice, Carcinoma, Pancreatic Ductal genetics, DNA Damage, Pancreatic Neoplasms genetics, Protein-Arginine N-Methyltransferases genetics, RNA genetics, Repressor Proteins genetics
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer that has remained clinically challenging to manage. Here we employ an RNAi-based in vivo functional genomics platform to determine epigenetic vulnerabilities across a panel of patient-derived PDAC models. Through this, we identify protein arginine methyltransferase 1 (PRMT1) as a critical dependency required for PDAC maintenance. Genetic and pharmacological studies validate the role of PRMT1 in maintaining PDAC growth. Mechanistically, using proteomic and transcriptomic analyses, we demonstrate that global inhibition of asymmetric arginine methylation impairs RNA metabolism, which includes RNA splicing, alternative polyadenylation, and transcription termination. This triggers a robust downregulation of multiple pathways involved in the DNA damage response, thereby promoting genomic instability and inhibiting tumor growth. Taken together, our data support PRMT1 as a compelling target in PDAC and informs a mechanism-based translational strategy for future therapeutic development.Statement of significancePDAC is a highly lethal cancer with limited therapeutic options. This study identified and characterized PRMT1-dependent regulation of RNA metabolism and coordination of key cellular processes required for PDAC tumor growth, defining a mechanism-based translational hypothesis for PRMT1 inhibitors., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
24. Targeting the αv integrin/TGF-β axis improves natural killer cell function against glioblastoma stem cells.
- Author
-
Shaim H, Shanley M, Basar R, Daher M, Gumin J, Zamler DB, Uprety N, Wang F, Huang Y, Gabrusiewicz K, Miao Q, Dou J, Alsuliman A, Kerbauy LN, Acharya S, Mohanty V, Mendt M, Li S, Lu J, Wei J, Fowlkes NW, Gokdemir E, Ensley EL, Kaplan M, Kassab C, Li L, Ozcan G, Banerjee PP, Shen Y, Gilbert AL, Jones CM, Bdiwi M, Nunez-Cortes AK, Liu E, Yu J, Imahashi N, Muniz-Feliciano L, Li Y, Hu J, Draetta G, Marin D, Yu D, Mielke S, Eyrich M, Champlin RE, Chen K, Lang FF, Shpall EJ, Heimberger AB, and Rezvani K
- Subjects
- Animals, Female, Glioblastoma genetics, Glioblastoma pathology, Glioblastoma therapy, Heterografts, Humans, Integrins genetics, Killer Cells, Natural pathology, Male, Mice, Neoplasm Proteins genetics, Neoplasm Transplantation, Neoplastic Stem Cells pathology, Receptor, Transforming Growth Factor-beta Type II genetics, Receptor, Transforming Growth Factor-beta Type II immunology, Transforming Growth Factor beta genetics, Glioblastoma immunology, Integrins immunology, Killer Cells, Natural immunology, Neoplasm Proteins immunology, Neoplastic Stem Cells immunology, Transforming Growth Factor beta immunology
- Abstract
Glioblastoma multiforme (GBM), the most aggressive brain cancer, recurs because glioblastoma stem cells (GSCs) are resistant to all standard therapies. We showed that GSCs, but not normal astrocytes, are sensitive to lysis by healthy allogeneic natural killer (NK) cells in vitro. Mass cytometry and single-cell RNA sequencing of primary tumor samples revealed that GBM tumor-infiltrating NK cells acquired an altered phenotype associated with impaired lytic function relative to matched peripheral blood NK cells from patients with GBM or healthy donors. We attributed this immune evasion tactic to direct cell-to-cell contact between GSCs and NK cells via αv integrin-mediated TGF-β activation. Treatment of GSC-engrafted mice with allogeneic NK cells in combination with inhibitors of integrin or TGF-β signaling or with TGFBR2 gene-edited allogeneic NK cells prevented GSC-induced NK cell dysfunction and tumor growth. These findings reveal an important mechanism of NK cell immune evasion by GSCs and suggest the αv integrin/TGF-β axis as a potentially useful therapeutic target in GBM.
- Published
- 2021
- Full Text
- View/download PDF
25. Discovery of IPN60090, a Clinical Stage Selective Glutaminase-1 (GLS-1) Inhibitor with Excellent Pharmacokinetic and Physicochemical Properties.
- Author
-
Soth MJ, Le K, Di Francesco ME, Hamilton MM, Liu G, Burke JP, Carroll CL, Kovacs JJ, Bardenhagen JP, Bristow CA, Cardozo M, Czako B, de Stanchina E, Feng N, Garvey JR, Gay JP, Do MKG, Greer J, Han M, Harris A, Herrera Z, Huang S, Giuliani V, Jiang Y, Johnson SB, Johnson TA, Kang Z, Leonard PG, Liu Z, McAfoos T, Miller M, Morlacchi P, Mullinax RA, Palmer WS, Pang J, Rogers N, Rudin CM, Shepard HE, Spencer ND, Theroff J, Wu Q, Xu A, Yau JA, Draetta G, Toniatti C, Heffernan TP, and Jones P
- Subjects
- Administration, Oral, Animals, Cell Line, Tumor, Dogs, Drug Evaluation, Preclinical, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacokinetics, Glutaminase genetics, Glutaminase metabolism, Half-Life, Hepatocytes cytology, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Inhibitory Concentration 50, Male, Mice, Microsomes metabolism, Protein Binding, Rats, Rats, Sprague-Dawley, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Structure-Activity Relationship, Triazoles chemistry, Triazoles metabolism, Enzyme Inhibitors chemistry, Glutaminase antagonists & inhibitors, Triazoles pharmacokinetics
- Abstract
Inhibition of glutaminase-1 (GLS-1) hampers the proliferation of tumor cells reliant on glutamine. Known glutaminase inhibitors have potential limitations, and in vivo exposures are potentially limited due to poor physicochemical properties. We initiated a GLS-1 inhibitor discovery program focused on optimizing physicochemical and pharmacokinetic properties, and have developed a new selective inhibitor, compound 27 (IPN60090), which is currently in phase 1 clinical trials. Compound 27 attains high oral exposures in preclinical species, with strong in vivo target engagement, and should robustly inhibit glutaminase in humans.
- Published
- 2020
- Full Text
- View/download PDF
26. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells.
- Author
-
Manzo T, Prentice BM, Anderson KG, Raman A, Schalck A, Codreanu GS, Nava Lauson CB, Tiberti S, Raimondi A, Jones MA, Reyzer M, Bates BM, Spraggins JM, Patterson NH, McLean JA, Rai K, Tacchetti C, Tucci S, Wargo JA, Rodighiero S, Clise-Dwyer K, Sherrod SD, Kim M, Navin NE, Caprioli RM, Greenberg PD, Draetta G, and Nezi L
- Subjects
- Acyl-CoA Dehydrogenase, Long-Chain biosynthesis, Acyl-CoA Dehydrogenase, Long-Chain genetics, Animals, CD8-Positive T-Lymphocytes pathology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Down-Regulation, Fatty Acids genetics, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Lymphocytes, Tumor-Infiltrating pathology, Mice, Mice, Mutant Strains, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Pancreas pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, CD8-Positive T-Lymphocytes metabolism, Carcinoma, Pancreatic Ductal metabolism, Fatty Acids metabolism, Lymphocytes, Tumor-Infiltrating metabolism, Pancreas metabolism, Pancreatic Neoplasms metabolism, Tumor Microenvironment
- Abstract
CD8+ T cells are master effectors of antitumor immunity, and their presence at tumor sites correlates with favorable outcomes. However, metabolic constraints imposed by the tumor microenvironment (TME) can dampen their ability to control tumor progression. We describe lipid accumulation in the TME areas of pancreatic ductal adenocarcinoma (PDA) populated by CD8+ T cells infiltrating both murine and human tumors. In this lipid-rich but otherwise nutrient-poor TME, access to using lipid metabolism becomes particularly valuable for sustaining cell functions. Here, we found that intrapancreatic CD8+ T cells progressively accumulate specific long-chain fatty acids (LCFAs), which, rather than provide a fuel source, impair their mitochondrial function and trigger major transcriptional reprogramming of pathways involved in lipid metabolism, with the subsequent reduction of fatty acid catabolism. In particular, intrapancreatic CD8+ T cells specifically exhibit down-regulation of the very-long-chain acyl-CoA dehydrogenase (VLCAD) enzyme, which exacerbates accumulation of LCFAs and very-long-chain fatty acids (VLCFAs) that mediate lipotoxicity. Metabolic reprogramming of tumor-specific T cells through enforced expression of ACADVL enabled enhanced intratumoral T cell survival and persistence in an engineered mouse model of PDA, overcoming one of the major hurdles to immunotherapy for PDA., Competing Interests: Disclosures: Dr. Manzo, Dr. Anderson, Dr. Bates, Dr. Greenberg, and Dr. Nezi reported a patent to US Application No. 62/756,467 pending. Dr. McLean reported a patent to US application pending. Our laboratory is a Waters Center of Innovation (Waters Corporation) and an Agilent Thought Leader laboratory. These relationships did not influence the research described in the present manuscript. Dr. Wargo reported "other" from Genentech, GlaxoSmithKline, BMS, Merck, Illumina, and personal fees from AstraZeneca outside the submitted work; in addition, Dr. Wargo had a patent to PCT/US17/53.717 issued, "MD Anderson." Dr. Greenberg reported grants from Juno Therapeutics and personal fees from Juno Therapeutics during the conduct of the study; personal fees from Rapt Therapeutics, Elpiscience, Celsius, and Nextech outside the submitted work; and had a patent to Juno Therapeutics licensed. Dr. Draetta reported personal fees from Biovelocita, Nurix, Blueprint Medicines, Frontier Medicines, Orionis Biosciences, Tessa Therapeutics, Helsinn, Forma Therapeutics, Symphogen, Alligator, Taiho Pharmaceutical Co., and FIRC Institute of Molecular Oncology outside the submitted work. No other disclosures were reported., (© 2020 Manzo et al.)
- Published
- 2020
- Full Text
- View/download PDF
27. Yap1 Activation Enables Bypass of Oncogenic Kras Addiction in Pancreatic Cancer.
- Author
-
Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, Zhong Y, Wu CJ, Sadanandam A, Hu B, Chang Q, Chu GC, Al-Khalil R, Jiang S, Xia H, Fletcher-Sananikone E, Lim C, Horwitz GI, Viale A, Pettazzoni P, Sanchez N, Wang H, Protopopov A, Zhang J, Heffernan T, Johnson RL, Chin L, Wang YA, Draetta G, and DePinho RA
- Published
- 2019
- Full Text
- View/download PDF
28. Pooled library screening with multiplexed Cpf1 library.
- Author
-
Liu J, Srinivasan S, Li CY, Ho IL, Rose J, Shaheen M, Wang G, Yao W, Deem A, Bristow C, Hart T, and Draetta G
- Subjects
- CRISPR-Associated Protein 9 genetics, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Gene Editing, Humans, RNA, Guide, CRISPR-Cas Systems chemistry, CRISPR-Cas Systems genetics, Gene Library
- Abstract
Capitalizing on the inherent multiplexing capability of AsCpf1, we developed a multiplexed, high-throughput screening strategy that minimizes library size without sacrificing gene targeting efficiency. We demonstrated that AsCpf1 can be used for functional genomics screenings and that an AsCpf1-based multiplexed library performs similarly as compared to currently available monocistronic CRISPR/Cas9 libraries, with only one vector required for each gene. We construct the smallest whole-genome CRISPR knock-out library, Mini-human, for the human genome (n = 17,032 constructs targeting 16,977 protein-coding genes), which performs favorably compared to conventional Cas9 libraries.
- Published
- 2019
- Full Text
- View/download PDF
29. Metabolic reprogramming toward oxidative phosphorylation identifies a therapeutic target for mantle cell lymphoma.
- Author
-
Zhang L, Yao Y, Zhang S, Liu Y, Guo H, Ahmed M, Bell T, Zhang H, Han G, Lorence E, Badillo M, Zhou S, Sun Y, Di Francesco ME, Feng N, Haun R, Lan R, Mackintosh SG, Mao X, Song X, Zhang J, Pham LV, Lorenzi PL, Marszalek J, Heffernan T, Draetta G, Jones P, Futreal A, Nomie K, Wang L, and Wang M
- Subjects
- Adenine analogs & derivatives, Animals, Cell Line, Tumor, DNA Copy Number Variations genetics, Drug Resistance, Neoplasm genetics, Lymphoma, Mantle-Cell genetics, Mice, Mutation genetics, Piperidines, Pyrazoles pharmacology, Pyrazoles therapeutic use, Pyrimidines pharmacology, Pyrimidines therapeutic use, Signal Transduction drug effects, Transcriptome genetics, Exome Sequencing, Lymphoma, Mantle-Cell drug therapy, Lymphoma, Mantle-Cell metabolism, Molecular Targeted Therapy, Oxidative Phosphorylation drug effects
- Abstract
Metabolic reprogramming is linked to cancer cell growth and proliferation, metastasis, and therapeutic resistance in a multitude of cancers. Targeting dysregulated metabolic pathways to overcome resistance, an urgent clinical need in all relapsed/refractory cancers, remains difficult. Through genomic analyses of clinical specimens, we show that metabolic reprogramming toward oxidative phosphorylation (OXPHOS) and glutaminolysis is associated with therapeutic resistance to the Bruton's tyrosine kinase inhibitor ibrutinib in mantle cell lymphoma (MCL), a B cell lymphoma subtype with poor clinical outcomes. Inhibition of OXPHOS with a clinically applicable small molecule, IACS-010759, which targets complex I of the mitochondrial electron transport chain, results in marked growth inhibition in vitro and in vivo in ibrutinib-resistant patient-derived cancer models. This work suggests that targeting metabolic pathways to subvert therapeutic resistance is a clinically viable approach to treat highly refractory malignancies., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
30. High-resolution clonal mapping of multi-organ metastasis in triple negative breast cancer.
- Author
-
Echeverria GV, Powell E, Seth S, Ge Z, Carugo A, Bristow C, Peoples M, Robinson F, Qiu H, Shao J, Jeter-Jones SL, Zhang X, Ramamoorthy V, Cai S, Wu W, Draetta G, Moulder SL, Symmans WF, Chang JT, Heffernan TP, and Piwnica-Worms H
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms secondary, Disease Models, Animal, Female, Humans, Liver Neoplasms genetics, Liver Neoplasms secondary, Lung Neoplasms genetics, Lung Neoplasms secondary, Mice, Mice, SCID, Neoplasm Metastasis pathology, Xenograft Model Antitumor Assays, Neoplasm Metastasis genetics, Triple Negative Breast Neoplasms complications, Triple Negative Breast Neoplasms genetics
- Abstract
Most triple negative breast cancers (TNBCs) are aggressively metastatic with a high degree of intra-tumoral heterogeneity (ITH), but how ITH contributes to metastasis is unclear. Here, clonal dynamics during metastasis were studied in vivo using two patient-derived xenograft (PDX) models established from the treatment-naive primary breast tumors of TNBC patients diagnosed with synchronous metastasis. Genomic sequencing and high-complexity barcode-mediated clonal tracking reveal robust alterations in clonal architecture between primary tumors and corresponding metastases. Polyclonal seeding and maintenance of heterogeneous populations of low-abundance subclones is observed in each metastasis. However, lung, liver, and brain metastases are enriched for an identical population of high-abundance subclones, demonstrating that primary tumor clones harbor properties enabling them to seed and thrive in multiple organ sites. Further, clones that dominate multi-organ metastases share a genomic lineage. Thus, intrinsic properties of rare primary tumor subclones enable the seeding and colonization of metastases in secondary organs in these models.
- Published
- 2018
- Full Text
- View/download PDF
31. Future cancer research priorities in the USA: a Lancet Oncology Commission.
- Author
-
Jaffee EM, Dang CV, Agus DB, Alexander BM, Anderson KC, Ashworth A, Barker AD, Bastani R, Bhatia S, Bluestone JA, Brawley O, Butte AJ, Coit DG, Davidson NE, Davis M, DePinho RA, Diasio RB, Draetta G, Frazier AL, Futreal A, Gambhir SS, Ganz PA, Garraway L, Gerson S, Gupta S, Heath J, Hoffman RI, Hudis C, Hughes-Halbert C, Ibrahim R, Jadvar H, Kavanagh B, Kittles R, Le QT, Lippman SM, Mankoff D, Mardis ER, Mayer DK, McMasters K, Meropol NJ, Mitchell B, Naredi P, Ornish D, Pawlik TM, Peppercorn J, Pomper MG, Raghavan D, Ritchie C, Schwarz SW, Sullivan R, Wahl R, Wolchok JD, Wong SL, and Yung A
- Subjects
- Biomedical Research methods, Forecasting, Humans, Medical Oncology trends, Neoplasms diagnosis, Precision Medicine trends, United States, Biomedical Research trends, Health Planning trends, Health Priorities, National Cancer Institute (U.S.) trends, Neoplasms therapy
- Abstract
We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US$2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. ILF2 Is a Regulator of RNA Splicing and DNA Damage Response in 1q21-Amplified Multiple Myeloma.
- Author
-
Marchesini M, Ogoti Y, Fiorini E, Aktas Samur A, Nezi L, D'Anca M, Storti P, Samur MK, Ganan-Gomez I, Fulciniti MT, Mistry N, Jiang S, Bao N, Marchica V, Neri A, Bueso-Ramos C, Wu CJ, Zhang L, Liang H, Peng X, Giuliani N, Draetta G, Clise-Dwyer K, Kantarjian H, Munshi N, Orlowski R, Garcia-Manero G, DePinho RA, and Colla S
- Subjects
- DNA Damage, DNA Repair, Homologous Recombination, Humans, Nuclear Factor 45 Protein genetics, Nuclear Factor 45 Protein metabolism, Splicing Factor U2AF metabolism, Tumor Cells, Cultured, Y-Box-Binding Protein 1 metabolism, Multiple Myeloma genetics, Nuclear Factor 45 Protein physiology, RNA Splicing genetics
- Abstract
Amplification of 1q21 occurs in approximately 30% of de novo and 70% of relapsed multiple myeloma (MM) and is correlated with disease progression and drug resistance. Here, we provide evidence that the 1q21 amplification-driven overexpression of ILF2 in MM promotes tolerance of genomic instability and drives resistance to DNA-damaging agents. Mechanistically, elevated ILF2 expression exerts resistance to genotoxic agents by modulating YB-1 nuclear localization and interaction with the splicing factor U2AF65, which promotes mRNA processing and the stabilization of transcripts involved in homologous recombination in response to DNA damage. The intimate link between 1q21-amplified ILF2 and the regulation of RNA splicing of DNA repair genes may be exploited to optimize the use of DNA-damaging agents in patients with high-risk MM., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
33. PRKCI promotes immune suppression in ovarian cancer.
- Author
-
Sarkar S, Bristow CA, Dey P, Rai K, Perets R, Ramirez-Cardenas A, Malasi S, Huang-Hobbs E, Haemmerle M, Wu SY, McGuire M, Protopopov A, Jiang S, Liu JF, Hirsch MS, Chang Q, Lazar AJ, Sood AK, Drapkin R, DePinho R, Draetta G, and Chin L
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Cell Cycle Proteins, Cell Movement genetics, Cytokines genetics, Female, Humans, Isoenzymes metabolism, Mice, Mice, Transgenic, Ovarian Neoplasms immunology, Ovarian Neoplasms physiopathology, Phosphoproteins metabolism, Protein Kinase C metabolism, T-Lymphocytes, Cytotoxic cytology, T-Lymphocytes, Cytotoxic immunology, Tumor Microenvironment immunology, Tumor Necrosis Factor-alpha metabolism, YAP-Signaling Proteins, Gene Expression Regulation, Neoplastic genetics, Immune Tolerance genetics, Isoenzymes genetics, Isoenzymes immunology, Ovarian Neoplasms genetics, Protein Kinase C genetics, Protein Kinase C immunology
- Abstract
A key feature of high-grade serous ovarian carcinoma (HGSOC) is frequent amplification of the 3q26 locus harboring PRKC-ι ( PRKCI ). Here, we show that PRKCI is also expressed in early fallopian tube lesions, called serous tubal intraepithelial carcinoma. Transgenic mouse studies establish PRKCI as an ovarian cancer-specific oncogene. Mechanistically, we show that the oncogenic activity of PRKCI relates in part to the up-regulation of TNFα to promote an immune-suppressive tumor microenvironment characterized by an abundance of myeloid-derived suppressor cells and inhibition of cytotoxic T-cell infiltration. Furthermore, system-level and functional analyses identify YAP1 as a downstream effector in tumor progression. In human ovarian cancers, high PRKCI expression also correlates with high expression of TNFα and YAP1 and low infiltration of cytotoxic T cells. The PRKCI-YAP1 regulation of the tumor immunity provides a therapeutic strategy for highly lethal ovarian cancer., (© 2017 Sarkar et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2017
- Full Text
- View/download PDF
34. Genomic deletion of malic enzyme 2 confers collateral lethality in pancreatic cancer.
- Author
-
Dey P, Baddour J, Muller F, Wu CC, Wang H, Liao WT, Lan Z, Chen A, Gutschner T, Kang Y, Fleming J, Satani N, Zhao D, Achreja A, Yang L, Lee J, Chang E, Genovese G, Viale A, Ying H, Draetta G, Maitra A, Wang YA, Nagrath D, and DePinho RA
- Subjects
- AMP-Activated Protein Kinases metabolism, Amino Acids, Branched-Chain metabolism, Animals, Biocatalysis, Carcinoma, Pancreatic Ductal enzymology, Carcinoma, Pancreatic Ductal psychology, Carcinoma, Pancreatic Ductal therapy, Humans, Ketoglutaric Acids metabolism, Malate Dehydrogenase genetics, Male, Mice, Minor Histocompatibility Antigens biosynthesis, Minor Histocompatibility Antigens genetics, Mitochondria enzymology, Mitochondria pathology, NADP biosynthesis, NADP metabolism, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms pathology, Pancreatic Neoplasms therapy, Pregnancy Proteins biosynthesis, Pregnancy Proteins genetics, Reactive Oxygen Species metabolism, Sterol Regulatory Element Binding Protein 1 metabolism, Transaminases biosynthesis, Transaminases genetics, Carcinoma, Pancreatic Ductal genetics, Gene Deletion, Malate Dehydrogenase deficiency, Pancreatic Neoplasms genetics
- Abstract
The genome of pancreatic ductal adenocarcinoma (PDAC) frequently contains deletions of tumour suppressor gene loci, most notably SMAD4, which is homozygously deleted in nearly one-third of cases. As loss of neighbouring housekeeping genes can confer collateral lethality, we sought to determine whether loss of the metabolic gene malic enzyme 2 (ME2) in the SMAD4 locus would create cancer-specific metabolic vulnerability upon targeting of its paralogous isoform ME3. The mitochondrial malic enzymes (ME2 and ME3) are oxidative decarboxylases that catalyse the conversion of malate to pyruvate and are essential for NADPH regeneration and reactive oxygen species homeostasis. Here we show that ME3 depletion selectively kills ME2-null PDAC cells in a manner consistent with an essential function for ME3 in ME2-null cancer cells. Mechanistically, integrated metabolomic and molecular investigation of cells deficient in mitochondrial malic enzymes revealed diminished NADPH production and consequent high levels of reactive oxygen species. These changes activate AMP activated protein kinase (AMPK), which in turn directly suppresses sterol regulatory element-binding protein 1 (SREBP1)-directed transcription of its direct targets including the BCAT2 branched-chain amino acid transaminase 2) gene. BCAT2 catalyses the transfer of the amino group from branched-chain amino acids to α-ketoglutarate (α-KG) thereby regenerating glutamate, which functions in part to support de novo nucleotide synthesis. Thus, mitochondrial malic enzyme deficiency, which results in impaired NADPH production, provides a prime 'collateral lethality' therapeutic strategy for the treatment of a substantial fraction of patients diagnosed with this intractable disease.
- Published
- 2017
- Full Text
- View/download PDF
35. Identification of potent and selective MTH1 inhibitors.
- Author
-
Petrocchi A, Leo E, Reyna NJ, Hamilton MM, Shi X, Parker CA, Mseeh F, Bardenhagen JP, Leonard P, Cross JB, Huang S, Jiang Y, Cardozo M, Draetta G, Marszalek JR, Toniatti C, Jones P, and Lewis RT
- Subjects
- DNA Repair Enzymes metabolism, Dose-Response Relationship, Drug, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Humans, Models, Molecular, Molecular Structure, Phosphoric Monoester Hydrolases metabolism, Structure-Activity Relationship, Substrate Specificity, DNA Repair Enzymes antagonists & inhibitors, Enzyme Inhibitors pharmacology, Phosphoric Monoester Hydrolases antagonists & inhibitors
- Abstract
Structure based design of a novel class of aminopyrimidine MTH1 (MutT homolog 1) inhibitors is described. Optimization led to identification of IACS-4759 (compound 5), a sub-nanomolar inhibitor of MTH1 with excellent cell permeability and good metabolic stability in microsomes. This compound robustly inhibited MTH1 activity in cells and proved to be an excellent tool for interrogation of the utility of MTH1 inhibition in the context of oncology., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
36. Structure-Guided Design of IACS-9571, a Selective High-Affinity Dual TRIM24-BRPF1 Bromodomain Inhibitor.
- Author
-
Palmer WS, Poncet-Montange G, Liu G, Petrocchi A, Reyna N, Subramanian G, Theroff J, Yau A, Kost-Alimova M, Bardenhagen JP, Leo E, Shepard HE, Tieu TN, Shi X, Zhan Y, Zhao S, Barton MC, Draetta G, Toniatti C, Jones P, Geck Do M, and Andersen JN
- Subjects
- Adaptor Proteins, Signal Transducing chemistry, Animals, Benzimidazoles pharmacokinetics, Carrier Proteins chemistry, DNA-Binding Proteins, Female, Humans, Methylation, Mice, Molecular Docking Simulation, Nuclear Proteins chemistry, Protein Binding, Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adaptor Proteins, Signal Transducing metabolism, Benzimidazoles chemistry, Benzimidazoles pharmacology, Carrier Proteins antagonists & inhibitors, Carrier Proteins metabolism, Drug Design, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins metabolism
- Abstract
The bromodomain containing proteins TRIM24 (tripartite motif containing protein 24) and BRPF1 (bromodomain and PHD finger containing protein 1) are involved in the epigenetic regulation of gene expression and have been implicated in human cancer. Overexpression of TRIM24 correlates with poor patient prognosis, and BRPF1 is a scaffolding protein required for the assembly of histone acetyltransferase complexes, where the gene of MOZ (monocytic leukemia zinc finger protein) was first identified as a recurrent fusion partner in leukemia patients (8p11 chromosomal rearrangements). Here, we present the structure guided development of a series of N,N-dimethylbenzimidazolone bromodomain inhibitors through the iterative use of X-ray cocrystal structures. A unique binding mode enabled the design of a potent and selective inhibitor 8i (IACS-9571) with low nanomolar affinities for TRIM24 and BRPF1 (ITC Kd = 31 nM and ITC Kd = 14 nM, respectively). With its excellent cellular potency (EC50 = 50 nM) and favorable pharmacokinetic properties (F = 29%), 8i is a high-quality chemical probe for the evaluation of TRIM24 and/or BRPF1 bromodomain function in vitro and in vivo.
- Published
- 2016
- Full Text
- View/download PDF
37. Development of novel cellular histone-binding and chromatin-displacement assays for bromodomain drug discovery.
- Author
-
Zhan Y, Kost-Alimova M, Shi X, Leo E, Bardenhagen JP, Shepard HE, Appikonda S, Vangamudi B, Zhao S, Tieu TN, Jiang S, Heffernan TP, Marszalek JR, Toniatti C, Draetta G, Tyler J, Barton M, Jones P, Palmer WS, Geck Do MK, and Andersen JN
- Abstract
Background: Proteins that 'read' the histone code are central elements in epigenetic control and bromodomains, which bind acetyl-lysine motifs, are increasingly recognized as potential mediators of disease states. Notably, the first BET bromodomain-based therapies have entered clinical trials and there is a broad interest in dissecting the therapeutic relevance of other bromodomain-containing proteins in human disease. Typically, drug development is facilitated and expedited by high-throughput screening, where assays need to be sensitive, robust, cost-effective and scalable. However, for bromodomains, which lack catalytic activity that otherwise can be monitored (using classical enzymology), the development of cell-based, drug-target engagement assays has been challenging. Consequently, cell biochemical assays have lagged behind compared to other protein families (e.g., histone deacetylases and methyltransferases)., Results: Here, we present a suite of novel chromatin and histone-binding assays using AlphaLISA, in situ cell extraction and fluorescence-based, high-content imaging. First, using TRIM24 as an example, the homogenous, bead-based AlphaScreen technology was modified from a biochemical peptide-competition assay to measure binding of the TRIM24 bromodomain to endogenous histone H3 in cells (AlphaLISA). Second, a target agnostic, high-throughput imaging platform was developed to quantify the ability of chemical probes to dissociate endogenous proteins from chromatin/nuclear structures. While overall nuclear morphology is maintained, the procedure extracts soluble, non-chromatin-bound proteins from cells with drug-target displacement visualized by immunofluorescence (IF) or microscopy of fluorescent proteins. Pharmacological evaluation of these assays cross-validated their utility, sensitivity and robustness. Finally, using genetic and pharmacological approaches, we dissect domain contribution of TRIM24, BRD4, ATAD2 and SMARCA2 to chromatin binding illustrating the versatility/utility of the in situ cell extraction platform., Conclusions: In summary, we have developed two novel complementary and cell-based drug-target engagement assays, expanding the repertoire of pharmacodynamic assays for bromodomain tool compound development. These assays have been validated through a successful TRIM24 bromodomain inhibitor program, where a micromolar lead molecule (IACS-6558) was optimized using cell-based assays to yield the first single-digit nanomolar TRIM24 inhibitor (IACS-9571). Altogether, the assay platforms described herein are poised to accelerate the discovery and development of novel chemical probes to deliver on the promise of epigenetic-based therapies.
- Published
- 2015
- Full Text
- View/download PDF
38. The SMARCA2/4 ATPase Domain Surpasses the Bromodomain as a Drug Target in SWI/SNF-Mutant Cancers: Insights from cDNA Rescue and PFI-3 Inhibitor Studies.
- Author
-
Vangamudi B, Paul TA, Shah PK, Kost-Alimova M, Nottebaum L, Shi X, Zhan Y, Leo E, Mahadeshwar HS, Protopopov A, Futreal A, Tieu TN, Peoples M, Heffernan TP, Marszalek JR, Toniatti C, Petrocchi A, Verhelle D, Owen DR, Draetta G, Jones P, Palmer WS, Sharma S, and Andersen JN
- Subjects
- Binding, Competitive, Catalysis, Cell Line, Tumor, Chromatin metabolism, Chromosomal Proteins, Non-Histone genetics, DNA Helicases chemistry, DNA Helicases deficiency, DNA, Complementary genetics, Gene Knockout Techniques, Genetic Complementation Test, Humans, Lung Neoplasms pathology, Microarray Analysis, Neoplasms genetics, Nuclear Proteins chemistry, Nuclear Proteins deficiency, Protein Structure, Tertiary, RNA Interference, RNA, Small Interfering pharmacology, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, Sarcoma, Synovial genetics, Sarcoma, Synovial pathology, Transcription Factors chemistry, Transcription Factors genetics, Azabicyclo Compounds pharmacology, Chromatin Assembly and Disassembly drug effects, Chromosomal Proteins, Non-Histone deficiency, DNA Helicases antagonists & inhibitors, Molecular Targeted Therapy, Neoplasm Proteins antagonists & inhibitors, Neoplasms drug therapy, Nuclear Proteins antagonists & inhibitors, Pyridines pharmacology, Transcription Factors antagonists & inhibitors, Transcription Factors deficiency
- Abstract
The SWI/SNF multisubunit complex modulates chromatin structure through the activity of two mutually exclusive catalytic subunits, SMARCA2 and SMARCA4, which both contain a bromodomain and an ATPase domain. Using RNAi, cancer-specific vulnerabilities have been identified in SWI/SNF-mutant tumors, including SMARCA4-deficient lung cancer; however, the contribution of conserved, druggable protein domains to this anticancer phenotype is unknown. Here, we functionally deconstruct the SMARCA2/4 paralog dependence of cancer cells using bioinformatics, genetic, and pharmacologic tools. We evaluate a selective SMARCA2/4 bromodomain inhibitor (PFI-3) and characterize its activity in chromatin-binding and cell-functional assays focusing on cells with altered SWI/SNF complex (e.g., lung, synovial sarcoma, leukemia, and rhabdoid tumors). We demonstrate that PFI-3 is a potent, cell-permeable probe capable of displacing ectopically expressed, GFP-tagged SMARCA2-bromodomain from chromatin, yet contrary to target knockdown, the inhibitor fails to display an antiproliferative phenotype. Mechanistically, the lack of pharmacologic efficacy is reconciled by the failure of bromodomain inhibition to displace endogenous, full-length SMARCA2 from chromatin as determined by in situ cell extraction, chromatin immunoprecipitation, and target gene expression studies. Furthermore, using inducible RNAi and cDNA complementation (bromodomain- and ATPase-dead constructs), we unequivocally identify the ATPase domain, and not the bromodomain of SMARCA2, as the relevant therapeutic target with the catalytic activity suppressing defined transcriptional programs. Taken together, our complementary genetic and pharmacologic studies exemplify a general strategy for multidomain protein drug-target validation and in case of SMARCA2/4 highlight the potential for drugging the more challenging helicase/ATPase domain to deliver on the promise of synthetic-lethality therapy., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
39. Redox-Mediated Suberoylanilide Hydroxamic Acid Sensitivity in Breast Cancer.
- Author
-
Chiaradonna F, Barozzi I, Miccolo C, Bucci G, Palorini R, Fornasari L, Botrugno OA, Pruneri G, Masullo M, Passafaro A, Galimberti VE, Fantin VR, Richon VM, Pece S, Viale G, Di Fiore PP, Draetta G, Pelicci PG, Minucci S, and Chiocca S
- Subjects
- Antineoplastic Agents metabolism, Breast Neoplasms genetics, Breast Neoplasms pathology, Buthionine Sulfoximine pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxamic Acids metabolism, Hydroxamic Acids toxicity, Oxidation-Reduction drug effects, Primary Cell Culture, Vorinostat, Antineoplastic Agents pharmacology, Breast Neoplasms metabolism, Drug Resistance, Neoplasm drug effects, Hydroxamic Acids pharmacology
- Abstract
Aims: Vorinostat (suberoylanilide hydroxamic acid; SAHA) is a histone deacetylase inhibitor (HDACi) approved in the clinics for the treatment of T-cell lymphoma and with the potential to be effective also in breast cancer. We investigated the responsiveness to SAHA in human breast primary tumors and cancer cell lines., Results: We observed a differential response to drug treatment in both human breast primary tumors and cancer cell lines. Gene expression analysis of the breast cancer cell lines revealed that genes involved in cell adhesion and redox pathways, especially glutathione metabolism, were differentially expressed in the cell lines resistant to SAHA compared with the sensitive ones, indicating their possible association with drug resistance mechanisms. Notably, such an association was also observed in breast primary tumors. Indeed, addition of buthionine sulfoximine (BSO), a compound capable of depleting cellular glutathione, significantly enhanced the cytotoxicity of SAHA in both breast cancer cell lines and primary breast tumors., Innovation: We identify and validate transcriptional differences in genes involved in redox pathways, which include potential predictive markers of sensitivity to SAHA., Conclusion: In breast cancer, it could be relevant to evaluate the expression of antioxidant genes that may favor tumor resistance as a factor to consider for potential clinical application and treatment with epigenetic drugs (HDACis).
- Published
- 2015
- Full Text
- View/download PDF
40. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer.
- Author
-
Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, Zhong Y, Wu CJ, Sadanandam A, Hu B, Chang Q, Chu GC, Al-Khalil R, Jiang S, Xia H, Fletcher-Sananikone E, Lim C, Horwitz GI, Viale A, Pettazzoni P, Sanchez N, Wang H, Protopopov A, Zhang J, Heffernan T, Johnson RL, Chin L, Wang YA, Draetta G, and DePinho RA
- Subjects
- Adenocarcinoma pathology, Animals, Carcinoma, Pancreatic Ductal pathology, Cell Cycle, Cell Cycle Proteins, Cell Line, Tumor, DNA Replication, DNA-Binding Proteins metabolism, Disease Models, Animal, E2F Transcription Factors metabolism, Humans, Mice, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins metabolism, TEA Domain Transcription Factors, Transcription Factors metabolism, YAP-Signaling Proteins, ras Proteins metabolism, Adaptor Proteins, Signal Transducing metabolism, Adenocarcinoma metabolism, Carcinoma, Pancreatic Ductal metabolism, Pancreatic Neoplasms metabolism, Phosphoproteins metabolism, Proto-Oncogene Proteins p21(ras) metabolism
- Abstract
Activating mutations in KRAS are among the most frequent events in diverse human carcinomas and are particularly prominent in human pancreatic ductal adenocarcinoma (PDAC). An inducible Kras(G12D)-driven mouse model of PDAC has established a critical role for sustained Kras(G12D) expression in tumor maintenance, providing a model to determine the potential for and the underlying mechanisms of Kras(G12D)-independent PDAC recurrence. Here, we show that some tumors undergo spontaneous relapse and are devoid of Kras(G12D) expression and downstream canonical MAPK signaling and instead acquire amplification and overexpression of the transcriptional coactivator Yap1. Functional studies established the role of Yap1 and the transcriptional factor Tead2 in driving Kras(G12D)-independent tumor maintenance. The Yap1/Tead2 complex acts cooperatively with E2F transcription factors to activate a cell cycle and DNA replication program. Our studies, along with corroborating evidence from human PDAC models, portend a novel mechanism of escape from oncogenic Kras addiction in PDAC., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
41. Oncology drug discovery: planning a turnaround.
- Author
-
Toniatti C, Jones P, Graham H, Pagliara B, and Draetta G
- Subjects
- Clinical Trials as Topic, Humans, Neoplasms pathology, Precision Medicine, Antineoplastic Agents therapeutic use, Drug Discovery methods, Neoplasms drug therapy
- Abstract
We have made remarkable progress in our understanding of the pathophysiology of cancer. This improved understanding has resulted in increasingly effective targeted therapies that are better tolerated than conventional cytotoxic agents and even curative in some patients. Unfortunately, the success rate of drug approval has been limited, and therapeutic improvements have been marginal, with too few exceptions. In this article, we review the current approach to oncology drug discovery and development, identify areas in need of improvement, and propose strategies to improve patient outcomes. We also suggest future directions that may improve the quality of preclinical and early clinical drug evaluation, which could lead to higher approval rates of anticancer drugs.
- Published
- 2014
- Full Text
- View/download PDF
42. PDK1 attenuation fails to prevent tumor formation in PTEN-deficient transgenic mouse models.
- Author
-
Ellwood-Yen K, Keilhack H, Kunii K, Dolinski B, Connor Y, Hu K, Nagashima K, O'Hare E, Erkul Y, Di Bacco A, Gargano D, Shomer NH, Angagaw M, Leccese E, Andrade P, Hurd M, Shin MK, Vogt TF, Northrup A, Bobkova EV, Kasibhatla S, Bronson RT, Scott ML, Draetta G, Richon V, Kohl N, Blume-Jensen P, Andersen JN, and Kraus M
- Subjects
- Animals, Gene Knockdown Techniques, Gene Silencing, Leukemia, Experimental enzymology, Leukemia, Experimental genetics, Male, Mice, Mice, Transgenic, Neoplasms, Experimental genetics, Oncogene Protein v-akt antagonists & inhibitors, Oncogene Protein v-akt metabolism, PTEN Phosphohydrolase genetics, Phosphorylation, Prostatic Neoplasms enzymology, Prostatic Neoplasms genetics, Protein Serine-Threonine Kinases genetics, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, RNA Interference, Neoplasms, Experimental enzymology, PTEN Phosphohydrolase deficiency, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
PDK1 activates AKT suggesting that PDK1 inhibition might suppress tumor development. However, while PDK1 has been investigated intensively as an oncology target, selective inhibitors suitable for in vivo studies have remained elusive. In this study we present the results of in vivo PDK1 inhibition through a universally applicable RNAi approach for functional drug target validation in oncogenic pathway contexts. This approach, which relies on doxycycline-inducible shRNA expression from the Rosa26 locus, is ideal for functional studies of genes like PDK1 where constitutive mouse models lead to strong developmental phenotypes or embryonic lethality. We achieved more than 90% PDK1 knockdown in vivo, a level sufficient to impact physiological functions resulting in hyperinsulinemia and hyperglycemia. This phenotype was reversible on PDK1 reexpression. Unexpectedly, long-term PDK1 knockdown revealed a lack of potent antitumor efficacy in 3 different mouse models of PTEN-deficient cancer. Thus, despite efficient PDK1 knockdown, inhibition of the PI3K pathway was marginal suggesting that PDK1 was not a rate limiting factor. Ex vivo analysis of pharmacological inhibitors revealed that AKT and mTOR inhibitors undergoing clinical development are more effective than PDK1 inhibitors at blocking activated PI3K pathway signaling. Taken together our findings weaken the widely held expectation that PDK1 represents an appealing oncology target., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
43. Met activation in non-small cell lung cancer is associated with de novo resistance to EGFR inhibitors and the development of brain metastasis.
- Author
-
Benedettini E, Sholl LM, Peyton M, Reilly J, Ware C, Davis L, Vena N, Bailey D, Yeap BY, Fiorentino M, Ligon AH, Pan BS, Richon V, Minna JD, Gazdar AF, Draetta G, Bosari S, Chirieac LR, Lutterbach B, and Loda M
- Subjects
- Brain Neoplasms drug therapy, Brain Neoplasms genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Enzyme Activation, ErbB Receptors genetics, Female, Humans, In Situ Hybridization, Fluorescence, Mutation, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-met genetics, Survival Rate, Brain Neoplasms secondary, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Drug Resistance, Neoplasm, ErbB Receptors antagonists & inhibitors, Proto-Oncogene Proteins c-met metabolism
- Abstract
Most non-small cell lung cancer (NSCLC) patients harboring activating epidermal growth factor receptor (EGFR) mutations respond to tyrosine kinase inhibitor (TKI) therapy. However, about 30% exhibit primary resistance to EGFR TKI therapy. Here we report that Met protein expression and phosphorylation were associated with primary resistance to EGFR TKI therapy in NSCLC patients harboring EGFR mutations, implicating Met as a de novo mechanism of resistance. In a separate patient cohort, Met expression and phosphorylation were also associated with development of NSCLC brain metastasis and were selectively enriched in brain metastases relative to paired primary lung tumors. A similar metastasis-specific activation of Met occurred in vitro in the isogenous cell lines H2073 and H1993, which are derived from the primary lung tumor and a metastasis, respectively, from the same patient. We conclude that Met activation is found in NSCLC before EGFR-targeted therapy and is associated with both primary resistance to EGFR inhibitor therapy and with the development of metastases. If confirmed in larger cohorts, our analysis suggests that patient tumors harboring both Met activation and EGFR mutation could potentially benefit from early intervention with a combination of EGFR and Met inhibitors.
- Published
- 2010
- Full Text
- View/download PDF
44. Down-regulation of the Notch pathway mediated by a gamma-secretase inhibitor induces anti-tumour effects in mouse models of T-cell leukaemia.
- Author
-
Tammam J, Ware C, Efferson C, O'Neil J, Rao S, Qu X, Gorenstein J, Angagaw M, Kim H, Kenific C, Kunii K, Leach KJ, Nikov G, Zhao J, Dai X, Hardwick J, Scott M, Winter C, Bristow L, Elbi C, Reilly JF, Look T, Draetta G, Van der Ploeg L, Kohl NE, Strack PR, and Majumder PK
- Subjects
- Amyloid beta-Peptides blood, Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Apoptosis, Cell Differentiation, Cell Line, Tumor, Colon cytology, Colon drug effects, Cyclic S-Oxides administration & dosage, Cyclic S-Oxides adverse effects, Down-Regulation, Drug Administration Schedule, Humans, Intestinal Mucosa cytology, Intestinal Mucosa drug effects, Membrane Potential, Mitochondrial drug effects, Mice, Mice, Nude, Mitochondrial Proteins biosynthesis, Mitochondrial Proteins genetics, Neoplasm Transplantation, Peptide Fragments blood, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Receptor, Notch1 genetics, Signal Transduction, Thiadiazoles administration & dosage, Thiadiazoles adverse effects, Transplantation, Heterologous, Amyloid Precursor Protein Secretases antagonists & inhibitors, Antineoplastic Agents pharmacology, Cyclic S-Oxides pharmacology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Receptor, Notch1 physiology, Thiadiazoles pharmacology
- Abstract
Background and Purpose: gamma-Secretase inhibitors (GSIs) block NOTCH receptor cleavage and pathway activation and have been under clinical evaluation for the treatment of malignancies such as T-cell acute lymphoblastic leukaemia (T-ALL). The ability of GSIs to decrease T-ALL cell viability in vitro is a slow process requiring >8 days, however, such treatment durations are not well tolerated in vivo. Here we study GSI's effect on tumour and normal cellular processes to optimize dosing regimens for anti-tumour efficacy., Experimental Approach: Inhibition of the Notch pathway in mouse intestinal epithelium was used to evaluate the effect of GSIs and guide the design of dosing regimens for xenograft models. Serum Abeta(40) and Notch target gene modulation in tumours were used to evaluate the degree and duration of target inhibition. Pharmacokinetic and pharmacodynamic correlations with biochemical, immunohistochemical and profiling data were used to demonstrate GSI mechanism of action in xenograft tumours., Key Results: Three days of >70% Notch pathway inhibition was sufficient to provide an anti-tumour effect and was well tolerated. GSI-induced conversion of mouse epithelial cells to a secretory lineage was time- and dose-dependent. Anti-tumour efficacy was associated with cell cycle arrest and apoptosis that was in part due to Notch-dependent regulation of mitochondrial homeostasis., Conclusions and Implications: Intermittent but potent inhibition of Notch signalling is sufficient for anti-tumour efficacy in these T-ALL models. These findings provide support for the use of GSI in Notch-dependent malignancies and that clinical benefits may be derived from transient but potent inhibition of Notch.
- Published
- 2009
- Full Text
- View/download PDF
45. FBW7 mutations in leukemic cells mediate NOTCH pathway activation and resistance to gamma-secretase inhibitors.
- Author
-
O'Neil J, Grim J, Strack P, Rao S, Tibbitts D, Winter C, Hardwick J, Welcker M, Meijerink JP, Pieters R, Draetta G, Sears R, Clurman BE, and Look AT
- Subjects
- Amino Acid Sequence, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid Precursor Protein Secretases metabolism, Apoptosis, Cell Cycle, Cell Line, Tumor, F-Box-WD Repeat-Containing Protein 7, Humans, Molecular Sequence Data, Protein Structure, Tertiary, RNA, Messenger metabolism, Receptors, Notch metabolism, Cell Cycle Proteins genetics, Enzyme Inhibitors pharmacology, F-Box Proteins genetics, Gene Expression Regulation, Neoplastic, Leukemia genetics, Leukemia metabolism, Mutation, Receptors, Notch genetics, Ubiquitin-Protein Ligases genetics
- Abstract
gamma-secretase inhibitors (GSIs) can block NOTCH receptor signaling in vitro and therefore offer an attractive targeted therapy for tumors dependent on deregulated NOTCH activity. To clarify the basis for GSI resistance in T cell acute lymphoblastic leukemia (T-ALL), we studied T-ALL cell lines with constitutive expression of the NOTCH intracellular domain (NICD), but that lacked C-terminal truncating mutations in NOTCH1. Each of the seven cell lines examined and 7 of 81 (8.6%) primary T-ALL samples harbored either a mutation or homozygous deletion of the gene FBW7, a ubiquitin ligase implicated in NICD turnover. Indeed, we show that FBW7 mutants cannot bind to the NICD and define the phosphodegron region of the NICD required for FBW7 binding. Although the mutant forms of FBW7 were still able to bind to MYC, they do not target it for degradation, suggesting that stabilization of both NICD and its principle downstream target, MYC, may contribute to transformation in leukemias with FBW7 mutations. In addition, we show that all seven leukemic cell lines with FBW7 mutations were resistant to the MRK-003 GSI. Most of these resistant lines also failed to down-regulate the mRNA levels of the NOTCH targets MYC and DELTEX1 after treatment with MRK-003, implying that residual NOTCH signaling in T-ALLs with FBW7 mutations contributes to GSI resistance.
- Published
- 2007
- Full Text
- View/download PDF
46. Apoptosis in T cell acute lymphoblastic leukemia cells after cell cycle arrest induced by pharmacological inhibition of notch signaling.
- Author
-
Lewis HD, Leveridge M, Strack PR, Haldon CD, O'neil J, Kim H, Madin A, Hannam JC, Look AT, Kohl N, Draetta G, Harrison T, Kerby JA, Shearman MS, and Beher D
- Subjects
- Cell Cycle drug effects, Cell Line, Tumor, Cell Survival drug effects, Cyclic S-Oxides pharmacology, Flow Cytometry, Humans, Leukemia-Lymphoma, Adult T-Cell enzymology, Leukemia-Lymphoma, Adult T-Cell metabolism, Receptors, Notch metabolism, Signal Transduction drug effects, Thiadiazoles pharmacology, Amyloid Precursor Protein Secretases antagonists & inhibitors, Apoptosis drug effects, Enzyme Inhibitors pharmacology, Leukemia-Lymphoma, Adult T-Cell drug therapy, Leukemia-Lymphoma, Adult T-Cell pathology, Receptors, Notch antagonists & inhibitors
- Abstract
In this report, inhibitors of the gamma-secretase enzyme have been exploited to characterize the antiproliferative relationship between target inhibition and cellular responses in Notch-dependent human T cell acute lymphoblastic leukemia (T-ALL) cell lines. Inhibition of gamma-secretase led to decreased Notch signaling, measured by endogenous NOTCH intracellular domain (NICD) formation, and was associated with decreased cell viability. Flow cytometry revealed that decreased cell viability resulted from a G(0)/G(1) cell cycle block, which correlated strongly to the induction of apoptosis. These effects associated with inhibitor treatment were rescued by exogenous expression of NICD and were not mirrored when a markedly less active enantiomer was used, demonstrating the gamma-secretase dependency and specificity of these responses. Together, these data strengthen the rationale for using gamma-secretase inhibitors therapeutically and suggest that programmed cell death may contribute to reduction of tumor burden in the clinic.
- Published
- 2007
- Full Text
- View/download PDF
47. Characterization of the BUD31 gene of Saccharomyces cerevisiae.
- Author
-
Masciadri B, Areces LB, Carpinelli P, Foiani M, Draetta G, and Fiore F
- Subjects
- Animals, Caenorhabditis elegans, Cell Division physiology, Cell Size, Gene Expression Regulation, Fungal physiology, Humans, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae growth & development, Saccharomyces cerevisiae Proteins genetics, Sequence Homology, Amino Acid, Species Specificity, Xenopus laevis, RNA Splicing physiology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins metabolism
- Abstract
A number of genes have been identified in the fully sequenced genome of Saccharomyces cerevisiae that appear to be conserved throughout evolution and the function of which remains poorly understood. In this manuscript we describe the initial characterization of yeast BUD31 gene. cDNA sequences highly related to BUD31 have been identified in human, Xenopus laevis, and Caenorhabditis elegans. With the aim of further understanding its function, we generated a BUD31-null yeast strain and characterized its phenotype: bud31l mutant cells showed severe cytoskeletal abnormalities, with dramatic effects on actin distribution and bud formation. We also proceeded to identify interacting proteins using the tandem affinity-purification method, coupled to mass spectrometry: Bud3lp was found in complex with proteins involved in mRNA splicing. We propose that the observed phenotypes for bud31-null strain could be the result of defective splicing and indicate a first functional role for Bud3lp and its homologs.
- Published
- 2004
- Full Text
- View/download PDF
48. Ras-GAP SH3 domain binding protein (G3BP) is a modulator of USP10, a novel human ubiquitin specific protease.
- Author
-
Soncini C, Berdo I, and Draetta G
- Subjects
- Amino Acid Sequence, Base Sequence, Carrier Proteins genetics, DNA Helicases, DNA, Complementary, Endopeptidases classification, Endopeptidases genetics, Humans, Molecular Sequence Data, Molecular Weight, Poly-ADP-Ribose Binding Proteins, RNA Helicases, RNA Recognition Motif Proteins, Ubiquitin Thiolesterase, Carrier Proteins metabolism, Endopeptidases metabolism, Ubiquitins metabolism
- Abstract
Degradation of cellular proteins through ubiquitination is a fundamental strategy for regulating biological pathways. De-ubiquitination, i.e. the removal of ubiquitin from proteins and peptides to which ubiquitin is attached, is catalyzed by processing proteases known as de-ubiquitinating enzymes. We are studying the biology of a family of de-ubiquitinating enzymes, the mammalian ubiquitin-specific proteases (USPs), some of which appear to play a role in growth control. Given the fact that the modes of regulation of USPs and of their substrate specificity are poorly understood, we decided to attempt the identification of USP interacting proteins. Using the yeast two-hybrid system (2HS), we have isolated a cDNA clone whose product specifically interacts with USP10 but not with other USP baits tested. The isolated clone encodes a protein known to interact with the Ras-GTPase activating protein (G3BP). This interaction was further confirmed by performing a 2HS with G3BP, which led to the isolation of USP10 encoding cDNAs. We validated the interaction between the two proteins by performing in vitro binding assays and immunoprecipitations in human cells. G3BP does not appear to be a substrate of USP10; it rather inhibits the ability of USP10 to disassemble ubiquitin chains. The USP10/G3BP complex appears to co-immunoprecipitate with ubiquitinated species that could be substrates of USP10.
- Published
- 2001
- Full Text
- View/download PDF
49. Down-regulation of p27 is associated with development of colorectal adenocarcinoma metastases.
- Author
-
Thomas GV, Szigeti K, Murphy M, Draetta G, Pagano M, and Loda M
- Subjects
- Adenocarcinoma metabolism, Aged, Colorectal Neoplasms metabolism, Cyclin-Dependent Kinase Inhibitor p27, Female, Humans, Immunoenzyme Techniques, Ki-67 Antigen metabolism, Liver Neoplasms metabolism, Male, Microtubule-Associated Proteins genetics, Middle Aged, Mutation, Neoplasm Staging, RNA, Messenger metabolism, Retrospective Studies, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Adenocarcinoma secondary, Cell Cycle Proteins, Colorectal Neoplasms pathology, Down-Regulation, Liver Neoplasms secondary, Microtubule-Associated Proteins metabolism, Tumor Suppressor Proteins
- Abstract
The cyclin-dependent kinase inhibitor p27 is a negative regulator of the cell cycle and a potential tumor suppressor gene. Because we had previously demonstrated that loss of p27 protein is associated with aggressive behavior in colorectal adenocarcinomas, we used immunohistochemistry and in situ hybridization to evaluate the potential role of alterations in p27 expression in primary and metastatic colorectal adenocarcinomas. Parallel immunostaining was performed for Ki-67 and p53. We evaluated 13 cases of metachronous and 23 cases of synchronous primary and metastatic colorectal tumor pairs. In the synchronous subgroup (Stage IV tumors), 57% of the primary tumor and metastases pairs did not express p27 protein and the remainder were low expressors. In the metachronous subgroup, 54% of the primary tumors were low expressors and the remainder high expressors of p27 protein. There was a significant reduction in the expression of p27 in the metachronous metastases (mean positive cells: 14.5%) when compared to the corresponding primary tumors (mean positive cells: 41.8%), P = 0.0023. All the primary and metastatic tumors in the metachronous subgroup showed high levels of p27 mRNA expression. There was no association between loss of p27 and either Ki-67 count or p53 expression. Because p27 is known to be up-regulated when epithelial cells are grown in suspension, the down-regulation of p27 in circulating tumor cells may confer the ability to grow in an environment of altered extracellular matrix or intercellular adhesion properties, two situations which may facilitate metastases.
- Published
- 1998
- Full Text
- View/download PDF
50. Cdc25 protein phosphatases in cell proliferation.
- Author
-
Draetta G and Eckstein J
- Subjects
- Animals, Cell Cycle Proteins metabolism, Cell Line, Cell Line, Transformed, Enzyme Activation, Humans, Models, Molecular, Phosphoprotein Phosphatases metabolism, Protein Tyrosine Phosphatases metabolism, cdc25 Phosphatases, Cell Cycle Proteins genetics, Cell Division genetics, Neoplasms enzymology, Phosphoprotein Phosphatases genetics
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.