173 results on '"Signal transduction inhibitor"'
Search Results
2. A phase I trial to determine safety and pharmacokinetics of ASLAN002, an oral MET superfamily kinase inhibitor, in patients with advanced or metastatic solid cancers.
- Author
-
Roohullah, Aflah, Cooper, Adam, Lomax, Anna J., Aung, Jennifer, Barge, Alan, Chow, Lilian, McHale, Mark, Desai, Jayesh, Whittle, James R., Tran, Ben, de Souza, Paul, and Horvath, Lisa G.
- Subjects
ATRIAL fibrillation ,CLINICAL trials ,CONSTIPATION ,DOSE-effect relationship in pharmacology ,FATIGUE (Physiology) ,METASTASIS ,NAUSEA ,ORAL drug administration ,TUMORS ,TUMOR classification ,PROTEIN-tyrosine kinase inhibitors ,TREATMENT effectiveness ,DISEASE incidence ,THERAPEUTICS - Abstract
Background The MET tyrosine kinase and its ligand, hepatocyte growth factor (HGF) also known as scatter factor, are associated with tumourigenesis and metastasis by promotion of scattering, proliferation, angiogenesis, motility and invasion. ASLAN-002 is a potent inhibitor of MET as well as related kinases. A phase I dose escalation study was conducted to determine the safety and pharmacokinetics of ASLAN-002 in patients with advanced cancer. Methods Patients with advanced or metastatic solid tumours, who had progressed on standard therapy or for whom standard therapy was not known, were administered ASLAN-002 orally. The starting dose was 100 mg once daily (QD) with subsequent cohorts to receive doses of 200 mg QD, 300 mg QD, 450 mg QD, 600 mg QD, 300 mg twice daily (BID), 450 mg BID, and 600 mg BID. Results Forty patients were included across 7 dose cohorts. Cohort 8 (600 mg BID) was not opened due to the lack of appreciable pharmacokinetic (PK) differences between 300 mg BID and 450 mg BID and higher incidences of grade 3 or 4 adverse events (AE) in Cohort 7 (450 mg BID). Fifteen patients (37.5%) experienced a grade 3 or 4 AE. The most commonly reported AEs were nausea (55%), fatigue (47.5%) and constipation (30%). One dose limiting toxicity (DLT) of atrial fibrillation was observed with 450 mg BID. Conclusions ASLAN-002 is well tolerated at 300 mg BID and is the recommended dose for future phase II studies (RP2D). Clinical Trials Registry Number:
NCT01721148 . [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
3. Anti-Inflammatory Drugs in the 21st Century
- Author
-
Rainsford, K.D., Harris, J. Robin, editor, Biswas, B. B., editor, Quinn, P., editor, Harris, Randall E., editor, Bittman, R., editor, Dasgupta, D., editor, Engelhardt, H., editor, Flohe, L., editor, Herrmann, H., editor, Holzenburg, A., editor, Nasheuer, H-P., editor, Rottem, S., editor, Wyss, M., editor, and Zwickl, P., editor
- Published
- 2007
- Full Text
- View/download PDF
4. Signal Transduction Inhibitors in Chronic Myeloid Leukemia
- Author
-
Deininger, Michael W. N., Melo, Junia V., and Goldman, John M.
- Published
- 2007
- Full Text
- View/download PDF
5. Innovative Treatment Strategies for High-Grade Gliomas
- Author
-
Conrad, Charles A., Heimberger, Amy B., Buzdar, Aman U., editor, Freedman, Ralph S., editor, DeMonte, Franco, editor, Gilbert, Mark R., editor, Mahajan, Anita, editor, and McCutcheon, Ian E., editor
- Published
- 2007
- Full Text
- View/download PDF
6. Signal Transduction Pathways : A Goldmine for Therapeutic Targets
- Author
-
Workman, Paul, Teicher, Beverly A., editor, Sebti, Saïd M., editor, and Hamilton, Andrew D., editor
- Published
- 2001
- Full Text
- View/download PDF
7. Cancer Drugs and Cancer Drug Development for the New Millennium
- Author
-
Dancey, J., Arbuck, S., Khayat, David, editor, and Hortobagyi, Gabriel N., editor
- Published
- 2000
- Full Text
- View/download PDF
8. A familial germline mutation in KIT associated with achalasia, mastocytosis and gastrointestinal stromal tumors shows response to kinase inhibitors
- Author
-
Alison L. Halpern, Robert J. Torphy, L. Michael Glode, Cosimo G. Sciotto, William A. Robinson, and Martin D. McCarter
- Subjects
Adult ,Male ,Cancer Research ,Gastrointestinal Stromal Tumors ,Signal transduction inhibitor ,medicine.disease_cause ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Child ,Molecular Biology ,Germ-Line Mutation ,Mutation ,GiST ,biology ,CD117 ,Infant ,Imatinib ,Exons ,Middle Aged ,digestive system diseases ,Pedigree ,Esophageal Achalasia ,Proto-Oncogene Proteins c-kit ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Signal transduction ,Mastocytosis ,medicine.drug - Abstract
Background Activating mutations of the tyrosine kinase receptor KIT have been described in both mastocytosis and gastrointestinal stromal tumors (GIST), but are usually found in separate domains and often respond differently to signal transduction inhibitors. We describe here a large family with both GIST, mastocytosis, and achalasia. Affected family members have a unique activating mutation in exon 9 of KIT which show promise to a novel signal transduction inhibitor. Methods Clinical data was collected from 15 family members, 7 of whom were variably affected with GIST, achalasia and mastocytosis. DNA was prepared from WBC of 12 subjects (6 affected and 6 unaffected) and exons 9, 11, 13 and 17 of KIT were amplified by PCR and directly sequenced. Results A unique activating single base pair mutation in the extracellular domain of KIT was found in all 6 affected subjects resulting in a K>I amino acid change at codon 509. Conclusions In the family reported here, a unique mutation in the extracellular domain leads to receptor activation resulting in GIST and mastocytosis as well as achalasia. Initial data suggests that this activation can be suppressed by signal transduction inhibitors and these patients may benefit from such therapy.
- Published
- 2019
9. High-Complexity shRNA Libraries and PI3 Kinase Inhibition in Cancer: High-Fidelity Synthetic Lethality Predictions
- Author
-
Laila Karra, Damia Romero-Moya, Yvonne Thus, Kevin Shannon, Jeroen P. Roose, Marthe F. Lindenbergh, Michael T. McManus, Anica M. Wandler, Matthew J. Hangauer, Marsilius Mues, and Olga Ksionda
- Subjects
0301 basic medicine ,Medical Physiology ,T-cell leukemia ,Signal transduction inhibitor ,Synthetic lethality ,Small hairpin RNA ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,shRNA ,PI3 kinase ,Neoplasms ,inhibitors ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Kinase ,leukemia ,Hematology ,Duvelisib ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Signal transduction ,Biotechnology ,Signal Transduction ,screen ,Biology ,Small Interfering ,vincristine ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Rare Diseases ,medicine ,cancer ,Animals ,Humans ,Protein Kinase Inhibitors ,GDC0941 ,Animal ,Cancer ,medicine.disease ,signaling pathways ,synthetic lethality ,Disease Models, Animal ,030104 developmental biology ,chemistry ,lcsh:Biology (General) ,Disease Models ,Cancer research ,RNA ,Biochemistry and Cell Biology ,preclinical mouse trials ,Synthetic Lethal Mutations ,030217 neurology & neurosurgery - Abstract
Summary: Deregulated signal transduction is a cancer hallmark, and its complexity and interconnectivity imply that combination therapy should be considered, but large data volumes that cover the complexity are required in user-friendly ways. Here, we present a searchable database resource of synthetic lethality with a PI3 kinase signal transduction inhibitor by performing a saturation screen with an ultra-complex shRNA library containing 30 independent shRNAs per gene target. We focus on Ras-PI3 kinase signaling with T cell leukemia as a screening platform for multiple clinical and experimental reasons. Our resource predicts multiple combination-based therapies with high fidelity, ten of which we confirmed with small molecule inhibitors. Included are biochemical assays, as well as the IPI145 (duvelisib) inhibitor. We uncover the mechanism of synergy between the PI3 kinase inhibitor GDC0941 (pictilisib) and the tubulin inhibitor vincristine and demonstrate broad synergy in 28 cell lines of 5 cancer types and efficacy in preclinical leukemia mouse trials. : Mues et al. present a web browser-based, searchable database of their synthetic lethal screen to identify a potentially potent combination therapy in cancer. They validated their screen with ten small molecule inhibitors in leukemia and four solid tumor types and in a T cell leukemia mouse model preclinical trial. Keywords: cancer, synthetic lethality, shRNA, screen, PI3 kinase, inhibitors, signaling pathways, leukemia, preclinical mouse trials, GDC0941, vincristine
- Published
- 2019
10. Effectively Intervening Epithelial-Mesenchymal Transition of Retinal Pigment Epithelial Cells With a Combination of ROCK and TGF-β Signaling Inhibitors
- Author
-
Junjie Luo, Jieping Zhang, Jingyao Chen, Jing-Ying Xu, Dandan Liu, Binxin Wu, Caixia Jin, Zi Wei Kang, Guo-Tong Xu, Chen Yi, Qingjian Ou, Jingfa Zhang, Haibin Tian, Xiaoxu Leng, Jian Feng He, Furong Gao, Kexin Fang, Lixia Lu, and Juan Wang
- Subjects
0301 basic medicine ,TGF-β ,Proliferative vitreoretinopathy ,Epithelial-Mesenchymal Transition ,Pyridines ,PVR ,Signal transduction inhibitor ,Retinal Pigment Epithelium ,Rats, Sprague-Dawley ,Transforming Growth Factor beta1 ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Dispase ,ROCK ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,Enzyme Inhibitors ,Cells, Cultured ,Retina ,medicine.diagnostic_test ,Vitreoretinopathy, Proliferative ,EMT ,Retinal ,medicine.disease ,Embryonic stem cell ,Amides ,eye diseases ,Cell biology ,Rats ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Retinal Cell Biology ,030220 oncology & carcinogenesis ,embryonic structures ,Pyrazoles ,sense organs ,RPE ,Electroretinography - Abstract
Purpose Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is a key pathological event in proliferative retinal diseases such as proliferative vitreoretinopathy (PVR). This study aimed to explore a new method to reverse EMT in RPE cells to develop an improved therapy for proliferative retinal diseases. Methods In vitro, human embryonic stem cell-derived RPE cells were passaged and cultured at low density for an extended period of time to establish an EMT model. At different stages of EMT after treatment with known molecules or combinations of molecules, the morphology was examined, transepithelial electrical resistance (TER) was measured, and expression of RPE- and EMT-related genes were examined with RT-PCR, Western blotting, and immunofluorescence. In vivo, a rat model of EMT in RPE cells was established via subretinal injection of dispase. Retinal function was examined by electroretinography (ERG), and retinal morphology was examined. Results EMT of RPE cells was effectively induced by prolonged low-density culture. After EMT occurred, only the combination of the Rho-associated coiled-coil containing protein kinase (ROCK) inhibitor Y27632 and the TGF-β receptor inhibitor RepSox (RY treatment) effectively suppressed and reversed the EMT process, even in cells in an intermediate state of EMT. In dispase-treated Sprague-Dawley rats, RY treatment maintained the morphology of RPE cells and the retina and preserved retinal function. Conclusions RY treatment might promote mesenchymal-epithelial transition (MET), the inverse process of EMT, to maintain the epithelial-like morphology and function of RPE cells. This combined RY therapy could be a new strategy for treating proliferative retinal diseases, especially those involving EMT of RPE cells.
- Published
- 2021
11. Folate decorated polymeric micelles for targeted delivery of the kinase inhibitor dactolisib to cancer cells
- Author
-
Shi, Haili, van Steenbergen, Mies J., Lou, Bo, Liu, Yanna, Hennink, Wim E., Kok, Robbert J., Shi, Haili, van Steenbergen, Mies J., Lou, Bo, Liu, Yanna, Hennink, Wim E., and Kok, Robbert J.
- Abstract
One of the main challenges in clinical translation of polymeric micelles is retention of the drug in the nanocarrier system upon its systemic administration. Core crosslinking and coupling of the drug to the micellar backbone are common strategies to overcome these issues. In the present study, polymeric micelles were prepared for tumor cell targeting of the kinase inhibitor dactolisib which inhibits both the mammalian Target of Rapamycin (mTOR) kinase and phosphatidylinositol-3-kinase (PI3K). We employed platinum(II)-based linker chemistry to couple dactolisib to the core of poly(ethylene glycol)-b-poly(acrylic acid) (PEG-b-PAA) polymeric micelles. The formed dactolisib-PEG-PAA unimers are amphiphilic and self-assemble in an aqueous milieu into core–shell polymeric micelles. Folate was conjugated onto the surface of the micelles to yield folate-decorated polymeric micelles which can target folate receptor over-expressing tumor cells. Fluorescently labeled polymeric micelles were prepared using a lissamine-platinum complex linked in a similar manner as dactolisib. Dactolisib polymeric micelles showed good colloidal stability in water and released the coupled drug in buffers containing chloride or glutathione. Folate decorated micelles were avidly internalized by folate-receptor-positive KB cells and displayed targeted cellular cytotoxicity at 50–75 nM IC50. In conclusion, we have prepared a novel type of folate-receptor targeted polymeric micelles in which platinum(II) linker chemistry modulates drug retention and sustained release of the coupled inhibitor dactolisib.
- Published
- 2020
12. Sensitivity of pancreatic cancer cells to chemotherapeutic drugs, signal transduction inhibitors and nutraceuticals can be regulated by WT-TP53
- Author
-
Lucio Cocco, Linda S. Steelman, Giuseppe Montalto, Shaw M. Akula, Stephen L. Abrams, James A. McCubrey, Saverio Candido, Alberto M. Martelli, Agnieszka Gizak, Stefano Ratti, Dariusz Rakus, Massimo Libra, Melchiorre Cervello, and S.L. Abrams, S.M. Akula, A.M. Martelli, L. Cocco, S. Ratti, M. Libra, S. Candido, G.Montalto, M. Cervello, A. Gizak, D. Rakus, L.S. Steelman, J.A. McCubrey.
- Subjects
Male ,0301 basic medicine ,Drug ,Cancer Research ,miRs ,endocrine system diseases ,media_common.quotation_subject ,Signal transduction inhibitors ,Targeted therapeutic ,Antineoplastic Agents ,Malignancy ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Chloroquine ,Pancreatic cancer ,Genetics ,medicine ,Humans ,TP53 ,Molecular Biology ,neoplasms ,Signal transduction inhibitor ,Targeted therapeutics ,Cell Proliferation ,media_common ,target therapeutics ,Cell growth ,business.industry ,Cancer ,medicine.disease ,digestive system diseases ,Metformin ,Pancreatic Neoplasms ,030104 developmental biology ,030220 oncology & carcinogenesis ,Dietary Supplements ,Mutation ,Cancer research ,Molecular Medicine ,Female ,KRAS ,Tumor Suppressor Protein p53 ,business ,Signal Transduction ,medicine.drug ,Drug sensitivity - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic malignancy. Approximately 85% of pancreatic cancers are classified as PDACs. The survival of PDAC patients is very poor and only 5–10% of patients survive 5 years after diagnosis. Mutations at the KRAS and TP53 gene are frequently observed in PDAC patients. The PANC-28 cell line lacks wild-type (WT) TP53. In the following study, we have investigated the effects of restoration of WT TP53 activity on the sensitivity of PANC-28 pancreatic cancer cells to various drugs which are used to treat PDAC patients as well as other cancer patients. In addition, we have examined the effects of signal transduction inhibitors which target critical pathways frequently deregulated in cancer. The effects of the anti-diabetes drug metformin and the anti-malarial drug chloroquine were also examined as these drugs may be repurposed to treat other diseases. Finally, the effects of certain nutraceuticals which are used to treat various ailments were also examined. Introduction of WT-TP53 activity in PANC-28 PDAC cells, can increase their sensitivity to various drugs. Attempts are being made clinically to increase TP53 activity in various cancer types which will often inhibit cell growth by multiple mechanisms.
- Published
- 2021
13. Characterization of VPS34-IN1, a selective inhibitor of Vps34, reveals that the phosphatidylinositol 3-phosphate-binding SGK3 protein kinase is a downstream target of class III phosphoinositide 3-kinase.
- Author
-
BAGO, Ruzica, MALIK, Nazma, MUNSON, Michael J., PRESCOTT, Alan R., DAVIES, Paul, SOMMER, Eeva, SHPIRO, Natalia, WARD, Richard, CROSS, Darren, GANLEY, Ian G., and ALESSI, Dario R.
- Subjects
- *
PHOSPHATIDYLINOSITOL 3-kinases , *ENZYME inhibitors , *ENDOSOMES , *CELL membranes , *CELLULAR signal transduction - Abstract
The Vps34 (vacuolar protein sorting 34) class III PI3K (phosphoinositide 3-kinase) phosphorylates PtdIns (phosphatidylinositol) at endosomal membranes to generate PtdIns(3)P that regulates membrane trafficking processes via its ability to recruit a subset of proteins possessing PtdIns(3)P-binding PX (phox homology) and FYVE domains. In the present study, we describe a highly selective and potent inhibitor of Vps34, termed VPS34-IN1, that inhibits Vps34 with 25 nM IC50 in vitro, but does not significantly inhibit the activity of 340 protein kinases or 25 lipid kinases tested that include all isoforms of class I as well as class II PI3Ks. Administration of VPS34-IN1 to cells induces a rapid dose-dependent dispersal of a specific PtdIns(3)Pbinding probe from endosome membranes, within 1 min, without affecting the ability of class I PI3K to regulate Akt. Moreover, we explored whether SGK3 (serum- and glucocorticoid-regulated kinase-3), the only protein kinase known to interact specifically with PtdIns(3)P via its N-terminal PX domain, might be controlled by Vps34. Mutations disrupting PtdIns(3)P binding ablated SGK3 kinase activity by suppressing phosphorylation of the T-loop [PDK1 (phosphoinositide-dependent kinase 1) site] and hydrophobic motif (mammalian target of rapamycin site) residues. VPS34-IN1 induced a rapid ~50-60% loss of SGK3 phosphorylation within 1 min. VPS34-IN1 did not inhibit activity of the SGK2 isoform that does not possess a PtdIns(3)P-binding PX domain. Furthermore, class I PI3K inhibitors (GDC-0941 and BKM120) that do not inhibit Vps34 suppressed SGK3 activity by ~40%. Combining VPS34-IN1 and GDC-0941 reduced SGK3 activity ~80-90%. These data suggest SGK3 phosphorylation and hence activity is controlled by two pools of PtdIns(3)P. The first is produced through phosphorylation of PtdIns by Vps34 at the endosome. The second is due to the conversion of class I PI3K product, PtdIns(3,4,5)P3 into PtdIns(3)P, via the sequential actions of the PtdIns 5-phosphatases [SHIP1/2 (Src homology 2-domain-containing inositol phosphatase 1/2)] and PtdIns 4-phosphatase [INPP4B (inositol polyphosphate 4- phosphatase type II)]. VPS34-IN1 will be a useful probe to delineate physiological roles of the Vps34. Monitoring SGK3 phosphorylation and activity could be employed as a biomarker of Vps34 activity, in an analogous manner by which Akt is used to probe cellular class I PI3K activity. Combining class I (GDC-0941) and class III (VPS34-IN1) PI3K inhibitors could be used as a strategy to better analyse the roles and regulation of the elusive class II PI3K. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
14. EXTH-56. DISRUPTION OF FUNCTIONAL NETWORKS IN GLIOBLASTOMA TO OVERCOME THERAPY RESISTANCE
- Author
-
Ulrich G. Hofmann, Matthias Schneider, Dieter Henrik Heiland, Juergen Beck, Oliver Schnell, Kevin Joseph, and Lea Vollmer
- Subjects
Cancer Research ,Tumor microenvironment ,Necrosis ,Central nervous system ,Signal transduction inhibitor ,Biology ,Preclinical Experimental Therapeutics ,Meclofenamic acid ,medicine.anatomical_structure ,Oncology ,Downregulation and upregulation ,Apoptosis ,Cancer research ,medicine ,Neurology (clinical) ,Signal transduction ,medicine.symptom ,medicine.drug - Abstract
Neurobeachin plays a crucial role when it comes to the formation, maturation, and differentiation of functional synapses, within the central nervous system (CNS). Glioblastoma has cellular compartments that have been shown to form functional synapses, integrating themselves to existing neuronal networks. We present evidence that GBM cells form electrophysiologically active networks, mediated by both electrical and chemical synapses during their growth. These networks carry information about changes in the tumor microenvironment, similar to those observed in neuronal networks. We present data about signature signaling patterns, dependent on stimuli, leading to transcriptional changes. These patterns are conducted through the network by means of electrical/chemical synapses, mediated by Neurobeachin. When the tumor network was acutely treated with a signaling inhibitor, Meclofenamate, functional synapses were altered, leading to an abolishment of electrical signaling, resulting in loss of biological organization. These results were validated in both extracellular electrophysiological recordings and Ca2+ imaging experiments. The formation of these functional networks plays a vital role in the resistance to alkylating agents or harsh metabolic imbalances, which was completely lost when the network was disrupted. Acute downregulation of Neurobeachin expression causes electrophysiological disruption of the GBM network, leading to loss of connectivity, terminally resulting in cell apoptosis/necrosis, transcriptionally validated by means of RNA-seq. This combined therapeutic approach could lead to potential alternative therapy, where the basic biological functionality of the GBM is targeted.
- Published
- 2020
15. Aloin promotes osteogenesis of bone-marrow-derived mesenchymal stem cells via the ERK1/2-dependent Runx2 signaling pathway
- Author
-
Kaipeng Jing, Peng Li, Guihe Lv, Zhuming Chen, Junchao Kong, Bo Wei, Jinsong Wei, Juanhua Quan, Jiaqi Chu, and Shuai Huang
- Subjects
Emodin ,Aloin ,Core Binding Factor Alpha 1 Subunit ,Signal transduction inhibitor ,01 natural sciences ,Bone remodeling ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,stomatognathic system ,Bone Marrow ,Osteogenesis ,medicine ,Animals ,Extracellular Signal-Regulated MAP Kinases ,Cells, Cultured ,Flavonoids ,010405 organic chemistry ,Chemistry ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Rats ,0104 chemical sciences ,Cell biology ,RUNX2 ,010404 medicinal & biomolecular chemistry ,medicine.anatomical_structure ,Molecular Medicine ,Alkaline phosphatase ,Bone marrow ,Signal transduction ,Signal Transduction - Abstract
Osteoporosis is characterized by low bone mass and the degeneration of bone structure, conditions which increase the risk of fracture. Aloin has been shown to affect bone metabolism, but its role in osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) remains unclear. The aim of our study was to determine whether aloin promotes the proliferation and osteogenic differentiation of BMSCs and, if so, whether it acts via activation of the ERK1/2-Runx2 signaling pathway. We found that the different concentrations of aloin tested had no obvious cytotoxic effects on the viability of BMSCs. Under osteogenic induction conditions, aloin increased cellular alkaline phosphatase activity, promoted BMSC mineralization, and increased osteogenic-related gene expression. In addition, treating the BMSCs with the signal transduction inhibitor PD98059 (ERK1/2) effectively attenuated Runx2 activation in these cells and also suppressed osteoblastic differentiation. Overall, our study demonstrates that aloin promotes osteogenic differentiation of BMSCs through activation of the ERK1/2-Runx2 signaling pathway.
- Published
- 2018
16. First-in-human study of LY3039478, an oral Notch signaling inhibitor in advanced or metastatic cancer
- Author
-
Ute Ohnmacht, C. Le Tourneau, Gerard J. Oakley, Christophe Massard, Analia Azaro, Jordi Rodon, Debashis Sarker, Karim A. Benhadji, Bharvin K. R. Patel, Ulrik Lassen, Claire Smith, Eunice Yuen, and Jean-Charles Soria
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,Maximum Tolerated Dose ,Colorectal cancer ,medicine.medical_treatment ,Notch signaling pathway ,Administration, Oral ,Antineoplastic Agents ,colorectal cancer ,Signal transduction inhibitor ,Young Adult ,03 medical and health sciences ,metastatic cancer ,0302 clinical medicine ,Breast cancer ,Neoplasms ,medicine ,Humans ,Response Evaluation Criteria in Solid Tumors ,Aged ,LY3039478 ,Chemotherapy ,Receptors, Notch ,business.industry ,Hematology ,Benzazepines ,Middle Aged ,medicine.disease ,Notch inhibition ,030104 developmental biology ,Oncology ,Notch proteins ,Tolerability ,Î 3-secretase inhibitors ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Amyloid Precursor Protein Secretases ,business - Abstract
Background Deregulated Notch signaling due to mutation or overexpression of ligands and/or receptors is implicated in various human malignancies. γ-Secretase inhibitors inhibit Notch signaling by preventing cleavage of transmembrane domain of Notch protein. LY3039478 is a novel, potent Notch inhibitor decreases Notch signaling and its downstream biologic effects. In this first-in-human study, we report the safety, pharmacokinetic (PK) profile, pharmacodynamic effects, and antitumor activity of LY3039478 in patients with advanced or metastatic cancer. Methods This phase I, open-label, multicenter, nonrandomized, and dose-escalation phase study determined and confirmed the recommended phase II dose of LY3039478 (oral dose: 2.5–100 mg, thrice weekly (TIW) on a 28-day cycle). The primary objectives are to determine (part A) and confirm (part B) a recommended phase II dose that may be safely administered to patients with advanced or metastatic cancer, and secondary objectives include evaluation of safety, tolerability, PK parameters, and preliminary antitumor activity of LY3039478. Results A total of 110 patients were treated with LY3039478 monotherapy between 31 October 2012 and 15 July 2016. Dose-limiting toxicities were thrombocytopenia, colitis, and nausea. Most adverse events were gastrointestinal. The recommended phase II dose was 50 mg TIW, because of its better tolerability compared with 75 mg. The PKs of LY3039478 appeared dose proportional. Pharmacodynamic data indicate an ∼80% inhibition of plasma Aβ, and >50% inhibition of Notch-regulated genes hairy and enhancer of split-1, cyclin D1, and Notch-regulated ankyrin repeat at 45–100-mg dose. Clinical activity (tumor necrosis, metabolic response, or tumor shrinkage) was observed in patients with breast cancer, leiomyosarcoma, and adenoid cystic carcinoma. Conclusion Potent inhibition of Notch signaling by LY3039478 was well tolerated at doses associated with target engagement, and demonstrated evidence of clinical activity in heavily pretreated patients. Further investigation with LY3039478 as monotherapy and in combination with targeted agent or chemotherapy is ongoing. Clinicaltrials.gov ID NCT01695005.
- Published
- 2018
17. Co-Administration of Fish Oil With Signal Transduction Inhibitors Has Anti-Migration Effects in Breast Cancer Cell Lines, in vitro
- Author
-
Stephen Edward Hiscox, Charles Martin Heard, Zoe Davison, and Robert Ian Nicholson
- Subjects
0301 basic medicine ,Chemistry ,Cancer ,Motility ,Cell migration ,Signal transduction inhibitor ,Fish oil ,medicine.disease ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,PD98059 ,Cell culture ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,LY294002 ,Signal transduction ,Omega 3 fatty acid - Abstract
Background:\ud \ud There is an urgent need for new therapies to treat cancer metastasis. Fish oil, with high omega 3 fatty acid content, has shown anticancer activity and signal transduction inhibitors have shown anti-metastatic properties.\ud \ud \ud Objective:\ud \ud To provide preliminary in vitro data on the anti-migration potential of signal transduction inhibitors and co-administered fish oil.\ud \ud \ud Methods:\ud \ud MCF-7, TamR and FasR breast cancer cell lines were used to determine the effects of combinations of PD98059, LY294002 and fish oil in growth assays. Modulations of p-Src and COX-2, both mediators of motility and invasion, were then determined by Western blotting and IHC to ascertain effects on migration potential.\ud \ud \ud Results:\ud \ud Migration rates for the three cell lines examined were ranked: FasR>TamR>MCF-7 (p
- Published
- 2018
18. Estudo exploratório da administração intranasal do monoterpeno álcool perílico em pacientes com glioma recidivante
- Author
-
Clóvis Orlando Da Fonseca ', Clóvis Orlando Da Fonseca, Cerli Rocha Gattass, Rafael Linden, Maria da Gloria Carvalho, Débora Futuro, and Thereza Quirico-santo
- Subjects
medicine.medical_specialty ,Tumor size ,business.industry ,Perillyl alcohol ,Anaplastic oligodendroglioma ,Signal transduction inhibitor ,medicine.disease ,Gastroenterology ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,Nasal administration ,Progression-free survival ,business ,Standard therapy ,Glioblastoma - Abstract
Objetivo: Conduzimos estudo de fase I/II para avaliar atividadeantitumoral do álcool perílico (AP) por via inalatóriaem pacientes com gliomas recidivantes. Pacientes e Métodos: Trinta e sete pacientes com glioma recidivanteapós tratamento convencional foram matriculados,dos quais 29 com glioblastoma (GBM), 5 com astrocitomaanaplásico (AA) e 3 com oligodendroglioma anaplásico (OA).O objetivo foi determinar a sobrevida livre de progressão no6º mês e a toxicidade do AP administrado 4 vezes ao dia empacientes refratários ao tratamento convencional. Resultados: Os resultados mostraram no 6º mês de tratamento, resposta parcial em 1 paciente (3,4%) com GBM e 1paciente (33,3%) com OA; doença estável em 13 pacientes(44,8%) com GBM, 3 pacientes (60%) com AA e 1 paciente(33,3%) com OA; progressão da doença em 15 pacientes(51,7%) com GBM, 2 pacientes (40%) com AA e 1 paciente(33,3%) com OA. A sobrevida livre de progressão (somatóriode resposta parcial e doença estável) foi de 48,2% para pacientescom GBM, 60% para pacientes com AA e 66,6% parapacientes com OA. Conclusão: O presente trabalho mostrou pela primeira vez,que a administração intranasal de um inibidor da transduçãodo sinal, álcool perílico, é uma estratégia segura, não invasiva,de baixo custo, e a regressão do volume tumoral em algunspacientes é sugestivo de atividade antitumoral.
- Published
- 2018
19. Elevated SGK1 predicts resistance of breast cancer cells to Akt inhibitors.
- Author
-
SOMMER, Eeva M., DRY, Hannah, CROSS, Darren, GUICHARD, Sylvie, DAVIES, Barry R., and ALESSI, Dario R.
- Subjects
- *
SGK protein , *BREAST cancer , *PROTEIN kinase B regulation , *CLINICAL trials , *GENETIC mutation , *RAPAMYCIN - Abstract
The majority of human cancers harbour mutations promoting activation of the Akt protein kinase, and Akt inhibitors are being evaluated in clinical trials. An important question concerns the understanding of the innate mechanisms that confer resistance of tumour cells to Akt inhibitors. SGK (serum- and glucocorticoid-regulated kinase) is closely related to Akt and controlled by identical upstream regulators {PI3K (phosphoinositide 3-kinase), PDK1 (phosphoinositide-dependent kinase 1) andmTORC2 [mTOR (mammalian target of rapamycin) complex 2]}. Mutations that trigger activation of Akt would also stimulate SGK. Moreover, Akt and SGK possess analogous substrate specificities and are likely to phosphorylate overlapping substrates to promote proliferation. To investigatewhether cancers possessing high SGK activity could possess innate resistance to Akt-specific inhibitors (that do not target SGK), we analysed SGK levels and sensitivity of a panel of breast cancer cells towards two distinct Akt inhibitors currently in clinical trials (AZD5363 and MK-2206). This revealed a number of Aktinhibitor- resistant lines displaying markedly elevated SGK1 that also exhibited significant phosphorylation of the SGK1 substrate NDRG1 [N-Myc (neuroblastoma-derived Myc) downstreamregulated gene 1]. In contrast, most Akt-inhibitor-sensitive cell lines displayed low/undetectable levels of SGK1. Intriguingly, despite low SGK1 levels, several Akt-inhibitor-sensitive cells showed marked NDRG1 phosphorylation that was, unlike in the resistant cells, suppressed by Akt inhibitors. SGK1 knockdown markedly reduced proliferation of Akt-inhibitor-resistant, but not -sensitive, cells. Furthermore, treatment of Akt-inhibitor-resistant cells with an mTOR inhibitor suppressed proliferation and led to inhibition of SGK1. The results of the present study suggest that monitoring SGK1 levels as well as responses of NDRG1 phosphorylation to Akt inhibitor administration could have a use in predicting the sensitivity of tumours to compounds that target Akt. Our findings highlight the therapeutic potential that SGK inhibitors or dual Akt/SGK inhibitors might have for treatment of cancers displaying elevated SGK activity. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
20. Resistance to endocrine therapy in breast cancer.
- Author
-
Kurebayashi, Junichi
- Subjects
- *
BREAST cancer treatment , *HORMONE therapy , *SELECTIVE estrogen receptor modulators , *PROGESTERONE receptors , *CANCER cell growth - Abstract
Endocrine therapy is the treatment of choice for patients with breast cancer expressing estrogen receptor (ER) and/or progesterone receptor. The efficacy of endocrine therapy is well established in the prevention, adjuvant and metastatic settings. However, either de novo or acquired resistance is frequently observed. Much effort has been made to elucidate the mechanisms of action underlying resistance to endocrine therapy in breast cancer, and several possible explanations have been suggested. Our previous studies have indicated that combined treatment with an antiestrogen, fulvestrant, and an inhibitor of the HER2 signaling pathway, trastuzumab, or an inhibitor of the HER1 signaling pathway, gefitinib, leads to an additive antitumor effect in breast cancer cells expressing ER and HER2 or HER1, respectively. It has also been suggested that the HER1 or HER2 signaling pathway is upregulated during the development of antiestrogen-resistant growth in breast cancer cells. These findings suggest that signal transduction inhibitors are effective for the treatment of antiestrogen-resistant breast cancer. A hypoxic microenvironment has been shown to promote malignant progression in cancer cells. Our previous study and others have suggested that hypoxia posttranscriptionally reduces ER expression and decreases sensitivity to hormonal agents in breast cancer cells. Our preliminary study has also shown that a hypoxic cytotoxin, tirapazamine, increases ER expression in breast cancer xenografts. Differential antitumor activity of tirapazamine on tumor cells under normoxic or hypoxic conditions may cause this phenomenon. These findings suggest that hypoxic cytotoxins may retard the development of endocrine resistance induced by hypoxia. Molecular mechanisms responsible for endocrine resistance in breast cancer are reviewed and possible therapeutic strategies against this resistance are discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
21. TT232, A Novel Signal Transduction Inhibitory Compound in the Therapy of Cancer and Inflammatory Diseases.
- Author
-
SZOKOLÓCZI, ORSOLYA, SCHWAB, RICHÁRD, PETÁK, ISTVÁN, ÖRFI, LÁSZLÓ, PAP, ÁKOS, EBERLE, ALEX N., SZÜTS, TAMÁS, and KÉRI1, GYÖRGY
- Subjects
SOMATOSTATIN ,GENE fusion ,GENETIC transduction ,CANCER treatment ,THERAPEUTICS ,PYRUVATE kinase ,CELL physiology ,CANCER vaccines - Abstract
TT-232 is a structural analogue of somatostatin exhibiting strong and selective growth-inhibitory effects, inhibition of neurogenic inflammation, as well as general anti-inflammatory and analgesic potential without the wide-ranging endocrine side effects of the parent hormone and its “traditional” analogues. The anti-inflammatory action of TT-232 is mediated through the SSTR4 receptor, and its antitumor activity is mediated through the SSTR1 receptor and by the tumor-specific isoform of pyruvate kinase. Its mechanism of action is in line with a new era of molecular medicine called signal transduction therapy, where “false” intracellular or intercellular communication is inhibited or corrected without interfering with basic cell functions and machinery. TT232 has passed phase I clinical trials without toxicity and significant side effects, and phase II studies are running for oncological and anti-inflammatory indications, respectively. This compound has the perspective to become the first drug in molecularly targeted therapy of inflammation where a combined effect of anti-inflammatory, analgesic, and neurogenic inflammation-inhibiting activity can be achieved. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
22. Grb7-based molecular therapeutics in cancer.
- Author
-
Pero, Stephanie C., Daly, Roger J., and Krag, David N.
- Abstract
Traditional anti-cancer drugs preferentially kill rapidly growing tumour cells rather than normal cells. However, most of these drugs have no preferential selection towards cancer cells and are taken up by the whole body, resulting in significant adverse side effects. Therapeutic molecules that could specifically inhibit undesirable phenotypes are an attractive way of eliminating cancer cells. There is a widespread effort to develop inhibitors against signal transduction molecules that play a key role in the proliferative, migratory and invasive properties of a cancer cell. Grb7 is an adaptor-type signalling protein that is recruited via its Src-homology 2 (SH2) domain to a variety of tyrosine kinases. Grb7 is overexpressed in breast, oesophageal and gastric cancers, and may contribute to the invasive potential of cancer cells. Molecular interactions involving Grb7 therefore provide attractive targets for therapeutic intervention. [ABSTRACT FROM PUBLISHER]
- Published
- 2003
- Full Text
- View/download PDF
23. Novel chemical genetic approaches to the discovery of signal transduction inhibitors
- Author
-
Shokat, Kevan and Velleca, Mark
- Subjects
- *
CHEMISTRY , *BIOCHEMICAL genetics , *TARGETED drug delivery - Abstract
Concurrent advances in both high-throughput chemistry and genomics have given rise to the field of chemical genetics as a discipline for elucidating and validating drug targets, and generating novel therapeutics. Indeed, chemical genetic approaches to drug discovery have now been applied to several important drug target classes, especially those involved in signal transduction. Chemical genetics is distinct from the broader term ‘chemogenomics’ which is defined as the description of all possible drugs against all possible targets (reviewed in ). This review covers several ‘orthogonal’ chemical genetic approaches and focuses on a unique analog sensitive kinase technology and its applications to kinase drug discovery. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
24. Abilities of berberine and chemically modified berberines to interact with metformin and inhibit proliferation of pancreatic cancer cells
- Author
-
Ramiro Mendonça Murata, Massimo Libra, Matilde Y. Follo, Giuseppe Montalto, Saverio Candido, Agnieszka Gizak, Kvin Lertpiriyapong, Heng-Liang Lin, Melchiorre Cervello, Pedro Luiz Rosalen, Paolo Lombardi, Giulia Ramazzotti, Weifeng Mao, Dariusz Rakus, Stefano Ratti, Alberto M. Martelli, James A. McCubrey, Linda S. Steelman, Severino Matias de Alencar, Shaw M. Akula, Stephen L. Abrams, Bruno Bueno-Silva, S.M. Akula, S. Candido, M. Libra, S.L. Abrams, L.S. Steelman, K. Lertpiriyapong, G. Ramazzotti, S. Ratti, M.Y. Follo, A.M. Martelli, R.M. Murata, P.L. Rosalen, B. Bueno-Silva, S. Matias de Alencar, G. Montalto, M. Cervello, A. Gizak, D. Rakus, W. Mao, H.-L. Lin, P. Lombardi, J.A. McCubrey., Akula, Shaw M, Candido, Saverio, Libra, Massimo, Abrams, Stephen L, Steelman, Linda S, Lertpiriyapong, Kvin, Ramazzotti, Giulia, Ratti, Stefano, Follo, Matilde Y, Martelli, Alberto M, Murata, Ramiro M, Rosalen, Pedro L, Bueno-Silva, Bruno, Matias de Alencar, Severino, Montalto, Giuseppe, Cervello, Melchiorre, Gizak, Agnieszka, Rakus, Dariusz, Mao, Weifeng, Lin, Heng-Liang, Lombardi, Paolo, and McCubrey, James A
- Subjects
0301 basic medicine ,Cancer Research ,Settore MED/09 - Medicina Interna ,endocrine system diseases ,Berberine ,Signal transduction inhibitors ,Blood sugar ,Pharmacology ,AMP-Activated Protein Kinases ,PDAC ,TP53 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,METFORMINA ,Pancreatic cancer ,Diabetes mellitus ,Genetics ,medicine ,Humans ,Signal transduction inhibitor ,Molecular Biology ,Cell Proliferation ,business.industry ,Cancer ,AMPK ,medicine.disease ,Metformin ,Neoplasm Proteins ,Pancreatic Neoplasms ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Molecular Medicine ,business ,medicine.drug - Abstract
Pancreatic cancer is devastating cancer worldwide with few if any truly effective therapies. Pancreatic cancer has an increasing incidence and may become the second leading cause of death from cancer. Novel, more effective therapeutic approaches are needed as pancreatic cancer patients usually survive for less than a year after being diagnosed. Control of blood sugar levels by the prescription drug metformin in diseases such as diabetes mellitus has been examined in association with pancreatic cancer. While the clinical trials remain inconclusive, there is hope that certain diets and medications may affect positively the outcomes of patients with pancreatic and other cancers. Other natural compounds may share some of the effects of metformin. One “medicinal” fruit consumed by millions worldwide is berberine (BBR). Metformin and BBR both activate AMP-activated protein kinase (AMPK) which is a key mediator of glucose metabolism. Glucose metabolism has been shown to be very important in cancer and its significance is increasing. In the following studies, we have examined the effects of metformin, BBR and a panel of modified BBRs (NAX compounds) and chemotherapeutic drugs on the growth of four different human pancreatic adenocarcinoma cell lines (PDAC). Interestingly, the effects of metformin could be enhanced by BBR and certain modified BBRs. Upon restoration of WT-TP53 activity in MIA-PaCa-2 cells, an altered sensitivity to the combination of certain NAX compounds and metformin was observed compared to the parental cells which normally lack WT-TP53. Certain NAX compounds may interact with WT-TP53 and metformin treatment to alter the expression of key molecules involved in cell growth. These results suggest a therapeutic approach by combining certain pharmaceutical drugs and nutraceuticals to suppress the growth of cancer cells.
- Published
- 2019
25. RAS/RAF/MEK/ERK, PI3K/PTEN/AKT/mTORC1 and TP53 pathways and regulatory miRs as therapeutic targets in hepatocellular carcinoma
- Author
-
Giuseppe Montalto, Shaw M. Akula, Stephen L. Abrams, Melchiorre Cervello, Giuseppa Augello, Linda S. Steelman, Maria Rita Emma, Antonina Azzolina, Antonella Cusimano, James A. McCubrey, Akula S.M., Abrams S.L., Steelman L.S., Emma M.R., Augello G., Cusimano A., Azzolina A., Montalto G., Cervello M., and McCubrey J.A.
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Carcinoma, Hepatocellular ,Hepatocellular carcinma ,medicine.medical_treatment ,Clinical Biochemistry ,Antineoplastic Agents ,mTORC1 ,signal transduction inhibitors ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,microRNA ,medicine ,PTEN ,Animals ,Humans ,cancer ,Molecular Targeted Therapy ,TP53 ,HCC ,RAS/RAF/MEK/ERK ,Protein kinase B ,PI3K/AKT/mTOR pathway ,miRNA ,Pharmacology ,biology ,business.industry ,Kinase ,Liver Neoplasms ,Mir ,hepatocellular carcinoma ,targeted therapy ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,signal transduction inhibitor ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,PI3K/PTEN/AKT ,business ,Signal Transduction - Abstract
Introduction: Hepatocellular carcinoma (HCC) is a significant problem globally because of viral infections and the increasing incidence of obesity and fatty liver disease. However, it is difficult to treat because its inherent genetic heterogeneity results in activation of numerous signaling pathways. Kinases have been targeted for decades with varying results, but the development of therapeutic resistance is a major challenge. Areas covered: The key roles of the RAS/RAF/MEK/ERK, PI3K/PTEN/AKT/mTORC1, TP53 microRNAs (miRs) as therapeutic targets are discussed and we suggests novel approaches for targeting miRs or their downstream targets to combat HCC. We performed literature searches using the Medline Database from 2000 to the present. Expert opinion: The involvement of RAS/RAF/MEK/ERK, PI3K/PTEN/AKT/mTORC and TP53 pathways as drivers of the disease and drug resistance is a challenge. Moreover, miRs regulate the expression of key genes in these pathways. What we and others are proposing is the prospect of targeting miRs and their downstream targets to improve conventional approaches to treat HCC. Combination approaches are often promising because multiple signaling pathways are deregulated due to diverse mutations and events.
- Published
- 2019
26. Abilities of berberine and chemically modified berberines to inhibit proliferation of pancreatic cancer cells
- Author
-
Stefano Ratti, Kvin Lertpiriyapong, Ramiro Mendonça Murata, Matilde Y. Follo, Dariusz Rakus, Pedro Luiz Rosalen, Saverio Candido, Linda S. Steelman, Paolo Lombardi, Agnieszka Gizak, Lucio Cocco, James A. McCubrey, Weifeng Mao, Alberto M. Martelli, Stephen L. Abrams, Giuseppe Montalto, Massimo Libra, Melchiorre Cervello, Abrams SL, Follo MY, Steelman LS, Lertpiriyapong K, Cocco L, Ratti S, Martelli AM, Candido S, Libra M, Murata RM, Rosalen PL, Montalto G, Cervello M, Gizak A, Rakus D, Mao W, Lombardi P, McCubrey JA., Abrams, Stephen L, Follo, Matilde Y, Steelman, Linda S, Lertpiriyapong, Kvin, Cocco, Lucio, Ratti, Stefano, Martelli, Alberto M, Candido, Saverio, Libra, Massimo, Murata, Ramiro M, Rosalen, Pedro L, Montalto, Giuseppe, Cervello, Melchiorre, Gizak, Agnieszka, Rakus, Dariusz, Mao, Weifeng, Lombardi, Paolo, and McCubrey, James A
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Settore MED/09 - Medicina Interna ,Berberine ,DNA damage ,Population ,Signal transduction inhibitors ,Apoptosis ,Inflammation ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Pancreatic cancer ,Genetics ,medicine ,Humans ,education ,Chemotherapeutic drug ,Molecular Biology ,Signal transduction inhibitor ,Aged ,education.field_of_study ,business.industry ,Cell Cycle ,Autophagy ,Cancer ,PDAC ,DNA, Neoplasm ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Molecular Medicine ,Chemotherapeutic drugs ,medicine.symptom ,business ,DNA Damage ,Signal Transduction - Abstract
Berberine (BBR) is a common nutraceutical consumed by millions worldwide. BBR has many different effects on human health, e.g., diabetes, diarrhea, inflammation and now more recently it has been proposed to have potent anti-cancer effects. BBR has been shown to suppress the growth of cancer cells more than normal cells. BBR has been proposed to exert its growth-inhibitory effects by many different biochemical mechanisms including: suppression of cell cycle progression, induction of reactive oxygen species, induction of apoptosis and autophagy and interactions with DNA potentially leading to DNA damage, and altered gene expression. Pancreatic cancer is a leading cancer worldwide associated with a poor prognosis. As our population ages, pancreatic cancer has an increasing incidence and will likely become the second leading cause of death from cancer. There are few truly-effective therapeutic options for pancreatic cancer. Surgery and certain chemotherapeutic drugs are used to treat pancreatic cancer patients. Novel approaches to treat pancreatic cancer patients are direly needed as they usually survive for less than a year after being diagnosed. In the following manuscript, we discuss the abilities of BBR and certain chemically- modified BBRs (NAX compounds) to suppress growth of pancreatic cancer cells.
- Published
- 2019
27. Ibrutinib-induced pneumonitis in patients with chronic lymphocytic leukemia
- Author
-
Lauren E. Strelec, Sunita D. Nasta, Jakub Svoboda, Sarah Brooks, Prioty Islam, Chadi Nabhan, Anthony R. Mato, Catherine Daniel, Alex Ganetsky, David L. Porter, Stephen J. Schuster, and Adam H. Kaye
- Subjects
business.industry ,Chronic lymphocytic leukemia ,Immunology ,breakpoint cluster region ,Cell Biology ,Hematology ,Signal transduction inhibitor ,030204 cardiovascular system & hematology ,medicine.disease ,Biochemistry ,03 medical and health sciences ,Pneumonia ,chemistry.chemical_compound ,Leukemia ,0302 clinical medicine ,chemistry ,immune system diseases ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Ibrutinib ,medicine ,Signal transduction ,business ,Pneumonitis - Abstract
To the editor: Chronic lymphocytic leukemia (CLL) is a B-cell lymphoproliferative disorder partly dependent on the B-cell receptor (BCR) signaling pathway. The novel BCR signal transduction inhibitor ibrutinib has emerged as an effective therapeutic agent for CLL.[1][1] Ibrutinib covalently binds
- Published
- 2016
28. SRC/ABL inhibition disrupts CRLF2-driven signaling to induce cell death in B-cell acute lymphoblastic leukemia
- Author
-
Sarno, J, Savino, A, Buracchi, C, Palmi, C, Pinto, S, Bugarin, C, Jager, A, Bresolin, S, Barber, R, Silvestri, D, Israeli, S, Dyer, M, Cazzaniga, G, Nolan, G, Biondi, A, Davis, K, Gaipa, G, Sarno, Jolanda, Savino, Angela M., Buracchi, Chiara, Palmi, Chiara, Pinto, Stefania, Bugarin, Cristina, Jager, Astraea, Bresolin, Silvia, Barber, Ruth C., Silvestri, Daniela, Israeli, Shai, Dyer, Martin J. S., Cazzaniga, Giovanni, Nolan, Garry P., Biondi, Andrea, Davis, Kara L., Gaipa, Giuseppe, Sarno, J, Savino, A, Buracchi, C, Palmi, C, Pinto, S, Bugarin, C, Jager, A, Bresolin, S, Barber, R, Silvestri, D, Israeli, S, Dyer, M, Cazzaniga, G, Nolan, G, Biondi, A, Davis, K, Gaipa, G, Sarno, Jolanda, Savino, Angela M., Buracchi, Chiara, Palmi, Chiara, Pinto, Stefania, Bugarin, Cristina, Jager, Astraea, Bresolin, Silvia, Barber, Ruth C., Silvestri, Daniela, Israeli, Shai, Dyer, Martin J. S., Cazzaniga, Giovanni, Nolan, Garry P., Biondi, Andrea, Davis, Kara L., and Gaipa, Giuseppe
- Abstract
Children with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) overexpressing the CRLF2 gene (hiCRLF2) have poor prognosis. CRLF2 protein overexpression leads to activated JAK/STAT signaling and trials are underway using JAK inhibitors to overcome treatment failure. Pre-clinical studies indicated limited efficacy of single JAK inhibitors, thus additional pathways must be targeted in hiCRLF2 cells. To identify additional activated networks, we used single-cell mass cytometry to examine 15 BCP-ALL primary patient samples. We uncovered a coordinated signaling network downstream of CRLF2 characterized by co-activation of JAK/STAT, PI3K, and CREB pathways. This CRLF2-driven network could be more effectively disrupted by SRC/ABL inhibition than single-agent JAK or PI3K inhibition, and this could be demonstrated even in primary minimal residual disease (MRD) cells. Our study suggests SCR/ABL inhibition as effective in disrupting the cooperative functional networks present in hiCRLF2 BCP-ALL patients, supporting further investigation of this strategy in pre-clinical studies.
- Published
- 2018
29. Vandetanib has antineoplastic activity in anaplastic thyroid cancer, in vitro and in vivo
- Author
-
Concettina La Motta, Francesca Ragusa, Teresa Di Desidero, Ilaria Ruffilli, Simona Piaggi, Guido Bocci, Alessandro Antonelli, Poupak Fallahi, Gabriele Materazzi, Armando Patrizio, Paola Orlandi, Anna Fioravanti, Enke Baldini, Silvia Martina Ferrari, Giusy Elia, Salvatore Ulisse, Paolo Miccoli, and Sabrina Rosaria Paparo
- Subjects
0301 basic medicine ,Cancer Research ,Anaplastic thyroid cancer ,Signal transduction inhibitor ,Thyroid Carcinoma, Anaplastic ,Vandetanib ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,in vivo studies ,Piperidines ,Cell Movement ,tyrosine kinase inhibitors ,Cyclin D1 ,Epidermal growth factor receptor ,Phosphorylation ,Mitogen-Activated Protein Kinase 1 ,in vitro studies ,Mitogen-Activated Protein Kinase 3 ,biology ,General Medicine ,primary anaplastic thyroid cancer cells ,oncology ,cancer research ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Vascular endothelial growth factor ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Tyrosine kinase ,medicine.drug ,Cell Survival ,Antineoplastic Agents ,In Vitro Techniques ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Thyroid Neoplasms ,Protein kinase B ,Cell Proliferation ,Dose-Response Relationship, Drug ,Cell growth ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,chemistry ,Quinazolines ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-akt - Abstract
The antitumor activity of vandetanib [a multiple signal transduction inhibitor including the RET tyrosine kinase, epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF) receptor (VEGFR), ERK and with antiangiogenic activity], in primary anaplastic thyroid cancer (ATC) cells, in the human cell line 8305C [undifferentiated thyroid cancer (TC)] and in an ATC‑cell line (AF), was investigated in the present study. Vandetanib (1 and 100 nM; 1, 10, 25 and 50 µM) was tested by WST‑1, apoptosis, migration and invasion assays: in primary ATC cells, in the 8305C continuous cell line, and in AF cells; and in 8305C cells in CD nu/nu mice. Vandetanib significantly reduced ATC cell proliferation (P
- Published
- 2018
30. Introduction of WT-TP53 into pancreatic cancer cells alters sensitivity to chemotherapeutic drugs, targeted therapeutics and nutraceuticals
- Author
-
Linda S. Steelman, Pedro Luiz Rosalen, Paolo Lombardi, Marco Falasca, Giuseppe Montalto, Ramiro Mendonça Murata, Lucio Cocco, Massimo Libra, Li V. Yang, Stefano Ratti, James A. McCubrey, Kvin Lertpiriyapong, Melchiorre Cervello, Saverio Candido, Alberto M. Martelli, Stephen L. Abrams, Abrams S.L., Lertpiriyapong K., Yang L.V., Martelli A.M., Cocco L., Ratti S., Falasca M., Murata R.M., Rosalen P.L., Lombardi P., Libra M., Candido S., Montalto G., Cervello M., Steelman L.S., and McCubrey J.A.
- Subjects
0301 basic medicine ,Cancer Research ,Paclitaxel ,endocrine system diseases ,medicine.medical_treatment ,Targeted therapeutic ,Irinotecan ,Deoxycytidine ,Targeted therapy ,Glycogen Synthase Kinase 3 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Pancreatic cancer ,Genetics ,medicine ,Humans ,Doxorubicin ,TP53 ,Signal transduction inhibitor ,neoplasms ,Molecular Biology ,Cell Proliferation ,Cisplatin ,Chemotherapy ,business.industry ,Pancreatic Neoplasm ,medicine.disease ,Gemcitabine ,digestive system diseases ,Pancreatic Neoplasms ,Oxaliplatin ,030104 developmental biology ,chemistry ,Fluorouracil ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,business ,Drug sensitivity ,Human ,Signal Transduction ,medicine.drug - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5–10%. Mutations at the TP53 gene are readily detected in pancreatic tumors isolated from PDAC patients. We have investigated the effects of restoration of wild-type (WT) TP53 activity on the sensitivity of pancreatic cancer cells to: chemotherapy, targeted therapy, as well as, nutraceuticals. Upon introduction of the WT-TP53 gene into the MIA-PaCa-2 pancreatic cancer cell line, the sensitivity to drugs used to treat pancreatic cancer cells such as: gemcitabine, fluorouracil (5FU), cisplatin, irinotecan, oxaliplatin, and paclitaxel increased significantly. Likewise, the sensitivity to drugs used to treat other cancers such as: doxorubicin, mitoxantrone, and 4 hydroxy tamoxifen (4HT) also increased upon introduction of WT-TP53 into MIA-PaCa-2 cells. Furthermore, the sensitivity to certain inhibitors which target: PI3K/mTORC1, PDK1, SRC, GSK-3, and biochemical processes such as proteasomal degradation and the nutraceutical berberine as increased upon introduction of WT-TP53. Furthermore, in some cases, cells with WT-TP53 were more sensitive to the combination of drugs and suboptimal doses of the MDM2 inhibitor nutlin-3a. However, TP53-independent effects of nutlin-3a were observed upon treatment with either a proteasomal or a PI3K/mTOR inhibitor. These studies indicate the sensitizing effects that WT-TP53 can have in PDAC cells which normally lack WT-TP53 to various therapeutic agents and suggest approaches to improve PDAC therapy.
- Published
- 2018
31. Introduction of WT-TP53 into pancreatic cancer cells with mutant TP53 alters sensitivity of chemotherapeutic drugs, targeted therapeutics and nutraceuticals
- Author
-
S. L. Abrams, K. Lertpiriyapong, L. V. Yang, A. M. Martelli, L. Cocco, S. Ratti, M. Falasca, R. M. Murata, P. L. Rosalen, P. Lombardi, M. Libra, G. Montalto, M. Cervello, L. S. Steelman, J. A. McCubrey., and S.L. Abrams, K. Lertpiriyapong, L.V. Yang, A.M. Martelli, L. Cocco, S. Ratti, M. Falasca, R.M. Murata, P.L. Rosalen, P. Lombardi, M. Libra, G. Montalto, M. Cervello, L.S. Steelman, J.A. McCubrey.
- Subjects
signal transduction inhibitor ,endocrine system diseases ,TP53 ,drug sensitivity ,neoplasms ,digestive system diseases ,targeted therapeutic - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5–10%. Mutations at the TP53 gene are readily detected in pancreatic tumors isolated from PDAC patients. We have investigated the effects of restoration of wild-type (WT) TP53 activity on the sensitivity of pancreatic cancer cells to: chemotherapy, targeted therapy, as well as, nutraceuticals. Upon introduction of the WT-TP53 gene into the MIA-PaCa-2 pancreatic cancer cell line, the sensitivity to drugs used to treat pancreatic cancer cells such as: gemcitabine, fluorouracil (5FU), cisplatin, irinotecan, oxaliplatin, and paclitaxel increased significantly. Likewise, the sensitivity to drugs used to treat other cancers such as: doxorubicin, mitoxantrone, and 4 hydroxy tamoxifen (4HT) also increased upon introduction of WT-TP53 into MIA-PaCa-2 cells. Furthermore, the sensitivity to certain inhibitors which target: PI3K/mTORC1, PDK1, SRC, GSK-3, and biochemical processes such as proteasomal degradation and the nutraceutical berberine as increased upon introduction of WT-TP53. Furthermore, in some cases, cells with WT-TP53 were more sensitive to the combination of drugs and suboptimal doses of the MDM2 inhibitor nutlin-3a. However, TP53-independent effects of nutlin-3a were observed upon treatment with either a proteasomal or a PI3K/mTOR inhibitor. These studies indicate the sensitizing effects that WT-TP53 can have in PDAC cells which normally lack WT-TP53 to various therapeutic agents and suggest approaches to improve PDAC therapy.
- Published
- 2018
32. Metformin influences drug sensitivity in pancreatic cancer cells
- Author
-
Linda S. Steelman, Giuseppe Montalto, Pedro Luiz Rosalen, Massimo Libra, Melchiorre Cervello, Paolo Lombardi, Ramiro Mendonça Murata, Lucio Cocco, Dariusz Rakus, Pann-Gill Suh, Alberto M. Martelli, Stefano Ratti, James A. McCubrey, Agnieszka Gizak, Saverio Candido, Kvin Lertpiriyapong, Stephen L. Abrams, Matilde Y. Follo, S. Candido, S.L. Abrams, L. Steelman, K. Lertpiriyapong, A.M. Martelli, L. Cocco, S. Ratti, M.Y. Follo, R.M. Murata, P.L. Rosalen, P. Lombardi, G. Montalto, M. Cervello, A Gizak, D. Rakus, P.-G. Suh, M. Libra, J. A. McCubrey., Candido S., Abrams S.L., Steelman L., Lertpiriyapong K., Martelli A.M., Cocco L., Ratti S., Follo M.Y., Murata R.M., Rosalen P.L., Lombardi P., Montalto G., Cervello M., Gizak A., Rakus D., Suh P.-G., Libra M., and McCubrey J.A.
- Subjects
AMPK ,0301 basic medicine ,Cancer Research ,endocrine system diseases ,03 medical and health sciences ,Pancreatic cancer ,Genetics ,Medicine ,Animals ,Humans ,Doxorubicin ,Drug Interactions ,Rapamycin ,Signal transduction inhibitor ,mTORC1 ,Molecular Biology ,Cisplatin ,Sirolimus ,Animal ,business.industry ,Pancreatic Neoplasm ,Cancer ,medicine.disease ,Gemcitabine ,Metformin ,Pancreatic Neoplasms ,030104 developmental biology ,Drug Interaction ,Docetaxel ,Diabetes Mellitus, Type 2 ,Cancer research ,Molecular Medicine ,business ,Human ,medicine.drug ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5–10% after diagnosis and treatment. Pancreatic cancer has been associated with type II diabetes as the frequency of recently diagnosed diabetics that develop pancreatic cancer within a 10-year period of initial diagnosis of diabetes in increased in comparison to non-diabetic patients. Metformin is a very frequently prescribed drug used to treat type II diabetes. Metformin acts in part by stimulating AMP-kinase (AMPK) and results in the suppression of mTORC1 activity and the induction of autophagy. In the following studies, we have examined the effects of metformin in the presence of various chemotherapeutic drugs, signal transduction inhibitors and natural products on the growth of three different PDAC lines. Metformin, by itself, was not effective at suppressing growth of the pancreatic cancer cell lines at concentration less than 1000 nM, however, in certain PDAC lines, a suboptimal dose of metformin (250 nM) potentiated the effects of various chemotherapeutic drugs used to treat pancreatic cancer (e.g., gemcitabine, cisplatin, 5-fluorouracil) and other cancer types (e.g., doxorubicin, docetaxel). Furthermore, metformin could increase anti-proliferative effects of mTORC1 and PI3K/mTOR inhibitors as well as natural products such as berberine and the anti-malarial drug chloroquine in certain PDAC lines. Thus, metformin can enhance the effects of certain drugs and signal transduction inhibitors which are used to treat pancreatic and various other cancers.
- Published
- 2018
33. Circulating Wnt/β-catenin signalling inhibitors and uraemic vascular calcifications
- Author
-
Chih Yu Yang, Zee-Fen Chang, Oscar K. Lee, An Hang Yang, Yat Pang Chau, Wu Chang Yang, and Ann Chen
- Subjects
Genetic Markers ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Signal transduction inhibitor ,chemistry.chemical_compound ,Renal Dialysis ,Internal medicine ,medicine ,Humans ,Prospective Studies ,Vascular Calcification ,Wnt Signaling Pathway ,Adaptor Proteins, Signal Transducing ,Uremia ,Transplantation ,Proportional hazards model ,business.industry ,Hazard ratio ,Wnt signaling pathway ,Middle Aged ,Cross-Sectional Studies ,Endocrinology ,chemistry ,Cardiovascular Diseases ,Parathyroid Hormone ,Nephrology ,Bone Morphogenetic Proteins ,Intercellular Signaling Peptides and Proteins ,Sclerostin ,Female ,Hemodialysis ,Animal studies ,Aortic valve calcification ,business ,Biomarkers - Abstract
BACKGROUND The process of vascular calcification has been associated with the canonical Wnt/β-catenin signalling pathway in cell cultures and animal studies. The relationship between circulating Wnt/β-catenin inhibitors and vascular calcification in dialysis patients is unknown. The aim of this study was to investigate the associations between serum dickkopf-1 (Dkk-1) and sclerostin, two circulating inhibitors of the Wnt/β-catenin signalling pathway, and the severity of aortic calcification (AoC) and cardiovascular outcomes in dialysis patients. METHODS This was a prospective observational cohort study. One hundred and twenty-five patients on maintenance haemodialysis participated in the study. Serum levels of Dkk-1 and sclerostin were measured. AoC scores were calculated from plain films of both posterior-anterior and lateral views. The patients were followed up for 2 years or until death or withdrawal. RESULTS The circulating sclerostin level was inversely associated with the severity of AoC (P = 0.035) and indicators of the bone turnover rate including serum alkaline phosphatase (ALP) (r = -0.235, P = 0.008) and intact parathyroid hormone (r = -0.523, P < 0.001). Furthermore, Cox regression analysis indicated that the patients with high circulating sclerostin levels were less likely to experience future cardiovascular events [1 pmol/L sclerostin increase, hazard ratio 0.982 (95% CI, 0.967-0.996), P = 0.015] after adjusting for a propensity score. In contrast, serum Dkk-1 was not associated with AoC and clinical outcomes. CONCLUSIONS In long-term haemodialysis patients, circulating sclerostin but not Dkk-1 is inversely associated with AoCs and future cardiovascular events. Our findings suggest that sclerostin, as a bone-related protein, might act as a communicator between uraemic bone and vasculature.
- Published
- 2015
34. MEDU-46. IDENTIFICATION AND CHARACTERIZATION OF TUMOR-INITIATING CELLS IN RECURRENT MEDULLOBLASTOMA
- Author
-
Liguo Zhang and Xuelian He
- Subjects
Medulloblastoma ,Cancer Research ,business.industry ,Signal transduction inhibitor ,Tumor initiation ,Recurrent Medulloblastoma ,medicine.disease ,Tumor Initiating Cells ,Abstracts ,Oncology ,Tumor progression ,Cancer research ,medicine ,Tumor growth ,Neurology (clinical) ,Stem cell ,business - Abstract
Medulloblastoma represents the most common childhood brain tumor. Despite aggressive multimodal therapy, a significant proportion of surviving patients suffer from severe treatment-related side effects, including tumor relapse. Thus, there is an urgent need for novel therapeutic modalities that can improve patient survival while minimizing tumor recurrence. Our recent study identified GNAS as a novel tumor-suppressor gene in Sonic hedgehog-driven medulloblastomas (Ref.1). Low expression or mutation of GNAS was tightly correlated with poor prognosis of Sonic hedgehog-group tumors and their resistance to the treatment of Sonic hedgehog signaling inhibitors alone. Ablation of the single GNAS gene in anatomically-distinct progenitors in the ventricular zone of the cerebellum and dorsal brainstem is sufficient to induce medulloblastoma formation, recapitulating their human counterparts. Gsα activation suppresses Sonic hedgehog signaling by regulating both the cAMP-dependent PKA pathway and ciliary trafficking of Hedgehog pathway components. Elevation of a Gsα effector, cAMP, effectively inhibits tumor growth in GNAS mutants. Strikingly, we find that in the drug-resistant tumor lesion and recurrent tumors after radiation therapy, there is an increase of Olig2 expression and Olig2+ progenitor cells. High levels of Olig2 are correlated with poor prognosis of medulloblastomas in human patients. We show that elimination of the mitotic Olig2-expressing cells blocks tumor progression in Olig2-TK transgenic mice. In addition, cell type-specific deletion of Olig2 halts tumor progression. Moreover, we find that Olig2 activates gene regulatory networks inferred for tumor cell stemness and regulates chromatin accessibility of oncogenic factors by altering epigenome landscapes. Furthermore, our single cell analyses reveal that Olig2-expressing progenitors are the distinct tumor-initiating cells during the onset of primary and recurrent medulloblastoma, highlighting potential therapeutic avenues for treating recurrent medulloblastomas by targeting Olig2-expressing progenitors. Ref.1: He, X. et al., “The G protein-alpha subunit Ga is a tumor suppressor in Sonic hedgehog-driven medulloblastoma”. Nat Med 20: 1035–1042.
- Published
- 2017
35. Efficacy of Therapies Following Venetoclax Discontinuation in CLL: Focus on B-Cell Receptor Signal Transduction Inhibitors and Cellular Therapies
- Author
-
Amber C. King, Michael Y. Choi, Guilherme Fleury Perini, Sirin Khajavian, Mazyar Shadman, Kentson Lam, Jason C. Lee, Bita Fakhri, Jeffrey J. Pu, Danielle M. Brander, Pratik Shah, Colleen Dorsey, Bruce D. Cheson, Kayla Bigelow, Talha Munir, Neil Bailey, Ryan Jacobs, Stephen J. Schuster, Thomas D. Rodgers, Hanna Weissbrot, Satyen H. Gohil, Lindsey E. Roeker, Andrew D. Zelenetz, Andrea Sitlinger, Pallawi Torka, Kate J Whitaker, Chaitra S. Ujjani, Nicolas Martinez-Calle, Christopher P. Fox, Brian T. Hill, Alan P Skarbnik, Paul M. Barr, Chadi Nabhan, Javier Pinilla Ibarz, Anthony R. Mato, Krista Isaac, Christine A. Garcia, Ariel F Grajales-Cruz, Allison M. Winter, Maryam Sarraf Yazdy, Toby A. Eyre, John M. Pagel, Jacqueline C. Barrientos, John N. Allan, Erica B. Bhavsar, Othman S. Akhtar, Julie Goodfriend, Helen Parry, Nicole Lamanna, Craig A. Portell, Timothy J Voorhees, Catherine C. Coombs, Rachael Pocock, and Joanna Rhodes
- Subjects
business.industry ,Venetoclax ,Immunology ,B-cell receptor ,Cell Biology ,Hematology ,Signal transduction inhibitor ,Biochemistry ,Discontinuation ,Cell therapy ,chemistry.chemical_compound ,chemistry ,Ibrutinib ,Cancer research ,Medicine ,Signal transduction ,business ,Idelalisib - Abstract
Introduction: Venetoclax (VEN) based therapy has become a standard of care in front line and relapsed-refractory (R/R) CLL based on favorable efficacy and toxicity. Whereas prospective data regarding activity of therapies following ibrutinib (IBR) or idelalisib (IDE) are available in the settings of progression (VEN, non-covalent BTKi) and intolerance (acalabrutinib), how best to manage patients (pts) who discontinue (dc) VEN remains a key unanswered question. With the increased use of VEN in early lines of therapy (LOT; CLL 14, MURANO), the activity of BTK inhibitors (BTKi) and cellular therapies following VEN becomes a critical issue. No prospective study has addressed this question, and currently reported VEN clinical trials have limited information about subsequent treatments. While recent data describe VEN resistance mechanisms (Guieze 2018, Blombery 2019), the impact of VEN resistance on efficacy of post VEN therapies is unknown. To address this gap, we conducted an international study to identify a large cohort of pts who dc VEN and have been subsequently treated. Methods: We conducted an IRB approved multicenter (31 US, EU, South American sites, in partnership with UK CLL Forum and CORE registry), retrospective cohort study of CLL pts who dc VEN for any reason. We examined demographics, dc reasons, responses, survival, adverse events (AEs) and activity of post VEN therapies. Primary endpoints were overall response rate (ORR) and progression free survival (PFS) for the post VEN treatments stratified by treatment type (BTKi, PI3Ki and cellular therapy: CAR-T or alloHSCT). ORR was defined by iwCLL criteria and PFS was defined from VEN dc to disease progression (PD), death, or last follow up for next treatment. Pts were further stratified by BTKi (resistant / intolerant) and PI3Ki exposure prior to VEN. PFS-2 was defined as time from VEN start to tumor progression on IBR or death from any cause. Results: 326 CLL pts who dc VEN in the front line (4%) and R/R settings (96%) were identified. The cohort was 69% male, 87% white, median (med) age 66 (38-91) at VEN start, 27% treated with VEN based combinations (n=88, med 6 cycles anti-CD20 abs). Pre VEN prognostic features: 82% IGHV unmutated (n tested=166), 47% del17p (n=306), 45% TP53 mut (n=217), 39% complex karyotype (n=273), 23% BTK mut (n=79), 18% NOTCH1 mut (n=103), 10% PLCγ2 mut (n=74). Pts received med 3 therapies (0-11) prior to VEN; 40% were BTKi naïve (n=130), 60% were BTKi exposed (196) and 81% were IDE naïve (n=263). Most common reasons for VEN dc were PD (38%), AE (20%), Richter's transformation (RT, 14%), 8% pt preference, and HSCT 5%. Of 326 pts who dc VEN, 188 (58%) were treated with a subsequent LOT, 61 are alive and untreated and 77 died prior to a subsequent LOT. Post VEN sequencing analyses focused on BTKi, PI3Ki and cellular therapy (CAR-T or alloHSCT) activities following VEN dc (Table1). ORR to BTKi was 84% (n=44) vs. 54% (n=30, p Conclusions: In the largest experience of therapies following VEN dc in CLL, we demonstrated that therapy selection following VEN requires consideration of prior novel agent exposure and reasons for discontinuation. For BTKi naïve pts, selection of a covalently binding BTKi results in high ORR and durable remissions. PFS-2 data provide reassurance for using VEN prior to IBR. For BTKi exposed pts, BTK inhibition is not effective in the setting of BTKi resistance but should be considered if prior BTKi intolerance. PI3K inhibition following VEN does not appear to result in durable remissions even in PI3Ki naïve pts, suggesting possible overlap in resistance mechanisms (BTK or VEN with PI3K). We conclude that BTKi in naïve or previously responsive pts and alloHSCT following VEN appear to be the most effective strategies with durable responses. These data suggest that a number of effective regimens exist for post VEN pts, providing support for VEN use earlier in the course of CLL. Disclosures Mato: Acerta: Consultancy; LOXO: Consultancy, Research Funding; DTRM Biopharma: Research Funding; Janssen: Consultancy; Gilead: Research Funding; Pharmacyclics: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Johnson & Johnson: Consultancy, Research Funding; TG Therapeutics: Consultancy, Other: DSMB member , Research Funding; AbbVie: Consultancy, Research Funding; Sunesis: Consultancy, Research Funding; AstraZeneca: Consultancy, Research Funding; Celgene: Consultancy. Roeker:AbbVie: Equity Ownership; Abbott Laboratories: Equity Ownership. Eyre:Gilead: Consultancy, Other: Research support, Speakers Bureau; Roche: Honoraria; Abbvie: Honoraria, Other: Travel to Conferences; Janssen: Honoraria, Other: Travel to Conferences ; Takeda: Other: Travel to Conferences . Jacobs:Pharmacyclics LLC, an AbbVie Company: Research Funding, Speakers Bureau; AstraZeneca: Speakers Bureau; AbbVie: Consultancy, Speakers Bureau; Genentech: Speakers Bureau; JUNO: Consultancy; Gilead: Consultancy; TG Therapeutics: Honoraria, Research Funding. Hill:TG therapeutics: Research Funding; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genentech: Consultancy, Research Funding; Kite: Consultancy, Honoraria; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AstraZeneca: Consultancy, Honoraria; Celegene: Consultancy, Honoraria, Research Funding; Seattle Genetics: Consultancy, Honoraria; Takeda: Research Funding; Amgen: Research Funding. Lamanna:Celgene: Consultancy; Infinity/ Verastem: Research Funding; Ming: Research Funding; TG Therapeutics: Research Funding; Oncternal: Research Funding. Brander:Tolero: Research Funding; MEI: Research Funding; BeiGene: Research Funding; DTRM Biopharma: Research Funding; Genentech: Consultancy, Honoraria, Research Funding; Teva: Consultancy, Honoraria; TG Therapeutics: Consultancy, Honoraria, Research Funding; AstraZeneca: Consultancy, Research Funding; Novartis: Consultancy; Pharmacyclics LLC, an AbbVie Company: Consultancy; AbbVie: Consultancy, Honoraria, Research Funding; Acerta: Research Funding. Shadman:AbbVie: Consultancy, Research Funding; Astra Zeneca: Consultancy; BeiGene: Research Funding; TG Therapeutic: Research Funding; ADC Therapeutics: Consultancy; Atara Biotherapeutics: Consultancy; Verastem: Consultancy; Acerta Pharma: Research Funding; Sunesis: Research Funding; Mustang Bio: Research Funding; Celgene: Research Funding; Pharmacyclics: Consultancy, Research Funding; Sound Biologics: Consultancy; Genentech: Consultancy, Research Funding; Gilead: Consultancy, Research Funding. Ujjani:AstraZeneca: Consultancy; Genentech: Consultancy; Rigel: Consultancy; Gilead: Consultancy; Abbvie: Research Funding; Pharmacyclics: Research Funding. Perini:Janssen: Other: Advisory Board; Abbvie: Other: Advisory Board; AstraZeneca: Other: Advisory Board. Pinilla Ibarz:Sanofi: Speakers Bureau; Bayer: Speakers Bureau; Novartis: Consultancy; Bristol-Myers Squibb: Consultancy; Takeda: Consultancy, Speakers Bureau; Abbvie: Consultancy, Speakers Bureau; Janssen: Consultancy, Speakers Bureau; Teva: Consultancy; TG Therapeutics: Consultancy. Barrientos:Pharmacyclics: Consultancy, Research Funding; Abbvie: Consultancy, Research Funding; Janssen: Consultancy; Gilead: Consultancy; Genentech: Consultancy. Skarbnik:Seattle Genetics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Genentech: Honoraria, Speakers Bureau; CLL Society: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Speakers Bureau; Novartis: Speakers Bureau; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pharmacyclics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Verastem Oncology: Honoraria, Research Funding, Speakers Bureau; Kite Pharma: Honoraria, Speakers Bureau; Gilead Sciences: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Consultancy, Honoraria, Speakers Bureau; Acerta: Research Funding. Pagel:AstraZeneca: Consultancy; Gilead Sciences: Consultancy; Pharmacyclics: Consultancy. Choi:Abbvie: Consultancy, Research Funding, Speakers Bureau; Genentech: Consultancy, Speakers Bureau; Pharmacyclics: Consultancy, Research Funding, Speakers Bureau; Rigel: Consultancy, Research Funding; Gilead: Consultancy, Speakers Bureau; Oncternal: Research Funding. Coombs:H3 Biomedicine: Research Funding. Barr:Janssen: Consultancy; Astra Zeneca: Consultancy, Research Funding; Merck: Consultancy; Verastem: Consultancy; Gilead: Consultancy; Genentech: Consultancy; Pharmacyclics LLC, an AbbVie company: Consultancy, Research Funding; Celgene: Consultancy; TG Therapeutics: Consultancy, Research Funding; Seattle Genetics: Consultancy; AbbVie: Consultancy. Portell:Xencor: Research Funding; Roche/Genentech: Research Funding; Infinity: Research Funding; TG Therapeutics: Research Funding; AbbVie: Research Funding; Pharmacyclics: Consultancy; Janssen: Consultancy; Genentech: Consultancy, Research Funding; Amgen: Consultancy; Bayer: Consultancy; BeiGene: Consultancy, Research Funding; Kite: Consultancy, Research Funding; Acerta/AstraZeneca: Research Funding. Schuster:AstraZeneca: Honoraria; AbbVie: Honoraria, Research Funding; Acerta: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Loxo Oncology: Honoraria; Pfizer: Honoraria; Nordic Nanovector: Honoraria; Pharmacyclics: Honoraria, Research Funding; Novartis: Honoraria, Patents & Royalties: Combination Therapies of CAR and PD-1 Inhibitors with royalties paid to Novartis, Research Funding; Merck: Honoraria, Research Funding; Genentech: Honoraria, Research Funding; Gilead: Honoraria, Research Funding. Martinez-Calle:ABBVIE: Other: Travel support. Munir:AbbVie: Honoraria; Alexion: Honoraria; Gilead: Honoraria; Janssen: Honoraria; Novartis: Honoraria; Roche: Honoraria; Morphosys: Consultancy, Membership on an entity's Board of Directors or advisory committees; Sunesis: Consultancy; Pharmacyclics: Other: TBC; Acerta: Membership on an entity's Board of Directors or advisory committees. Nabhan:Aptitude Health: Employment. King:Astrazeneca: Other: Advisory board; Genentech: Other: Advisory Board ; Incyte: Other: Advisory Board. Zelenetz:Karyopharm: Consultancy, Membership on an entity's Board of Directors or advisory committees; MEI Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astra-Zeneca: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pharmacyclics/AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Beigene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Beigene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Verastem: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pharmacyclics/AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees; Verastem: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; DAVA Oncology: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; DAVA Oncology: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees; Genentech/Roche: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Consultancy, Membership on an entity's Board of Directors or advisory committees; MEI Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astra-Zeneca: Consultancy, Membership on an entity's Board of Directors or advisory committees; Genentech/Roche: Consultancy, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Consultancy, Membership on an entity's Board of Directors or advisory committees; Morphosys: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Morphosys: Consultancy, Membership on an entity's Board of Directors or advisory committees. Cheson:Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Symbios: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Trillium: Research Funding; TG Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Seattle Genetics: Research Funding; Bristol Myers Squibb: Research Funding; Portola: Research Funding; Kite: Research Funding; Gilead: Research Funding; Epizyme: Research Funding; Morphosys: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Membership on an entity's Board of Directors or advisory committees; Acerta: Consultancy, Research Funding. Fox:Gilead: Consultancy; Janssen: Consultancy; Celgene: Consultancy; AbbVie: Consultancy; Sunesis: Consultancy; Takeda Pharmaceuticals: Consultancy; Atara Biotherapeutics: Consultancy; Adienne: Other: Travel Support. Allan:Sunesis Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Consultancy; Janssen: Consultancy, Honoraria; Pharmacyclics LLC, an AbbVie company: Consultancy; Verastem Oncology, Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees; AbbVie, Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Acerta Pharma: Consultancy.
- Published
- 2019
36. Identification of Genotype-Specific Therapeutic Vulnerabilities By Comparative Dynamic BH3 Profiling Analysis of Human and Murine CLL
- Author
-
Donna Neuberg, Matthew S. Davids, Heather Joyal, Rebecca Valentin, Elisa Ten Hacken, Shanye Yin, Laura Z. Rassenti, Anthony Letai, Emanuela M. Ghia, Lili Wang, Catherine J. Wu, Thomas J. Kipps, Salma Parvin, and Ruben D. Carrasco
- Subjects
Trametinib ,Oncology ,Sorafenib ,medicine.medical_specialty ,business.industry ,Chronic lymphocytic leukemia ,Immunology ,Cell Biology ,Hematology ,Signal transduction inhibitor ,medicine.disease ,Biochemistry ,Duvelisib ,chemistry.chemical_compound ,MTOR Kinase Inhibitor AZD8055 ,chemistry ,Internal medicine ,Ibrutinib ,Medicine ,business ,Idelalisib ,medicine.drug - Abstract
Mouse models that reflect human cancer genetics are valuable tools for dissecting disease mechanisms and for discovering novel therapeutic vulnerabilities, although such models for chronic lymphocytic leukemia (CLL) have been largely lacking until now. The most commonly used CLL model, the TCL1 mouse, is based on constitutive B-cell restricted expression of the human TCL1 oncogene, but does not reflect CLL genetics per se. Novel mouse models, such as the MDR mice (modeling del[13q]) and our recently developed Sf3b1mut/Atmdel double mutant mice, are able to recapitulate the expression of CLL driver mutations and develop CLL-like disease with a rather delayed onset (~18 months), mimicking the indolent nature of human CLL. To understand the utility and limitations of these murine CLL models as preclinical platforms for drug screening, we compared BH3 profiles of murine and genetically-matched human CLLs in response to a panel of drugs including 3 BCL-2 signaling inhibitors (BCL-2i, including BCL-2 inhibitor ABT-199, BCL-2/XL inhibitor ABT-263, BCL-XL inhibitor A133 and MCL1 inhibitor S63845) and 7 BCR signaling inhibitors (BCRi, including PI3K inhibitors idelalisib, duvelisib, umbralisib, the mTOR kinase inhibitor AZD8055, the MEK1/MEK2 inhibitor trametinib, the BTK inhibitor ibrutinib and the BRAF inhibitor sorafenib). BH3 profiling is based on exposure of cellular mitochondria to synthetic peptides that mimic the BH3 domains of pro-apoptotic BCL-2 family members followed by measurement of mitochondrial permeabilization using flow-cytometry based detection of cytochrome-c loss. We measured apoptotic priming (BIM peptide), and the dependence on BCL-2 or MCL-1 (BAD and MS1 peptides, respectively) after drug exposure. The assay was miniaturized into 384 well plates, where OP9 stromal cells (used as a support to limit human and murine CLL spontaneous apoptosis) were coated at a 1:10 ratio compared to CLL cells, before treatment with drugs (experiments were performed in duplicate). As a starting point, to determine whether primary CLLs with different mutated drivers have distinct therapeutic vulnerabilities, we profiled 6 primary human CLLs (4 carrying del[13q] and 2 carrying del[13q], [11q] and mutant SF3B1). We observed a general increase in BCL-2 dependence in response to the aggregate set of 7 BCRi as compared to 3 BCL-2i (ABT-199, ABT-263 and A133) (p Similar analyses were performed using splenocytes from NSG recipient mice, transplanted with murine CLLs carrying either MDR or Sf3b1mut/Atmdel mutations. Reduced activity of the MCL1 inhibitor S63845 was observed in murine CLLs compared to human CLLs (p=0.024, Mann-Whitney U test), consistent with the higher affinity of this compound for human, rather than murine MCL1. Notably, Sf3b1mut/Atmdel CLL cells were more resistant to the panel of BCRi than MDR cells (p=0.046, the Van Elteren test) and their dependence on BCL-2 for survival after BCRi was likewise diminished (p=0.018, the Van Elteren test), similar to human CLLs carrying del(13q)/(11q)-SF3B1. These results support the use of the Sf3b1mut/Atmdel line as a valuable platform for the exploration of therapeutic vulnerabilities of primary CLLs carrying the same genetic lesions. Altogether, these results suggest that both MDR and Sf3b1mut/Atmdel mouse lines are valuable tools for predicting therapeutic sensitivities of genetically matched primary CLLs. We are currently extending this approach to the testing of additional drugs, and to the validation of the most relevant compounds in larger cohorts of primary CLLs. Disclosures Valentin: Abbvie Inc: Other: Travel reimbursement; Roche: Other: Travel reimbursement. Kipps:Pharmacyclics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Membership on an entity's Board of Directors or advisory committees, Research Funding; Genentech, Inc.: Membership on an entity's Board of Directors or advisory committees, Research Funding; Velos-Bio: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Research Funding; Jannsen Pharmaceutical Companies of Johnson & Johnson: Honoraria, Membership on an entity's Board of Directors or advisory committees; AstraZeneca, Inc.: Membership on an entity's Board of Directors or advisory committees; Verastem: Membership on an entity's Board of Directors or advisory committees. Neuberg:Pharmacyclics: Research Funding; Madrigal Pharmaceuticals: Equity Ownership; Celgene: Research Funding. Davids:Acerta Pharma, Ascentage Pharma, Genentech, MEI pharma, Pharmacyclics, Surface Oncology, TG Therapeutics, Verastem: Research Funding; Research to Practice: Honoraria; AbbVie, Acerta Pharma, Adaptive, Biotechnologies, Astra-Zeneca, Genentech, Gilead Sciences, Janssen, Pharmacyclics, TG therapeutics: Membership on an entity's Board of Directors or advisory committees; AbbVie, Astra-Zeneca, Genentech, Janssen, MEI, Pharmacyclics, Syros Pharmaceuticals, Verastem: Consultancy. Letai:Zeno Pharmaceuticals, Vivid Bioscience, Flash Therapeutics, Dialectic Therapeutics: Membership on an entity's Board of Directors or advisory committees, Other: Cofounder or Advisory Board member; AbbVie, AstraZeneca, Novartis: Consultancy, Research Funding. Wu:Neon Therapeutics: Other: Member, Advisory Board; Pharmacyclics: Research Funding.
- Published
- 2019
37. Expression of IL-17RA promotes cancer stem-like properties of colorectal cancer cells by Stat3 activation
- Author
-
C.-Y. Yang, Chien-Hsing Lin, and Jeng-Kai Jiang
- Subjects
business.industry ,Angiogenesis ,LGR5 ,Cancer ,Hematology ,Signal transduction inhibitor ,Stem cell marker ,medicine.disease ,Oncology ,SOX2 ,Cancer research ,Medicine ,Epithelial–mesenchymal transition ,Stem cell ,business - Abstract
Background Colorectal cancer stem cells (CSCs) serve crucial functions in tumor relapse, metastasis and therapy failure. Interleukin 17 receptor A (IL-17RA) is a potent mediator in the pathogenesis and progression of colorectal cancer. Our previous study showed that IL-17RA could promote angiogenesis, tumor growth and metastasis. In addition, IL-17RA was correlated with CRC recurrence. Recent study showed IL-17RA play a potential role in self-renewal in glioma stem cells. This study aims to evaluate the functional role and mechanism of IL-17RA in colorectal CSCs. Methods In this study, IL-17RA stable overexpression cells were used to examine the change of sphere formation, proliferation, CSC markers and epithelial-mesenchymal transition biomarkers and the regulation mechanism of signaling pathway. The specific IL-17RA signaling inhibitors were used to evaluate the function of the IL-17RA signaling affecting the CSC markers, epithelial-mesenchymal transition gene expression and sphere formation. Results We observed high IL-17RA expression can significantly promote the self-renewal by increasing stem cell markers CD133, ALDH1, Lgr5 and Sox2 expression and the ability to form tumorsphere in SW480 and SW620 cells. In additional, IL-17RA overexpression markedly increased chemoresistant ability and mesenchymal markers Vimentin, Slug, Snail, Zeb1expression. The STAT3 inhibitor remarkably decreased the CD133 stem cell gene, drug resistance and tumorsphere ability. Conclusions IL-17RA can enhance the self-renewal ability via activating the STAT3 pathway. Therefore, we expect that IL-17RA could serves as a prognosis marker and a potential therapeutic target in CRC. Legal entity responsible for the study Chih-Yung Yang. Funding Has not received any funding. Disclosure All authors have declared no conflicts of interest.
- Published
- 2019
38. Inositol Deficiency Induces Autophagy Impairing via PI3K/Akt/mTOR/p70S6K Signaling (P11-032-19)
- Author
-
Jianhua Wang and Jin Guo
- Subjects
Maternal, Perinatal and Pediatric Nutrition ,Nutrition and Dietetics ,Proto-Oncogene Proteins c-akt ,Autophagy ,Medicine (miscellaneous) ,Signal transduction inhibitor ,Cell biology ,chemistry.chemical_compound ,chemistry ,P70S6 kinase ,Inositol ,Signal transduction ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Food Science - Abstract
OBJECTIVES: Our previous case-control study found that maternal MI deficiency was associated with an increased risk of NTDs. Bioinformatics analysis showed that PI3K/Akt/mTOR/p70S6K signaling pathway might be one of the important regulatory mechanisms of inositol deficiency-induced NTDs. So, we intended to explore the possible mechanisms of PI3K/Akt/mTOR/p70S6K signaling in inositol deficiency-induced neural tube defects (NTDs) in this study. METHODS: The activity of the PI3-kinase in MI deficiency NE-4C cells was detected by the PI3-kinase ELISA kit. Using the method of western blot, we analyzed the activity of the PI3K/Akt/mTore/p70S6K signaling pathway and the level of LC3B. And the LC3B levels were also detected by electron microscopy. RESULTS: The result showed that PI3K activity was significantly higher in the Li2CO3-treated group than in the control cells (P
- Published
- 2019
39. EPEN-11. PROTEASOME AND HDAC INHIBITORS ARE CYTOTOXIC AGAINST RELA-FUSION PEDIATRIC EPENDYMOMA CELLS
- Author
-
Ian F. Pollack, Marissa Campagna, Rajeev Kumar, Gary Kohanbash, Xiangjuan Ma, Alberto Broniscer, Sameer Agnihotri, James Felker, and Lauren McCarl
- Subjects
Ependymoma ,Cancer Research ,business.industry ,Signal transduction inhibitor ,medicine.disease ,NFKB1 ,chemistry.chemical_compound ,Oncology ,Proteasome ,chemistry ,Panobinostat ,medicine ,Cancer research ,Cytotoxic T cell ,Pediatric ependymoma ,Neurology (clinical) ,Histone deacetylase ,business - Abstract
Ependymomas are the third most common pediatric brain tumor. Nine ependymoma subgroups have been described based on tumor location and genetics. Of supratentorial ependymomas, the C11orf95-RELA fusion (ST-EPN-RELA) subgroup is the most aggressive. The RELA encoded protein is an NF-κB transcription factor family member and activates NF-κB signaling. It has been reported that NF-κB and other pathways are important for ST-EPN-RELA tumors. We therefore hypothesized that ST-EPN-RELA cells may be sensitive to NF-κB signaling cascade inhibitors. To test our hypothesis we evaluated the cytotoxicity of multiple NF-kB signaling inhibitors on DKFZ-EP1NS RELA-fusion positive ependymoma cells and on our established patient-derived ST-EPN-RELA line (CPITT-1). Cells were treated with a single drug or combinations of drugs at concentrations ranging from 10 µM- 0.01 nM for 3–4 days and a WST-1 assay was used to assess viability. Of multiple inhibitors initially tested, Trichostatin A, a histone deacetylase (HDAC) inhibitor, in combination with MG-132, a proteasome inhibitor, displayed the highest level of cytotocity against both RELA-fusion lines. These findings led us to test clinically applicable drugs, including Marizomib, a brain penetrant proteasome inhibitor, and HDAC inhibitors, Panobinostat and Valproic acid. For DKFZ-EP1NS cells, the IC(50) for Marizomib alone was 40.82 nM, Marizomib in combination with Panobinostat had an IC(50) of 7.4 nM, and Marizomib in combination with Valproic Acid had an IC(50) of 44.19. For CPITT-1 cells, the IC(50) for Marizomib alone was 25.8nM, Marizomib in combination with Panobinostat had and IC(50) of 3.07 nM, and Marizomib in combination with Valproic Acid had an IC(50) of 27.04 nM. Overall, we observed that combination of proteasome and HDAC inhibitors are highly toxic to the RELA-fusion ependymoma cells tested. Further studies are warranted to evaluate the use these inhibitors for treating ST-EPN-RELA tumors.
- Published
- 2019
40. c-KIT-expressing Ewing tumour cells are insensitive to imatinib mesylate (STI571).
- Author
-
Hotfilder, Marc, Lanvers, Claudia, Jürgens, Heribert, Boos, Joachim, and Vormoor, Josef
- Subjects
EWING'S sarcoma ,IMATINIB ,PROTEIN-tyrosine kinase inhibitors ,METHANESULFONATES ,PLATELET-derived growth factor ,BLOOD proteins ,CANCER cell proliferation - Abstract
Purpose. In order to determine whether Ewing tumour patients may be potential candidates for imatinib mesylate therapy, we analysed the expression of the currently known imatinib mesylate-sensitive tyrosine kinases and tested sensitivity to imatinib mesylate in a panel of eight Ewing tumour cell lines in vitro. Methods. Expression of the different tyrosine kinases was assessed by flow cytometry and RT-PCR. Sensitivity to imatinib mesylate was analysed using a standard MTT proliferation assay. Results. Flow cytometric and RT-PCR analyses in a panel of eight Ewing tumour cell lines demonstrated expression of several imatinib mesylate-sensitive tyrosine kinases, including c-KIT, platelet-derived growth factor receptor, c-ABL and c-ARG. However, in the MTT proliferation assay, all eight Ewing tumour cell lines were found to be resistant to imatinib mesylate at concentrations ranging from 0.1 to 10 µM. Conclusions. Despite the expression of imatinib mesylate-sensitive tyrosine kinases, Ewing tumour cells proved resistant to imatinib mesylate in vitro. This observation has implications for the selection of patients for experimental therapy with imatinib mesylate. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
41. Elevated SGK1 predicts resistance of breast cancer cells to Akt inhibitors
- Author
-
Eeva Sommer, Dario R. Alessi, Barry R. Davies, Darren Cross, Sylvie Guichard, and Hannah Dry
- Subjects
Molecular Sequence Data ,Breast Neoplasms ,Signal transduction inhibitor ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,mTORC2 ,Immediate early protein ,Immediate-Early Proteins ,Predictive Value of Tests ,Cell Line, Tumor ,Animals ,Humans ,Amino Acid Sequence ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sheep ,Cell growth ,Kinase ,Cell Biology ,Growth Inhibitors ,HEK293 Cells ,Drug Resistance, Neoplasm ,Cancer research ,Phosphorylation ,Female ,Proto-Oncogene Proteins c-akt - Abstract
The majority of human cancers harbour mutations promoting activation of the Akt protein kinase, and Akt inhibitors are being evaluated in clinical trials. An important question concerns the understanding of the innate mechanisms that confer resistance of tumour cells to Akt inhibitors. SGK (serum- and glucocorticoid-regulated kinase) is closely related to Akt and controlled by identical upstream regulators {PI3K (phosphoinositide 3-kinase), PDK1 (phosphoinositide-dependent kinase 1) and mTORC2 [mTOR (mammalian target of rapamycin) complex 2]}. Mutations that trigger activation of Akt would also stimulate SGK. Moreover, Akt and SGK possess analogous substrate specificities and are likely to phosphorylate overlapping substrates to promote proliferation. To investigate whether cancers possessing high SGK activity could possess innate resistance to Akt-specific inhibitors (that do not target SGK), we analysed SGK levels and sensitivity of a panel of breast cancer cells towards two distinct Akt inhibitors currently in clinical trials (AZD5363 and MK-2206). This revealed a number of Akt-inhibitor-resistant lines displaying markedly elevated SGK1 that also exhibited significant phosphorylation of the SGK1 substrate NDRG1 [N-Myc (neuroblastoma-derived Myc) downstream-regulated gene 1]. In contrast, most Akt-inhibitor-sensitive cell lines displayed low/undetectable levels of SGK1. Intriguingly, despite low SGK1 levels, several Akt-inhibitor-sensitive cells showed marked NDRG1 phosphorylation that was, unlike in the resistant cells, suppressed by Akt inhibitors. SGK1 knockdown markedly reduced proliferation of Akt-inhibitor-resistant, but not -sensitive, cells. Furthermore, treatment of Akt-inhibitor-resistant cells with an mTOR inhibitor suppressed proliferation and led to inhibition of SGK1. The results of the present study suggest that monitoring SGK1 levels as well as responses of NDRG1 phosphorylation to Akt inhibitor administration could have a use in predicting the sensitivity of tumours to compounds that target Akt. Our findings highlight the therapeutic potential that SGK inhibitors or dual Akt/SGK inhibitors might have for treatment of cancers displaying elevated SGK activity.
- Published
- 2013
42. Role of CXCR4 in the Pathogenesis of Acute Myeloid Leukemia
- Author
-
Sigal Tavor and Amnon Peled
- Subjects
Receptors, CXCR4 ,Stromal cell ,Medicine (miscellaneous) ,Antineoplastic Agents ,Signal transduction inhibitor ,Review ,Biology ,CXCR4 ,Immune system ,AML ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Bone marrow ,Progenitor cell ,Stem Cell Niche ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,neoplasms ,Myeloid leukemia ,CXCL12 ,medicine.disease ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Cellular Microenvironment ,Immunology ,Microenvironment ,Signal Transduction - Abstract
The Chemokine receptor CXCR4 and its ligand stromal derived factor-1 (SDF-1/CXCL12) are important players involved in cross-talk between leukemia cells and the bone marrow (BM) microenvironment. CXCR4 expression is associated with poor prognosis in AML patients with and without the mutated FLT3 gene. CXCL12 which is constrictively secreted from the BM stroma and AML cells is critical for the survival and retention of AML cells within the BM. In vitro, CXCR4 antagonists were shown to inhibit the migration of AML cells in response to CXCL12. In addition, such antagonists were shown to inhibit the survival and colony forming potential of AML cells and abrogate the protective effects of stromal cells on chemotherapy-induced apoptosis in AML cells. In vivo, using immune deficient mouse models, CXCR4 antagonists were found to induce the mobilization of AML cells and progenitor cells into the circulation and enhance anti leukemic effects of chemotherapy. The hypothesis that CXCL12/CXCR4 interactions contribute to the resistance of AML cells to signal transduction inhibitor- and chemotherapy-induced apoptosis is currently being tested in a series of Phase I/II studies in humans.
- Published
- 2013
43. Outcomes of Follicular Lymphoma (FL) with Early Progression (EP): Does Choice of Second Line Therapy Impact the Course of Disease?
- Author
-
Ahmad Hanif, Francisco J. Hernandez-Ilizaliturri, Pallawi Torka, Kris Attwood, and Sumera Khan
- Subjects
Oncology ,Brachial Plexus Neuritis ,medicine.medical_specialty ,business.industry ,Surrogate endpoint ,medicine.medical_treatment ,Immunology ,Follicular lymphoma ,Cell Biology ,Hematology ,Signal transduction inhibitor ,Immunotherapy ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Internal medicine ,medicine ,Rituximab ,Biological response modifiers ,business ,medicine.drug - Abstract
Background: The spectrum of FL varies from an indolent disease course spanning decades to early transformation and death. Longitudinal studies have identified a subset of high risk patients (pts) who progress within 24 months of frontline therapy. It is still unclear how best to treat these patients and as yet, there is no standard 2nd line therapy for FL. Given the multitude of treatments available and the heterogenous disease course, it is challenging to compare outcomes of 2nd line therapy. It is unclear if the efficacy of second-line therapy in FL is influenced by the type of therapy, disease biology (early vs. late progression) or both. We conducted a single institute, retrospective study to determine the clinical benefit of 2nd line therapy in FL and to evaluate if any particular type of therapy was associated with improved outcomes in FL patients with early progression (EP). Methods: All patients with relapsed/refractory FL treated at our Institute between 1990 and 2014 were included. Patients were included if they received anti-CD20 monoclonal antibody (mAb)-based therapy in the first-line setting and completed both, first-line and second-line therapy at our Institution. Demographic, clinical, pathological and outcomes data was collected by retrospective chart review. Clinical endpoints included overall response rate (ORR), progression-free survival (PFS), time to next treatment (TTNT), and overall survival (OS). Treatments were divided in two groups for comparison: immunotherapy alone (anti-CD20 mAb) (IT) or chemo-immunotherapy (CIT). Differences in clinical outcomes were analyzed between FL patients with early progression after 1st line therapy (EP, i.e. progression ≤ 2yrs) vs. FL with late relapse (LP, i.e. progression > 2yrs). Comparisons were made using the Mann-Whitney U and Person chi-square tests as appropriate. Survival outcomes were assessed using standard Kaplan-Meier methods. All analyses were conducted in SAS v9.4 (Cary, NC) at a significance level of 0.05. Results: A total of 537 newly diagnosed FL pts were identified of which 291 pts received 1st line therapy at our Institute. IT or CIT was given to 19.6% and 80.4% pts respectively. IT treated pts were older (median age: 61y vs 57y, p=0.033) and had lower FLIPI scores (1.5 vs 2.1, p Conclusion: FL pts with LR have excellent outcomes to 2nd line therapy and treatment selection could be determined by the agent(s) toxicity profile. In contrast, EP predicts poor clinical outcomes with short duration of response to standard IT or CIT. Therapeutic approaches incorporating clinically available targeted agents (i.e. immunomodulatory agents of B-cell receptor signaling inhibitors) or novel agents in the context of clinical trials, may provide a more effective disease control in this subgroup. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
44. Signal Transduction Inhibitor Therapy for Lymphoma
- Author
-
Mamta Gupta and Thomas E. Witzig
- Subjects
Everolimus ,Lymphoma ,TOR Serine-Threonine Kinases ,Chronic lymphocytic leukemia ,Receptors, Antigen, B-Cell ,Syk ,Antineoplastic Agents ,Hematology ,Signal transduction inhibitor ,Biology ,medicine.disease ,Fostamatinib ,Article ,BCL10 ,chemistry.chemical_compound ,Enzastaurin ,chemistry ,hemic and lymphatic diseases ,medicine ,Cancer research ,Humans ,Protein Kinase Inhibitors ,Diffuse large B-cell lymphoma ,Signal Transduction ,medicine.drug - Abstract
Current research in lymphoma is focused on two areas of lymphoma biology—the signal transduction pathways used to maintain the growth of malignant lymphocytes and the role of the tumor microenvironment in lymphoma growth and survival. This review focuses on three signaling pathways: the phosphatidylinositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway, the B-cell receptor/spleen tyrosine kinase (BCR/Syk) pathway, and the protein kinase C-beta (PKC-β) pathway, known to be important to lymphoma cells. The mTOR inhibitors temsirolimus and everolimus have demonstrated antitumor activity in all types of lymphoma, the Syk inhibitor fostamatinib has activity in diffuse large B-cell lymphoma and chronic lymphocytic leukemia, and the PKC-β inhibitor enzastaurin is being used as consolidation therapy after remission in diffuse large B-cell lymphoma. This review discusses the biology behind the development of each new agent and the results of initial clinical trials. The goal is to provide the hematologist/oncologist background information on these new agents and understand their current and potential role in the management of patients.
- Published
- 2010
45. Use of nuclear-localized androgen receptor splice variant 7 protein in CTCs after 1st androgen receptor signaling inhibitor (ARSi) as a predictive biomarker for overall survival on a second ARSi or taxane chemotherapy in metastatic castration-resistant prostate cancer (mCRPC)
- Author
-
Ryan Dittamore, M. Hulling, Emily Carbone, Howard I. Scher, and Ryon P. Graf
- Subjects
Chemotherapy ,Taxane ,business.industry ,medicine.medical_treatment ,Hematology ,Signal transduction inhibitor ,Androgen Receptor Splice Variant 7 ,medicine.disease ,Chemotherapy regimen ,Androgen receptor ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Circulating tumor cell ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,030217 neurology & neurosurgery - Published
- 2018
46. Folate decorated polymeric micelles for targeted delivery of the kinase inhibitor dactolisib to cancer cells.
- Author
-
Shi H, van Steenbergen MJ, Lou B, Liu Y, Hennink WE, and Kok RJ
- Subjects
- A549 Cells, Antineoplastic Agents chemistry, Antineoplastic Agents metabolism, Cell Survival, Drug Compounding, Drug Liberation, Drug Stability, Folic Acid chemistry, Folic Acid Transporters metabolism, Humans, Imidazoles chemistry, Imidazoles metabolism, Lung Neoplasms enzymology, Lung Neoplasms pathology, Micelles, Phosphoinositide-3 Kinase Inhibitors chemistry, Phosphoinositide-3 Kinase Inhibitors metabolism, Quinolines chemistry, Quinolines metabolism, Signal Transduction, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases metabolism, Acrylic Resins chemistry, Antineoplastic Agents pharmacology, Drug Carriers, Folic Acid metabolism, Imidazoles pharmacology, Lung Neoplasms drug therapy, Phosphoinositide-3 Kinase Inhibitors pharmacology, Polyethylene Glycols chemistry, Quinolines pharmacology
- Abstract
One of the main challenges in clinical translation of polymeric micelles is retention of the drug in the nanocarrier system upon its systemic administration. Core crosslinking and coupling of the drug to the micellar backbone are common strategies to overcome these issues. In the present study, polymeric micelles were prepared for tumor cell targeting of the kinase inhibitor dactolisib which inhibits both the mammalian Target of Rapamycin (mTOR) kinase and phosphatidylinositol-3-kinase (PI3K). We employed platinum(II)-based linker chemistry to couple dactolisib to the core of poly(ethylene glycol)-b-poly(acrylic acid) (PEG-b-PAA) polymeric micelles. The formed dactolisib-PEG-PAA unimers are amphiphilic and self-assemble in an aqueous milieu into core-shell polymeric micelles. Folate was conjugated onto the surface of the micelles to yield folate-decorated polymeric micelles which can target folate receptor over-expressing tumor cells. Fluorescently labeled polymeric micelles were prepared using a lissamine-platinum complex linked in a similar manner as dactolisib. Dactolisib polymeric micelles showed good colloidal stability in water and released the coupled drug in buffers containing chloride or glutathione. Folate decorated micelles were avidly internalized by folate-receptor-positive KB cells and displayed targeted cellular cytotoxicity at 50-75 nM IC
50 . In conclusion, we have prepared a novel type of folate-receptor targeted polymeric micelles in which platinum(II) linker chemistry modulates drug retention and sustained release of the coupled inhibitor dactolisib., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
47. Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML
- Author
-
Elihu H. Estey, Ismael Samudio, Olga Frolova, Sergej Konoplev, Xiaoyang Ling, Mark J. Levis, Michael Andreeff, Zhihong Zeng, Marina Konopleva, Joshua B. Rubin, Rui Yu Wang, Yue Xi Shi, and Robert R. Negrin
- Subjects
Receptors, CXCR4 ,Stromal cell ,Myeloid ,Pyridines ,Blotting, Western ,Immunology ,Antineoplastic Agents ,Apoptosis ,Signal transduction inhibitor ,Biology ,Biochemistry ,Immunoenzyme Techniques ,Mice ,Cell Movement ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Cells, Cultured ,Cell Proliferation ,Mice, Inbred BALB C ,Leukemia, Experimental ,Myeloid Neoplasia ,Chemotaxis ,Myeloid leukemia ,Cell Biology ,Hematology ,Flow Cytometry ,medicine.disease ,Chemokine CXCL12 ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Drug Resistance, Neoplasm ,Mutation ,Fms-Like Tyrosine Kinase 3 ,Cancer research ,Bone marrow ,Stromal Cells - Abstract
SDF-1α/CXCR4 signaling plays a key role in leukemia/bone marrow microenvironment interactions. We previously reported that bone marrow–derived stromal cells inhibit chemotherapy-induced apoptosis in acute myeloid leukemia (AML). Here we demonstrate that the CXCR4 inhibitor AMD3465 antagonized stromal-derived factor 1α (SDF-1α)–induced and stroma-induced chemotaxis and inhibited SDF-1α–induced activation of prosurvival signaling pathways in leukemic cells. Further, CXCR4 inhibition partially abrogated the protective effects of stromal cells on chemotherapy-induced apoptosis in AML cells. Fetal liver tyrosine kinase-3 (FLT3) gene mutations activate CXCR4 signaling, and coculture with stromal cells significantly diminished antileukemia effects of FLT3 inhibitors in cells with mutated FLT3. Notably, CXCR4 inhibition increased the sensitivity of FLT3-mutated leukemic cells to the apoptogenic effects of the FLT3 inhibitor sorafenib. In vivo studies demonstrated that AMD3465, alone or in combination with granulocyte colony-stimulating factor, induced mobilization of AML cells and progenitor cells into circulation and enhanced antileukemic effects of chemotherapy and sorafenib, resulting in markedly reduced leukemia burden and prolonged survival of the animals. These findings indicate that SDF-1α/CXCR4 interactions contribute to the resistance of leukemic cells to signal transduction inhibitor– and chemotherapy-induced apoptosis in systems mimicking the physiologic microenvironment. Disruption of these interactions with CXCR4 inhibitors represents a novel strategy of sensitizing leukemic cells by targeting their protective bone marrow microenvironment.
- Published
- 2009
48. Signal Transduction Inhibition of APCs Diminishes Th17 and Th1 Responses in Experimental Autoimmune Encephalomyelitis
- Author
-
Tianhong Wang, Katharine A. Whartenby, Donald Small, Mario Skarica, David Kardian, Peter A. Calabresi, and Erin McCadden
- Subjects
Encephalomyelitis, Autoimmune, Experimental ,medicine.medical_treatment ,T cell ,Blotting, Western ,Immunology ,Carbazoles ,Antigen-Presenting Cells ,Signal transduction inhibitor ,Biology ,Article ,Mice ,T-Lymphocyte Subsets ,Signal Transduction Inhibition ,medicine ,Animals ,Immunology and Allergy ,Enzyme Inhibitors ,Furans ,Antigen-presenting cell ,Microglia ,Interleukin-17 ,Experimental autoimmune encephalomyelitis ,Brain ,Th1 Cells ,medicine.disease ,Chemotaxis, Leukocyte ,Cytokine ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Cytokines ,Female ,Interleukin 17 ,Signal Transduction - Abstract
IL-17- and IFN-γ-secreting T cells play an important role in autoimmune responses in multiple sclerosis and the model system experimental autoimmune encephalomyelitis (EAE). Dendritic cells (DCs) in the periphery and microglia in the CNS are responsible for cytokine polarization and expansion of this T cell subset. Our results indicate that in vivo administration of a signal transduction inhibitor that targets DCs to mice with EAE led to a decrease in CNS infiltration of pathogenic Ag-specific T cells. Since this approach does not target T cells directly, we assessed the effects on the APCs that are involved in generating the T cell responses. Since in EAE and multiple sclerosis, both microglia and peripheral DCs are likely to contribute to disease, we utilized a bone marrow chimera system to distinguish between these two populations. These studies show that peripheral DCs are the primary target but that microglia are also modestly affected by CEP-701, as numbers and activation states of the cells in the CNS are decreased after therapy. Our results also showed a decrease in secretion of TNF-α, IL-6, and IL-23 by DCs as well as a decrease in expression of costimulatory molecules. We further determined that levels of phospho-Stat1, Stat3, Stat5, and NF-κB, which are signaling molecules that have been implicated in these pathways, were decreased. Thus, use of this class of signal transduction inhibitors may represent a novel method to treat autoimmunity by dampening the autoreactive polarizing condition driven by DCs.
- Published
- 2009
49. Differential Effect of Imatinib and Synergism of Combination Treatment with Chemotherapeutic Agents in Malignant Glioma Cells
- Author
-
Xiaoqing Tan, Nikolai G. Rainov, Alf Giese, Yucui Dong, Huan Ren, Ting Chao Chou, and Baofeng Yang
- Subjects
medicine.drug_class ,Apoptosis ,Signal transduction inhibitor ,Toxicology ,Piperazines ,Tyrosine-kinase inhibitor ,Cell Movement ,Cell Line, Tumor ,hemic and lymphatic diseases ,Glioma ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Receptors, Platelet-Derived Growth Factor ,neoplasms ,Cell Proliferation ,Pharmacology ,Temozolomide ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Cell Cycle ,Drug Synergism ,Imatinib ,General Medicine ,Cell cycle ,Flow Cytometry ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Proto-Oncogene Proteins c-kit ,Pyrimidines ,Imatinib mesylate ,Benzamides ,Imatinib Mesylate ,Cancer research ,business ,Tyrosine kinase ,Signal Transduction ,medicine.drug - Abstract
Imatinib mesylate (STI571, Gleevec) is a signal transduction inhibitor and novel anti-cancer agent. It selectively inhibits aberrantly activated tyrosine kinases in malignant cells, for example, bcr-abl in leukaemia, platelet-derived growth factor receptor and stem cell factor receptor (c-Kit) in solid cancers including malignant glioma. However, recently published clinical studies with imatinib monotherapy in patients with malignant glioma demonstrated only very modest anti-tumour activity. The aim of this study was to investigate the biological activity of imatinib, its cellular mechanisms of action and its synergism with other chemotherapeutic agents in human malignant glioma cells in culture. Expression of PDGF/R and c-Kit was analyzed by RT-PCR. Proliferation was measured by MTT assays and drug synergy was assessed by the Chou-Talalay method. Cell cycle and apoptosis were analyzed by flow cytometry and migration by monolayer migration assays. Multi-immunoblot was performed on imatinib-treated and control malignant glioma cells. Results indicate that imatinib is more effective in inhibiting cell colony formation and migration rather than proliferation. Imatinib treatment caused cell cycle arrest of glioma cells in G0-G1 or G2/M, with significant elevation of a few cyclin-dependent kinases. Furthermore, imatinib acted synergistically with chemotherapy agents, such as the DNA alkylating agent, temozolomide, and riboneucleotide reductase inhibitors, for example, hydroxyurea at varied effective dose levels. In conclusion, imatinib exerts varied biological effects on malignant glioma cells in culture. Synergistic interaction of imatinib with chemotherapy agents may be related to cell cycle control mechanisms and could be potentially important in a clinical setting.
- Published
- 2009
50. Sorafenib in Lung Cancer: Clinical Developments and Future Directions
- Author
-
George R. Blumenschein
- Subjects
Male ,Niacinamide ,Oncology ,Sorafenib ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Lung Neoplasms ,Platelet-derived growth factor ,Bevacizumab ,Pyridines ,Angiogenesis ,Signal transduction inhibitor ,NSCLC ,Risk Assessment ,Metastasis ,VEGFR ,chemistry.chemical_compound ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Lung cancer ,neoplasms ,Neoplasm Staging ,Randomized Controlled Trials as Topic ,Neovascularization, Pathologic ,Vascular Endothelial Growth Factors ,business.industry ,Phenylurea Compounds ,Benzenesulfonates ,medicine.disease ,Survival Analysis ,TKI ,Vascular endothelial growth factor ,Receptors, Vascular Endothelial Growth Factor ,Treatment Outcome ,Proangiogenic ,chemistry ,Raf kinase ,Female ,business ,Follow-Up Studies ,Forecasting ,medicine.drug - Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death in the United States. Angiogenesis, primarily mediated through vascular endothelial growth factor (VEGF), is one of the key steps in tumor growth and metastasis. VEGF is now a validated target for NSCLC based on the results of the Eastern Cooperative Oncology Group trial E4599 which showed that the addition of bevacizumab, a VEGF monoclonal antibody, to cytotoxic chemotherapy improves survival compared with chemotherapy alone in patients with metastatic NSCLC. As NSCLC has complex and integrated signaling pathways, a rational approach is to target more than one of these pathways concurrently. Sorafenib, which is approved for the treatment of renal cell carcinoma, is a multitargeted signal transduction inhibitor that inhibits raf-kinases, VEGF receptor-2, platelet derived growth factor receptor-B, and c-kit. In a phase II monotherapy trial in patients with previously treated NSCLC, sorafenib demonstrated activity with a disease control rate and survival rate comparable to other small molecules. Additionally, sorafenib has shown preliminary activity in combination with chemotherapy and with epidermal growth factor receptor inhibitors. Future directions will include the development of rational combinations either with cytotoxic compounds or biologically targeted compounds and the identification of subsets of patients that might benefit from the other targets of sorafenib.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.