Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Curr Probl Cancer ; 45(3): 100705, 2021 06.
Article in English | MEDLINE | ID: mdl-33495025

ABSTRACT

Nilotinib has been used as a third-line drug for gastrointestinal stromal tumors (GISTs) after a failure of sunitinib. In this study, we aimed to evaluate the efficacy of nilotinib in different genomic subtypes of GISTs. We searched the English articles through EMBASE, Cochrane Library and PubMed Database regarding to the use of nilotinib on GISTs, which published up to February 15, 2019. Inclusion criteria were: GISTs patients received nilotinib in a clinical trial and had detailed genetic subtype records (such as KIT exon 9, KIT exon 11, or PDGFRA mutations, or wild-type). The clinical benefit rate was used to assess the efficacy of nilotinib. A total of 3 studies involving 218 GISTs were included in this meta-analysis. The overall OR (KIT group vs WT group) was 3.26 (95% CI: 1.14-9.28; P = 0.027, Pheterogeneity = 0.613). The overall OR in KIT exon 11 group vs WT group was 5.30 (95% CI: 1.79-15.68; P = 0.003, Pheterogeneity = 0.409). The overall OR in KIT exon 9 group vs WT group was 0.13 (95% CI: 0.02-0.86; P = 0.035, Pheterogeneity = 0.229). The overall OR in KIT exon 11 group vs exon 9 group was 9.96 (95% CI: 0.39-254.66; P < 0.0001, Pheterogeneity = 0.024). Different genotypes of GISTs showed different responses to nilotinib, and KIT exon 11-mutant GISTs mostly benefited from nilotinib, followed by KIT exon 9-mutant or WT one.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Pyrimidines/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Female , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Genotype , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/genetics , Treatment Outcome , Young Adult
2.
J Urol ; 194(6): 1797-805, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26055827

ABSTRACT

PURPOSE: Bladder outlet obstruction is a finding in many urological disorders, leading to bladder wall hyperplasia. We investigated platelet derived growth factor and its receptor in human bladder smooth muscle cells and urothelial cells exposed to hydrostatic pressure or PDGF in vitro. MATERIALS AND METHODS: Bladder smooth muscle cells and urothelial cells were exposed to elevated hydrostatic pressure for 1 hour. The expression of PDGF and PDGFR was evaluated using reverse transcriptase-polymerase chain reaction and Western blot analysis. Pressure or PDGF induced proliferation of bladder smooth muscle cells with or without pretreatment with lovastatin or imatinib was measured by enzyme-linked immunosorbent assay. PDGFRα was knocked down with siRNA. RESULTS: After hydrostatic pressure bladder smooth muscle cells showed increased PDGFRα and ß expression. PDGF was not expressed in bladder smooth muscle cells. Urothelial cells showed no expression of PDGFR but PDGF expression was noted. Western blot analysis of bladder smooth muscle cells revealed a pressure induced increase in PDGFR in the membrane fraction. Phosphorylation of PDGFR occurred with pressure induction. Bladder smooth muscle cell proliferation was increased in pressure and PDGF mediated fashion. Pretreatment with lovastatin or imatinib prevented proliferation. There was no cell proliferation after PDGFRα knockdown. CONCLUSIONS: Increased expression and phosphorylation of PDGFR in bladder smooth muscle cells after hydrostatic pressure suggests a pivotal role of the PDGF pathway in pressure induced hyperplasia of bladder smooth muscle cells. PDGF expressed in urothelial cells may act in a paracrine way. Cholesterol depletion, inhibition of receptor tyrosine kinase activity and knockdown of PDGFRα in bladder smooth muscle cells prevent pressure and PDGF mediated cell proliferation. Targeting PDGFR seems a promising way to influence pressure induced bladder wall hyperplasia.


Subject(s)
Muscle, Smooth/pathology , Paracrine Communication/physiology , Platelet-Derived Growth Factor/physiology , Urinary Bladder/pathology , Urodynamics/physiology , Blotting, Western , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Proliferation/physiology , Child , Gene Knockdown Techniques , Humans , Hydrostatic Pressure , Hyperplasia , Imatinib Mesylate/pharmacology , In Vitro Techniques , Lovastatin/pharmacology , Muscle, Smooth/drug effects , Paracrine Communication/drug effects , Paracrine Communication/genetics , Phosphorylation/physiology , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , Urinary Bladder/drug effects , Urinary Bladder Neck Obstruction/genetics , Urinary Bladder Neck Obstruction/pathology , Urinary Bladder Neck Obstruction/physiopathology
3.
Intern Med ; 54(6): 637-42, 2015.
Article in English | MEDLINE | ID: mdl-25786456

ABSTRACT

Obtaining a precise characterization of eosinophilia is crucial, as successful treatment relies on the underlying etiology of the disease. Platelet-derived growth factor receptor alpha-related disorders were first specified in 2008 as a distinct group of clonal eosinophilic disorders with exceptional responsiveness to imatinib. We herein present the case of a man with myeloid neoplasm and eosinophilia in whom a definitive diagnosis could not be adequately made based on histopathological features who was ultimately diagnosed only after extensive molecular analyses and successfully treated with imatinib. In addition, we discuss the diagnostic and therapeutic approaches to treating patients presenting with eosinophilia.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzamides/administration & dosage , Eosinophilia/drug therapy , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/drug therapy , Oncogene Proteins, Fusion/isolation & purification , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Receptor, Platelet-Derived Growth Factor alpha/isolation & purification , mRNA Cleavage and Polyadenylation Factors/isolation & purification , Adult , Eosinophilia/metabolism , Follow-Up Studies , Humans , Imatinib Mesylate , Leukemia, Myeloid/metabolism , Male , Oncogene Proteins, Fusion/drug effects , Oncogene Proteins, Fusion/metabolism , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Treatment Outcome , mRNA Cleavage and Polyadenylation Factors/drug effects , mRNA Cleavage and Polyadenylation Factors/metabolism
4.
Stroke ; 46(1): 221-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25395417

ABSTRACT

BACKGROUND AND PURPOSE: Translational research is beginning to reveal the importance of trophic factors as a therapy for cellular brain repair. The purpose of this study was to analyze whether brain-derived neurotrophic factor (BDNF) administration could mediate oligodendrogenesis and remyelination after white matter injury in subcortical stroke. METHODS: Ischemia was induced in rats by injection of endothelin-1. At 24 hours, 0.4 µg/kg of BDNF or saline was intravenously administered to the treatment and control groups, respectively. Functional evaluation, MRI, and fiber tract integrity on tractography images were analyzed. Proliferation (KI-67) and white matter repair markers (A2B5, 2',3'-cyclic-nucleotide 3'-phosphodiesterase [CNPase], adenomatous polyposis coli [APC], platelet-derived growth factor receptor alpha [PDGFR-α], oligodendrocyte marker O4 [O4], oligodendrocyte transcription factor [Olig-2], and myelin basic protein [MBP]) were analyzed at 7 and 28 days. RESULTS: The BDNF-treated animals showed less functional deficit at 28 days after treatment than the controls (P<0.05). Although T2-MRI did not show differences in lesion size at 7 and 28 days between groups, diffusion tensor imaging tractography analysis revealed significantly better tract connectivity at 28 days in the BDNF group than in the controls (P<0.05). Increased proliferation of oligodendrocyte progenitors was observed in treated animals at 7 days (P<0.05). Finally, the levels of white matter repair markers (A2B5, CNPase, and O4 at 7 days; Olig-2 and MBP at 28 days) were higher in the BDNF group than in the controls (P<0.05). CONCLUSIONS: BDNF administration exerted better functional outcome, oligodendrogenesis, remyelination, and fiber connectivity than controls in rats subjected to subcortical damage in ischemic stroke.


Subject(s)
Brain Ischemia/pathology , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation/drug effects , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Stroke/pathology , White Matter/drug effects , 2',3'-Cyclic-Nucleotide Phosphodiesterases/drug effects , Adenomatous Polyposis Coli Protein/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/drug effects , Brain/drug effects , Brain/pathology , Brain Ischemia/complications , Diffusion Tensor Imaging , Magnetic Resonance Imaging , Myelin Basic Protein/drug effects , Myelin Sheath/pathology , Myelin Sheath/physiology , Rats , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Stroke/etiology , White Matter/pathology
5.
Int Forum Allergy Rhinol ; 4(11): 909-14, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25256824

ABSTRACT

BACKGROUND: The pathogenesis of human chronic rhinosinusitis with nasal polyps (CRSwNP) comprising eosinophilic CRSwNP (ECRSwNP) and non-eosinophilic (nECRSwNP) is not completely understood. Recent evidence has suggested that platelet-derived growth factor receptor alpha (PDGFRα) is implicated in cell growth, transformation, proliferation, migration, and vascular permeability and platelet-derived growth factor-A (PDGF-A) is a specific ligand for PDGFRα. However, little is known about their roles in CRSwNP. Therefore, we aimed to investigate the expression and role of PDGFRα and PDGF-A in CRSwNP. METHODS: PDGFRα protein expression was investigated by immunohistochemistry method and messenger RNA (mRNA) expression of PDGFRα and PDGF-A were assessed by real-time polymerase chain reaction (PCR) in CRSwNP patients and control subjects. Moreover, the effects of various stimulators with different concentrations and time on PDGFRα were evaluated on nasal explant culture. RESULTS: Quantitative analysis of immunostaining for PDGFRα showed an obvious elevation in immunolabeling of PDGFRα in CRSwNP groups compared with controls. Furthermore, PDGFRα protein was significantly stronger expressed in ECRSwNP group than nECRSwNP group and atopic patients showed stronger expression of PDGFRα protein than nonatopic patients. The mRNA of PDGFRα and PDGF-A were overexpressed in CRSwNP, especially in ECRSwNP. PDGFRα mRNA expression was closely related to PDGF-A mRNA. In nasal explant culture and stimulation, PDGFRα mRNA was augmented by interleukin 4 (IL-4), IL-5, or IL-1ß respectively, but suppressed by IL-27. CONCLUSION: PDGFRα may play a pivotal role in the pathophysiology of ECRSwNP and nECRSwNP by combining with PDGF-A. IL-4, IL-5, or IL-1ß may be critical for PDGFRα gene expression.


Subject(s)
Eosinophilia/etiology , Nasal Polyps/etiology , Platelet-Derived Growth Factor/physiology , Receptor, Platelet-Derived Growth Factor alpha/physiology , Rhinitis/etiology , Sinusitis/etiology , Adult , Chronic Disease , Female , Humans , Immunohistochemistry , Interleukins/pharmacology , Male , Paranasal Sinuses/metabolism , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
Arthritis Res Ther ; 14(3): R145, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22697462

ABSTRACT

INTRODUCTION: Recently, several studies assessing the clinical efficacy of rituximab (RTX) in systemic sclerosis (SSc) have reported encouraging results. We aimed at exploring whether RTX exerts its beneficial effects on fibrosis through attenuation of platelet-derived growth factor receptor (PDGFR) pathway activation. METHODS: We immunohistochemically assessed skin biopsies obtained from eight patients with SSc prior to and 6 months following RTX treatment, three control SSc patients (at the same time points) and three healthy subjects. We assessed the expression of platelet-derived growth factor, PDGFR and phosphorylated (activated) PDGFR. RESULTS: We found a strong correlation of PDGFRα and PDGFRß expression on spindle-like cells and collagen deposition in SSc biopsies (r = 0.97 and r = 0.96 for PDGFRα and PDGFRß, respectively; P < 0.0001 for both), indicating a strong link between PDGFR expression and fibrosis. Expression of PDGFRα and PDGFRß in the papillary dermis significantly decreased following RTX administration (mean ± standard error of the mean at baseline vs. 6 months, respectively: PDGFRα, 42.05 ± 5.03 vs. 26.85 ± 3.00, P = 0.004; and PDGFRß, 37.14 ± 4.94 vs. 24.01 ± 3.27, P = 0.012). Similarly, expression of phosphorylated PDGFRα and PDGFRß in the papillary dermis significantly decreased following RTX administration (P = 0.006 and P = 0.013 for phospho-PDGFRα and phospho-PDGFRß, respectively). No changes in platelet-derived growth factor tissue expression or serum levels were found following RTX treatment. CONCLUSION: RTX may favorably affect skin fibrosis through attenuation of PDGFR expression and activation, a finding that supports a disease-modifying role of RTX in SSc. Large-scale, multicenter studies are needed to further explore the efficacy of RTX in SSc.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antirheumatic Agents/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Scleroderma, Diffuse/metabolism , Adult , B-Lymphocytes/drug effects , Female , Fibrosis , Humans , Immunohistochemistry , Lymphocyte Depletion/methods , Male , Middle Aged , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor beta/drug effects , Receptors, Platelet-Derived Growth Factor/metabolism , Rituximab , Scleroderma, Diffuse/drug therapy , Scleroderma, Diffuse/immunology , Skin/metabolism , Skin/pathology
7.
Expert Opin Ther Targets ; 15(7): 803-15, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21385119

ABSTRACT

INTRODUCTION: The biological complexity of gastrointestinal stromal tumors (GISTs) and the concomitant increase in patients' life expectancy have enhanced the need for new therapeutic options to overcome the development of primary and secondary resistance to tyrosine kinase inhibitors. Aided by more sophisticated molecular biology techniques, researchers have recently sought to identify new therapeutic targets with a defined role in GISTs pathogenesis and a potential application in clinical practice. AREAS COVERED: The first aim of this review is to describe new targets and drugs in GISTs, alone or in combination, both in pre-clinical and clinical settings. The second aim is to discuss the criticism in this field, the role of molecular biology, and future perspectives in light of the recent development of more sophisticated whole-genomic technologies. EXPERT OPINION: Several targets involved in GIST pathogenesis have been identified and novel biological drugs have recently been developed, offering new treatment options in the scenario of GIST therapy. However, the identification of new therapeutic targets represents a long process and requires a global overview of the problem and a multi-step approach to convert an initial intuition or casual finding into a systematic analytical process.


Subject(s)
Antineoplastic Agents/pharmacology , Gastrointestinal Stromal Tumors/drug therapy , Proto-Oncogene Proteins c-kit/drug effects , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Antineoplastic Agents/therapeutic use , Humans
8.
Clin. transl. oncol. (Print) ; 12(10): 670-676, oct. 2010. ilus, tab
Article in English | IBECS | ID: ibc-124356

ABSTRACT

Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract. GISTs are characterised by the expression of KIT, a type III tyrosine kinase receptor, and the presence of mutations in KIT or PDGFRA in about 80-85% of cases. The primary treatment for GIST is surgery, which cures most patients with low- or intermediate-risk tumours. The introduction of the kinase inhibitor imatinib mesylate, and sunitinib in second line, against KIT and PDGFRA has provided the first evidence of directed therapy in GIST. The aim of this review is to highlight the growing evidence that KIT and PDGFRA genotyping provides valuable information for the clinical management of GIST patients. We show that KIT and PDGFRA genotyping has emerged as one of the principal factors in the evaluation of GISTs, particularly in those tumours that are clearly malignant or have a high risk of recurrence. In addition to helping establish the diagnosis of GIST in unusual cases, genotyping can be very useful to physicians and patients in deciding on imatinib dose, in estimating the likelihood and duration of benefit, and potentially in selecting second-line therapies (AU)


Subject(s)
Humans , Male , Female , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/genetics , Antineoplastic Agents/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Genotype , Mutation , Piperazines/therapeutic use , Pyrimidines/therapeutic use
9.
Dig Dis Sci ; 55(12): 3315-27, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20725786

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are relatively common mesenchymal tumors of the digestive tract characterized by c-KIT mutations. This is a comprehensive review of the current data of the literature on the various aspects of the diagnosis and treatment of these tumors. METHODS: The stomach is the most commonly involved site for these tumors in the digestive tract. Computed tomography and endoscopy can usually establish the diagnosis. The study of certain specific immunohistochemical markers may contribute to better characterization of these tumors. RESULTS: Surgical resection of GISTs has been the most effective therapy. In addition, targeted therapy with tyrosine kinase inhibitors may reduce the development of recurrence or decrease the disease progression in patients with metastatic disease. CONCLUSIONS: The introduction of tyrosine kinase inhibitors has resulted in significant improvement in the overall prognosis of these patients. Furthermore, preoperative imatinib can decrease tumor volume and is associated with complete surgical resection in locally advanced primary GISTs.


Subject(s)
Gastrointestinal Stromal Tumors/therapy , Antineoplastic Agents/therapeutic use , Benzamides , Biomarkers, Tumor , Diagnosis, Differential , Disease Progression , Endoscopy, Gastrointestinal , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/epidemiology , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate , Immunohistochemistry , Neoplasm Recurrence, Local/epidemiology , Piperazines/therapeutic use , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/physiology , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/physiology
10.
Biochem Pharmacol ; 80(5): 575-83, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20385106

ABSTRACT

Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal (GI) tract and are caused by activating KIT or PDGFRA mutations. GISTs can be successfully treated with the small molecule kinase inhibitor imatinib mesylate (Gleevec, Novartis) with response rates of up to 85%. However, complete responses are rare, and most patients will develop imatinib resistance over time. Recent results have shown that although imatinib effectively stimulates apoptotic cell death in sensitive GIST cells, a considerable proportion of cells does not undergo apoptosis, but instead enters a state of quiescence. Quiescence is characterized by a reversible withdrawal from the cell division cycle, during which the cells remain alive and metabolically active. It is conceivable that quiescence not only plays a pivotal role in the emergence of residual disease but also in creating a pool of tumor cells that survive continuous small molecule therapy and may hence represent the "seeds" for the outgrowth of resistant clones. This review will summarize the current knowledge about GIST biology and treatment response to imatinib including the induction of cellular quiescence in GIST. In addition, we will highlight future strategies to design more effective treatment options to overcome these problems with an aim towards cure of this hitherto untreatable tumor entity.


Subject(s)
Antineoplastic Agents/therapeutic use , Gastrointestinal Stromal Tumors/drug therapy , Piperazines/therapeutic use , Proto-Oncogene Proteins c-kit/drug effects , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Antineoplastic Agents/pharmacology , Benzamides , Gastrointestinal Stromal Tumors/genetics , Humans , Imatinib Mesylate , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/genetics
11.
J Exp Clin Cancer Res ; 29: 173, 2010 Dec 30.
Article in English | MEDLINE | ID: mdl-21192792

ABSTRACT

BACKGROUND: Primary and secondary drug resistance to imatinib and sunitinib in patients with gastrointestinal stromal tumors (GISTs) has led to a pressing need for new therapeutic strategies such as drug combinations. Most GISTs are caused by mutations in the KIT receptor, leading to upregulated KIT tyrosine kinase activity. Imatinib and nilotinib directly inhibit the kinase activity of KIT, while RAD001 (everolimus) inhibits mTOR. We report a preclinical study on drug combinations in a xenograft model of GIST in which effects on tumor dimensions and metabolic activity were assessed by small animal PET imaging. METHODS: Rag2-/-; γcommon -/- male mice were injected s.c. into the right leg with GIST 882. The animals were randomized into 6 groups of 6 animals each for different treatment regimens: No therapy (control), imatinib (150 mg/kg b.i.d.) by oral gavage for 6 days, then once/day for another 7 days, everolimus (10 mg/kg/d.) by oral gavage, everolimus (10 mg/kg/d.) + imatinib (150 mg/kg b.i.d.) by oral gavage for 6 days, then once/day for another 7 days, nilotinib (75 mg/kg/d.) by oral gavage, nilotinib (75 mg/kg/d.) + imatinib (150 mg/kg b.i.d) by oral gavage for 6 days, then once/day for another 7 days. Tumor growth control was evaluated by measuring tumor volume (cm3). Small animal PET (GE Explore tomography) was used to evaluate tumor metabolism and performed in one animal per group at base-line then after 4 and 13 days of treatment. RESULTS: After a median latency time of 31 days, tumors grew in all animals (volume 0,06-0,15 cm3) and the treatments began at day 38 after cell injection. Tumor volume control (cm3) after 13 days of treatment was > 0.5 for imatinib alone and nilotinib alone, and < 0.5 for the 2 combinations of drugs and for everolimus alone. The baseline FDG uptake was positive in all animals. FDG/SUV/TBR was strongly reduced over time by everolimus both as a single agent and in combination with imatinib respectively: 3.1 vs. 2.3 vs. 1.9 and 2.5 vs 2.3 vs 0. CONCLUSIONS: As single agents, all drugs showed an anti-tumor effect in GIST xenografts but everolimus was superior. The everolimus plus imatinib combination appeared to be the most active regimen both in terms of inhibiting tumor growth and tumor metabolism. The integration of everolimus in GIST treatment merits further investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gastrointestinal Stromal Tumors/diagnostic imaging , Gastrointestinal Stromal Tumors/drug therapy , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Benzamides , Enzyme Inhibitors/pharmacology , Everolimus , Fluorodeoxyglucose F18 , Humans , Imatinib Mesylate , Male , Mice , Mice, Knockout , Piperazines/administration & dosage , Positron-Emission Tomography , Proto-Oncogene Proteins c-kit/drug effects , Pyrimidines/administration & dosage , Radiopharmaceuticals , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/drug effects , Xenograft Model Antitumor Assays
13.
Int Arch Allergy Immunol ; 146 Suppl 1: 7-10, 2008.
Article in English | MEDLINE | ID: mdl-18504399

ABSTRACT

BACKGROUND: Acetylation and deacetylation of proteins occur in cells in response to various stimuli, and are reversibly catalyzed by histone acetyltransferase and histone deacetylase (HDAC), respectively. EoL-1 cells have an FIP1L1-PDGFRA fusion gene that causes transformation of eosinophilic precursor cells into leukemia cells. The HDAC inhibitors apicidin and n-butyrate suppress the proliferation of EoL-1 cells and induce differentiation into eosinophils by a decrease in the protein level of FIP1L1-PDGFRalpha without affecting the mRNA level for FIP1L1-PDGFRA. In this study, we analyzed the mechanism by which the protein level of FIP1L1-PDGFRalpha is decreased by apicidin and n-butyrate. METHODS: EoL-1 cells were incubated in the presence of the HDAC inhibitors apicidin, trichostatin A or n-butyrate. The protein levels of FIP1L1-PDGFRalpha and phosphorylated eIF-2alpha were determined by Western blotting. Actinomycin D and cycloheximide were used to block RNA synthesis and protein synthesis, respectively, in the chasing experiment of the amount of FIP1L1-PDGFRalpha protein. RESULTS: When apicidin- and n-butyrate-treated EoL-1 cells were incubated in the presence of actinomycin D, the decrease in the protein level of FIP1L1-PDGFRalpha was significantly enhanced when compared with controls. In contrast, the protein levels were not changed by cycloheximide among these groups. Apicidin and n-butyrate induced the continuous phosphorylation of eIF-2alpha for up to 8 days. CONCLUSIONS: The decrease in the level of FIP1L1-PDGFRalpha protein by continuous inhibition of HDAC may be due to the decrease in the translation rate of FIP1L1-PDGFRA.


Subject(s)
Butyrates/pharmacology , Enzyme Inhibitors/pharmacology , Eosinophils/drug effects , Eukaryotic Initiation Factor-2/metabolism , Histone Deacetylase Inhibitors , Hypereosinophilic Syndrome/drug therapy , Oncogene Proteins, Fusion/analysis , Peptides, Cyclic/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/analysis , mRNA Cleavage and Polyadenylation Factors/analysis , Acetylation/drug effects , Blotting, Western , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Eosinophils/metabolism , Eukaryotic Initiation Factor-2/analysis , Eukaryotic Initiation Factor-2/drug effects , Histone Deacetylases/metabolism , Humans , Hypereosinophilic Syndrome/metabolism , Oncogene Proteins, Fusion/drug effects , Oncogene Proteins, Fusion/metabolism , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/metabolism , mRNA Cleavage and Polyadenylation Factors/drug effects , mRNA Cleavage and Polyadenylation Factors/metabolism
14.
Rev Med Interne ; 29(1): 65-7, 2008 Jan.
Article in French | MEDLINE | ID: mdl-18036706

ABSTRACT

Hypereosinophilic syndromes (HES) are a heterogeneous group of disorders characterized by marked peripheral blood and tissue eosinophilia resulting in organ damage. Recent advances in molecular biology have led to the identification of a FIP1L1-PDGFRA fusion gene as a recurrent abnormality in some patients with HES. This fusion gene results from a cryptic 4q12 interstitial deletion involving an 800 kb region. Recent reports indicate that this subtype of HES is imatinib responsive with rapid and complete haematological remissions. Here we report two patients successfully treated with imatinib.


Subject(s)
Hypereosinophilic Syndrome/drug therapy , Oncogene Proteins, Fusion/drug effects , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/drug effects , mRNA Cleavage and Polyadenylation Factors/drug effects , Adult , Benzamides , Humans , Hypereosinophilic Syndrome/genetics , Imatinib Mesylate , Male , Middle Aged , Treatment Outcome
15.
Curr Opin Pulm Med ; 13(5): 422-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17940488

ABSTRACT

PURPOSE OF REVIEW: Hypereosinophilic syndrome is increasingly recognized as a heterogeneous group of disorders, in some cases with precisely defined pathogenesis, which has led to changes in diagnostic approaches and therapeutic strategies. An update on causes and modern therapy is presented here. RECENT FINDINGS: Clonal eosinophilias belong to the group of myeloid malignancies. Karyotypically occult FIP1L1- platelet-derived growth factor receptor alpha and beta rearranged eosinophilic disorders respond to imatinib mesylate with almost 100% efficacy. If standard therapies fail, the FIP1L1- platelet-derived growth factor receptor-negative cases of hypereosinophilic syndrome should also be considered for treatment with imatinib. The recognition of acquired resistance to imatinib has aroused interest in developing new tyrosine kinase inhibitors. Other subgroups of clonal eosinophilias have been molecularly defined, but the curative verification of pathogenetic relevance has not been certified. Hypereosinophilic syndrome patients with abnormal T-cell populations have benefited from treatment with anti IL-5 monoclonal antibodies. SUMMARY: The FIP1L1- platelet-derived growth factor receptor alpha and beta-positive patients, and those with abnormal T-cell populations are currently the only clearly defined treatable subgroups of hypereosinophilic syndrome. The FIP1L1- platelet-derived growth factor receptor alpha-negative responders to imatinib pose a question as to the existence of subentities with unrecognized tyrosine kinases-based mutation. The search for such cases and other treatable subgroups of hypereosinophilic syndrome has already begun.


Subject(s)
Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/drug therapy , Antibodies, Monoclonal/therapeutic use , Benzamides , Humans , Imatinib Mesylate , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pulmonary Eosinophilia/diagnosis , Pulmonary Eosinophilia/drug therapy , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/physiology
16.
Br J Cancer ; 96(1): 180-1, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17213828
17.
Leuk Res ; 30(12): 1499-505, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16682077

ABSTRACT

Idiopathic hypereosinophilic syndrome (HES) is a myeloproliferative disorder characterized by tissue involvement and organ dysfunction due to abnormal eosinophil proliferation. In a subset of patients, this is caused by the FIP1L1-PDGFR-alpha fusion tyrosine kinase. Cumulative evidence indicates that the Bcr-Abl tyrosine kinase inhibitor imatinib mesylate (Gleevec) is active for the treatment of patients with HES, particularly those expressing the FIP1L1-PDGFR-alpha oncoprotein. The novel tyrosine kinase inhibitor AMN107 was initially developed as a potent Bcr-Abl inhibitor based on the molecular structure of imatinib. We tested the in vitro efficacy of imatinib and AMN107 in the EOL-1 cell line and in cells from a patient with HES harboring the FIP1L1-PDGFR-alpha fusion kinase. AMN107 was as potent as imatinib in inducing apoptosis and inhibiting proliferation of EOL-1 cells, with IC(50) values of 0.54 and 0.20 nM, respectively. In addition, both drugs inhibited the phosphorylation of PDGFR-alpha tyrosine kinase with equivalent efficacy. We conclude that AMN107 and imatinib are active and equipotent against cells expressing the FIP1L1-PDGFR-alpha fusion gene.


Subject(s)
Antineoplastic Agents/pharmacology , Oncogene Proteins, Fusion/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/drug effects , mRNA Cleavage and Polyadenylation Factors/drug effects , Apoptosis/drug effects , Benzamides , Caspase 3/metabolism , Caspase Inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/antagonists & inhibitors , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Humans , Hypereosinophilic Syndrome/metabolism , Imatinib Mesylate , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphorylation , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
18.
Am J Respir Cell Mol Biol ; 34(6): 746-53, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16439802

ABSTRACT

Bronchial smooth muscle cell (BSMC) hyperplasia is a typical feature of airway remodeling and contributes to airway obstruction and hyperresponsiveness in asthma. Fibroblast growth factor 2 (FGF-2) and transforming growth factor beta1 (TGF-beta1) are sequentially upregulated in asthmatic airways after allergic challenge. Whereas FGF-2 induces BSMC proliferation, the mitogenic effect of TGF-beta1 remains controversial, and the effect of sequential FGF-2 and TGF-beta1 co-stimulation on BSMC proliferation is unknown. This study aimed to assess the individual and sequential cooperative effects of FGF-2 and TGF-beta1 on human BSMC proliferation and define the underlying mechanisms. Mitogenic response was measured using crystal violet staining and [3H]-thymidine incorporation. Steady-state mRNA and protein levels were measured by semiquantitative RT-PCR, Western blot, and ELISA, respectively. TGF-beta1 (0.1-20 ng/ml) alone had no effect on BSMC proliferation, but increased the proliferative effect of FGF-2 (2 ng/ml) in a concentration-dependent manner (up to 6-fold). Two distinct platelet-derived growth factor receptor (PDGFR) inhibitors, AG1296 and Inhibitor III, as well as a neutralizing Ab against PDGFRalpha, partially blocked the synergism between these two growth factors. In this regard, TGF-beta1 increased PDGF-A and PDGF-C mRNA expression as well as PDGF-AA protein expression. Moreover, FGF-2 pretreatment increased the mRNA and protein expression of PDGFRalpha and the proliferative effect of exogenous PDGF-AA (140%). Our data suggest that FGF-2 and TGF-beta1 synergize in BSMC proliferation and that this synergism is partially mediated by a PDGF loop, where FGF-2 and TGF-beta1 upregulate the receptor (PDGFRalpha) and the ligands (PDGF-AA and PDGF-CC), respectively. This powerful synergistic effect may thus contribute to the hyperplastic phenotype of BSMC in remodeled asthmatic airways.


Subject(s)
Bronchi/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Myocytes, Smooth Muscle/drug effects , Transforming Growth Factor beta/pharmacology , Adult , Bronchi/cytology , Bronchi/metabolism , Cells, Cultured , DNA/biosynthesis , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression Regulation/drug effects , Humans , Infant , Lymphokines/genetics , Lymphokines/metabolism , Male , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Time Factors , Tissue Plasminogen Activator/biosynthesis , Tissue Plasminogen Activator/genetics , Transforming Growth Factor beta1 , Tyrphostins/pharmacology
19.
Verh K Acad Geneeskd Belg ; 67(3): 169-76, 2005.
Article in English | MEDLINE | ID: mdl-16089297

ABSTRACT

The identification of the FIP1L1-PDGFRA fusion gene provides a molecular explanation for the pathogenesis of approximately half of the patients with the hypereosinophilic syndrome (HES). A diagnostic test to identify FIP1L1-PDGFRA positive HES cases (subsequently reclassified as chronic eosinophilic leukemia, CEL) is now available. FIP1L1-PDGFR alpha is a novel therapeutic target of the kinase inhibitor imatinib (Glivec, Novartis), which provides the basis for the treatment of these patients with this drug. FIP1L1-PDGFRA positive CEL patients respond very well to imatinib therapy, some of which are remarkable responses with normalization of the blood counts within 2 weeks after start of the therapy. Imatinib is well tolerated with minimal side effects, and most CEL patients respond to low doses of imatinib (100 mg/day), being important for lowering both the cost of therapy and drug related toxicity. All imatinib treated FIP1L1-PDGFRA positive CEL patients achieve hematological and cytogenetic remission, and the majority of patients also achieve a molecular remission with the fusion gene no longer detectable in blood, even by the most sensitive PCR techniques.


Subject(s)
Antineoplastic Agents/therapeutic use , Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/genetics , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/drug effects , mRNA Cleavage and Polyadenylation Factors/drug effects , Benzamides , Chronic Disease , Humans , Imatinib Mesylate , Oncogene Proteins, Fusion , Receptor, Platelet-Derived Growth Factor alpha/genetics , Treatment Outcome , mRNA Cleavage and Polyadenylation Factors/genetics
20.
J Steroid Biochem Mol Biol ; 94(1-3): 189-96, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15862965

ABSTRACT

Calcitriol, a hormonal form of Vitamin D, regulates growth of normal and cancer cells of various origins by modulation of peptide growth factors signaling. Platelet-Derived Growth Factor (PDGF) signaling pathway is involved in prostate cancer progression. We studied the expression of PDGF receptors in human prostate primary stromal cells and cancer epithelial cell lines and growth response to PDGF-BB isoform. We found that the expression of PDGF receptors and PDGF-BB-mediated cell growth are regulated by calcitriol in prostate cells. Quantitative RT-PCR analysis revealed a lower level of mRNA for PDGF receptors in LNCaP and PC-3 cells than in primary stromal cells. Western blotting showed a high amount of PDGFRalpha and beta proteins in primary stromal cells that could not be detected in LNCaP, which may explain the resistance of LNCaP cells to growth-promoting effect of PDGF-BB. Addition of Epidermal Growth Factor (EGF) to the culture medium induces the expression of PDGFRbeta and restores responsiveness of LNCaP to PDGF-BB to some extent. Calcitriol down-regulates PDGFRbeta expression and negatively regulates PDGF-mediated cell growth. Calcitriol does not affect PDGFRalpha and PDGF-B mRNA expression. We suggest that inhibition of PDGFRbeta expression by calcitriol might reduce responsiveness of prostate cells to mitogenic action of PDGF-BB.


Subject(s)
Calcitriol/pharmacology , Cell Division/drug effects , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/physiology , Prostate/physiology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Base Sequence , Becaplermin , Cell Line, Tumor , DNA Primers , Humans , Male , Prostate/drug effects , Prostatic Neoplasms , Proto-Oncogene Proteins c-sis , RNA, Messenger/drug effects , RNA, Messenger/genetics , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor beta/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...