Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Ann Oncol ; 21(3): 562-567, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19767315

ABSTRACT

BACKGROUND: The purpose of this study is to investigate the prognostic role of insulin-like growth factor receptor 1 (IGF1R) expression in surgically resected non-small-cell lung cancer (NSCLC). Patient characteristics and methods: This retrospective study was conducted in 369 stage I-II-IIIA, surgically resected, NSCLC patients. Patients exposed to anti-epidermal growth factor receptor (EGFR) agents were excluded. IGF1R expression was evaluated by immunohistochemistry in tissue microarray sections. RESULTS: A positive IGF1R expression (score > or = 100) was observed in 282 cases (76.4%) and was significantly associated with squamous cell histology (P = 0.04) and with grade III differentiation (P = 0.02). No difference in survival was observed between the positive and negative group when score 100 was used as cut-off for discriminating a positive versus a negative IGF1R result (52 versus 48 months, P = 0.99) or when median value of IGF1R expression was used (45 versus 55 months, P = 0.36). No difference in survival was observed between IGF1R-positive and -negative patients in a subgroup of stage I-II adenocarcinoma (n = 137) with known EGFR mutation and copy number status. CONCLUSIONS: IGF1R expression does not represent a prognostic factor in resected NSCLC patients. Patients with squamous cell carcinoma overexpress IGF1R more frequently than patients with nonsquamous histology, justifying the different sensitivity to anti-IGF1R agents observed in clinical trials.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Receptor, IGF Type 1/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/surgery , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Adenocarcinoma, Bronchiolo-Alveolar/mortality , Adenocarcinoma, Bronchiolo-Alveolar/surgery , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/surgery , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/surgery , Cohort Studies , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/surgery , Male , Middle Aged , Neoplasm Staging , Retrospective Studies , Survival Rate , Tissue Array Analysis , Treatment Outcome
2.
Expert Rev Anticancer Ther ; 20(9): 743-753, 2020 09.
Article in English | MEDLINE | ID: mdl-32755244

ABSTRACT

INTRODUCTION: Patients treated with third-generation EGFR TKIs will develop resistance to treatment at a certain point. Early detection of resistance occurrence could allow more options for treatment. AREAS COVERED: We discuss the development of third-generation EGFR TKIs, focusing on osimertinib and discuss the most common resistance mechanisms under evaluation. We also debate how this resistance can be detected; particularly we review the possible application of liquid biopsy in this scenario. Lastly we discuss available treatment options when resistance occurs, with an eye on ongoing trials and possible future developments. EXPERT OPINION: As resistance will ultimately develop, a strict instrumental follow-up as per international guidelines is required with the aim of detecting this resistance in an early phase. Detecting an oligoprogression could allow the integration of local ablative therapies while further delaying the need for a systemic therapy change. By exploiting the increasing potentiality of liquid biopsy, in the near future, physicians could be able to understand why a patient develops resistance and therefore can choose the best possible individualized treatment option.


Subject(s)
Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Acrylamides/administration & dosage , Acrylamides/pharmacology , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Disease Progression , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Humans , Liquid Biopsy , Lung Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology
3.
Crit Rev Oncol Hematol ; 148: 102894, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32062314

ABSTRACT

Oncogene-driven non small cell lung cancer (NSCLC) is a distinct entity in thoracic oncology. The availability of effective target therapies, like EGFR inhibitors or ALK inhibitors, have revolutionized the prognosis of these patients. However, despite an initial response in the majority of patients, drug resistance ultimately occurs. In some cases, this resistance develops in few clonal cells (oligoprogression), so that a local ablation of these resistant deposits could allow to maintain the same systemic therapy and possibly to prolong patients' survival. For these purposes, stereotactic body radiation therapy (SBRT) is an ideal local ablative treatment, because it is effective, non invasive and with limited side effects. In this review, we aim to analyze available clinical data to verify whether SBRT can allow these patients to continue with existing target therapy longer, delay the switch to other systemic therapies and improve their outcome modifying the natural history of the disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/radiotherapy , Radiosurgery , Carcinoma, Non-Small-Cell Lung/pathology , Catheter Ablation , Disease Progression , Humans , Lung Neoplasms/pathology , Neoplasm Metastasis , Neoplasm Recurrence, Local , Neoplasm Staging , Oncogenes , Protein Kinase Inhibitors , Treatment Outcome
4.
Ann Oncol ; 20(2): 298-304, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18836087

ABSTRACT

BACKGROUND: MET amplification has been detected in approximately 20% of non-small-cell lung cancer patients (NSCLC) with epidermal growth factor receptor (EGFR) mutations progressing after an initial response to tyrosine kinase inhibitor (TKI) therapy. PATIENTS AND METHODS: We analyzed MET gene copy number using FISH in two related NSCLC cell lines, one sensitive (HCC827) and one resistant (HCC827 GR6) to gefitinib therapy and in two different NSCLC patient populations: 24 never smokers or EGFR FISH-positive patients treated with gefitinib (ONCOBELL cohort) and 182 surgically resected NSCLC not exposed to anti-EGFR agents. RESULTS: HCC827 GR6-resistant cell line displayed MET amplification, with a mean MET copy number >12, while sensitive HCC827 cell line had a mean MET copy number of 4. In the ONCOBELL cohort, no patient had gene amplification and MET gene copy number was not associated with outcome to gefitinib therapy. Among the surgically resected patients, MET was amplified in 12 cases (7.3%) and only four (2.4%) had a higher MET copy number than the resistant HCC827 GR6 cell line. CONCLUSIONS: MET gene amplification is a rare event in patients with advanced NSCLC. The development of anti-MET therapeutic strategies should be focused on patients with acquired EGFR-TKI resistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Dosage , Lung Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Quinazolines/therapeutic use , Receptors, Growth Factor/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Cell Line, Tumor , Clinical Trials, Phase II as Topic , Cohort Studies , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gefitinib , Gene Expression Regulation, Neoplastic , Genes, erbB-1/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Middle Aged , Proto-Oncogene Proteins c-met , Survival Analysis
5.
J Cell Biol ; 107(5): 1623-8, 1988 Nov.
Article in English | MEDLINE | ID: mdl-2903166

ABSTRACT

UV irradiation of quiescent human fibroblasts immediately triggers the appearance of the nuclear protein cyclin/proliferating cell nuclear antigen (PCNA) as detected by indirect immunofluorescent staining after methanol fixation. This was found to be independent of new synthesis of cyclin/PCNA by two-dimensional gel analysis and cycloheximide treatment. The intensity of the immunofluorescent staining of cyclin/PCNA observed in UV-irradiated cells corresponded with the UV dose used and with the DNA repair synthesis detected by autoradiography. The nuclear staining remains as long as DNA repair activity is detected in the cells. By extracting the UV-irradiated quiescent cells with Triton X-100 and fixing with formaldehyde, it was possible to demonstrate by indirect immunofluorescence rapid changes in the cyclin/PCNA population after irradiation, a small proportion (5-10%) of which is tightly associated to the nucleus as determined by high salt extraction. By incubating at low temperature and depleting the ATP pools of the cells before UV irradiation, we have demonstrated that the changes in cyclin/PCNA distribution observed involve at least two different nuclear associations.


Subject(s)
DNA Repair , Nuclear Proteins/physiology , Aphidicolin , Autoradiography , Cells, Cultured , Cycloheximide , DNA/biosynthesis , Diterpenes , Fluorescent Antibody Technique , Humans , Interphase , Nuclear Proteins/biosynthesis , Proliferating Cell Nuclear Antigen , Protein Biosynthesis , Ultraviolet Rays
6.
Br J Cancer ; 99(1): 83-9, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18577988

ABSTRACT

The impact of KRAS mutations on cetuximab sensitivity in epidermal growth factor receptor fluorescence in situ hybridisation-positive (EGFR FISH+) metastatic colorectal cancer patients (mCRC) has not been previously investigated. In the present study, we analysed KRAS, BRAF, PI3KCA, MET, and IGF1R in 85 mCRC treated with cetuximab-based therapy in whom EGFR status was known. KRAS mutations (52.5%) negatively affected response only in EGFR FISH+ patients. EGFR FISH+/KRAS mutated had a significantly lower response rate (P=0.04) than EGFR FISH+/KRAS wild type patients. Four EGFR FISH+ patients with KRAS mutations responded to cetuximab therapy. BRAF was mutated in 5.0% of patients and none responded to the therapy. PI3KCA mutations (17.7%) were not associated to cetuximab sensitivity. Patients overexpressing IGF1R (74.3%) had significantly longer survival than patients with low IGF1R expression (P=0.006), with no difference in response rate. IGF1R gene amplification was not detected, and only two (2.6%) patients, both responders, had MET gene amplification. In conclusion, KRAS mutations are associated with cetuximab failure in EGFR FISH+ mCRC, even if it does not preclude response. The rarity of MET and IGF1R gene amplification suggests a marginal role in primary resistance. The potential prognostic implication of IGF1R expression merits further evaluation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Cetuximab , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Nuclear Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met , Proto-Oncogene Proteins p21(ras) , Receptors, Growth Factor/genetics , Receptors, Somatomedin/genetics , Transcription Factors/genetics
7.
Mol Cell Biol ; 8(3): 1345-51, 1988 Mar.
Article in English | MEDLINE | ID: mdl-3367910

ABSTRACT

We have previously reported that antibodies to phosphotyrosine recognize the phosphorylated forms of platelet-derived growth factor (PDGF) and epidermal growth factor (EGF) receptors (Zippel et al., Biochim. Biophys. Acta 881:54-61, 1986, and Sturani et al., Biochem. Biophys. Res. Commun. 137:343-350, 1986). In this report, the time course of receptor phosphorylation is investigated. In normal human fibroblasts, ligand-induced phosphorylation of PDGF and EGF receptors is followed by rapid dephosphorylation. However, in A431 cells the tyrosine-phosphorylated form of EGF receptor persists for many hours after EGF stimulation, allowing a detailed analysis of the conditions affecting receptor phosphorylation and dephosphorylation. In A431 cells, the number of receptor molecules phosphorylated on tyrosine was quantitated and found to be about 10% of total EGF receptors. The phosphorylated receptor molecules are localized on the cell surface, and they are rapidly dephosphorylated upon removal of EGF from binding sites by a short acid wash of intact cells and upon a mild treatment with trypsin. ATP depletion also results in rapid dephosphorylation, indicating that continuous phosphorylation-dephosphorylation reactions occur in the ligand-receptor complex at steady state. Phorbol 12-myristate 13-acetate added shortly before EGF reduces the rate and the final extent of receptor phosphorylation. Moreover, it also reduces the amount of phosphorylated receptors if it is added after EGF. Down-regulation of protein kinase C by chronic treatment with phorbol dibutyrate increases the receptor phosphorylation induced by EGF, suggesting a homologous feedback regulation of EGF receptor functions.


Subject(s)
ErbB Receptors/metabolism , Tyrosine/metabolism , Carcinoma, Squamous Cell , Electrophoresis, Polyacrylamide Gel , Fibroblasts , Humans , Hydrogen-Ion Concentration , Immunoassay , Kinetics , Phosphorylation , Protein Kinase C/metabolism , Trypsin/pharmacology , Tumor Cells, Cultured
8.
Int J Biol Markers ; 22(1 Suppl 4): S10-23, 2007.
Article in English | MEDLINE | ID: mdl-17520577

ABSTRACT

The epidermal growth factor receptor (EGFR) plays a key role in cancer development and progression in several human malignancies including non-small cell lung cancer (NSCLC). Several strategies aimed at inhibiting the EGFR have been investigated in the last years, including the use of small tyrosine kinase inhibitors (TKIs) directed against the intracellular domain of the receptor and monoclonal antibodies targeting its extracellular portion. Subgroups of patients who are more likely to respond to TKIs have been identified based on both clinical and biological features. Never-smoking history has emerged as the most relevant clinical characteristic predictive of response to TKIs in NSCLC, while presence of drug-sensitive EGFR mutations and EGFR gene gain represent critical biological variables associated with an improved outcome for patients exposed to these agents. Recent studies have highlighted the existence of biological factors involved in intrinsic and acquired resistance to TKIs, including k-ras, HER-2 and EGFR exon 20 mutations. Increasing knowledge of EGFR biology and drug-receptor interactions will allow to identify individuals who are likely to derive a clinical benefit from the proposed targeted therapy, sparing refractory patients expensive and potentially toxic treatment.


Subject(s)
Biomarkers/analysis , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/drug effects , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cetuximab , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Erlotinib Hydrochloride , Gefitinib , Gene Dosage , Humans , Immunohistochemistry , Phosphoproteins/analysis , Predictive Value of Tests , Proto-Oncogene Proteins c-akt/analysis , Quinazolines/therapeutic use , Receptor, ErbB-2/analysis , Receptor, ErbB-3/analysis
9.
Leukemia ; 31(10): 2020-2028, 2017 10.
Article in English | MEDLINE | ID: mdl-28232670

ABSTRACT

Neomorphic mutations in isocitrate dehydrogenase 1 (IDH1) are frequently found in several human cancer types including acute myeloid leukemia (AML) and lead to the production of high levels of the oncometabolite (R)-2-hydroxyglutarate (R-2HG). Here we report the characterization of BAY1436032, a novel pan-mutant IDH1 inhibitor, both in vitro and in vivo. BAY1436032 specifically inhibits R-2HG production and colony growth, and induces myeloid differentiation of AML cells carrying IDH1R132H, IDH1R132C, IDH1R132G, IDH1R132L and IDH1R132S mutations. In addition, the compound impacts on DNA methylation and attenuates histone hypermethylation. Oral administration of BAY1436032 led to leukemic blast clearance, myeloid differentiation, depletion of leukemic stem cells and prolonged survival in two independent patient-derived xenograft IDH1 mutant AML mouse models. Together, BAY1436032 is highly effective against all major types of IDH1 mutant AML.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Isocitrate Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Glutarates/metabolism , Histone Code/drug effects , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Methylation/drug effects , Mice , Molecular Targeted Therapy , Mutation , Mutation, Missense , Myeloid Cells/drug effects , Myelopoiesis/drug effects , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Point Mutation , Protein Processing, Post-Translational/drug effects , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
10.
Ann Oncol ; 17 Suppl 5: v37-46, 2006 May.
Article in English | MEDLINE | ID: mdl-16807461

ABSTRACT

Gemcitabine, a pyrimidine nucleoside antimetabolite, is one of the most promising new cytotoxic agents. The drug has shown activity in a variety of solid tumors, but appears to be most active in the treatment of non-small cell lung cancer. In this disease, several Italian investigators have evaluated gemcitabine in phase II and III clinical trials. Due to preclinical synergism with cisplatin, the Italian Lung Cancer Project played an important role to assess the efficacy and activity of the gemcitabine-cisplatin combination along with the best doses and schedule to adopt, thus leading to gemcitabine approval for first line treatment of advanced non-small cell lung cancer. Several Italian studies have also investigated gemcitabine non-platinum based combinations, gemcitabine in third generation platinum-based triplets and gemcitabine as second line therapy, but all these studies led to conflicting and inconclusive results. The low toxicity profile makes the drug a valid option for unfit and elderly patients. The Multicenter Italian Lung Cancer in the Elderly Study was a phase III randomized trial conducted in elderly patients with advanced non-small cell lung cancer that showed that single agent gemcitabine is at least as effective as either single agent vinorelbine or the combination of gemcitabine and vinorelbine. In the neoadjuvant treatment of stage III disease, a number of phase II studies with third generation platinum-based doublets or triplets have been conducted by Italian investigators with encouraging results. Current clinical trials are addressing the role of gemcitabine in combination with new targeted therapies. Future studies should be designed in order to identify subgroups of patients who are more likely to benefit from gemcitabine chemotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxycytidine/analogs & derivatives , Lung Neoplasms/drug therapy , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Disease Progression , Drug Administration Routes , Humans , Italy , Lung Neoplasms/pathology , Neoplasm Staging , Randomized Controlled Trials as Topic , Gemcitabine
11.
Clin Oncol (R Coll Radiol) ; 28(1): 13-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26385822

ABSTRACT

AIMS: The aim of this observational study was the evaluation of toxicity, local control and overall survival in non-small cell lung cancer (NSCLC) oligometastatic patients who had undergone stereotactic ablative body radiotherapy (SABR) for lung metastatic lesions. MATERIALS AND METHODS: SABR was carried out in oligometastatic patients with controlled primary tumour (adequate pulmonary function). We adopted the following dose prescriptions according to the site and the maximum diameter of the lung lesions: 60 Gy in three fractions for peripheral lesions with diameter ≤ 2 cm, 48 Gy in four fractions for peripheral lesions between 2 and 5 cm and 60 Gy in eight fractions for central lesions. A radiological response was defined according to RECIST criteria. Toxicity was recorded according to the Common Toxicity Criteria version 4.0. RESULTS: Between October 2010 and December 2014, 60 NSCLC patients with 90 lung lesions in total were treated at our institution. A radiological response was obtained in most patients. No pulmonary toxicity grade 4, chest pain or rib fracture occurred. The median follow-up from diagnosis was 28 months (range 5.4-104.5 months). The local control at 2 years was 88.9%. Overall survival at 1 and 2 years was 94.5 and 74.6%, respectively. CONCLUSION: SABR is well tolerated with a good radiological response and toxicity profile. Discussion within a multidisciplinary team is crucial to identify the oligometastatic patients who would probably benefit from ablative local therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Non-Small-Cell Lung/surgery , Lung Neoplasms/surgery , Neoplasm Metastasis/therapy , Radiosurgery/methods , Adult , Aged , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Radiosurgery/adverse effects , Treatment Outcome
12.
Biochim Biophys Acta ; 881(1): 54-61, 1986 Mar 19.
Article in English | MEDLINE | ID: mdl-2418884

ABSTRACT

Antibodies against the synthetic hapten azobenzyl phosphonate which specifically crossreact with phosphotyrosine have been produced and used to detect the proteins phosphorylated in tyrosine following exposure of intact quiescent Swiss 3T3 fibroblasts to the platelet-derived growth factor (PDGF). Western blotting of sodium dodecyl sulfate-polyacrylamide gel electrophoresis-fractionated proteins followed by decoration with phosphotyrosine antibodies and 125I-labeled protein A have been used. The major tyrosine-phosphorylated component was a 170 kDa protein. The following lines of evidence suggest that this protein is the PDGF receptor in its tyrosine-phosphorylated form: (a) both proteins have the same (170 kDa) molecular weight; (b) the phosphorylated 170 kDa protein was detectable only in cell lines bearing the PDGF receptor; (c) the phosphorylation of the 170 kDa protein required PDGF and was dose-dependent. Kinetic studies showed that the phosphorylation of the receptor was maximal after 5-10 min at 37 degrees C and was followed by a rapid decrement of the band. The loss of the 170 kDa component was not prevented by inhibitors of membrane internalization and of lysosomal proteinases, while it was inhibited by lowering the temperature to 5 degrees C. In PDGF-stimulated cells, phosphotyrosine antibodies detected also a minor 36 kDa component phosphorylated at tyrosine.


Subject(s)
Antibodies , Receptors, Cell Surface/metabolism , Tyrosine/analogs & derivatives , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Antibodies/immunology , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Chloroquine/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Immunologic Techniques , Methylamines/pharmacology , Mice , Molecular Weight , Phenylglyoxal/pharmacology , Phosphorylation , Phosphotyrosine , Platelet-Derived Growth Factor/pharmacology , Receptors, Platelet-Derived Growth Factor , Tyrosine/immunology , Tyrosine/metabolism
13.
Biochim Biophys Acta ; 971(3): 351-7, 1988 Oct 07.
Article in English | MEDLINE | ID: mdl-2844295

ABSTRACT

The conditions affecting the appearance and disappearance of platelet-derived growth factor (PDGF) receptors from the pool of active cell surface-associated receptors were studied. Receptor molecules were revealed in intact Swiss 3T3 fibroblasts by their ability to bind 125I-labeled PDGF and, due to their property to become phosphorylated in tyrosine following ligand binding, by antibodies to phosphotyrosine. PDGF receptor molecules were found to be quite scarce in exponentially growing fibroblasts as compared to quiescent cells. When growing cells were either shifted to a medium containing plasma or received suramin in the culture medium, cell surface-associated PDGF receptors largely increased. This process required about 12 h. Incubation of quiescent cells in serum, but not in plasma, induced a slow decrement of ligand-activatable receptors. In the presence of PDGF the rate of receptor removal from the cell surface was very rapid and was a function of the PDGF concentration. Quiescent cells deprived of cell-surface receptors by incubation with PDGF reexpressed PDGF receptors in about 14 h.


Subject(s)
Cell Membrane/metabolism , Platelet-Derived Growth Factor/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Division , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Kinetics , Mice , Phosphorylation , Receptors, Cell Surface/isolation & purification , Receptors, Platelet-Derived Growth Factor
14.
Gene ; 161(2): 287-92, 1995 Aug 19.
Article in English | MEDLINE | ID: mdl-7665095

ABSTRACT

A reporter plasmid coding for the low-molecular-weight (low-M(r)) form of single-chain urokinase-type plasminogen activator (low-M(r) u-PA; Leu144-Leu411) has been constructed and used to analyze promoter activity. Vectors containing the low-M(r) u-PA cDNA coupled to hormone-responsive promoters were introduced to mammalian cells. Following hormone treatment, the activity of the secreted reporter protein was determined in aliquots of cell culture supernatants. The assay is based on plasminogen activation by low-M(r) u-PA and subsequent cleavage of the plasmin-specific tripeptide substrate, S-2251. The resulting chromophore, p-nitroanilide, was quantified colorimetrically at 405 nm. Transient and stable expression of the low-M(r) u-PA reporter gene in different eukaryotic cells demonstrates the suitability of the system for the quantification of the activity of eukaryotic promoter elements in a rapid and highly sensitive manner, while maintaining cell integrity.


Subject(s)
Urokinase-Type Plasminogen Activator/genetics , Animals , Base Sequence , Cell Line , DNA, Complementary/genetics , Gene Expression , Genes, Reporter , Genetic Vectors , HeLa Cells , Humans , L Cells , Mice , Molecular Sequence Data , Molecular Weight , Plasmids/genetics , Promoter Regions, Genetic , Transfection , Urokinase-Type Plasminogen Activator/chemistry
15.
Brain Res Bull ; 36(5): 443-52, 1995.
Article in English | MEDLINE | ID: mdl-7712206

ABSTRACT

This article investigates the distribution of neurotransmitters, neuropeptides, and related receptors in the vestibular nuclei complex (VNC) of the adult rat by means of immunohistochemistry, in situ hybridization, and quantitative receptor autoradiography. The entire complex proves to be rich in muscarinic receptors and it shows a high density of imipramine and benzodiazepine binding sites. Peptidergic neurons and a few positive fibers are described in the caudal part of the VNC. In particular, the medial vestibular nucleus contains a number of neurons expressing both the enkephalin mRNA and peptide. This nucleus and the lateral vestibular nucleus are also rich in opiate receptors. Substance P, thyrotropin releasing hormone, and neurotensin receptors are also found in the medial and in the spinal vestibular nuclei. In spite of the presence of alpha 2 catecholaminergic receptors, no thyrosine-hydroxylase-immuno-reactive elements are seen in the caudal VNC. The possible functional meaning of these data is discussed.


Subject(s)
Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Neurotransmitter/metabolism , Vestibular Nuclei/metabolism , Animals , Autoradiography , Base Sequence , Immunohistochemistry , In Situ Hybridization , Male , Molecular Sequence Data , Oligonucleotide Probes/genetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
16.
Int J Biol Markers ; 22(4): 10-23, 2007.
Article in English | MEDLINE | ID: mdl-28207110

ABSTRACT

The epidermal growth factor receptor (EGFR) plays a key role in cancer development and progression in several human malignancies including non-small cell lung cancer (NSCLC). Several strategies aimed at inhibiting the EGFR have been investigated in the last years, including the use of small tyrosine kinase inhibitors (TKIs) directed against the intracellular domain of the receptor and monoclonal antibodies targeting its extracellular portion. Subgroups of patients who are more likely to respond to TKIs have been identified based on both clincal and biological features. Never-smoking history has emerged as the most relevant clinical characteristic predictive of response to TKIs in NSCLC, while presence of drugsensitive EGFR mutations and EGFR gene gain represent critical biological variables associated with an improved outcome for patients exposed to these agents. Recent studies have highlighted the existence of biological factors involved in intrinsic and acquired resistance to TKIs, including k-ras, HER-2 and EGFR exon 20 mutations. Increasing knowledge of EGFR biology and drug-receptor interactions will allow to identify individuals who are likely to derive a clinical benefit from the proposed targeted therapy, sparing refractory patients expensive and potentially toxic treatment.

17.
Ann Oncol ; 17(7): 1120-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16600976

ABSTRACT

BACKGROUND: The aim of the study was to assess whether loss of PTEN and expression of insulin-like growth factor receptor 1 (IGFR-1) could be responsible for intrinsic resistance to the tyrosine kinase inhibitor (TKI) gefitinib. PATIENTS AND METHODS: One hundred and twenty-four gefitinib-treated patients with advanced non-small-cell lung cancer (NSCLC) were analyzed for PTEN and IGFR-1 expression by immunohistochemistry. RESULTS: IGFR-1 was evaluated in 77 patients and resulted positive in 30 (39.0%). IGFR-1 expression was not significantly associated with clinical or biological characteristics. No difference in response to gefitinib treatment (16.7% versus 12.8%, P = 0.74) and time to progression (2.6 versus 3.06 months, P = 0.83) was observed between IGFR-1+ and IGFR-1-. Median survival was significantly longer in IGFR-1+ patients (17.8 versus 7.3 months, P = 0.013). PTEN expression was successfully evaluated in 93 cases. Loss of PTEN was detected in 19 tumors (20.4%) and was not associated with any clinical or biological characteristic. No difference in terms of response, time to progression and survival was observed between PTEN+ and PTEN- patients. In multivariable analysis IGFR-1 negative status was significantly associated with higher risk of death (hazard ratio 2.21, P = 0.012). CONCLUSIONS: IGFR-1 expression and loss of PTEN are not associated with intrinsic resistance to gefitinib. Clinical relevance of these two biomarkers as determinant for acquired resistance, and the prognostic role of IGFR-1 expression in patients not exposed to TKIs should be evaluated further.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Lung Neoplasms/mortality , PTEN Phosphohydrolase/metabolism , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Receptor, IGF Type 1/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance , Female , Gefitinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Middle Aged , Multivariate Analysis , Patient Selection , Survival Analysis
18.
Future Oncol ; 1(1): 7-17, 2005 Feb.
Article in English | MEDLINE | ID: mdl-16555971

ABSTRACT

Gemcitabine, a pyrimidine nucleoside antimetabolite, is one of the most promising new cytotoxic agents. The drug has shown activity in a variety of solid tumors, and has been approved for the treatment of non-small cell lung cancer, pancreatic, bladder, and breast cancer. Recent data showed that gemcitabine is also active against ovarian cancer. Gemcitabine has a good toxicity profile, with myelosuppression being the most common side effect, while non-hematological events are relatively uncommon. The low toxicity profile makes the drug a valid option for unfit and elderly patients. Due to the synergistic activity with other chemotherapeutic compounds, mainly cisplatinum, several trials have been conducted to evaluate the efficacy and tolerability of gemcitabine in combination with other cytotoxic agents. Current clinical trials are evaluating the role of gemcitabine in combination with new targeted therapies.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxycytidine/adverse effects , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Interactions , Female , Humans , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Urinary Bladder Neoplasms/drug therapy , Gemcitabine
19.
Br J Cancer ; 93(12): 1334-40, 2005 Dec 12.
Article in English | MEDLINE | ID: mdl-16288303

ABSTRACT

In non-small-cell lung cancer (NSCLC), sensitivity to tyrosine kinase inhibitors (TKIs) is associated with activating mutations and genomic gain of the epidermal growth factor receptor (EGFR). Preclinical data suggested that HER3 overexpression increases sensitivity to TKIs. A total of 82 NSCLC patients treated with gefitinib (250 mg), and previously evaluated for EGFR and HER2 status by fluorescence in situ hybridisation (FISH) and DNA sequencing, and for Phospho-Akt status by immunohistochemistry, were investigated for HER3 genomic gain by FISH. Patients with high polysomy and gene amplification were considered as HER3 FISH positive (+). HER3 FISH+ pattern was significantly associated with female gender (P=0.02) and never smoking history (P=0.02). Patients with HER3+ tumours (26.8%) had a significantly longer time to progression (3.7 vs 2.7, P=0.04) than patients with HER3- tumours, but not a significantly better response rate or survival. Patients with EGFR+/HER3+ tumours had higher objective response rate (36.4 vs 9.9%, P=0.03) and time to progression (7.7 vs 2.7 months, P=0.03) than patients with EGFR- and/or HER3- tumours, but no significantly longer survival. No difference in response was observed according to HER3 status in patients with EGFR+ tumours. Patients with HER2+/HER3+ tumours had similar outcome as patients with HER2- and/or HER3- tumours. Significantly different clinical end points were not observed between patients with HER3+/P-Akt+ and HER3- and/or P-Akt- tumours. Genomic gain for HER3 is not a marker for response or resistance to TKI therapy in advanced NSCLC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor, ErbB-3/biosynthesis , Biomarkers, Tumor/analysis , Disease Progression , Drug Resistance, Neoplasm , ErbB Receptors , Female , Gefitinib , Gene Amplification , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Predictive Value of Tests , Prognosis , Receptor, ErbB-3/analysis , Receptor, ErbB-3/genetics , Sex Factors , Survival Analysis
20.
Cell Biol Int Rep ; 10(7): 575-82, 1986 Jul.
Article in English | MEDLINE | ID: mdl-2425987

ABSTRACT

In normal mouse fibroblasts exponentially growing at suboptimal serum concentration the population of cells having a presynthetic (G1) DNA content is heterogeneous, since there are present both competent and non-competent cells (Sturani et al, 1984). We show here that SV40 transformed 3T3 fibroblasts, which have lost growth control and serum requirement, lack the G1 non-competent phase. These findings are confirmed by the different kinetics of exit from the G1 compartment of transformed and normal cells. In addition a positive correlation is reported between the fraction of non-competent cells and the average abundance of the phosphorylable PDGF receptor on the cell surface of normal cells.


Subject(s)
Cell Cycle/drug effects , Fibroblasts/cytology , Platelet-Derived Growth Factor/pharmacology , Receptors, Cell Surface/physiology , Animals , Cell Transformation, Viral , Culture Media , Interphase , Mice , Phosphorylation , Phosphotyrosine , Receptors, Platelet-Derived Growth Factor , Simian virus 40 , Tyrosine/analogs & derivatives , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL