Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Eur J Cancer ; 106: 225-233, 2019 01.
Article in English | MEDLINE | ID: mdl-30528807

ABSTRACT

BACKGROUND: To explore the activity of axitinib in advanced solitary fibrous tumour (SFT). PATIENTS AND METHODS: In this investigator-driven phase II study on axitinib in advanced and progressive SFT, patients received axitinib, 5 mg bis in day (BID), until progression or limiting toxicity. Pathologic diagnosis was centrally reviewed, distinguishing malignant SFT (M-SFT) and high-grade/dedifferentiated SFT (HG/D-SFT) subtypes. The primary end-point was the overall response rate (ORR) by Choi criteria (Choi). Secondary end-points were response by Response Evaluation Criteria in Solid Tumours (RECIST), progression-free survival (PFS) and overall survival (OS). RESULTS: From April 2015 and October 2017, 17 eligible patients entered the study (metastatic: 17; SFT subtype: 13 M-SFT, 4 HG/D-SFT; prior treatment: 9 antiangiogenics, 5 cytotoxics). All patients were evaluable for response. The best Choi response was seven partial response (PR) (ORR, 41.2%), six stable disease (SD) and four progressions. Choi-ORR was 54% (7/13) when only M-SFTs were considered. Four of seven responsive patients were pretreated with pazopanib. No responses were detected in HG/D-SFT. Best RECIST response was one PR (5.9%), 14 SD and two progressions. Toxicity was as expected. Median Choi-PFS was 5.1 (interquartile range [IQR]: 2.5-14.8) months. Median Choi-PFS was 14.8 (IQR: 5.1-18.0) and 2.8 (IQR: 2.0-5.9) months for patients responsive and non-responsive by Choi, respectively (p = 0.0416). At a 14.4-month median follow-up, median OS was 25.3 months. CONCLUSION: This study showed that axitinib is active in progressive advanced SFT. One-half of patients carrying the malignant variant of the disease responded, with a >12-month median progression arrest. Responses were better detected with Choi and seen even in patients resistant to other antiangiogenics. Tolerability was good.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Axitinib/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Solitary Fibrous Tumors/drug therapy , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Axitinib/adverse effects , Disease Progression , Female , Humans , Italy , Male , Middle Aged , Progression-Free Survival , Protein Kinase Inhibitors/adverse effects , Response Evaluation Criteria in Solid Tumors , Solitary Fibrous Tumors/mortality , Solitary Fibrous Tumors/secondary , Time Factors
2.
Eur J Cancer ; 76: 84-92, 2017 05.
Article in English | MEDLINE | ID: mdl-28284173

ABSTRACT

BACKGROUND: Preclinical models that mimic pathological and molecular features of solitary fibrous tumour (SFT) represent an important tool to select effective regimes and novel compounds to be tested in the clinic. This study was aimed at developing two preclinical models of SFT, assessing their predictive value in the clinic and selecting potential novel effective treatments. MATERIAL AND METHODS: Two dedifferentiated-SFT (D-SFT) models obtained from patients' biopsies were grown in immunodeficient mice. The antitumour activity on these models of doxorubicin, dacarbazine (DTIC), ifosfamide (monotherapy or combination), trabectedin and eribulin was tested. Twelve SFT patients were treated with doxorubicin and DTIC. Response by RECIST, progression-free survival and overall survival were retrospectively evaluated, distinguishing malignant-SFT (M-SFT) and D-SFT. RESULTS: Two D-SFT patient-derived xenografts (PDXs) that represent the first available preclinical in vivo models of SFT were developed and characterised. Doxorubicin/DTIC, DTIC/ifosfamide, doxorubicin/ifosfamide combinations consistently induced better antitumour activity than the single-agents. Particularly, doxorubicin/DTIC combination caused a max tumour volume inhibition >80% in both models. Doxorubicin/DTIC combo showed activity also in the case-series. Best RECIST responses were: 6 responses (M-SFT = 2 of 7, D-SFT = 4 of 5), 1 stable disease, 5 progressions, with a 6-month median progression-free survival (M-SFT = 6, D-SFT = 10 months). The PDXs were very sensitive to trabectedin and eribulin. CONCLUSION: Doxorubicin plus DTIC combination was effective in our two D-SFT mice models and appeared to be active also in the clinic, especially in high-grade D-SFT patients. Among additional drugs tested in the PDXs, trabectedin and eribulin were highly effective, providing a rational to test these drugs in D-SFT patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cerebellar Neoplasms/drug therapy , Kidney Neoplasms/drug therapy , Meningeal Neoplasms/drug therapy , Pleural Neoplasms/drug therapy , Retroperitoneal Neoplasms/drug therapy , Soft Tissue Neoplasms/drug therapy , Solitary Fibrous Tumors/drug therapy , Adult , Aged , Animals , Blotting, Western , Cerebellar Neoplasms/mortality , Dacarbazine/administration & dosage , Dioxoles/administration & dosage , Disease-Free Survival , Doxorubicin/administration & dosage , Female , Furans/administration & dosage , Humans , Ifosfamide/administration & dosage , Ketones/administration & dosage , Kidney Neoplasms/mortality , Male , Meningeal Neoplasms/mortality , Mice, SCID , Middle Aged , Pleural Neoplasms/mortality , Response Evaluation Criteria in Solid Tumors , Retroperitoneal Neoplasms/mortality , Retrospective Studies , Soft Tissue Neoplasms/mortality , Solitary Fibrous Tumors/mortality , Survival Rate , Tetrahydroisoquinolines/administration & dosage , Trabectedin , Xenograft Model Antitumor Assays
3.
BMC Cancer ; 17(1): 107, 2017 Feb 06.
Article in English | MEDLINE | ID: mdl-28166781

ABSTRACT

BACKGROUND: Desmoplastic small round cell tumor (DSRCT) is a rare and highly aggressive disease, that can be described as a member of the family of small round blue cell tumors. The molecular diagnostic marker is the t(11;22)(p13;q12) translocation, which creates an aberrant transcription factor, EWS-WT1, that underlies the oncogenesis of DSRCT. Current treatments are not very effective so new active drugs are needed. Trabectedin, now used as a single agent for the treatment of soft tissue sarcoma, was reported to be active in some pre-treated DSRCT patients. Using JN-DSRCT-1, a cell line derived from DSRCT expressing the EWS-WT1 fusion protein, we investigated the ability of trabectedin to modify the function of the chimeric protein, as in other sarcomas expressing fusion proteins. After detailed characterization of the EWS-WT1 transcripts structure, we investigated the mode of action of trabectedin, looking at the expression and function of the oncogenic chimera. METHODS: We characterized JN-DSRCT-1 cells using cellular approaches (FISH, Clonogenicity assay) and molecular approaches (Sanger sequencing, ChIP, GEP). RESULTS: JN-DSRCT-1 cells were sensitive to trabectedin at nanomolar concentrations. The cell line expresses different variants of EWS-WT1, some already identified in patients. EWS-WT1 mRNA expression was affected by trabectedin and chimeric protein binding on its target gene promoters was reduced. Expression profiling indicated that trabectedin affects the expression of genes involved in cell proliferation and apoptosis. CONCLUSIONS: The JN-DSRCT-1 cell line, in vitro, is sensitive to trabectedin: after drug exposure, EWS-WT1 chimera expression decreases as well as binding on its target promoters. Probably the heterogeneity of chimera transcripts is an obstacle to precisely defining the molecular mode of action of drugs, calling for further cellular models of DSRCT, possibly growing in vivo too, to mimic the biological complexity of this disease.


Subject(s)
Desmoplastic Small Round Cell Tumor/drug therapy , Dioxoles/pharmacology , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/drug effects , Tetrahydroisoquinolines/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Desmoplastic Small Round Cell Tumor/metabolism , Desmoplastic Small Round Cell Tumor/physiopathology , Dioxoles/therapeutic use , Humans , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein EWS , Tetrahydroisoquinolines/therapeutic use , Trabectedin , WT1 Proteins
4.
J Invest Dermatol ; 137(2): 484-493, 2017 02.
Article in English | MEDLINE | ID: mdl-27608549

ABSTRACT

Dermatofibrosarcoma protuberans (DFSP), although rare, is the most frequent skin sarcoma. Here, we focus on DFSP carrying the fibrosarcomatous transformation (FS-DFSP). FS-DFSP responds to imatinib (IM); however, tumor relapse often occurs. In a series of 21 pre- and post-treatment FS-DFSP samples, the present study explored the events that occur at the tumor site during IM therapy. Gene expression profile and immunohistochemistry analyses documented the occurrence of IM-induced senescence phenotype in the tumor cells and showed the accumulation of activated CD3+ T cells and CD163+CD14+ myeloid cells expressing the CD209 marker in post-therapy lesions. In post-IM specimens, the pathological response and tumor apoptosis were tightly associated with T-cell infiltration, thus suggesting the presence of an ongoing anti-tumor response, which was further confirmed by in vitro functional assays with CD3+ T cells isolated from an IM-responding FS-DFSP lesion. The integration of targeted therapies with immune therapies is currently under investigation to achieve longer tumor control. Our data outline the in situ immunological effects of IM and classify IM-treated FS-DFSP as potentially sensitive to immunotherapy, thus providing the rationale for further investigations of combination treatment for this soft-tissue sarcoma.


Subject(s)
Adaptive Immunity/drug effects , Antineoplastic Agents/therapeutic use , Dermatofibrosarcoma/drug therapy , Imatinib Mesylate/therapeutic use , Skin Neoplasms/drug therapy , Apoptosis/drug effects , Cell Adhesion Molecules/analysis , Dermatofibrosarcoma/immunology , Dermatofibrosarcoma/pathology , Histocompatibility Antigens Class I/analysis , Humans , Lectins, C-Type/analysis , Myeloid Cells/drug effects , Receptors, Cell Surface/analysis , Skin Neoplasms/immunology , T-Lymphocytes/immunology
5.
Eur J Cancer ; 69: 158-165, 2016 12.
Article in English | MEDLINE | ID: mdl-27821319

ABSTRACT

BACKGROUND: Pre-clinical and clinical evidence suggests a rationale for the use of anti-angiogenic agents, including sorafenib, in recurrent and/or metastatic salivary gland carcinomas (RMSGCs). This study evaluates the activity of sorafenib in patients with RMSGCs and also investigates whether the activity of sorafenib could be related to its main tailored targets (i.e. BRAF, vascular endothelial growth factor receptor 2 [VEGFR2], platelet-derived growth factor receptor α [PDGFRα] and ß, RET, KIT). PATIENTS AND METHODS: Patients received sorafenib at 400 mg BID. The primary end-point was response rate (RR) including complete response or partial response (PR); secondary end-points included RR according to Choi criteria, disease control rate (DCR), overall survival (OS), and progression-free survival (PFS). RESULTS: Thirty-seven patients (19 adenoid cystic cancers, ACC) were enrolled. Six PRs were recorded. RR was 16% (95% confidence interval [CI]: 6-32; 11% in ACC and 22% in non-ACC). Choi criteria could be applied in 30 out of 37 cases with a RR of 50% (95% CI: 31-69%); DCR was 76% (95% CI: 59-88%). Incidence of ≥G3 adverse events was 29.7%. Median PFS and OS for the entire population were 5.9 months and 23.4 months, respectively. Median PFS and OS were 8.9 and 26.4 months for ACC versus 4.2 and 12.3 months for non-ACC patients. All the cases showed expression of PDGFRß in the stroma and VEGFR2 in endothelial cells; PDGFRα positivity was found in the stroma of four (27%) cases. All except for two cases showed no PDGFRß, VEGFR2 and PDGFRα expression in the tumour cells. KIT expression was restricted to ACC and a weak RET expression was limited to one adenocarcinoma, not otherwise specified (NOS). No BRAF mutation was found. No correlation was observed between the sorafenib activity and the expression of its markers although all six responders (two ACC, one adenocarcinoma, NOS, one salivary duct cancer [SDC], one high-grade mucoepidermoid [HG-MEC] and one poorly-differentiated cancer) are enriched in the stromal component showing a PDGFRß immunodecoration. In ACCs, immunohistochemistry revealed MYB protein expression in 15/16 cases (94%) and the MYB-NFIB fusion oncogene was observed in 9/14 (64%). CONCLUSIONS: Sorafenib is the first anti-angiogenic agent to demonstrate activity in RMSGC patients, particularly in some histotypes such as HG-MEC, SDC and adenocarcinoma, NOS. The PDGFRß-positive rich stromal component characterising these histotypes and the lack of correlation between the activity of sorafenib and its targets suggests anti-angiogenic effect as the prevalent mechanism of action of sorafenib in SGCs.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Carcinoma, Adenoid Cystic/drug therapy , Carcinoma, Mucoepidermoid/drug therapy , Myoepithelioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Salivary Gland Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Adult , Aged , Carcinoma, Adenoid Cystic/metabolism , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Adenoid Cystic/secondary , Carcinoma, Mucoepidermoid/metabolism , Carcinoma, Mucoepidermoid/pathology , Carcinoma, Mucoepidermoid/secondary , Diarrhea/chemically induced , Disease-Free Survival , Drug Eruptions/etiology , Fatigue/chemically induced , Hand-Foot Syndrome/etiology , Humans , Hypertension/chemically induced , Immunohistochemistry , Male , Middle Aged , Myoepithelioma/metabolism , Myoepithelioma/pathology , Myoepithelioma/secondary , Neoplasm Metastasis , Neoplasm Recurrence, Local/metabolism , Niacinamide/therapeutic use , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology , Sorafenib , Survival Rate , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult
7.
Eur J Cancer ; 50(17): 3021-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25269954

ABSTRACT

BACKGROUND: To explore the activity of pazopanib in solitary fibrous tumour (SFT). PATIENTS AND METHODS: In a preclinical study, we compared the activity of pazopanib, sorafenib, sunitinib, regorafenib, axitinib and bevacizumab in a dedifferentiated-SFT (DSFT) xenotransplanted into Severe Combined Immunodeficiency (SCID) mice. Antiangiogenics were administered at their reported optimal doses when mean tumour volume (TV) was 80 mm(3). Drug activity was assessed as TV inhibition percentage (TVI%). From May 2012, six consecutive patients with advanced SFT received pazopanib, on a national name-based programme. In one case sunitinib was administered after pazopanib failure. RESULTS: In the xenograft model, pazopanib showed the lowest antitumour activity (21%TVI), while regorafenib was the most active (95%TVI). Sorafenib, bevacizumab, sunitinib were markedly active (78/70/65%TVI). Axitinib was marginally active (51%TVI). In the retrospective case-series, three patients carried malignant-SFT (MSFT), three DSFT. Best Response Evaluation Criteria in Solid Tumour (RECIST) responses were: three stable disease (SD), all MSFT, three progressive disease (PD), all DSFT, corresponding to one partial response (PR), two SD, three PD by Choi criteria. Median-progression-free survival was 3 months (range 1-15). In one patient, sunitinib was started after pazopanib failure, with a response. CONCLUSIONS: In dedifferentiated-SFT xenograft pazopanib induced a marginal antitumour activity, while regorafenib appeared the most active and promising agent. When administered in patients, pazopanib showed a modest activity in terms of tumour growth stabilisation, observed only in non-dedifferentiated cases.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Pyrimidines/pharmacology , Solitary Fibrous Tumors/drug therapy , Sulfonamides/pharmacology , Administration, Oral , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/administration & dosage , Axitinib , Bevacizumab , Humans , Imidazoles/pharmacology , Indazoles/pharmacology , Indoles/pharmacology , MAP Kinase Signaling System/physiology , Male , Mice, SCID , Middle Aged , Neoplasm Transplantation , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Pyrimidines/administration & dosage , Pyrroles/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Sorafenib , Sulfonamides/administration & dosage , Sunitinib , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Eur J Cancer ; 50(9): 1657-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24703573

ABSTRACT

BACKGROUND: Extraskeletal myxoid chondrosarcoma (EMC) is a rare soft tissue sarcoma, marked by NR4A3 rearrangement. Herein we report on the activity of sunitinib in a series of 10 patients, strengthening what initially observed in two cases. PATIENTS AND METHODS: From July 2011, 10 patients with progressive metastatic translocated EMC have been consecutively treated with sunitinib 37.5mg/day, on a named-use basis. In an attempt to interpret the activity of sunitinib in EMC, genotype/phenotype correlations were carried out by fluorescence in situ hybridization (FISH) analyses. Moreover, transcriptome, immunohistochemical and biochemical analyses of a limited set of samples were performed focusing on some putative targets of sunitinib. RESULTS: Eight of 10 patients are still on therapy. Six patients had a Response Evaluation Criteria in Solid Tumours (RECIST) partial response (PR), two were stable, two progressed. Positron emission tomography (PET) was consistent in 6/6 evaluable cases. One patient underwent surgery after sunitinib, with evidence of a pathologic response. At a median follow-up of 8.5 months (range 2-28), no secondary resistance was detected. Median progression free survival (PFS) has not been reached. Interestingly, all responsive cases turned out to express the typical EWSR1-NR4A3 fusion, while refractory cases carried the alternative TAF15-NR4A3 fusion. Among putative sunitinib targets, only RET was expressed and activated in analysed samples. CONCLUSIONS: This report confirms the therapeutic activity of sunitinib in EMC. Genotype/phenotype analyses support a correlation between response and EWSR1-NR4A3 fusion. Involvement of RET deserves further investigation.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Bone Neoplasms , Chondrosarcoma/drug therapy , Indoles/therapeutic use , Neoplasms, Connective and Soft Tissue/drug therapy , Pyrroles/therapeutic use , Adult , Aged , Calmodulin-Binding Proteins/genetics , Chondrosarcoma/genetics , Chondrosarcoma/secondary , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Rearrangement/genetics , Genotype , Humans , Male , Middle Aged , Neoplasms, Connective and Soft Tissue/genetics , Neoplasms, Connective and Soft Tissue/secondary , Phenotype , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-ret/drug effects , RNA-Binding Protein EWS , RNA-Binding Proteins/genetics , Receptor, Platelet-Derived Growth Factor beta/drug effects , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Sunitinib , TATA-Binding Protein Associated Factors/genetics , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/drug effects , fms-Like Tyrosine Kinase 3/drug effects
9.
Leukemia ; 26(2): 349-55, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21836613

ABSTRACT

The association of genetic lesions detected by fluorescence in situ hybridization (FISH) with survival was analyzed in 1069 patients with newly presenting myeloma treated in the Medical Research Council Myeloma IX trial, with the aim of identifying patients associated with the worst prognosis. A comprehensive FISH panel was performed, and the lesions associated with short progression-free survival and overall survival (OS) in multivariate analysis were +1q21, del(17p13) and an adverse immunoglobulin heavy chain gene (IGH) translocation group incorporating t(4;14), t(14;16) and t(14;20). These lesions frequently co-segregated, and there was an association between the accumulation of these adverse FISH lesions and a progressive impairment of survival. This observation was used to define a series of risk groups based on number of adverse lesions. Taking this approach, we defined a favorable risk group by the absence of adverse genetic lesions, an intermediate group with one adverse lesion and a high-risk group defined by the co-segregation of >1 adverse lesion. This genetic grouping was independent of the International Staging System (ISS) and so was integrated with the ISS to identify an ultra-high-risk group defined by ISS II or III and >1 adverse lesion. This group constituted 13.8% of patients and was associated with a median OS of 19.4 months.


Subject(s)
Models, Theoretical , Multiple Myeloma/pathology , Humans , In Situ Hybridization, Fluorescence , Multiple Myeloma/genetics , Prognosis , Survival Analysis , Translocation, Genetic
10.
Leukemia ; 20(9): 1610-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16826223

ABSTRACT

In myeloma, the prognostic impact of different strategies used to detect chromosome 13 deletion (Delta13) remains controversial. To address this, we compared conventional cytogenetics and interphase fluorescence in situ hybridization (iFISH) in a large multicenter study (n=794). The ability to obtain abnormal metaphases was associated with a poor prognosis, which was worse if Delta13, p53 deletion or t(4;14) was present, but only Delta13 remained significant on multivariate analysis. Patients with Delta13, by either cytogenetics or iFISH, had a poor prognosis. However, when cases with Delta13 detectable by both cytogenetics and iFISH were separated from those detected by iFISH only, the poor prognosis of iFISH-detectable Delta13 disappeared; their outcome matched that of patients with no detectable Delta13 (P=0.115). Addition of ploidy status to iFISH-Delta13 did not affect the prognostic value of the test. Indeed both cytogenetics and iFISH Delta13 divided both hyperdiploidy and nonhyperdiploidy into two groups with similar prognoses, indicating that the poor prognosis of ploidy is entirely due to its association with Delta13. We conclude that Delta13 detected by metaphase analysis is a critical prognostic factor in myeloma. Absence of Delta13, even in those patients yielding only normal or no metaphases, is associated with a relatively good prognosis.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 13 , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Genes, Immunoglobulin Heavy Chain , Genes, p53 , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Ploidies , Prognosis , Survival Analysis , Translocation, Genetic
11.
J Pathol ; 208(5): 615-23, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16470538

ABSTRACT

Chondrosarcomas represent 20% of all primary bone sarcomas, and many studies have attempted to unravel molecular targets for future development of new therapies. The aim of this study was to investigate the expression/activation of PDGFRalpha, PDGFRbeta and KIT receptor tyrosine kinases (RTKs) as potential therapeutic targets in conventional central primary chondrosarcomas (CCS). The expression of PDGFRalpha, PDGFRbeta and KIT RTKs was detected in 16 CCSs using immunohistochemistry (IHC), and their level of expression and activation status were analysed by immunoprecipitation and western blot experiments. PDGFRalpha, PDGFRbeta and KIT cDNAs were screened to verify the presence of activating mutations and the presence of the cognate ligands was analysed by means of RT-PCR. RTK gene amplification was further studied by means of fluorescence in situ hybridization (FISH) analysis. The immunophenotyping and biochemical analyses showed that the CCSs co-expressed PDGFRalpha and PDGFRbeta, with the latter showing definitively greater protein expression and phosphorylation levels. PDGFRbeta was expressed but not activated in control healthy joint cartilage, in line with no PDGFB detection. Conversely, the KIT gene product did not seem to play a relevant role. These findings, in the absence of activating mutations or an abnormal genomic profile and the presence of PDGFA and PDGFB expression, are consistent with an autocrine/paracrine loop activation of the corresponding receptors. The CCS gene profile described here offers a rationale for the use of RTK inhibitors alone or in combination with chemotherapy, and supports further investigation of RTKs and their downstream signals.


Subject(s)
Bone Neoplasms/metabolism , Chondrosarcoma/metabolism , Neoplasm Proteins/metabolism , Bone Neoplasms/genetics , Chondrosarcoma/genetics , Gene Expression Profiling/methods , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Ligands , Neoplasm Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
14.
Cytopathology ; 13(5): 317-25, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12421448

ABSTRACT

Detection of bladder cancer by multitarget multicolour FISH: comparative analysis on archival cytology and paraffin-embedded tissue We have evaluated the possibility of using the same specimen for both cytological diagnosis and multitarget multicolour FISH (MtMcFISH) analysis in order to determine whether the routinely processed specimens used for diagnosis were also suitable for this ancillary procedure. For this purpose 18 positive samples (11 voided urine and seven bladder washings) were selected, together with a representative section of the corresponding immediately previous or subsequent histological specimens. Two negative cytology slides were added as negative controls. FISH analysis revealed a normal pattern for each probe in the two negative controls and an abnormal pattern in the 18 positive cases. In the latter the same FISH alterations were found in the cytology samples and in the corresponding histological sections, and superimposable cytological/histological features were observed in two cases where two different histology samples were analyzed. The results clearly show that MtMcFISH may be successfully applied to destained routinely processed cytology slides.


Subject(s)
Carcinoma, Transitional Cell/pathology , Cytodiagnosis/methods , In Situ Hybridization, Fluorescence , Urinary Bladder Neoplasms/pathology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/urine , DNA, Neoplasm/analysis , Humans , Neoplasm Staging , Reagent Kits, Diagnostic , Retrospective Studies , Specimen Handling/methods , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...