Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Leukemia ; 32(2): 383-390, 2018 02.
Article in English | MEDLINE | ID: mdl-28761118

ABSTRACT

The Dutch-Belgian Cooperative Trial Group for Hematology Oncology Group-65/German-speaking Myeloma Multicenter Group-HD4 (HOVON-65/GMMG-HD4) phase III trial compared bortezomib (BTZ) before and after high-dose melphalan and autologous stem cell transplantation (HDM, PAD arm) compared with classical cytotoxic agents prior and thalidomide after HDM (VAD arm) in multiple myeloma (MM) patients aged 18-65 years. Here, the long-term follow-up and data on second primary malignancies (SPM) are presented. After a median follow-up of 96 months, progression-free survival (censored at allogeneic transplantation, PFS) remained significantly prolonged in the PAD versus VAD arm (hazard ratio (HR)=0.76, 95% confidence interval (95% CI) of 0.65-0.89, P=0.001). Overall survival (OS) was similar in the PAD versus VAD arm (HR=0.89, 95% CI: 0.74-1.08, P=0.24). The incidence of SPM were similar between the two arms (7% each, P=0.73). The negative prognostic effects of the cytogenetic aberration deletion 17p13 (clone size ⩾10%) and renal impairment at baseline (serum creatinine >2 mg dl-1) on PFS and OS remained abrogated in the PAD but not VAD arm. OS from first relapse/progression was similar between the study arms (HR=1.02, P=0.85). In conclusion, the survival benefit with BTZ induction/maintenance compared with classical cytotoxic agents and thalidomide maintenance is maintained without an increased risk of SPM.


Subject(s)
Bortezomib/administration & dosage , Multiple Myeloma/drug therapy , Adolescent , Adult , Aged , Chromosome Aberrations/drug effects , Female , Follow-Up Studies , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Melphalan/therapeutic use , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Prognosis , Progression-Free Survival , Thalidomide/therapeutic use , Transplantation, Autologous/methods , Young Adult
3.
Leukemia ; 31(10): 2048-2056, 2017 10.
Article in English | MEDLINE | ID: mdl-28196983

ABSTRACT

Recent developments in sequencing technologies led to the discovery of a novel form of genomic instability, termed chromothripsis. This catastrophic genomic event, involved in tumorigenesis, is characterized by tens to hundreds of simultaneously acquired locally clustered rearrangements on one chromosome. We hypothesized that leukemias developing in individuals with Ataxia Telangiectasia, who are born with two mutated copies of the ATM gene, an essential guardian of genome stability, would show a higher prevalence of chromothripsis due to the associated defect in DNA double-strand break repair. Using whole-genome sequencing, fluorescence in situ hybridization and RNA sequencing, we characterized the genomic landscape of Acute Lymphoblastic Leukemia (ALL) arising in patients with Ataxia Telangiectasia. We detected a high frequency of chromothriptic events in these tumors, specifically on acrocentric chromosomes, as compared with tumors from individuals with other types of DNA repair syndromes (27 cases total, 10 with Ataxia Telangiectasia). Our data suggest that the genomic landscape of Ataxia Telangiectasia ALL is clearly distinct from that of sporadic ALL. Mechanistically, short telomeres and compromised DNA damage response in cells of Ataxia Telangiectasia patients may be linked with frequent chromothripsis. Furthermore, we show that ATM loss is associated with increased chromothripsis prevalence in additional tumor entities.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/physiology , Ataxia Telangiectasia/genetics , Neoplasm Proteins/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Ataxia Telangiectasia/complications , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Child , Child, Preschool , Chromosomes, Human/ultrastructure , Chromothripsis , DNA Repair/genetics , DNA, Neoplasm/genetics , Female , Genome, Human , Genomic Instability , Humans , In Situ Hybridization, Fluorescence , Male , Mutation , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasms/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Neoplasm/genetics , Sequence Analysis, DNA , Sequence Analysis, RNA , Telomere Shortening/genetics , Transcriptome
4.
Leukemia ; 31(8): 1735-1742, 2017 08.
Article in English | MEDLINE | ID: mdl-28025584

ABSTRACT

Immunoglobulin light chain (AL) amyloidosis is characterized by tissue deposition of amyloid fibers derived from immunoglobulin light chain. AL amyloidosis and multiple myeloma (MM) originate from monoclonal gammopathy of undetermined significance. We wanted to characterize germline susceptibility to AL amyloidosis using a genome-wide association study (GWAS) on 1229 AL amyloidosis patients from Germany, UK and Italy, and 7526 healthy local controls. For comparison with MM, recent GWAS data on 3790 cases were used. For AL amyloidosis, single nucleotide polymorphisms (SNPs) at 10 loci showed evidence of an association at P<10-5 with homogeneity of results from the 3 sample sets; some of these were previously documented to influence MM risk, including the SNP at the IRF4 binding site. In AL amyloidosis, rs9344 at the splice site of cyclin D1, promoting translocation (11;14), reached the highest significance, P=7.80 × 10-11; the SNP was only marginally significant in MM. SNP rs79419269 close to gene SMARCD3 involved in chromatin remodeling was also significant (P=5.2 × 10-8). These data provide evidence for common genetic susceptibility to AL amyloidosis and MM. Cyclin D1 is a more prominent driver in AL amyloidosis than in MM, but the links to aggregation of light chains need to be demonstrated.


Subject(s)
Amyloidosis/genetics , Genome-Wide Association Study , Immunoglobulin Light Chains/metabolism , Multiple Myeloma/genetics , Adult , Aged , Aged, 80 and over , Cyclin D1/physiology , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
5.
Leukemia ; 29(10): 2015-23, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25971364

ABSTRACT

Mutations of the tumor suppressor p53 lead to chemotherapy resistance and a dismal prognosis in chronic lymphocytic leukemia (CLL). Whereas p53 targets are used to identify patient subgroups with impaired p53 function, a comprehensive assessment of non-coding RNA targets of p53 in CLL is missing. We exploited the impaired transcriptional activity of mutant p53 to map out p53 targets in CLL by small RNA sequencing. We describe the landscape of p53-dependent microRNA/non-coding RNA induced in response to DNA damage in CLL. Besides the key p53 target miR-34a, we identify a set of p53-dependent microRNAs (miRNAs; miR-182-5p, miR-7-5p and miR-320c/d). In addition to miRNAs, the long non-coding RNAs (lncRNAs) nuclear enriched abundant transcript 1 (NEAT1) and long intergenic non-coding RNA p21 (lincRNA-p21) are induced in response to DNA damage in the presence of functional p53 but not in CLL with p53 mutation. Induction of NEAT1 and lincRNA-p21 are closely correlated to the induction of cell death after DNA damage. We used isogenic lymphoma cell line models to prove p53 dependence of NEAT1 and lincRNA-p21. The current work describes the p53-dependent miRNome and identifies lncRNAs NEAT1 and lincRNA-p21 as novel elements of the p53-dependent DNA damage response machinery in CLL and lymphoma.


Subject(s)
Apoptosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics , Aged , Aged, 80 and over , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , DNA Damage , Female , Flow Cytometry , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
6.
Leukemia ; 29(8): 1721-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25787915

ABSTRACT

We aimed at demonstrating non-inferiority of bortezomib/cyclophosphamide/dexamethasone (VCD) compared to bortezomib/doxorubicin/dexamethasone (PAd) induction therapy with respect to very good partial response rates or better (⩾VGPR) in 504 newly diagnosed, transplant-eligible multiple myeloma patients. VCD was found to be non-inferior to PAd with respect to ⩾VGPR rates (37.0 versus 34.3%, P=0.001). The rates of progressive disease (PD) were 0.4% (VCD) versus 4.8% (PAd; P=0.003). In the PAd arm, 11 of 12 patients with PD had either renal impairment (creatinine ⩾2 mg/dl) at diagnosis or the cytogenetic abnormality gain 1q21, whereas no PD was observed in these subgroups in the VCD arm. Leukocytopenia/neutropenia (⩾3°) occurred more frequently in the VCD arm (35.2% versus 11.3%, P<0.001). Neuropathy rates (⩾2°) were higher in the PAd group (14.9 versus 8.4%, P=0.03). Serious adverse events, both overall and those related to thromboembolic events, were higher in the PAd group (32.7 versus 24.0%, P=0.04 and 2.8 versus 0.4%, P=0.04). Stem cell collection was not impeded by VCD. VCD is as effective as PAd in terms of achieving ⩾VGPR rates with fewer PD and has a favorable toxicity profile. Therefore, VCD is preferable to PAd as induction therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Adult , Aged , Boronic Acids/administration & dosage , Bortezomib , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Female , Follow-Up Studies , Hematopoietic Stem Cell Mobilization , Humans , Induction Chemotherapy , Male , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Neoplasm Staging , Prognosis , Prospective Studies , Pyrazines/administration & dosage , Remission Induction , Survival Rate
8.
Leukemia ; 28(9): 1902-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24535407

ABSTRACT

Previous studies demonstrated the relevance of focal lesions (FL) in whole-body magnetic resonance imaging (wb-MRI) at the initial workup of patients with smoldering multiple myeloma (SMM). The aim of this study was to assess the effects of longitudinal wb-MRIs on progression into multiple myeloma (MM). Sixty-three patients with SMM were analyzed who received at least two wb-MRIs for follow-up before progression into MM. Radiological progressive disease (MRI-PD) was defined as detection of new FL or increase in diameter of existing FL and a novel or progressive diffuse infiltration. Radiological stable disease (MRI-SD) was defined by no change compared with the prior MRI. Patients were followed-up every 3-6 months, including a serological and clinical evaluation. One Hundred and eighty-two wb-MRIs were analyzed. MRI-PD occurred in 31 patients (49%), and 25 (40%) patients developed MM. MRI-PD was highly significantly associated with progression into MM, regardless of findings at the initial MRI. In multivariate analysis, MRI-PD remained a risk factor, independent of relevant baseline parameters like serum monoclonal protein or ⩾95% aberrant plasma cells in the bone marrow. Patients with MRI-SD had no higher risk of progression, even when FL were present at the initial MRI. Therefore, MRI is suitable for the follow-up of patients with SMM.


Subject(s)
Magnetic Resonance Imaging/methods , Multiple Myeloma/pathology , Adult , Aged , Humans , Middle Aged , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies
9.
Clin Genet ; 85(4): 347-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23614707

ABSTRACT

Craniofrontonasal syndrome (CFNS) is an X-linked disorder caused by inactivating mutations in the gene for ephrin-B1 (EFNB1). Paradoxically it shows a more severe phenotype in females than in males. As a result of X inactivation cell populations with and without EFNB1 expression are found in EFNB1+/- females. This is thought to initiate a process termed cellular interference which may be responsible for the phenotype in females. We present a boy with severe clinical features of CFNS. In ∼42% of his blood cells we found a supernumerary ring X chromosome containing EFNB1 but lacking XIST. Mosaicism for cell populations with different levels of EFNB1 expression can explain the severe phenotype of this patient. In vitro experiments in Xenopus tissue showed that cells overexpress ephrinB1 cluster and sort out from wild-type cells. Our report provides further evidence that cellular interference contributes to the paradoxical inheritance pattern of CFNS.


Subject(s)
Craniofacial Abnormalities/genetics , Ephrin-B1/genetics , Animals , Chromosomes, Human, X , Ephrin-B1/metabolism , Humans , Infant , Male , Mosaicism , RNA, Long Noncoding/genetics , Syndrome , Xenopus/genetics
10.
Oncogene ; 33(27): 3506-18, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-23955078

ABSTRACT

The incidence of skin cancer is increasing worldwide and cutaneous squamous cell carcinomas (SCCs) are associated with considerable morbidity and mortality, particularly in immunosuppressed individuals ('carcinomatous catastrophy'). Yet, molecular mechanisms are still insufficiently understood. Besides ultraviolet (UV)-indicative mutations, chromosomal aberrations are prominent. As telomeres are essential in preserving chromosome integrity, and telomere erosion as well as aberrant spatial telomere distribution contribute to genomic instability, we first established telomere length profiles across the whole tissue and identified normal skin (10/30) harboring discrete epidermal sites (stem cell territories) of evenly short telomeres. Precancerous actinic keratoses (AKs) (17) and SCCs (27) expressed two telomere phenotypes: (i) tissue-wide evenly short to intermediate and (ii) longer and tissue-wide heterogeneous telomere lengths, suggesting two modes of initiation, with one likely to originate in the epidermal stem cells. Although tumor histotype, location, patient gender or age failed to distinguish the two SCC telomere phenotypes, as did telomerase activity, we found a trend for a higher degree of aberrant p53 and cyclin D1 expression with long/heterogeneous telomeres. In addition, we established an association for the short/homogeneous telomeres with a simpler and the heterogeneous telomeres with a more complex karyotype correlating also with distinct chromosomal changes. SCCs (13) from renal transplant recipients displayed the same telomere dichotomy, suggesting that both telomere subtypes contribute to 'carcinomatous catastrophy' under immunosuppression by selecting for a common set (3, 9p and 17q) and subtype-specific aberrations (e.g., 6p gain, 13q loss). As a second mechanism of telomere-dependent genomic instability, we investigated changes in telomere distribution with its most severe form of telomeric aggregates (TAs). We identified a telomere length-independent but progression-dependent increase in cells with small telomere associations in AKs (17/17) and additional TAs in SCCs (24/32), basal cell carcinomas (30/31) and malignant melanomas (15/15), and provide evidence for a reactive oxygen species-dependent mechanism in this UV-induced telomere organization-dependent genomic instability.


Subject(s)
Carcinoma, Squamous Cell/classification , Carcinoma, Squamous Cell/genetics , Skin Neoplasms/classification , Skin Neoplasms/genetics , Telomere/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Child , Cyclin D1/metabolism , Disease Progression , Genomic Instability/radiation effects , Humans , Male , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Middle Aged , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Telomerase/metabolism , Telomere/radiation effects , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Young Adult
11.
Oncogene ; 32(20): 2586-91, 2013 May 16.
Article in English | MEDLINE | ID: mdl-22751129

ABSTRACT

AVEN has been identified as an inhibitor of apoptosis, which binds to the adaptor protein, APAF-1, and thereby prevents apoptosome formation and mitochondrial apoptosis. Recent data have demonstrated high expression levels of AVEN messenger RNA in acute leukemias as well as a positive correlation between AVEN mRNA overexpression and poor prognosis in childhood acute lymphoblastic leukemia. On the basis of these data, we investigated the potential involvement of AVEN in tumorigenesis. First, we confirmed the overexpression of AVEN in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) patient samples. We then established a transgenic mouse model with T-cell-specific overexpression of AVEN, with which we demonstrated the oncogenic cooperation of AVEN with heterozygous loss of p53. Finally, we used a subcutaneous xenograft mouse model to show that AVEN knockdown in the T-ALL cell lines, MOLT-4 and CCRF-CEM, and in the acute myeloblastic leukemia cell line, Kasumi-1, leads to a halt in tumor growth owing to the increased apoptosis and decreased proliferation of tumor cells. Collectively, our data demonstrate that the anti-apoptotic molecule, AVEN, functions as an oncoprotein in hematopoietic neoplasms.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Membrane Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Genes, p53 , Humans , Lymphoma, T-Cell/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thymocytes/physiology , Xenograft Model Antitumor Assays
12.
Ther Umsch ; 69(12): 697-701, 2012 Dec.
Article in German | MEDLINE | ID: mdl-23188782

ABSTRACT

Fever of unknown origin (FUO) is a common medical diagnosis by exclusion. In these cases, fever is the predominant symptom of an underlying disease. We describe the case of a 60-year old patient with FUO. Intensive search for the causative disease was carried out. Unfortunately all the investigations remained fruitless. Eventually, the patient was discharged with the diagnosis of common variable immunodeficiency, based on hypogammaglobulinemia and Cytomegalovirus replication. Two weeks after discharge, the patient presented in the outpatient clinic with the typical symptoms of giant cell arteriitis (GCA). The diagnosis was confirmed by a repeated ultrasound imaging and biopsy findings. The clinical condition of the patient improved rapidly after beginning of treatment with steroids. This case illustrates the importance of a longitudinal observation of patients presenting with FUO if the diagnosis remains unclear after intensive investigations.


Subject(s)
Fever of Unknown Origin/etiology , Giant Cell Arteritis/complications , Giant Cell Arteritis/diagnosis , Biopsy , Diagnosis, Differential , Giant Cell Arteritis/pathology , Humans , Male , Temporal Arteries/pathology
13.
Clin Genet ; 82(5): 494-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-21954990

ABSTRACT

Silver-Russell syndrome (SRS) is a genetically heterogeneous disorder characterized by intrauterine and postnatal growth retardation, typical facial features and a spectrum of additional features including body and limb asymmetry and clinodactyly. Maternal uniparental disomy for chromosome 7 (upd(7)mat) was shown to occur in 5-10% of patients with SRS. Maternal UPD7 is clinically often associated with mild SRS. Parents of an affected child are given a negligible recurrence risk as all reported cases with upd(7)mat have been sporadic so far. In general, chromosomal rearrangements-like translocations increase the likelihood of uniparental disomy (UPD) for the chromosomes involved. However, SRS as the result of a upd(7)mat in association with an inherited chromosomal translocation involving chromosome 7 has only been reported once before. Here, we describe the second case of SRS with upd(7)mat due to a familial reciprocal translocation t(7;13). This emphasizes the importance of chromosome analysis in SRS patients with upd(7)mat to rule out chromosomal rearrangements despite their rare occurrence as they are of great relevance for genetic counseling of SRS families.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Silver-Russell Syndrome/genetics , Translocation, Genetic , Uniparental Disomy/genetics , Abnormalities, Multiple/genetics , Child, Preschool , Genetic Counseling , Genome, Human/genetics , Humans , Male , Microsatellite Repeats , Pedigree , Sequence Analysis, DNA
14.
Clin Genet ; 79(5): 489-94, 2011 May.
Article in English | MEDLINE | ID: mdl-20681991

ABSTRACT

The short stature homeobox (SHOX) gene is located in the pseudoautosomal region 1 of both sex chromosomes. Haploinsufficiency of SHOX leads to different phenotypes ranging from isolated short stature to Léri-Weill syndrome characterized by short stature, mesomelia and Madelung deformity. We describe a family with a SHOX deletion originally located on the Y chromosome and transmitted from father to daughter by crossover during meiosis. The male index patient presented with short stature, mesomelia and mild Madelung deformity. His father had a normal height but slightly disproportionate short legs. The sister of the index patient presented with marked Madelung deformity and normal height. A deletion of the SHOX gene was identified in the male index patient, his father and his sister. Metaphase fluorescence in situ hybridization (FISH) analyses showed a deletion of the SHOX gene on the Y chromosomes of the index patient and his father, and on the X chromosome of his sister, indicating that a meiotic crossover of the SHOX gene region between the X and Y chromosomes had occurred. The pseudoautosomal region 1 is a known recombination 'hot spot' in male meiosis. Published genetic maps indicate high recombination frequency of ∼40% for SHOX in male meiosis leading to pseudoautosomal inheritance.


Subject(s)
Chromosome Disorders/genetics , Child, Preschool , Female , Growth Disorders/genetics , Homeodomain Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Male , Osteochondrodysplasias/genetics , Pedigree , Short Stature Homeobox Protein
15.
Oncogene ; 28(44): 3866-79, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19718049

ABSTRACT

Pathogenesis of multiple myeloma is associated with an aberrant expression of pro-proliferative, pro-angiogenic and bone-metabolism-modifying factors by malignant plasma cells. Given the frequently long time span from diagnosis of early-stage plasma cell dyscrasias to overt myeloma and the mostly low proliferation rate of malignant plasma cells, we hypothesize these to similarly express a novel class of inhibitory factors of potential prognostic relevance. Bone morphogenic proteins (BMPs) represent possible candidates as they inhibit proliferation, stimulate bone formation and have an effect on the survival of cancer patients. We assessed the expression of BMPs and their receptors by Affymetrix DNA microarrays (n=779) including CD138-purified primary myeloma cell samples (n=635) of previously untreated patients. BMP6 is the only BMP expressed by malignant and normal plasma cells. Its expression is significantly lower in proliferating myeloma cells, myeloma cell lines or plasmablasts. BMP6 significantly inhibits the proliferation of myeloma cell lines, survival of primary myeloma cells and in vitro angiogenesis. A high BMP6 expression in primary myeloma cell samples delineates significantly superior overall survival for patients undergoing high-dose chemotherapy independent of conventional prognostic factors (International Staging System (ISS) stage, beta(2) microglobulin).


Subject(s)
Biomarkers, Tumor/biosynthesis , Bone Morphogenetic Protein 6/biosynthesis , Cell Proliferation , Gene Expression Regulation, Neoplastic , Multiple Myeloma/metabolism , Multiple Myeloma/mortality , Neoplasm Proteins/biosynthesis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/mortality , Disease-Free Survival , Female , Gene Expression Profiling , Humans , Male , Multiple Myeloma/pathology , Neovascularization, Pathologic/pathology , Oligonucleotide Array Sequence Analysis , Plasma Cells , Survival Rate
16.
J Clin Pathol ; 62(10): 892-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19541684

ABSTRACT

BACKGROUND: Papillary renal cell tumours (RCTs) have been described as a genetic entity. Recently, papillary RCTs have been divided into small (type 1) and large (type 2) cell tumours. Subsequent DNA analyses have resulted in controversial data regarding putative genetic changes marking type 1 and type 2 tumours. AIM: The aim of this study was to improve the original description that papillary RCT is a genetic entity regardless of the phenotypic variation. METHODS: DNA from 163 papillary RCTs, including 82 multiplex tumours from eight hereditary cases, was analysed for copy number changes by chromosomal comparative genomic hybridisation (CGH) and/or for allelic changes at chromosomes 7 and 17 by microsatellite analysis. The results of the genetic analysis were compared with the cytological characteristics of the tumours. RESULTS: The results showed alterations of chromosomes 7 and 17 at similar frequencies in papillary RCTs with characteristics ranging from small to large cell, nuclear grade 1 to 3, and 3 mm to 16 cm diameter. CONCLUSION: Trisomies of chromosomes 7 and 17 are specific genetic alterations in papillary RCTs irrespective of their size, grade and cellular differentiation.


Subject(s)
Carcinoma, Papillary/genetics , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 7/genetics , Kidney Neoplasms/genetics , Trisomy , Carcinoma, Papillary/pathology , Comparative Genomic Hybridization/methods , DNA, Neoplasm/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Kidney Neoplasms/pathology , Microsatellite Repeats/genetics , Phenotype
17.
Bone Marrow Transplant ; 40(11): 1033-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17891186

ABSTRACT

Lack of CD56 expression was reported to be associated with a poor prognosis in multiple myeloma (MM) patients treated with conventional chemotherapy. Aim of our retrospective study was to analyse whether CD56 expression on MM cells reveals as a prognostic factor in patients treated with high-dose chemotherapy. MM cells of 99 patients prior to treatment with high-dose chemotherapy were analysed for CD56 expression by flow cytometry. Multivariable analysis of event-free survival in these patients showed no statistically significant difference between the CD56(-) (n=28) and the CD56(+) (n=71) group. The lack of CD56 expression on MM cells of these patients correlated significantly with the presence of translocation (11;14) (t(11;14)) (estimated correlation coefficient=0.655 95%, confidence interval (0.481; 0.779)). In summary, our results indicate that lack of CD56 expression on MM cells is not a prognostic marker in patients treated with high-dose chemotherapy, but is associated with t(11;14).


Subject(s)
CD56 Antigen/metabolism , Melphalan/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Myeloablative Agonists/administration & dosage , Adult , Aged , Biomarkers , CD56 Antigen/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 17/genetics , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Hematopoietic Stem Cell Transplantation , Humans , Male , Middle Aged , Multiple Myeloma/pathology , Retrospective Studies , Translocation, Genetic/genetics , Transplantation, Autologous
18.
Oncogene ; 26(7): 1088-97, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-16909113

ABSTRACT

The molecular pathogenesis of pleomorphic xanthoastrocytoma (PXA), a rare astrocytic brain tumor with a relatively favorable prognosis, is still poorly understood. We characterized 50 PXAs by comparative genomic hybridization (CGH) and found the most common imbalance to be loss on chromosome 9 in 50% of tumors. Other recurrent losses affected chromosomes 17 (10%), 8, 18, 22 (4% each). Recurrent gains were identified on chromosomes X (16%), 7, 9q, 20 (8% each), 4, 5, 19 (4% each). Two tumors demonstrated amplifications mapping to 2p23-p25, 4p15, 12q13, 12q21, 21q21 and 21q22. Analysis of 10 PXAs with available high molecular weight DNA by high-resolution array-based CGH indicated homozygous 9p21.3 deletions involving the CDKN2A/p14(ARF)/CDKN2B loci in six tumors (60%). Interphase fluorescence in situ hybridization to tissue sections confirmed the presence of tumor cells with homozygous 9p21.3 deletions. Mutational analysis of candidate genes on 9q, PTCH and TSC1, revealed no mutations in PXAs with 9q loss and no evidence of TSC1 promoter methylation. However, PXAs consistently showed low TSC1 transcript levels. Taken together, our study identifies loss of chromosome 9 as the most common chromosomal imbalance in PXAs and suggests important roles for homozygous CDKN2A/p14(ARF)/CDKN2B deletion as well as low TSC1 mRNA expression in these tumors.


Subject(s)
Astrocytoma/genetics , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Deletion , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Proteins/deficiency , Adolescent , Adult , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p15/deficiency , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Female , Homozygote , Humans , Male , Middle Aged , RNA, Messenger/biosynthesis , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
19.
Leukemia ; 19(7): 1198-206, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15902298

ABSTRACT

Overexpression of BCR-ABL and P-glycoprotein (Pgp) are two of the known mechanisms of imatinib resistance. As combination therapy may allow to overcome drug resistance, we investigated the effect of combination treatment with imatinib and 17-allylamino-17-demethoxygeldanamycin (17-AAG), a heat-shock protein 90 (Hsp90) inhibitor, on different imatinib-sensitive and imatinib-resistant CML cell lines. In imatinib-sensitive cells, combination index (CI) values obtained using the method of Chou and Talalay indicated additive (CI=1) or marginally antagonistic (CI>1) effects following simultaneous treatment with imatinib and 17-AAG. In imatinib-resistant cells both drugs acted synergistically (CI<1). In primary chronic-phase CML cells additive or synergistic effects of the combination of imatinib plus 17-AAG were discernible. Annexin V/propidium iodide staining showed that the activity of imatinib plus 17-AAG is mediated by apoptosis. Combination treatment with imatinib plus 17-AAG was more effective in reducing the BCR-ABL protein level than 17-AAG alone. Monotherapy with 17-AAG decreased P-glycoprotein activity, which may increase intracellular imatinib levels and contribute to the sensitization of CML cells to imatinib. The results suggest that combination of imatinib and 17-AAG may be useful to overcome imatinib resistance in a clinical setting.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Fusion Proteins, bcr-abl/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Protein-Tyrosine Kinases/biosynthesis , Pyrimidines/pharmacology , Rifabutin/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Apoptosis/drug effects , Benzamides , Benzoquinones , Cell Proliferation/drug effects , Drug Synergism , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic/drug effects , Humans , Imatinib Mesylate , In Situ Hybridization, Fluorescence , Lactams, Macrocyclic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Phosphorylation , Protein-Tyrosine Kinases/analysis , RNA, Messenger/genetics , Rifabutin/pharmacology , Tumor Stem Cell Assay
20.
Neurogenetics ; 6(1): 29-35, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15655674

ABSTRACT

Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are common X-chromosomal recessive disorders caused by mutations in the dystrophin gene. Using the novel multiplex ligation-dependent probe amplification (MLPA) method we performed retrospective and prospective analyses in a total of 193 individuals. Deletions or duplications were identified in 14 out of 90 families previously tested negative by multiplex PCR or FISH analysis. Partially incorrect results were subsequently identified in two families: the loss of exon 38 signal in one case was due to a p.Q1802X nonsense mutation, whilst in another patient an apparent deletion of exon 37 (coinciding with a duplication of exons 46-53) was caused by a p.R1735C polymorphism. In one case we found a complex rearrangement involving a duplication of two regions: dupEX45-48 and dupEX54-55. We conclude that MLPA is a highly sensitive and rapid alternative to multiplex PCR. It can be used on blood samples, chorionic villi and paraffin-embedded tissue. The ease of detection of duplications and the application for female carrier analysis are clearly the main advantages of the method. However, apparent single exon deletions detected by MLPA should be checked by an independent method. Complex rearrangements such as double mutations on the same allele are rare.


Subject(s)
Dystrophin/genetics , Gene Duplication , Muscular Dystrophy, Duchenne/genetics , Mutation , Sequence Deletion , DNA Primers , Exons , Female , Gene Amplification , Gene Rearrangement , Genetic Carrier Screening , Humans , Male , Polymerase Chain Reaction , Trinucleotide Repeats
SELECTION OF CITATIONS
SEARCH DETAIL