Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Pathol Oncol Res ; 30: 1611590, 2024.
Article in English | MEDLINE | ID: mdl-38605929

ABSTRACT

Lung cancer is a paradigm for a genetically driven tumor. A variety of drugs were developed targeting specific biomarkers requiring testing for tumor genetic alterations in relevant biomarkers. Different next-generation sequencing technologies are available for library generation: 1) anchored multiplex-, 2) amplicon based- and 3) hybrid capture-based-PCR. Anchored multiplex PCR-based sequencing was investigated for routine molecular testing within the national Network Genomic Medicine Lung Cancer (nNGM). Four centers applied the anchored multiplex ArcherDX-Variantplex nNGMv2 panel to re-analyze samples pre-tested during routine diagnostics. Data analyses were performed by each center and compiled centrally according to study design. Pre-defined standards were utilized, and panel sensitivity was determined by dilution experiments. nNGMv2 panel sequencing was successful in 98.9% of the samples (N = 90). With default filter settings, all but two potential MET exon 14 skipping variants were identified at similar allele frequencies. Both MET variants were found with an adapted calling filter. Three additional variants (KEAP1, STK11, TP53) were called that were not identified in pre-testing analyses. Only total DNA amount but not a qPCR-based DNA quality score correlated with average coverage. Analysis was successful with a DNA input as low as 6.25 ng. Anchored multiplex PCR-based sequencing (nNGMv2) and a sophisticated user-friendly Archer-Analysis pipeline is a robust and specific technology to detect tumor genetic mutations for precision medicine of lung cancer patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/pathology , Kelch-Like ECH-Associated Protein 1/genetics , Multiplex Polymerase Chain Reaction , NF-E2-Related Factor 2/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Mutation/genetics , High-Throughput Nucleotide Sequencing , Biomarkers , DNA
3.
J Mol Diagn ; 22(11): 1333-1343, 2020 11.
Article in English | MEDLINE | ID: mdl-32822851

ABSTRACT

Persistent human papillomavirus (HPV) infection is associated with the development of oropharyngeal squamous cell carcinoma (OPSCC), and increasing incidences of OPSCC are reported. The generally favorable treatment outcome in patients with HPV-driven OPSCC has brought de-escalation of treatment into discussion. Nevertheless, 13% to 25% develop a relapse within two years after current standard treatment. New biomarkers are urgently required to monitor therapy response, tumor burden, and minimal residual disease during follow-up. This observational study examined 50 patients with OPSCC to investigate plasma cell-free (cf) HPV-DNA derived from tumor cells before therapy and during follow-up. Real-time quantitative PCR was applied to quantify the DNA concentration of HPV oncogenes E6 and E7. A total of 85.7% of pretreatment samples from patients with HPV-driven OPSCC (n = 28) were positive for at least one marker, and cfHPV-DNA concentration increased with tumor size. Virtually no signals were detected in HPV-negative OPSCC patients (n = 20; P ≤ 0.001). Patients without clinical evidence of recurrence had significantly reduced cfHPV-DNA concentrations after therapy (P ≤ 0.001). Conversely, cfHPV-DNA levels increased or remained above threshold in five patients who had residual disease or developed recurrence. In conclusion, plasma cfHPV-DNA detection correlates with the clinical course of disease in patients with HPV-driven OPSCC. Consequently, extensive clinical investigation should be considered if cfHPV-DNA is detected during follow-up of patients with HPV-driven OPSCC.


Subject(s)
Alphapapillomavirus/metabolism , Cell-Free Nucleic Acids/blood , DNA, Viral/blood , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/complications , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/complications , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/blood , Papillomavirus Infections/complications , Repressor Proteins/genetics , Squamous Cell Carcinoma of Head and Neck/blood , Squamous Cell Carcinoma of Head and Neck/complications , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Cell-Free Nucleic Acids/genetics , DNA, Viral/genetics , Female , Follow-Up Studies , Humans , Male , Middle Aged , Papillomavirus Infections/virology , Pilot Projects , Real-Time Polymerase Chain Reaction
4.
Mol Oncol ; 14(9): 2142-2162, 2020 09.
Article in English | MEDLINE | ID: mdl-32533757

ABSTRACT

A hallmark of ovarian high-grade serous carcinoma (HGSC) is its early and massive peritoneal dissemination via the peritoneal fluid. It is generally believed that tumor cells must breach the mesothelium of peritoneal organs to adhere to the underlying extracellular matrix (ECM) and initiate metastatic growth. However, the molecular mechanisms underlying these processes are only partially understood. Here, we have analyzed 52 matched samples of spheroids and solid tumor masses (suspected primary lesions and metastases) from 10 patients by targeted sequencing of 21 loci previously proposed as targets of HGSC driver mutations. This analysis revealed very similar patterns of genetic alterations in all samples. One exception was FAT3 with a strong enrichment of mutations in metastases compared with presumed primary lesions in two cases. FAT3 is a putative tumor suppressor gene that codes for an atypical cadherin, pointing a potential role in peritoneal dissemination in a subgroup of HGSC patients. By contrast, transcriptome data revealed clear and consistent differences between tumor cell spheroids from ascites and metastatic lesions, which were mirrored by the in vitro adherence of ascites-derived spheroids. The adhesion-induced transcriptional alterations in metastases and adherent cells resembled epithelial-mesenchymal transition, but surprisingly also included the upregulation of a specific subset of mesothelial genes, such as calretinin (CALB2) and podoplanin (PDPN). Consistent with this finding, calretinin staining was also observed in subsets of tumor cells in HGSC metastases, particularly at the invasive tumor edges. Intriguingly, a high expression of either CALB2 or PDPN was strongly associated with a poor clinical outcome. siRNA-mediated CALB2 silencing triggered the detachment of adherent HGSC cells in vitro and inhibited the adhesion of detached HGSC cells to collagen type I. Our data suggest that the acquisition of a mesenchymal-mesothelial phenotype contributes to cancer cell adhesion to the ECM of peritoneal organs and HGSC progression.


Subject(s)
Epithelium/pathology , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Up-Regulation/genetics , Apoptosis/genetics , Ascites/genetics , Ascites/pathology , Biomarkers, Tumor/metabolism , Cell Adhesion , Cell Line, Tumor , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Disease-Free Survival , Female , Humans , Neoplasm Grading , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Peritoneal Neoplasms/secondary , Polymorphism, Single Nucleotide/genetics , Spheroids, Cellular/pathology , Treatment Outcome
5.
Leukemia ; 34(1): 151-166, 2020 01.
Article in English | MEDLINE | ID: mdl-31431735

ABSTRACT

We used hybrid capture-targeted next-generation sequencing of circulating cell-free DNA (ccfDNA) of pediatric Hodgkin lymphoma (PHL) patients to determine pathogenic mechanisms and assess the clinical utility of this method. Hodgkin-Reed/Sternberg (HRS) cell-derived single nucleotide variants, insertions/deletions, translocations and VH-DH-JH rearrangements were detected in pretherapy ccfDNA of 72 of 96 patients. Number of variants per patient ranged from 1 to 21 with allele frequencies from 0.6 to 42%. Nine translocation breakpoints were detected. Genes involved in JAK/STAT, NFkB and PI3K signaling and antigen presentation were most frequently affected. SOCS1 variants, mainly deletions, were found in most circulating tumor (ct) DNAs, and seven of the nine translocation breakpoints involved SOCS1. Analysis of VH-DH-JH rearrangements revealed an origin of PHL HRS cells from partially selected germinal center B cells. Amounts of pretherapy ctDNA were correlated with metabolic tumor volumes. Furthermore, in all ccfDNA samples of 43 patients with early response assessment quantitative qPET < 3, indicative of a favorable clinical course, ctDNA was not detectable. In contrast, in five of six patients with qPET > 3, indicative of an unfavorable clinical course, ctDNA remained detectable. ccfDNA analysis of PHL is thus a suitable approach to determine pathogenic mechanisms and monitor therapy response.


Subject(s)
Circulating Tumor DNA/genetics , Hodgkin Disease/genetics , Adolescent , Child , Child, Preschool , Female , Genotype , Humans , Male
6.
Head Neck ; 40(6): 1109-1119, 2018 06.
Article in English | MEDLINE | ID: mdl-29522268

ABSTRACT

BACKGROUND: In head and neck squamous cell carcinoma (HNSCC), the occurrence of concurrent lung malignancies poses a significant diagnostic challenge because metastatic HNSCC is difficult to discern from second primary lung squamous cell carcinoma (SCC). However, this differentiation is crucial because the recommended treatments for metastatic HNSCC and second primary lung SCC differ profoundly. METHODS: We analyzed the origin of lung tumors in 32 patients with HNSCC using human papillomavirus (HPV) typing and targeted next generation sequencing of all coding exons of tumor protein 53 (TP53). RESULTS: Lung tumors were clearly identified as HNSCC metastases or second primary tumors in 29 patients, thus revealing that 16 patients had received incorrect diagnoses based on clinical and morphological data alone. CONCLUSION: The HPV typing and mutation analysis of all TP53 coding exons is a valuable diagnostic tool in patients with HNSCC and concurrent lung SCC, which can help to ensure that patients receive the most suitable treatment.


Subject(s)
Head and Neck Neoplasms/etiology , Lung Neoplasms/etiology , Neoplasms, Second Primary/diagnosis , Papillomaviridae/isolation & purification , Squamous Cell Carcinoma of Head and Neck/etiology , Tumor Suppressor Protein p53/genetics , Adult , Aged , Female , Head and Neck Neoplasms/pathology , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Mutation/genetics , Neoplasms, Second Primary/etiology , Squamous Cell Carcinoma of Head and Neck/pathology
7.
J Mol Diagn ; 20(3): 344-354, 2018 05.
Article in English | MEDLINE | ID: mdl-29471115

ABSTRACT

Myelodysplastic syndromes are hematological neoplasias in which immunohistologic examination of bone marrow trephines is important for a definite diagnosis. Unequivocal distinction from reactive bone marrow changes is, however, sometimes difficult. Because neoplastic clones in myelodysplastic syndrome carry mutations in recurrent genes, mutation detection by targeted next-generation sequencing may be a useful support for differential diagnosis. To elucidate the accuracy of this approach in the clinical diagnostic setting, we analyzed single and consecutive bone marrow trephines processed for immunohistologic examination from 145 patients by targeted next-generation sequencing of 12 genes recurrently mutated in myelodysplastic syndromes. Of 110 patients with immunohistologic unequivocal diagnosis, 41 of 47 with myelodysplastic syndrome carried mutations. In 14 consecutive samples available from these patients, remissions were accompanied by loss of mutations and ongoing disease with persisting mutations. Of 35 samples with indefinite immunohistologic appearance, 22 developed clinical unequivocal myelodysplastic syndrome in the further course, and 19 carried mutations already in the initial biopsy, which persisted in consecutive samples available from 13 patients. No mutation was detected in any initial and consecutive sample of 13 patients with indefinite immunohistologic appearance without clinical unequivocal myelodysplastic syndrome in the further course. We conclude that targeted next-generation sequencing is an accurate tool for differential diagnosis of myelodysplastic syndrome in the clinical diagnostic setting.


Subject(s)
Bone Marrow/pathology , High-Throughput Nucleotide Sequencing/methods , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Clone Cells , Diagnosis, Differential , Humans , Mutation/genetics , Mutation Rate , Myelodysplastic Syndromes/pathology
8.
Transfusion ; 55(6 Pt 2): 1538-44, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25873286

ABSTRACT

BACKGROUND: Fetal human platelet antigen (HPA) genotyping is required to determine whether the fetus is at risk and whether prenatal interventions to prevent fetal bleeding are required in pregnant women with a history of fetal and neonatal alloimmune thrombocytopenia (FNAIT). Methods for noninvasive genotyping of HPA alleles with the use of maternal plasma cell-free DNA were published recently but do lack internal controls to exclude false-negative results. STUDY DESIGN AND METHODS: Cell-free DNA was isolated from plasma of four pregnant women with a history of FNAIT caused by anti-HPA-1a and controls. A primer panel was designed to target sequences flanking single-nucleotide polymorphisms (SNPs)/exonic regions of ITGB3 (HPA-1), ITGA2B (HPA-3), ITGA2 (HPA-5), CD109 (HPA-15), RHD, RHCE, KEL, DARC, SLC14A1, GYPA, GYPB, and SRY. These regions and eight anonymous SNPs were massively parallel sequenced by semiconductor technology. RESULTS: The mean (±SD) number of reads for targeted SNPs was 5255 (±2838). Fetal DNA was detected at a median of 4.5 (range, 2-8) polymorphic loci. The mean fractional fetal DNA concentration in cell-free maternal plasma was 8.36% (range, 4.79%-15.9%). For HPA-1, nonmaternal ITGB3 sequences (c.176T, HPA-1a) were detected in all HPA-1ab fetuses. One HPA-1bb fetus was unequivocally identified, showing the pregnancy was not at risk of FNAIT. CONCLUSION: We have successfully established massively parallel sequencing as a novel reliable method for noninvasive genotyping of fetal HPA-1a alleles. This technique may also allow the safe detection of other fetal blood group polymorphisms frequently involved in FNAIT and hemolytic disease of the newborn.


Subject(s)
Antigens, Human Platelet/genetics , Fetal Diseases/diagnosis , Genotyping Techniques/methods , High-Throughput Nucleotide Sequencing , Prenatal Diagnosis/methods , Sequence Analysis, DNA/methods , Thrombocytopenia, Neonatal Alloimmune/diagnosis , Blood Chemical Analysis/methods , Case-Control Studies , Cell-Free System , Female , Fetal Diseases/genetics , Genotype , Humans , Infant, Newborn , Integrin beta3 , Polymorphism, Single Nucleotide , Pregnancy/blood , Reproducibility of Results , Thrombocytopenia, Neonatal Alloimmune/genetics
9.
Cancer Res ; 74(21): 6173-83, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25252913

ABSTRACT

Primary cardiac angiosarcomas are rare tumors with unfavorable prognosis. Pathogenic driver mutations are largely unknown. We therefore analyzed a collection of cases for genomic aberrations using SNP arrays and targeted next-generation sequencing (tNGS) of oncogenes and tumor-suppressor genes. Recurrent gains of chromosome 1q and a small region of chromosome 4 encompassing KDR and KIT were identified by SNP array analysis. Repeatedly mutated genes identified by tNGS were KDR with different nonsynonymous mutations, MLL2 with different nonsense mutations, and PLCG1 with a recurrent nonsynonymous mutation (R707Q) in the highly conserved autoinhibitory SH2 domain in three of 10 cases. PLCγ1 is usually activated by Y783 phosphorylation and activates protein kinase C and Ca(2+)-dependent second messengers, with effects on cellular proliferation, migration, and invasiveness. Ectopic expression of the PLCγ1-R707Q mutant in endothelial cells revealed reduced PLCγ1-Y783 phosphorylation with concomitant increased c-RAF/MEK/ERK1/2 phosphorylation, increased IP3 amounts, and increased Ca(2+)-dependent calcineurin activation compared with ectopic expressed PLCγ1-wild-type. Furthermore, cofilin, whose activation is associated with actin skeleton reorganization, showed decreased phosphorylation, and thus activation after expression of PLCγ1-R707Q compared with PLCγ1-wild-type. At the cellular level, expression of PLCγ1-R707Q in endothelial cells had no influence on proliferation rate, but increased apoptosis resistance and migration and invasiveness in in vitro assays. Together, these findings indicate that the PLCγ1-R707Q mutation causes constitutive activation of PLCγ1 and may represent an alternative way of activation of KDR/PLCγ1 signaling besides KDR activation in angiosarcomas, with implications for VEGF/KDR targeted therapies.


Subject(s)
Heart Neoplasms/genetics , Hemangiosarcoma/genetics , Neoplasm Invasiveness/genetics , Phospholipase C gamma/genetics , Apoptosis/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation, Neoplastic , Heart Neoplasms/pathology , Hemangiosarcoma/pathology , High-Throughput Nucleotide Sequencing , Humans , Mutation , Phospholipase C gamma/biosynthesis , Polymorphism, Single Nucleotide/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , src Homology Domains/genetics
10.
Pathol Res Pract ; 210(6): 369-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24674452

ABSTRACT

Myelodysplastic syndromes (MDS) are hematopoietic disorders characterized by ineffective hematopoiesis and progression to acute leukemia. In patients ineligible for hematopoietic stem cell transplantation, azacitidine is the only treatment shown to prolong survival. However, with the availability of a growing compendium of cancer biomarkers and related drugs, analysis of relevant genetic alterations for individual MDS patients might become part of routine evaluation. Therefore and in order to cover the entire bone marrow microenvironment involved in the pathogenesis of MDS, SNP array analysis and targeted next generation sequencing (tNGS) for the mostly therapy relevant 46 onco- and tumor-suppressor genes were performed on bone marrow biopsies from 29 MDS patients. In addition to the detection of mutations known to be associated with MDS in NRAS, KRAS, MPL, NPM1, IDH1, PTPN11, APC and MET, single nucleotide variants so far unrelated to MDS in STK11 (n=1), KDR (n=3), ATM (n=1) and JAK3 (n=2) were identified. Moreover, a recurrent microdeletion was detected in Xq26.3 (n=2), causing loss of PHF6 expression, a potential tumor suppressor gene, and the miR-424, which is involved in the development of acute myeloid leukemia. Finally, combined genetic aberrations affecting the VEGF/VEGFR pathway were found in the majority of cases demonstrating the diversity of mutations affecting different nodes of a particular signaling network as an intrinsic feature in MDS patients. We conclude that combined SNP array analyses and tNGS can identify established and novel therapy relevant genomic aberrations in MDS patients and track them in a clinical setting for individual therapy selection.


Subject(s)
Carrier Proteins/genetics , Chromosome Deletion , Chromosomes, Human, X , Genetic Testing/methods , High-Throughput Nucleotide Sequencing , Janus Kinase 3/genetics , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Bone Marrow/enzymology , Bone Marrow/pathology , Case-Control Studies , Female , Genetic Markers , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Male , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/pathology , Nucleophosmin , Phenotype , Repressor Proteins , Risk Factors , Signal Transduction/genetics
11.
PLoS One ; 9(3): e93067, 2014.
Article in English | MEDLINE | ID: mdl-24667836

ABSTRACT

Ewing sarcoma, the second most common bone tumor in children and young adults, is an aggressive malignancy with a strong potential to metastasize. Ewing sarcoma is characterised by translocations encoding fusion transcription factors with an EWSR1 transactivation domain fused to an ETS family DNA binding domain. microRNAs are post-transcriptional regulators of gene expression and aberrantly expressed microRNAs have been identified as tumor suppressors or oncogenes in most cancer types. To identify potential oncogenic and tumor suppressor microRNAs in Ewing sarcoma, we determined and compared the expression of 377 microRNAs in 40 Ewing sarcoma biopsies, 6 Ewing sarcoma cell lines and mesenchymal stem cells, the putative cellular origin of Ewing sarcoma, from 6 healthy donors. Of the 35 differentially expressed microRNAs identified (fold change >4 and q<0.05), 19 were higher and 16 lower expressed in Ewing sarcoma. In comparisons between Ewing sarcoma samples with EWS-FLI or EWS-ERG translocations, with differing dissemination characteristics and of primary samples and metastases no significantly differential expressed microRNAs were detected using various stringency criteria. For miR-31, the microRNA with lowest expression in comparison to mesenchymal stem cells, functional analyses were performed to determine its potential as a tumor suppressor in Ewing sarcoma. Two of four miR-31 transfected Ewing sarcoma cell lines showed a significantly reduced proliferation (19% and 33% reduction) due to increased apoptosis in one and increased length of G1-phase in the other cell line. All three tested miR-31 transfected Ewing sarcoma cell lines showed significantly reduced invasiveness (56% to 71% reduction). In summary, we identified 35 microRNAs differentially expressed in Ewing sarcoma and demonstrate that miR-31 affects proliferation and invasion of Ewing sarcoma cell lines in ex vivo assays.


Subject(s)
Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Adult , Cell Line, Tumor , Cell Proliferation , Humans , Neoplasm Invasiveness
15.
Am J Pathol ; 182(4): 1205-18, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462508

ABSTRACT

Despite recent advances in understanding the relevance of cell adhesion-related signaling in the pathogenesis of ischemic cardiomyopathy (ICM) in animal models, substantial questions remain unanswered in the human setting. We have previously shown that the neural cell adhesion molecule CD56 [neural cell adhesion molecule (NCAM1)] is specifically overexpressed in ICM; it was the aim of the current study to further elucidate the role of CD56 in the pathogenesis of human ICM. We used quantitative real-time PCR and IHC in human ICM and a rat model of coronary obstruction to demonstrate that CD56(140kD), the only extraneuronally expressed NCAM1 isoform with a cytoplasmic protein domain capable of inducing intracellular signaling, is the only up-regulated CD56 isoform in failing cardiomyocytes in human ICM in vivo. In subsequent analyses of the cellular effects of CD56(140kD) overexpression in the development of ICM using differential whole transcriptome expression analyses and functional in vitro cardiomyocyte cell culture assays, we further show that the up-regulation of CD56(140kD) is associated with profound gene expression changes, increased apoptosis, and reduced Ca(2+) signaling in failing human cardiomyocytes. Because apoptosis and Ca(2+)-related sarcomeric dysfunction are molecular hallmarks of ICM in humans, our results provide strong evidence that CD56(140kD) up-regulation plays a pivotal role in the pathogenesis of ICM and may be a target for future immunotherapeutic strategies in the treatment of this common and often fatal disease.


Subject(s)
CD56 Antigen/metabolism , Cardiomyopathies/pathology , Myocardial Ischemia/pathology , Animals , Apoptosis , CD56 Antigen/genetics , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomyopathies/complications , Cardiomyopathies/genetics , Cell Proliferation , Disease Models, Animal , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Molecular Weight , Mutant Proteins/metabolism , Myocardial Ischemia/complications , Myocardial Ischemia/genetics , Myocardium/metabolism , Myocardium/pathology , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Isoforms/metabolism , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
16.
Mol Cancer Ther ; 12(2): 173-83, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23243060

ABSTRACT

Hodgkin-Reed/Sternberg (HRS) cells of classical Hodgkin lymphoma show aberrant expression and activation of several receptor tyrosine kinases (RTK) in the majority of cases. Therefore, we tested whether tyrosine kinase inhibitors (TKI) already in clinical use or late stages of clinical trials have antiproliferative effects on HRS cell lines and evaluated the targets, affected signaling pathways, and mechanisms of cell death and resistance. Sorafenib and lestaurtinib had antiproliferative effects on HRS cell lines at concentrations achievable in patients. Sorafenib inhibited platelet-derived growth factor receptor (PDGFR) α, TRKA and RON, caused decreases in total and phosphorylated amounts of several signaling molecules, and provoked caspase-3-independent cell death, most likely due to endoplasmic reticulum stress as indicated by upregulation of GADD34 and GADD153 and phosphorylation of PERK. Lestaurtinib inhibited TRKA, PDGFRα, RON, and JAK2 and had only a cytostatic effect. Besides deactivation, lestaurtinib also caused activation of signaling pathways. It caused increases in CD30L and TRAIL expression, and CD30L/CD30 signaling likely led to the observed concomitant activation of extracellular signal-regulated kinase 1/2 and the alternative NF-κB pathway. These data disclose the possible use of sorafenib for the treatment of Hodgkin lymphoma and highlight NF-κB activation as a potential novel mechanism of resistance toward TKIs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carbazoles/pharmacology , Endoplasmic Reticulum Stress/drug effects , Hodgkin Disease/drug therapy , NF-kappa B/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Reed-Sternberg Cells/drug effects , Apoptosis/drug effects , Carbazoles/administration & dosage , Cell Line, Tumor , Drug Interactions , Drug Resistance, Neoplasm , Furans , Hodgkin Disease/enzymology , Hodgkin Disease/metabolism , Hodgkin Disease/pathology , Humans , Niacinamide/administration & dosage , Niacinamide/pharmacology , Phenylurea Compounds/administration & dosage , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Reed-Sternberg Cells/enzymology , Reed-Sternberg Cells/metabolism , Reed-Sternberg Cells/pathology , Signal Transduction/drug effects , Sorafenib
17.
Cardiovasc Pathol ; 21(6): 515-8, 2012.
Article in English | MEDLINE | ID: mdl-22377182

ABSTRACT

Hereditary hemorrhagic telangiectasia is an autosomal dominant vascular disease often manifesting with epistaxis, telangiectasia, and intraparenchymatous arteriovenous malformations. We report on the case of a 71-year-old man who was admitted to hospital due to a tricuspid valve insufficiency. During the following days, the patient developed liver and renal failure; the clinical condition worsened rapidly. Computed tomographic diagnostics revealed arteriovenous malformations in the lung and in the liver portal. Additionally, mucocutaneous telangiectasia in the mouth was found. Hereditary hemorrhagic telangiectasia was assumed; nevertheless, an effective treatment was impossible because of the patient's worse clinical state; he died a few days later. Autopsy affirmed the diagnosis of hereditary hemorrhagic telangiectasia; molecular genetic analysis revealed a heterozygous mutation in the ALK-1 gene. Despite its relatively high prevalence, hereditary hemorrhagic telangiectasia is not considered as a diagnosis as frequently as it should be, and clinicians need to be aware of the signs of hereditary hemorrhagic telangiectasia as well as the appropriate diagnostic workup.


Subject(s)
Heart Failure/diagnosis , Heart Ventricles/pathology , Telangiectasia, Hereditary Hemorrhagic/diagnosis , Tricuspid Valve Insufficiency/pathology , Ventricular Dysfunction, Right/diagnosis , Aged , Fatal Outcome , Heart Failure/etiology , Heart Failure/physiopathology , Humans , Male , Telangiectasia, Hereditary Hemorrhagic/complications , Telangiectasia, Hereditary Hemorrhagic/physiopathology , Ventricular Dysfunction, Right/physiopathology
18.
Blood ; 114(20): 4503-6, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19734449

ABSTRACT

STATs are constitutively activated in several malignancies. In primary mediastinal large B-cell lymphoma and Hodgkin lymphoma (HL), inactivating mutations in SOCS1, an inhibitor of JAK/STAT signaling, contribute to deregulated STAT activity. Based on indications that the SOCS1 mutations are caused by the B cell-specific somatic hypermutation (SHM) process, we analyzed B-cell non-HL and normal B cells for mutations in SOCS1. One-fourth of diffuse large B-cell lymphoma and follicular lymphomas carried SOCS1 mutations, which were preferentially targeted to SHM hotspot motifs and frequently obviously inactivating. Rare mutations were observed in Burkitt lymphoma, plasmacytoma, and mantle cell lymphoma but not in tumors of a non-B-cell origin. Mutations in single-sorted germinal center B cells were infrequent relative to other genes mutated as byproducts of normal SHM, indicating that SOCS1 inactivation in primary mediastinal large B-cell lymphoma, HL, diffuse large B-cell lymphoma, and follicular lymphoma is frequently the result of aberrant SHM.


Subject(s)
Lymphoma, B-Cell/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Humans , Mutation , Polymerase Chain Reaction , Suppressor of Cytokine Signaling 1 Protein
19.
Mol Immunol ; 46(4): 630-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18952293

ABSTRACT

Human peripheral blood (PB) B cells expressing only IgD and tonsillar IgD-secreting plasma cells carry highly mutated V(H) genes and show preferential Iglambda usage. To further characterize these peculiar cells and gain insight into their generation, we analysed rearranged V(H) and V(L) genes of single IgD-only lambda(+) PB B cells and IgD(+) plasma cells from four individuals each. We demonstrate that the high somatic hypermutation activity in these cells is not restricted to V(H) genes but also present in V(L) genes. Moreover, not only PB IgD-only B cells, as reported earlier, but also IgD-expressing plasma cells often belong to very large clones. Surprisingly, the V(H)3-30 gene segment was used in each PB donor by >30% of IgD-only cells and in 2 tonsils by >50% of IgD plasma cells, whereas it was used less frequent in other B cells. All these features fit to a model in which IgD-only cells develop in superantigen-driven germinal center reactions, in which B cells are activated by binding of antigens to constant parts of Cdelta and often lambda light chains and the V(H)3-30 segment, and are selected for deletion of Cmu. IgD-only B cells may hence represent a unique B lineage subset generated in response to particular antigens.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin D/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Plasma Cells/immunology , Somatic Hypermutation, Immunoglobulin , Amino Acid Sequence , B-Lymphocytes/metabolism , Gene Rearrangement/genetics , Gene Rearrangement/immunology , Humans , Immunoglobulin D/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Molecular Sequence Data , Mutation , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Plasma Cells/metabolism , Superantigens/genetics , Superantigens/immunology
20.
J Exp Med ; 205(10): 2251-68, 2008 Sep 29.
Article in English | MEDLINE | ID: mdl-18794340

ABSTRACT

The pathogenesis of nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL) and its relationship to other lymphomas are largely unknown. This is partly because of the technical challenge of analyzing its rare neoplastic lymphocytic and histiocytic (L&H) cells, which are dispersed in an abundant nonneoplastic cellular microenvironment. We performed a genome-wide expression study of microdissected L&H lymphoma cells in comparison to normal and other malignant B cells that indicated a relationship of L&H cells to and/or that they originate from germinal center B cells at the transition to memory B cells. L&H cells show a surprisingly high similarity to the tumor cells of T cell-rich B cell lymphoma and classical Hodgkin lymphoma, a partial loss of their B cell phenotype, and deregulation of many apoptosis regulators and putative oncogenes. Importantly, L&H cells are characterized by constitutive nuclear factor kappaB activity and aberrant extracellular signal-regulated kinase signaling. Thus, these findings shed new light on the nature of L&H cells, reveal several novel pathogenetic mechanisms in NLPHL, and may help in differential diagnosis and lead to novel therapeutic strategies.


Subject(s)
Gene Expression Profiling , Hodgkin Disease , Lymphocytes/immunology , Lymphoma, Follicular , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Biomarkers/metabolism , Diagnosis, Differential , Enzyme Activation , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Germinal Center/cytology , Hodgkin Disease/genetics , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Lymph Nodes/cytology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Microarray Analysis , NF-kappa B/metabolism , Phenotype , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...