Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Clin Exp Hematop ; 60(3): 87-96, 2020.
Article in English | MEDLINE | ID: mdl-32981916

ABSTRACT

MYC is a transcriptional factor that regulates growth and proliferation through cell cycle pathways. MYC alterations, in particular MYC rearrangements, are important in assessing the prognosis of aggressive B-cell lymphoma. In this study, we focused on the impact of nine major cell cycle genes for MYC-driven aggressive mature B-cell lymphoma and analyzed the mutational status using targeted next generation sequencing. Our 40 cases of aggressive mature B-cell lymphomas included 5 Burkitt lymphomas, 17 high-grade B-cell lymphomas and 18 diffuse large B-cell lymphomas with MYC breaks in 100%, 88% and 11%, respectively. Our data allowed a molecular classification into four categories partially independent from the histopathological diagnosis but correlating with the Ki-67 labelling index: (I) harboring TP53 and CDKN2A mutations, being highly proliferative, (II) with MYC rearrangement associated with MYC and/or ID3 mutations, being highly proliferative, (III) with MYC rearrangement combined with additional molecular changes, being highly proliferative, and (IV) with a diverse pattern of molecular alterations, being less proliferative. Taken together, we found that mutations of TP53, CDKN2A, MYC and ID3 are associated with highly proliferative B-cell lymphomas that could profit from novel therapeutic strategies.


Subject(s)
Cell Cycle , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins c-myc/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Humans , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/pathology , Mutation , Prognosis
2.
J Cancer Res Clin Oncol ; 144(3): 499-507, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29327244

ABSTRACT

PURPOSE: To analyze the impact of TNFα or IL2 on human lymphocytes in vitro and the anti-tumor and immune-modifying effects of L19-IL2 and L19-TNFα on subcutaneously growing J558L myeloma in immunocompetent mice. METHODS: PBMCs from three healthy volunteers were incubated with IL2, TNFα, or with IL2 plus addition of TNFα (final 20 h). BALB/c J558L mice with subcutaneous tumors were treated with intravenous L19-TNFα plus L19-IL2, or controls. Tumor growth and intra- and peri-tumoral tissues were analyzed for micro-vessel density, necrosis, immune cell composition, and PD1 or PD-L1 expressing cells. RESULTS: Exposure of PBMC in vitro to IL2, TNFα, or to IL2 over 3 and 5 days plus TNFα for the final 20 h resulted in an approximately 50 and 75% reduction of the CD25low effector cell/CD25high Treg cell ratio, respectively, compared to medium control. IL2 or TNFα increased the proportion of CD4- CD25low effector lymphocytes while reducing the proportion of CD4+ CD25low Teff cells. In the J558L myeloma model, tumor eradication was observed in 58, 42, 25, and 0% of mice treated with L19-TNFα plus L19-IL2, L19-TNFα, L19-IL2, and PBS, respectively. L19-TNFα/L19-IL2 combination caused tumor necrosis, capillary density doubling, peri-tumoral T cell and PD1+ T cell reduction (- 50%), and an increase in PD-L1+ myeloma cells. CONCLUSION: IL2, TNFα, or IL2 plus TNFα (final 20 h) increased the proportion of CD4- CD25low effector lymphocytes possibly indicating immune activation. L19-TNFα/L19-IL2 combination therapy eradicated tumors in J558L myeloma BALB/c mice likely via TNFα-induced tumor necrosis and L19-TNFα/L19-IL2-mediated local cellular immune reactions.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Interleukin-2/therapeutic use , Multiple Myeloma/therapy , Neovascularization, Pathologic/drug therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Drug Delivery Systems , Female , Immunotoxins/therapeutic use , Mice , Mice, Inbred BALB C , Multiple Myeloma/immunology , Multiple Myeloma/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Transplantation, Isogeneic
3.
Mol Oncol ; 10(8): 1232-44, 2016 10.
Article in English | MEDLINE | ID: mdl-27324824

ABSTRACT

Histone deacetylase (HDAC) inhibitors such as suberoylanilide hydroxamic acid (SAHA) are not commonly used in clinical practice for treatment of B-cell lymphomas, although a subset of patients with refractory or relapsed B-cell lymphoma achieved partial or complete remissions. Therefore, the purpose of this study was to identify molecular features that predict the response of B-cell lymphomas to SAHA treatment. We designed an integrative approach combining drug efficacy testing with exome and captured target analysis (DETECT). In this study, we tested SAHA sensitivity in 26 B-cell lymphoma cell lines and determined SAHA-interacting proteins in SAHA resistant and sensitive cell lines employing a SAHA capture compound (CC) and mass spectrometry (CCMS). In addition, we performed exome mutation analysis. Candidate validation was done by expression analysis and knock-out experiments. An integrated network analysis revealed that the Src tyrosine kinase Gardner-Rasheed feline sarcoma viral (v-fgr) oncogene homolog (FGR) is associated with SAHA resistance. FGR was specifically captured by the SAHA-CC in resistant cells. In line with this observation, we found that FGR expression was significantly higher in SAHA resistant cell lines. As functional proof, CRISPR/Cas9 mediated FGR knock-out in resistant cells increased SAHA sensitivity. In silico analysis of B-cell lymphoma samples (n = 1200) showed a wide range of FGR expression indicating that FGR expression might help to stratify patients, which clinically benefit from SAHA therapy. In conclusion, our comprehensive analysis of SAHA-interacting proteins highlights FGR as a factor involved in SAHA resistance in B-cell lymphoma.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Lymphoma, B-Cell/pathology , Proto-Oncogene Proteins/metabolism , src-Family Kinases/metabolism , Cell Line, Tumor , Gene Knockout Techniques , Gene Regulatory Networks/drug effects , Humans , Mass Spectrometry , Mutation/genetics , Reproducibility of Results , Vorinostat
4.
Br J Cancer ; 113(12): 1704-11, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26645239

ABSTRACT

BACKGROUND: Lung cancer is the leading cause of cancer-related deaths worldwide. The typical and atypical carcinoid (TC and AC), the large-cell neuroendocrine carcinoma (LCNEC) and the small-cell lung cancers (SCLC) are subgroups of pulmonary tumours that show neuroendocrine differentiations. With the rising impact of molecular pathology in routine diagnostics the interest for reliable biomarkers, which can help to differentiate these subgroups and may enable a more personalised treatment of patients, grows. METHODS: A collective of 70 formalin-fixed, paraffin-embedded (FFPE) pulmonary neuroendocrine tumours (17 TCs, 17 ACs, 19 LCNECs and 17 SCLCs) was used to identify biomarkers by high-throughput sequencing. Using the Illumina TruSeq Amplicon-Cancer Panel on the MiSeq instrument, the samples were screened for alterations in 221 mutation hot spots of 48 tumour-relevant genes. RESULTS: After filtering >26 000 detected variants by applying strict algorithms, a total of 130 mutations were found in 29 genes and 49 patients. Mutations in JAK3, NRAS, RB1 and VHL1 were exclusively found in SCLCs, whereas the FGFR2 mutation was detected in LCNEC only. KIT, PTEN, HNF1A and SMO were altered in ACs. The SMAD4 mutation corresponded to the TC subtype. We prove that the frequency of mutations increased with the malignancy of tumour type. Interestingly, four out of five ATM-mutated patients showed an additional alteration in TP53, which was by far the most frequently altered gene (28 out of 130; 22%). We found correlations between tumour type and IASLC grade for ATM- (P=0.022; P=0.008) and TP53-mutated patients (P<0.001). Both mutated genes were also associated with lymph node invasion and distant metastasis (P⩽0.005). Furthermore, PIK3CA-mutated patients with high-grade tumours showed a reduced overall survival (P=0.040) and the mutation frequency of APC and ATM in high-grade neuroendocrine lung cancer patients was associated with progression-free survival (PFS) (P=0.020). CONCLUSIONS: The implementation of high-throughput sequencing for the analysis of the neuroendocrine lung tumours has revealed that, even if these tumours encompass several subtypes with varying clinical aggressiveness, they share a number of molecular features. An improved understanding of the biology of neuroendocrine tumours will offer the opportunity for novel approaches in clinical management, resulting in a better prognosis and prediction of therapeutic response.


Subject(s)
Lung Neoplasms/genetics , Mutation , Neuroendocrine Tumors/genetics , Adult , Aged , Aged, 80 and over , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neuroendocrine Tumors/pathology , Paraffin Embedding , Young Adult
5.
J Cancer Res Clin Oncol ; 140(1): 35-43, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24132461

ABSTRACT

PURPOSE: The extradomain B of fibronectin (ED-B) is a promising vascular target for selective pharmacodelivery in cancer patients. We analyzed a large series of prostatectomies from patients with prostate cancer, hyperplastic prostate disease, and normal prostates to study extent and tumor-selectivity of ED-B expression. METHODS: Using immunohistology, 68 adenocarcinomas of the prostate or prostate cancer-inflicted lymph nodes, 4 samples of benign prostatic hyperplasia, and 6 normal prostate glands were studied for ED-B expressing newly formed blood vessels. Further, we treated an advanced prostate cancer patient with the anti-ED-B antibody (131)I-L19SIP to study in vivo target accessibility. RESULTS: ED-B-positive blood vessels were found significantly more frequent in prostate cancers as compared with peritumoral prostate tissues or normal prostate glands, independent of tumor differentiation. The ED-B-positive blood vessels' density was 97 (±23), 65 (±9), and 59 (±9)/mm(2) in G3, G2, and G1 prostate cancers, respectively, and 7 (±5)/mm(2) in normal prostate glands. In high-grade (G3) prostate cancers, also the peritumoral tissue showed a higher density of ED-B vessels than normal prostate glands. Similar results were obtained when ED-B-positive vessel density was expressed as a fraction of CD34-positive vessel density. Finally, selective uptake of ED-B-binding (131)I-L19SIP to tumor lesions was found in an advanced prostate cancer patient by whole-body planar scintigraphy. CONCLUSIONS: ED-B-positive blood vessels were found to a large extent in prostate cancer tissues, but only rarely in normal prostates or benign prostatic hyperplasia. Whole-body planar scintigraphy in a prostate cancer patient confirmed selective uptake of (131)I-L19SIP in the prostate cancer tissues, qualifying ED-B as a promising target for selective pharmacodelivery of anticancer agents in prostate cancer.


Subject(s)
Fibronectins/biosynthesis , Immunotoxins/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Antibodies/immunology , Blood Vessels/metabolism , Epitopes/analysis , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/metabolism , Paraffin Embedding , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Radioimmunodetection , Recombinant Fusion Proteins/immunology
6.
Lab Invest ; 93(6): 677-89, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23545938

ABSTRACT

CD30, a member of the tumor necrosis factor receptor (TNFR) superfamily, is consistently expressed by tumor cells of anaplastic large-cell lymphoma (ALCL). CD30 stimulation induces massive caspase-dependent cell death of ALCL cells in case of canonical NFκB inhibition or proteasome inhibition. However, CD30, a TNFR lacking a death domain (DD), is unable to recruit a death inducing complex containing TRADD (TNFR1-associated DD-protein) or FADD (FAS-associated DD-domain protein) together with the receptor-interacting protein 1 (RIP1) and caspase-8. Thus, the mechanism explaining CD30-induced cell death of lymphocytes remains obscure. Here, we demonstrate that blockage of RIP1 by siRNA or pharmacological inhibition of RIP1 by Necrostatin-1 almost completely prevented CD30-induced cell death. In addition, we revealed CD30-induced accumulation of RIP1 at the cytoplasma membrane of NFκB-inhibited ALCL cells by confocal laser scanning microscopy. Finally, primary ALCL cases can be subdivided into two groups based on the presence or absence of RIP1 as revealed by immunohistology. Taken together, our study identified RIP1 as a crucial mediator of CD30-induced cell death that bears features of apoptosis as well as necroptosis. RIP1 expression in ALCL tumor cells might eligible for the therapeutic application of CD30 antibodies in combination with NFκB/proteasome inhibitors that should result in CD30-induced cell death.


Subject(s)
Ki-1 Antigen/metabolism , Lymphoma, Large-Cell, Anaplastic/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Autocrine Communication , Baculoviral IAP Repeat-Containing 3 Protein , Benzoquinones , Cell Death , Cell Membrane/metabolism , HeLa Cells , Humans , Imidazoles , Indoles , Inhibitor of Apoptosis Proteins/metabolism , Lactams, Macrocyclic , NF-kappa B/metabolism , TNF Receptor-Associated Factor 1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases
7.
Histopathology ; 60(6B): E19-27, 2012 May.
Article in English | MEDLINE | ID: mdl-22393903

ABSTRACT

AIMS: A20 (TNFAIP3) is a nuclear factor-κB (NF-κB)-inducible component of tumour necrosis factor and Toll-like receptor intracellular signal transduction. It negatively regulates NF-κB, and has been identified as a tumour suppressor. Several studies have described A20 inactivation by deletion of the A20 locus at 6q23, inactivating mutations, and/or methylation of the A20 promoter in various lymphoma entities. METHODS AND RESULTS: We generated a monoclonal antibody against the C-terminus of A20 (Ber-A20) and investigated full-length A20 expression of normal lymphoid tissue and lymphomas for the first time. We identified loss of A20 expression in tumour cells of 24% of classical Hodgkin lymphoma, 27% of diffuse large B-cell lymphoma, 20% of chronic lymphocytic leukaemia, 19% of follicular lymphoma, 13% of mantle cell lymphoma and 8% of primary mediastinal B-cell lymphoma cases by immunohistology. Loss of A20 expression rarely occurred in T-cell non-Hodgkin lymphoma. CONCLUSIONS: Our data are in agreement with cytogenetic and molecular analyses. Among 21 cases of ocular adnexal marginal zone lymphomas with known A20 mutation status, we detected complete absence of A20 expression, whereas cases with wild-type A20 were weakly A20-positive. We demonstrate that A20 loss can be detected by immunohistology with a sensitivity similar to that of complex molecular and genetic methods.


Subject(s)
Antibodies, Monoclonal/chemistry , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Antibodies, Monoclonal/metabolism , DNA-Binding Proteins/immunology , Eye Neoplasms/metabolism , Hodgkin Disease/metabolism , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/immunology , Lymphoma, Non-Hodgkin/metabolism , Mutation , Nuclear Proteins/immunology , Tumor Necrosis Factor alpha-Induced Protein 3
8.
Lab Invest ; 92(2): 191-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21986812

ABSTRACT

Endogenous ß-galactose-binding lectins have many biological functions, but their biological significance in Hodgkin lymphoma (HL) and anaplastic large cell lymphoma (ALCL) remains unclear. By immunohistochemistry, we analyzed the expression of galectin-1 and galectin-3 in HL and ALCL cases as well as in cell lines, and investigated the pharmacological effects of galectin-1 treatment with and without CD30 pre-stimulation of HL and ALCL cell lines. The galectin-3-negative human embryonic kidney cell line (HEK-293) was transfected with galectin-3 cDNA. Galectin-3 is differentially expressed in HL and ALCL. CD30 stimulation of the ALCL cell line Karpas 299 activates NF-κB without induction of apoptosis. Galectin-1 treatment of Karpas 299 induces cell death, which is significantly increased by CD30 pre-stimulation. The CD30-mediated increase of galectin-1-induced cell death is to some extent caspase independent and does not influence the expression of tumor necrosis factor-associated factor 1 (TRAF1), TRAF2, and cellular inhibitor of apoptosis 2 protein (cIAP2), as revealed in Karpas 299 cells. In other cell lines except Karpas 299, CD30 pre-stimulation did not significantly enhance galectin-1-induced cell death. Galectin-3 transfection of HEK-293 cells resulted in cell surface expression of galectin-3, associated with marked cell aggregation. CD30-targeted therapy in combination with galectin-1 treatment may induce effective killing of ALCL cells but not of HL cells.


Subject(s)
Apoptosis/physiology , Galectin 1/physiology , Hodgkin Disease/pathology , Ki-1 Antigen/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Signal Transduction/physiology , Base Sequence , Cell Line , Cell Line, Tumor , DNA Primers , Flow Cytometry , Hodgkin Disease/metabolism , Humans , Immunohistochemistry , Lymphoma, Large-Cell, Anaplastic/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
PLoS One ; 6(11): e26837, 2011.
Article in English | MEDLINE | ID: mdl-22102868

ABSTRACT

BACKGROUND: MYC is a key transcription factor involved in central cellular processes such as regulation of the cell cycle, histone acetylation and ribosomal biogenesis. It is overexpressed in the majority of human tumors including aggressive B-cell lymphoma. Especially Burkitt lymphoma (BL) is a highlight example for MYC overexpression due to a chromosomal translocation involving the c-MYC gene. However, no genome-wide analysis of MYC-binding sites by chromatin immunoprecipitation (ChIP) followed by next generation sequencing (ChIP-Seq) has been conducted in BL so far. METHODOLOGY/PRINCIPAL FINDINGS: ChIP-Seq was performed on 5 BL cell lines with a MYC-specific antibody giving rise to 7,054 MYC-binding sites after bioinformatics analysis of a total of approx. 19 million sequence reads. In line with previous findings, binding sites accumulate in gene sets known to be involved in the cell cycle, ribosomal biogenesis, histone acetyltransferase and methyltransferase complexes demonstrating a regulatory role of MYC in these processes. Unexpectedly, MYC-binding sites also accumulate in many B-cell relevant genes. To assess the functional consequences of MYC binding, the ChIP-Seq data were supplemented with siRNA- mediated knock-downs of MYC in BL cell lines followed by gene expression profiling. Interestingly, amongst others, genes involved in the B-cell function were up-regulated in response to MYC silencing. CONCLUSION/SIGNIFICANCE: The 7,054 MYC-binding sites identified by our ChIP-Seq approach greatly extend the knowledge regarding MYC binding in BL and shed further light on the enormous complexity of the MYC regulatory network. Especially our observations that (i) many B-cell relevant genes are targeted by MYC and (ii) that MYC down-regulation leads to an up-regulation of B-cell genes highlight an interesting aspect of BL biology.


Subject(s)
Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , DNA, Neoplasm/genetics , High-Throughput Nucleotide Sequencing , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Binding Sites , Biomarkers, Tumor/genetics , Chromatin Immunoprecipitation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , RNA, Small Interfering/genetics , Tumor Cells, Cultured
10.
J Invest Dermatol ; 130(3): 826-40, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19890350

ABSTRACT

Death ligands, including TNF-alpha, CD95L/FasL, and TRAIL, mediate safeguard mechanisms against tumor growth and critically contribute to lymphocyte homeostasis. We investigated death receptor-mediated apoptosis and CD30/CD95 crosstalk in four CD30-positive cell lines of cutaneous anaplastic large-cell lymphoma (cALCL). Whereas CD95 stimulation strongly induced apoptosis in cALCL cells, the pro-apoptotic pathways of TNF-alpha and TRAIL were completely blocked at an early step. Expression of TNF receptor 1 was lost in three of four cell lines, providing an explanation for TNF-alpha unresponsiveness. TRAIL resistance may be explained by the consistent overexpression of cellular flice inhibitory protein (c-FLIP) (four of four cell lines) and frequent loss of the pro-apoptotic Bcl-2 protein Bid (three of four cell lines). Changes at the receptor-expression level were largely ruled out. CD30/CD95 crosstalk experiments showed that CD30 ligation leads to NF-kappaB-mediated c-FLIP upregulation in cALCL cells, which in turn conferred enhanced resistance to CD95-mediated apoptosis. Knockdown of c-FLIP by a lentiviral approach enhanced basic apoptosis rates in cALCL cells and diminished the CD30-mediated suppression of apoptosis, thus proving the significance of c-FLIP in this context. These in vitro findings may be indicative of the clinical situation of cALCL. Further clarifying the defects in apoptosis pathways in cutaneous lymphomas may lead to improved therapies for these disorders.


Subject(s)
Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Ki-1 Antigen/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Skin Neoplasms/pathology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspases/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Ki-1 Antigen/pharmacology , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/physiopathology , NF-kappa B/metabolism , RNA Interference , Receptor Cross-Talk/physiology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/physiopathology , Up-Regulation/drug effects , Up-Regulation/physiology , fas Receptor/metabolism
11.
Blood ; 113(10): 2265-74, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19131554

ABSTRACT

Current treatment of hematologic malignancies involves rather unspecific chemotherapy, frequently resulting in severe adverse events. Thus, modern clinical research focuses on compounds able to discriminate malignant from normal tissues. Being expressed in newly formed blood vessels of solid cancers but not in normal mature tissues, the extradomain B of fibronectin (ED-B FN) is a promising target for selective cancer therapies. Using immunohistology with a new epitope retrieval technique for paraffin-embedded tissues, ED-B FN expression was found in biopsies from more than 200 Hodgkin and non-Hodgkin lymphoma patients of nearly all entities, and in patients with myeloproliferative diseases. ED-B FN expression was nearly absent in normal lymph nodes (n = 10) and bone marrow biopsies (n = 9). The extent of vascular ED-B FN expression in lymphoma tissues was positively correlated with grade of malignancy. ED-B FN expression was enhanced in lymph nodes with severe lymphadenopathy and in some hyperplastic tonsils. The in vivo accessibility of ED-B FN was confirmed in 3 lymphoma patients, in whom the lymphoma lesions were visualized on scintigraphy with (131)I-labeled L19 small immunoprotein ((131)I-L19SIP). In 2 relapsed Hodgkin lymphoma patients(131)I-L19SIP radioimmunotherapy induced a sustained partial response, qualifying ED-B FN as a promising target for antibody-based lymphoma therapies.


Subject(s)
Antibodies/therapeutic use , Fibronectins/biosynthesis , Hodgkin Disease/radiotherapy , Radioimmunotherapy/methods , Recombinant Fusion Proteins/therapeutic use , Animals , Fluorescent Antibody Technique , Glucose-6-Phosphate/analogs & derivatives , Hodgkin Disease/metabolism , Humans , Immunohistochemistry , Mice , Mice, Nude , Positron-Emission Tomography , Protein Isoforms/biosynthesis , Tomography, Emission-Computed, Single-Photon
12.
Am J Pathol ; 172(2): 510-20, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18187570

ABSTRACT

High CD30 expression in classical Hodgkin's lymphoma and anaplastic large cell lymphoma (ALCL) suggests an important pathogenic role of this cytokine receptor. To test this hypothesis, we investigated CD30 signaling in Hodgkin's and ALCL cell lines by different approaches: 1) CD30 stimulation, 2) CD30 down-regulation, and 3) a combination of both. The effects were determined at the RNA (microarray and real-time quantitative RT-PCR), protein (electrophoretic mobility shift analysis, immunoblot, and flow cytometry), and cellular/functional (proliferation and apoptosis) levels. We demonstrate that Hodgkin's cells are virtually CD30 unresponsive. Neither CD30 stimulation nor CD30 silencing of Hodgkin's cells had any significant effect. In contrast, CD30 stimulation of ALCL cells activated nuclear transcription factor-kappaB (NF-kappaB), induced major transcriptional changes, and decreased proliferation. These effects could be abrogated by down-regulation of CD30. Stimulation of CD30 in ALCL cells, stably transfected with a dominant-negative NF-kappaB inhibitor, induced pronounced caspase activation and massive apoptosis. Our data indicate that 1) CD30 signaling is not effective in Hodgkin's cell lines but is effective in ALCL cell lines, 2) CD30 is probably not significantly involved in the pathogenesis of classical Hodgkin's lymphoma, and 3) CD30 stimulation triggers two competing effects in ALCL cells, namely activation of caspases and NF-kappaB-mediated survival. These data suggest that CD30-targeted therapy in ALCL should be combined with NF-kappaB inhibitors to induce effective cell killing.


Subject(s)
Gene Expression Regulation, Neoplastic , Hodgkin Disease/metabolism , Ki-1 Antigen/metabolism , Lymphoma, Large-Cell, Anaplastic/metabolism , Signal Transduction/physiology , Apoptosis/physiology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Electrophoretic Mobility Shift Assay , Enzyme Activation/physiology , Flow Cytometry , Gene Expression , Gene Expression Profiling , Humans , Immunoblotting , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection
13.
Leuk Lymphoma ; 48(6): 1179-86, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17577782

ABSTRACT

Although the therapy of Hodgkin lymphoma and anaplastic large cell lymphoma has been considerably improved during the last decades, high therapeutic toxicity, relapses, secondary tumors, and primary treatment failure(s) occur. Both malignancies are well suited for CD30-targeted immunotherapy because of their strong CD30 expression. We constructed an immunotoxin composed of a single chain variable fragment of a CD30 antibody fused to the human pancreatic ribonuclease, showing CD30-specific binding and ribonucleolytic activity resistant to the inhibitor RNasin. This immunotoxin revealed CD30-specific anti-tumor activity in BALB/c mice that were challenged with CD30-positive or CD30-negative syngeneic tumor cells.


Subject(s)
Cytotoxins/pharmacology , Immunotoxins/pharmacology , Ki-1 Antigen/immunology , Ribonuclease, Pancreatic/immunology , Animals , Antibody Specificity , Cell Line , Cell Proliferation/drug effects , Drosophila melanogaster , Female , Humans , Ki-1 Antigen/chemistry , Mice , Mice, Inbred BALB C , Neoplasm Transplantation/pathology , Protein Binding , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Ribonuclease, Pancreatic/chemistry , Transplantation, Isogeneic/pathology , Tumor Cells, Cultured
14.
J Invest Dermatol ; 127(8): 1898-904, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17392826

ABSTRACT

Lymphomatoid papulosis (LyP), primary cutaneous anaplastic large T-cell lymphoma (cALCL), and cutaneous infiltrates of systemic anaplastic large cell lymphoma (sALCL) are CD30-positive lymphoproliferative disorders of the skin that overlap clinically, histopathologically, immunophenotypically, and genetically but differ considerably in their prognosis. In particular, lesions of LyP regress spontaneously, whereas those of cALCL and sALCL persist and may progress and spread to extracutaneous sites. In contrast to patients with cALCL, LyP patients do not benefit from an aggressive radio- and/or chemotherapeutic approach. We generated a novel tumor necrosis factor receptor (TNFR)-associated factor 1 (TRAF1) antibody that recognizes a formalin-resistant epitope (Ber-TRAF1A) and investigated the expression of TRAF1, an intracellular component of TNFR signaling, in LyP and ALCL. We could show a strong TRAF1 expression in the tumor cells of most LyP cases (42/49, 84%). In contrast, tumor cells of primary and secondary cALCL revealed TRAF1 expression in only a few cases (3/41, 7%) as shown for sALCL without skin manifestation. The data indicate that TRAF1 expression reliably distinguishes LyP from primary or secondary cALCL. This might be of crucial diagnostic importance and has a strong impact on the treatment decision for patients with cALCL and LyP.


Subject(s)
Ki-1 Antigen/analysis , Lymphoma, Large B-Cell, Diffuse/chemistry , Lymphoma, T-Cell, Cutaneous/chemistry , Lymphoma, T-Cell/chemistry , Lymphomatoid Papulosis/metabolism , TNF Receptor-Associated Factor 1/analysis , Animals , Humans , Mice , Mice, Inbred BALB C , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 1/physiology
15.
J Mol Med (Berl) ; 84(2): 132-41, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16308685

ABSTRACT

In this study, the expression of activated caspase-3 by the tumor cells of classical Hodgkin lymphoma (cHL), the Hodgkin-Reed-Sternberg (HRS) cells, is confirmed. This raises the question why caspase-3 does not kill HRS cells. There are only a few molecules, which are able to directly inhibit caspase-3. One of them is cIAP2. We show that cIAP2 is expressed in the HRS cells in 20 of 23 cHL cases by in situ hybridization. Suppression experiments with cIAP2 antisense RNA show that down-regulation of cIAP2 significantly reduces apoptosis resistance in cHL cell lines. cIAP2 overexpression appears to be unique for HRS cells since the tumor cells of non-Hodgkin lymphomas are nearly cIAP2-negative. We demonstrate that cIAP2 is inducible by CD30 stimulation in cHL cell lines of T-cell origin and anaplastic large cell lymphoma cell lines, whereas cHL cell lines of B-cell origin constitutively express cIAP2. Inhibition of cIAP2 expression by cIAP2 antisense RNA decreases resistance to apoptosis. The results indicate that cIAP2 contributes to the apoptosis resistance of HRS cells, mainly by inhibiting effector caspases. According to these findings, a therapeutical application of inhibitors of apoptosis proteins antagonists in cHL appears promising.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Reed-Sternberg Cells/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , HeLa Cells , Humans , In Situ Hybridization , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , RNA, Antisense/pharmacology , Reed-Sternberg Cells/pathology
16.
J Exp Med ; 199(8): 1041-52, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15078899

ABSTRACT

Resistance to death receptor-mediated apoptosis is supposed to be important for the deregulated growth of B cell lymphoma. Hodgkin/Reed-Sternberg (HRS) cells, the malignant cells of classical Hodgkin's lymphoma (cHL), resist CD95-induced apoptosis. Therefore, we analyzed death receptor signaling, in particular the CD95 pathway, in these cells. High level CD95 expression allowed a rapid formation of the death-inducing signaling complex (DISC) containing Fas-associated death domain-containing protein (FADD), caspase-8, caspase-10, and most importantly, cellular FADD-like interleukin 1beta-converting enzyme-inhibitory protein (c-FLIP). The immunohistochemical analysis of the DISC members revealed a strong expression of CD95 and c-FLIP overexpression in 55 out of 59 cases of cHL. FADD overexpression was detectable in several cases. Triggering of the CD95 pathway in HRS cells is indicated by the presence of CD95L in cells surrounding them as well as confocal microscopy showing c-FLIP predominantly localized at the cell membrane. Elevated c-FLIP expression in HRS cells depends on nuclear factor (NF)-kappaB. Despite expression of other NF-kappaB-dependent antiapoptotic proteins, the selective down-regulation of c-FLIP by small interfering RNA oligoribonucleotides was sufficient to sensitize HRS cells to CD95 and tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Therefore, c-FLIP is a key regulator of death receptor resistance in HRS cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Carrier Proteins/physiology , Hodgkin Disease/pathology , Hodgkin Disease/physiopathology , Intracellular Signaling Peptides and Proteins , Reed-Sternberg Cells/pathology , Reed-Sternberg Cells/physiology , fas Receptor/physiology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Base Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 10 , Caspase 8 , Caspases/metabolism , Cell Line, Tumor , Cycloheximide/pharmacology , Fas-Associated Death Domain Protein , Humans , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Reed-Sternberg Cells/drug effects , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/physiology
17.
J Pathol ; 200(2): 229-39, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12754742

ABSTRACT

A20 and TRAF1 are two anti-apoptotic components of the intracellular signalling pathway of the tumour necrosis factor receptor (TNFR) family. Induction of apoptosis seems to be a main function of these receptors. It is astonishing that a member of this family, CD30, is overexpressed by highly proliferating tumours such as Hodgkin lymphoma (HL) and anaplastic large cell lymphoma (ALCL). It is known that CD30 stimulation leads to the apoptosis of ALCL tumour cells but not of Hodgkin-Reed-Sternberg (HRS) cells. We have already established the overexpression of TRAF1 in HRS cells. In this study we demonstrate that A20 is highly expressed in the HRS cells in 20/22 of cases of classical HL, in 4/4 cases of nodular lymphocyte-predominant HL (NLPHL), and in 2/2 cases of the anaplastic variant of diffuse large B cell lymphoma. In contrast, all other non-Hodgkin lymphomas, including ALCL, revealed either no A20 reactivity, or reactivity in less than 1% of all tumour cells. CD30 stimulation induced A20 and TRAF1 expression. This effect was most prominent in HL and ALCL cell lines with low basal expression levels of these molecules. Immunohistological studies of reactive lymphoid blasts in tonsillar tissue demonstrated that co-expression of CD30, A20, and TRAF1 also occurs in vivo. Cell lines with high basal A20 and TRAF1 expression were resistant to CD30-mediated apoptosis. The sensitivity to CD30-induced apoptosis was increased by inhibition of protein synthesis. TRAF1 transfection decreased CD30-induced apoptosis. Our data suggest that A20 and TRAF1 contribute to apoptosis resistance and, therefore, play an important role in the pathogenesis of classical HL.


Subject(s)
Hodgkin Disease/metabolism , Lymphoma, Large-Cell, Anaplastic/metabolism , Neoplasm Proteins/metabolism , Proteins/metabolism , Apoptosis , Cell Division , DNA-Binding Proteins , Hodgkin Disease/pathology , Humans , Hyperplasia/metabolism , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Ki-1 Antigen/physiology , Lymphoma, Large-Cell, Anaplastic/pathology , Nuclear Proteins , Palatine Tonsil/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TNF Receptor-Associated Factor 1 , Tumor Cells, Cultured , Tumor Necrosis Factor alpha-Induced Protein 3 , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...