Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
2.
Nature ; 565(7738): 240-245, 2019 01.
Article in English | MEDLINE | ID: mdl-30568303

ABSTRACT

Patients with glioblastoma currently do not sufficiently benefit from recent breakthroughs in cancer treatment that use checkpoint inhibitors1,2. For treatments using checkpoint inhibitors to be successful, a high mutational load and responses to neoepitopes are thought to be essential3. There is limited intratumoural infiltration of immune cells4 in glioblastoma and these tumours contain only 30-50 non-synonymous mutations5. Exploitation of the full repertoire of tumour antigens-that is, both unmutated antigens and neoepitopes-may offer more effective immunotherapies, especially for tumours with a low mutational load. Here, in the phase I trial GAPVAC-101 of the Glioma Actively Personalized Vaccine Consortium (GAPVAC), we integrated highly individualized vaccinations with both types of tumour antigens into standard care to optimally exploit the limited target space for patients with newly diagnosed glioblastoma. Fifteen patients with glioblastomas positive for human leukocyte antigen (HLA)-A*02:01 or HLA-A*24:02 were treated with a vaccine (APVAC1) derived from a premanufactured library of unmutated antigens followed by treatment with APVAC2, which preferentially targeted neoepitopes. Personalization was based on mutations and analyses of the transcriptomes and immunopeptidomes of the individual tumours. The GAPVAC approach was feasible and vaccines that had poly-ICLC (polyriboinosinic-polyribocytidylic acid-poly-L-lysine carboxymethylcellulose) and granulocyte-macrophage colony-stimulating factor as adjuvants displayed favourable safety and strong immunogenicity. Unmutated APVAC1 antigens elicited sustained responses of central memory CD8+ T cells. APVAC2 induced predominantly CD4+ T cell responses of T helper 1 type against predicted neoepitopes.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Glioblastoma/diagnosis , Glioblastoma/therapy , Precision Medicine/methods , Adult , Aged , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Female , Glioblastoma/immunology , HLA-A Antigens/immunology , Humans , Immunologic Memory/immunology , Male , Middle Aged , T-Lymphocytes, Helper-Inducer/immunology , Treatment Outcome
3.
Nature ; 547(7662): 222-226, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28678784

ABSTRACT

T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of ß2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/therapy , Mutation/genetics , Precision Medicine/methods , RNA/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/immunology , CD8 Antigens/immunology , Cancer Vaccines/therapeutic use , Epitopes/genetics , Epitopes/immunology , Humans , Immunotherapy/methods , Melanoma/genetics , Neoplasm Metastasis , Neoplasm Recurrence, Local/prevention & control , Nivolumab , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Vaccination , beta 2-Microglobulin/deficiency
4.
Nature ; 534(7607): 396-401, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281205

ABSTRACT

Lymphoid organs, in which antigen presenting cells (APCs) are in close proximity to T cells, are the ideal microenvironment for efficient priming and amplification of T-cell responses. However, the systemic delivery of vaccine antigens into dendritic cells (DCs) is hampered by various technical challenges. Here we show that DCs can be targeted precisely and effectively in vivo using intravenously administered RNA-lipoplexes (RNA-LPX) based on well-known lipid carriers by optimally adjusting net charge, without the need for functionalization of particles with molecular ligands. The LPX protects RNA from extracellular ribonucleases and mediates its efficient uptake and expression of the encoded antigen by DC populations and macrophages in various lymphoid compartments. RNA-LPX triggers interferon-α (IFNα) release by plasmacytoid DCs and macrophages. Consequently, DC maturation in situ and inflammatory immune mechanisms reminiscent of those in the early systemic phase of viral infection are activated. We show that RNA-LPX encoding viral or mutant neo-antigens or endogenous self-antigens induce strong effector and memory T-cell responses, and mediate potent IFNα-dependent rejection of progressive tumours. A phase I dose-escalation trial testing RNA-LPX that encode shared tumour antigens is ongoing. In the first three melanoma patients treated at a low-dose level, IFNα and strong antigen-specific T-cell responses were induced, supporting the identified mode of action and potency. As any polypeptide-based antigen can be encoded as RNA, RNA-LPX represent a universally applicable vaccine class for systemic DC targeting and synchronized induction of both highly potent adaptive as well as type-I-IFN-mediated innate immune mechanisms for cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunotherapy/methods , Melanoma/immunology , Melanoma/therapy , RNA/administration & dosage , Administration, Intravenous , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Antigens, Viral/genetics , Autoantigens/genetics , Autoantigens/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Clinical Trials, Phase I as Topic , Dendritic Cells/cytology , Disease Models, Animal , Drug Carriers/administration & dosage , Female , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Lymphocyte Activation/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , RNA/genetics , Static Electricity , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Toll-Like Receptor 7/immunology
5.
BMC Cancer ; 15: 663, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26450156

ABSTRACT

BACKGROUND: T-cell acute lymphoblastic leukemia (T-ALL) is a genetically heterogeneous disease with the need for treatment optimization. Previously, high expression of Insulin-like growth factor binding protein 7 (IGFBP7), a member of the IGF system, was identified as negative prognostic factor in adult T-ALL patients. Since aberrant IGFBP7 expression was observed in a variety of neoplasia and was relevant for prognosis in T-ALL, we investigated the functional role of IGFBP7 in Jurkat and Molt-4 cells as in vitro models for T-ALL. METHODS: Jurkat and Molt-4 cells were stably transfected with an IGFBP7 over-expression vector or the empty vector as control. Proliferation of the cells was assessed by WST-1 assays and cell cycle status was measured by flow-cytometry after BrDU/7-AAD staining. The effect of IGFBP7 over-expression on sensitivity to cytostatic drugs was determined in AnnexinV/7-AAD assays. IGF1-R protein expression was measured by Western Blot and flow-cytometric analysis. IGF1-R associated gene expression profiles were generated from microarray gene expression data of 86 T-ALL patients from the Microarrays Innovations in Leukemia (MILE) multicenter study. RESULTS: IGFBP7-transfected Jurkat cells proliferated less, leading to a longer survival in a nutrient-limited environment. Both IGFBP7-transfected Jurkat and Molt-4 cells showed an arrest in the G0/G1 cell cycle phase. Furthermore, Jurkat IGFBP7-transfected cells were resistant to vincristine and asparaginase treatment. Surface expression and whole protein measurement of IGF1-R protein expression showed a reduced abundance of the receptor after IGFBP7 transfection in Jurkat cells. Interestingly, combination of the IGF1-R inhibitor NPV-AEW541 restored sensitivity to vincristine in IGFBP7-transfected cells. Additionally, IGF1-R associated GEP revealed an up-regulation of important drivers of T-ALL pathogenesis and regulators of chemo-resistance and apoptosis such as NOTCH1, BCL-2, PRKCI, and TP53. CONCLUSION: This study revealed a proliferation inhibiting effect of IGFBP7 by G0/G1 arrest and a drug resistance-inducing effect of IGFBP7 against vincristine and asparaginase in T-ALL. These results provide a model for the previously observed association between high IGFBP7 expression and chemotherapy failure in T-ALL patients. Since the resistance against vincristine was abolished by IGF1-R inhibition, IGFBP7 could serve as biomarker for patients who may benefit from therapies including IGF1-R inhibitors in combination with chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Insulin-Like Growth Factor Binding Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression , Gene Expression Profiling , Humans , Jurkat Cells , Receptor, IGF Type 1 , Transcriptome
6.
Oncotarget ; 6(5): 2754-66, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25595890

ABSTRACT

Novel target discovery is warranted to improve treatment in adult T-cell acute lymphoblastic leukemia (T-ALL) patients. We provide a comprehensive study on mutations to enhance the understanding of therapeutic targets and studied 81 adult T-ALL patients. NOTCH1 exhibitedthe highest mutation rate (53%). Mutation frequencies of FBXW7 (10%), WT1 (10%), JAK3 (12%), PHF6 (11%), and BCL11B (10%) were in line with previous reports. We identified recurrent alterations in transcription factors DNM2, and RELN, the WNT pathway associated cadherin FAT1, and in epigenetic regulators (MLL2, EZH2). Interestingly, we discovered novel recurrent mutations in the DNA repair complex member HERC1, in NOTCH2, and in the splicing factor ZRSR2. A frequently affected pathway was the JAK/STAT pathway (18%) and a significant proportion of T-ALL patients harboured mutations in epigenetic regulators (33%), both predominantly found in the unfavourable subgroup of early T-ALL. Importantly, adult T-ALL patients not only showed a highly heterogeneous mutational spectrum, but also variable subclonal allele frequencies implicated in therapy resistance and evolution of relapse. In conclusion, we provide novel insights in genetic alterations of signalling pathways (e.g. druggable by γ-secretase inhibitors, JAK inhibitors or EZH2 inhibitors), present in over 80% of all adult T-ALL patients, that could guide novel therapeutic approaches.


Subject(s)
Biomarkers, Tumor/genetics , Mutation , Polymorphism, Single Nucleotide , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Aged , DNA Mutational Analysis , Drug Resistance, Neoplasm/genetics , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Phenotype , Precision Medicine , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Predictive Value of Tests , Recurrence , Reelin Protein , Treatment Outcome , Young Adult
7.
J Hematol Oncol ; 7: 51, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25023966

ABSTRACT

BACKGROUND: Risk stratification, detection of minimal residual disease (MRD), and implementation of novel therapeutic agents have improved outcome in acute lymphoblastic leukemia (ALL), but survival of adult patients with T-cell acute lymphoblastic leukemia (T-ALL) remains unsatisfactory. Thus, novel molecular insights and therapeutic approaches are urgently needed. METHODS: We studied the impact of B-cell CLL/lymphoma 11b (BCL11b), a key regulator in normal T-cell development, in T-ALL patients enrolled into the German Multicenter Acute Lymphoblastic Leukemia Study Group trials (GMALL; n = 169). The mutational status (exon 4) of BCL11b was analyzed by Sanger sequencing and mRNA expression levels were determined by quantitative real-time PCR. In addition gene expression profiles generated on the Human Genome U133 Plus 2.0 Array (affymetrix) were used to investigate BCL11b low and high expressing T-ALL patients. RESULTS: We demonstrate that BCL11b is aberrantly expressed in T-ALL and gene expression profiles reveal an association of low BCL11b expression with up-regulation of immature markers. T-ALL patients characterized by low BCL11b expression exhibit an adverse prognosis [5-year overall survival (OS): low 35% (n = 40) vs. high 53% (n = 129), P = 0.02]. Within the standard risk group of thymic T-ALL (n = 102), low BCL11b expression identified patients with an unexpected poor outcome compared to those with high expression (5-year OS: 20%, n = 18 versus 62%, n = 84, P < 0.01). In addition, sequencing of exon 4 revealed a high mutation rate (14%) of BCL11b. CONCLUSIONS: In summary, our data of a large adult T-ALL patient cohort show that low BCL11b expression was associated with poor prognosis; particularly in the standard risk group of thymic T-ALL. These findings can be utilized for improved risk prediction in a significant proportion of adult T-ALL patients, which carry a high risk of standard therapy failure despite a favorable immunophenotype.


Subject(s)
Biomarkers, Tumor/analysis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Biomarkers, Tumor/genetics , DNA Mutational Analysis , Female , Humans , Kaplan-Meier Estimate , Male , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Real-Time Polymerase Chain Reaction , Repressor Proteins/biosynthesis , Risk Factors , Transcriptome , Tumor Suppressor Proteins/biosynthesis
8.
Blood ; 121(23): 4749-52, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23603912

ABSTRACT

Early T-cell precursor (ETP) acute lymphoblastic leukemia (ALL) is a high-risk subgroup of T-lineage ALL characterized by specific stem cell and myeloid features. In adult ETP-ALL, no comprehensive studies on the genetic background have been performed to elucidate molecular lesions of this distinct subgroup. We performed whole-exome sequencing of 5 paired ETP-ALL samples. In addition to mutations in genes known to be involved in leukemogenesis (ETV6, NOTCH1, JAK1, and NF1), we identified novel recurrent mutations in FAT1 (25%), FAT3 (20%), DNM2 (35%), and genes associated with epigenetic regulation (MLL2, BMI1, and DNMT3A). Importantly, we verified the high rate of DNMT3A mutations (16%) in a larger cohort of adult patients with ETP-ALL (10/68). Mutations in epigenetic regulators support clinical trials, including epigenetic-orientated therapies, for this high-risk subgroup. Interestingly, more than 60% of adult patients with ETP-ALL harbor at least a single genetic lesion in DNMT3A, FLT3, or NOTCH1 that may allow use of targeted therapies.


Subject(s)
Biomarkers, Tumor/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Exome/genetics , Mutation/genetics , Precursor Cells, T-Lymphoid/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Aged , DNA Methyltransferase 3A , Epigenesis, Genetic , Female , Follow-Up Studies , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Prognosis , Receptor, Notch1/genetics , Young Adult , fms-Like Tyrosine Kinase 3/genetics
9.
Leuk Res ; 37(6): 647-56, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23522449

ABSTRACT

Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) has been identified as high-risk subgroup in acute T-cell lymphoblastic leukemia (T-ALL). To investigate the immature and myeloid nature of ETP-ALL we examined global microRNA (miRNA) expression in adult ETP-ALL. miRNA profiling of ETP-ALL (n=8), non-ETP T-ALL (n=6), and healthy controls was performed and results were validated in independent cohorts of 66 ETP-ALL and 111 non-ETP T-ALL using real-time RT-PCR. Furthermore, in vitro studies were performed on deregulated miRNAs in acute leukemia. We identified miR-221 and miR-222 as the most upregulated and six miRNAs (miR-151-3p, miR-19a, miR-20b, miR-342-3p, miR-363, and miR-576-3p) as downregulated in ETP-ALL compared to non-ETP T-ALL. In the validation cohorts, miR-221 and miR-222 were significantly upregulated in ETP-ALL, and miR-363 and miR-19a were downregulated in ETP-ALL. ETS1, downregulated in ETP-ALL, was identified as direct target of miR-222. In our in vitro studies miR-222 significantly inhibited proliferation, and caused cell cycle arrest and apoptosis in leukemic cells. In conclusion, our study revealed aberrant miRNA expression in ETP-ALL, with miR-221 and miR-222 as the most overexpressed miRNAs and implied a functional role for miR-222 in leukemic cells. Importantly, miR-222 may impact leukemogenesis by altering expression of the proto-oncogene ETS1 in acute leukemia.


Subject(s)
Gene Expression Profiling , MicroRNAs/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Aged , Case-Control Studies , Cell Line, Tumor , Cohort Studies , Gene Expression Regulation, Leukemic , Genetic Predisposition to Disease , Humans , Jurkat Cells , MicroRNAs/physiology , Microarray Analysis , Middle Aged , Precursor Cells, T-Lymphoid/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Mas , Young Adult
10.
Ann Hematol ; 92(6): 747-58, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23412561

ABSTRACT

Acute leukemias of ambiguous lineage represent a heterogeneous group of rare, poorly characterized leukemias with adverse outcome. No larger studies have yet performed a combined approach of molecular and clinical characterization of acute undifferentiated leukemia (AUL) and biphenotypic acute leukemia (BAL) in adults. Here we describe 16 adults with AUL and 26 with BAL and performed mutational as well as expression studies of genes with prognostic impact in acute leukemia (BAALC, ERG, MN1, WT1, and IGFBP7). AUL showed overexpression of these genes compared to T-lymphoblastic leukemia (T-ALL), B-precursor ALL, and to acute myeloid leukemia (AML). Genotype alterations were not detectable in AUL. BAL samples were characterized by frequent WT1 mutations (18 %) and BCR-ABL translocations (30 %). ALL-based treatment protocols induced complete remissions in 40 % and AML-like therapies in 22 % of AUL/BAL patients. The outcome in both groups was very poor; a long-term survival was only observed in patients undergoing allogeneic stem cell transplantation (SCT). Our findings indicate that AUL and BAL share important molecular and high-risk features of both myeloid and lymphoid leukemias. BAL patients exhibited genetic alterations, which can be targeted therapeutically. Importantly, ALL therapy might be more effective than AML protocols and AUL/BAL patients should be considered for allogeneic SCT.


Subject(s)
DNA, Neoplasm/genetics , Leukemia/pathology , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Lineage , Combined Modality Therapy , Female , Gene Expression Profiling , Genetic Association Studies , Humans , Immunophenotyping , Karyotyping , Leukemia/classification , Leukemia/drug therapy , Leukemia/genetics , Leukemia/surgery , Leukemia, Biphenotypic, Acute/drug therapy , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Biphenotypic, Acute/pathology , Leukemia, Biphenotypic, Acute/surgery , Lymphocytes/pathology , Male , Middle Aged , Mutation , Myeloid Cells/pathology , Neoplasm Proteins/genetics , Prognosis , Stem Cell Transplantation , Transplantation, Homologous , Treatment Outcome , Young Adult
11.
PLoS One ; 8(1): e53190, 2013.
Article in English | MEDLINE | ID: mdl-23359050

ABSTRACT

Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) has been identified as high-risk subgroup of acute T-lymphoblastic leukemia (T-ALL) with a high rate of FLT3-mutations in adults. To unravel the underlying pathomechanisms and the clinical course we assessed molecular alterations and clinical characteristics in a large cohort of ETP-ALL (n = 68) in comparison to non-ETP T-ALL adult patients. Interestingly, we found a high rate of FLT3-mutations in ETP-ALL samples (n = 24, 35%). Furthermore, FLT3 mutated ETP-ALL was characterized by a specific immunophenotype (CD2+/CD5-/CD13+/CD33-), a distinct gene expression pattern (aberrant expression of IGFBP7, WT1, GATA3) and mutational status (absence of NOTCH1 mutations and a low frequency, 21%, of clonal TCR rearrangements). The observed low GATA3 expression and high WT1 expression in combination with lack of NOTCH1 mutations and a low rate of TCR rearrangements point to a leukemic transformation at the pluripotent prothymocyte stage in FLT3 mutated ETP-ALL. The clinical outcome in ETP-ALL patients was poor, but encouraging in those patients with allogeneic stem cell transplantation (3-year OS: 74%). To further explore the efficacy of targeted therapies, we demonstrate that T-ALL cell lines transfected with FLT3 expression constructs were particularly sensitive to tyrosine kinase inhibitors. In conclusion, FLT3 mutated ETP-ALL defines a molecular distinct stem cell like leukemic subtype. These data warrant clinical studies with the implementation of FLT3 inhibitors in addition to early allogeneic stem cell transplantation for this high risk subgroup.


Subject(s)
Antineoplastic Agents/therapeutic use , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Adult , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
12.
Leuk Res ; 35(12): 1585-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21899885

ABSTRACT

Insulin-like growth factor (IGF) signaling plays an important role in many tumors and overexpression of IGF Binding Protein (IGFBP) 2 has been associated with adverse outcome in childhood leukemia. Here, we evaluated IGFBP2 mRNA expression and its prognostic implications in 99 adult acute myeloid leukemia (AML) patients by quantitative real-time RT-PCR. High IGFBP2 was associated with a high incidence of primary resistant disease (IGFBP2 high 65%, IGFBP2 low 32%; P=0.02) and was independently predictive for therapy resistance [OR 3.6 (95% CI 1.2-11); P=0.02] in multivariate analyses. Gene-expression profiling revealed an up-regulation of genes implicated in leukemogenesis (MYB, MEIS1, HOXB3, HOXA9) and genes associated with adverse outcome (ERG, WT1) in patients with high IGFBP2 expression. Thus, our data suggest a role of IGFBP2 and IGF signaling in chemoresistance of AML. Patients with high IGFBP2 expression might benefit from molecular therapies targeting the IGF pathway.


Subject(s)
Drug Resistance, Neoplasm/genetics , Insulin-Like Growth Factor Binding Protein 2/genetics , Leukemia, Myeloid, Acute/genetics , Adult , Age of Onset , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/physiology , Humans , Insulin-Like Growth Factor Binding Protein 2/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/epidemiology , Male , Middle Aged , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Somatomedins/metabolism , Somatomedins/physiology , Up-Regulation , Young Adult
13.
Cancer Sci ; 102(1): 253-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21040219

ABSTRACT

The important role of insulin-like growth factor binding protein 7 (IGFBP7) as a tumor suppressor in solid tumors has been revealed in several studies. Interestingly, in a recent study IGFBP7 was also shown to be aberrantly expressed in acute leukemia. Moreover, in acute T-lymphoblastic leukemia (T-ALL), high IGFBP7 expression predicts primary therapy resistance. In order to elucidate the mechanisms underlying aberrant IGFBP7 expression, we used pyrosequencing technology to investigate the DNA methylation of IGFBP7 in 109 T-ALL patient samples. Aberrant methylation was shown and hypomethylation was associated with an early immunophenotype and co-expression of the stem cell markers CD117 (P < 0.001) and CD34 (P < 0.001). In concordance, gene expression profiles of 86 T-ALL patients revealed upregulation of stem cell markers (CD34 and CD133) as well as genes associated with poor outcome and pathogenesis of leukemia (MN1, BAALC, FLT3) in the high IGFBP7 expression group. In conclusion, aberrant IGFBP7 expression is regulated by DNA methylation in acute leukemia. Hypomethylation of the gene is likely to characterize an immature and a more malignant subtype of the disease.


Subject(s)
DNA Methylation , Insulin-Like Growth Factor Binding Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Aged , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/analysis
14.
Haematologica ; 95(6): 942-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20435628

ABSTRACT

BACKGROUND: The role of the Wilms tumor 1 gene (WT1) in acute leukemias has been underscored by mutations found in acute myeloid leukemia identifying patients with inferior survival. Furthermore, aberrant expression of WT1 in acute myeloid leukemia was associated with an increased risk of relapse. No larger studies have performed a combined approach including WT1 mutation and expression analyses in acute T-lymphoblastic leukemia. DESIGN AND METHODS: We analyzed the WT1 mutations and the expression status in a total of 252 consecutive adult patients with newly diagnosed T-lymphoblastic leukemia, who were registered on the GMALL 06/99 and 07/03 protocols and had sufficient material available. The GMALL protocols included intensive chemotherapy as well as stem cell transplantation according to a risk-based model with indication for stem cell transplantation in first complete remission for early and mature T-lymphoblastic leukemia patients; patients with thymic T-lymphoblastic leukemia were allocated to a standard risk group and treated with intensive chemotherapy. RESULTS: Twenty of the 238 patients analyzed had WT1 mutations (WT1mut) in exon 7. WT1mut cases were characterized by immature features such as an early immunophenotype and higher WT1 expression. In thymic T-lymphoblastic leukemia, WT1mut patients had an inferior relapse-free survival compared to WT1 wild-type patients. T-lymphoblastic leukemia patients with aberrant WT1 expression (high or negative) showed a higher relapse rate and an inferior outcome compared to patients with intermediate WT1 expression. In the standard risk group of thymic T-lymphoblastic leukemia, aberrant WT1 expression was predictive for an inferior relapse-free survival as compared to patients with intermediate expression. In multivariate analysis, WT1 expression was of independent prognostic significance for relapse-free survival. CONCLUSIONS: WT1 mutations were associated with an inferior relapse-free survival in standard risk thymic T-lymphoblastic leukemia patients. Moreover, altered expression associated with inferior outcome also suggests a role of WT1 in T-lymphoblastic leukemia and the potential use of molecularly-based treatment stratification to improve outcome.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Wilms Tumor/physiology , Mutation/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Female , Humans , Male , Middle Aged , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Survival Rate/trends , Young Adult
15.
Blood ; 115(18): 3737-44, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20065290

ABSTRACT

Overexpression of BAALC is an adverse prognostic factor in adults with cytogenetically normal acute myeloid leukemia and T-cell acute lymphoblastic leukemia (ALL). Here, we analyzed the prognostic significance of BAALC in B-precursor ALL. BAALC MRNA expression was determined in 368 primary adult B-precursor ALL patients enrolled on the 06/99 and 07/03 GMALL trials. Patients were grouped into tertiles according to BAALC expression (T1-T3). Higher BAALC expression (T3 vs T2 vs T1) was associated with higher age (P < .001), a higher white blood cell count (P = .008), CD34 (P = .001), BCR-ABL (P < .001), and MLL-AF4 (P < .001). Higher BAALC expression predicted primary therapy resistance in the overall cohort (P = .002) and in the BCR-ABL(-) and MLL-AF4(-) subgroup (P = .01). In BCR-ABL(-) and MLL-AF4(-) patients, higher BAALC expression was associated with a shorter overall survival (OS; 5-year OS: T3, 38%; T2, 52%; T1, 70%; P = .004) and independently predicted OS in multivariate models (P = .03). Gene-expression profiling revealed an up-regulation of stem cell markers and genes involved in chemoresistance (TSPAN7 and LYN) in the high BAALC group. Thus, high BAALC expression is associated with an immature, chemoresistant leukemic phenotype and identifies patients with inferior OS. Determination of BAALC might contribute to risk assessment of molecularly undefined adult B-precursor ALL.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Neoplasm Proteins/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Female , Gene Expression Profiling , Humans , Immunophenotyping , Male , Middle Aged , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Survival Rate , Treatment Outcome , Young Adult
16.
Leuk Res ; 33(6): 817-22, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19108891

ABSTRACT

Expression of ERG is of prognostic significance in acute myeloid leukemia (AML) and T-lymphoblastic leukemia (T-ALL) pointing to its role in leukemogenesis. To unravel its transcriptional regulation we analyzed the expression of ERG specific isoforms. Expression of the two main isoforms ERG2 and ERG3 was found in AML and normal CD34+ cells, whereas T-ALL blasts only expressed ERG isoforms harboring exon 5 (ERG3) lacking expression of ERG2. Bisulfite sequencing revealed hypermethylation of a CpG island within the ERG2 promoter region in T-ALL. Treatment of the T-lymphoblastic cell line BE13 with decitabine led to re-expression of ERG2 and pyrosequencing showed concordant DNA hypomethylation, thus confirming a methylation regulated expression of ERG2. Moreover, the identification of a new ERG isoform (ERG3Deltaex12) suggests the association with different interaction partners and adds to the complexity of downstream pathways mediated by the expression of specific ERG transcripts in acute leukemia.


Subject(s)
Epigenesis, Genetic , Oncogene Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Isoforms/genetics , Trans-Activators/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , CpG Islands , DNA Methylation , DNA Primers , Decitabine , Exons , Polymerase Chain Reaction , Transcriptional Regulator ERG
17.
Int J Oncol ; 28(3): 755-66, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16465382

ABSTRACT

Histone deacetylase inhibitors (HDIs) are a promising new class of antineoplastic agents with the ability to induce apoptosis and growth arrest of cancer cells. In addition, HDIs have been suggested to enhance the anticancer efficacy of other therapeutic regimens, such as ionizing radiation (IR) or chemotherapy. The objective of this study was to evaluate the activity of HDIs against medulloblastoma cells when applied either as single agents or in combination with IR, cytostatics, or TRAIL. The HDIs, suberoyl anilide hydroxamic acid (SAHA), sodium butyrate, and trichostatin A, were examined for their effects on the medulloblastoma cell lines, DAOY and UW228-2. We found that treatment with HDIs induced the dissipation of mitochondrial membrane potential, activation of caspase-9 and -3 and, consequently, apoptotic cell death. Moreover, all three HDIs significantly enhanced the cytotoxic effects of IR in DAOY cells. Likewise, treatment with SAHA markedly augmented the cytotoxicity of etoposide, while it had no effect on vincristine-mediated cell death. HDIs also potently increased the killing efficiency of TRAIL. TRAIL-induced, but not SAHA-induced, cell killing could be prevented by the caspase-8 inhibitor, z-IEDT-fmk. We conclude that HDIs may be useful for the treatment of medulloblastoma as monotherapy and particularly when given in combination with IR, appropriate cytostatics, or TRAIL.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Acetylation/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/pharmacology , Butyrates/pharmacology , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Line, Tumor , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Synergism , Enzyme Activation/drug effects , Etoposide/pharmacology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Medulloblastoma/metabolism , Medulloblastoma/pathology , Medulloblastoma/physiopathology , Membrane Glycoproteins/pharmacology , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/pharmacology , Vincristine/pharmacology , Vorinostat
SELECTION OF CITATIONS
SEARCH DETAIL
...