Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 22(19)2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34639011

ABSTRACT

The prognosis of elderly AML patients is still poor due to chemotherapy resistance. The Hedgehog (HH) pathway is important for leukemic transformation because of aberrant activation of GLI transcription factors. MBZ is a well-tolerated anthelmintic that exhibits strong antitumor effects. Herein, we show that MBZ induced strong, dose-dependent anti-leukemic effects on AML cells, including the sensitization of AML cells to chemotherapy with cytarabine. MBZ strongly reduced intracellular protein levels of GLI1/GLI2 transcription factors. Consequently, MBZ reduced the GLI promoter activity as observed in luciferase-based reporter assays in AML cell lines. Further analysis revealed that MBZ mediates its anti-leukemic effects by promoting the proteasomal degradation of GLI transcription factors via inhibition of HSP70/90 chaperone activity. Extensive molecular dynamics simulations were performed on the MBZ-HSP90 complex, showing a stable binding interaction at the ATP binding site. Importantly, two patients with refractory AML were treated with MBZ in an off-label setting and MBZ effectively reduced the GLI signaling activity in a modified plasma inhibitory assay, resulting in a decrease in peripheral blood blast counts in one patient. Our data prove that MBZ is an effective GLI inhibitor that should be evaluated in combination to conventional chemotherapy in the clinical setting.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Mebendazole/pharmacology , Proteasome Endopeptidase Complex/metabolism , Tubulin Modulators/pharmacology , Zinc Finger Protein GLI1/metabolism , Case-Control Studies , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Proteolysis , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/metabolism , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/chemistry
2.
Oncoimmunology ; 8(12): e1674605, 2019.
Article in English | MEDLINE | ID: mdl-31741778

ABSTRACT

Immune checkpoints are intensively investigated as targets in cancer therapy. T-cell immunoreceptor with immunoglobulin (Ig) and ITIM domains (TIGIT) and its ligand poliovirus receptor (PVR) are recently emerging as novel promising targets in immunotherapy. Here, we show that high expression of PVR represents an independent prognostic marker being associated with poor outcome for breast cancer patients. Furthermore, PVR mRNA, as well as protein expression, is associated with more aggressive breast cancer subtypes such as HER2 positive and triple-negative breast cancer. In vitro, blocking TIGIT or PVR resulted in enhanced immune cell-mediated lysis of breast cancer cell lines SKBR-3, MDA-MB-231, MDA-MB-468, and BT549 and additionally increased the cytotoxic effects of a bispecific T cell engager BiTE® antibody construct targeting EGFR. Taken together, our data identify the immune checkpoint factor PVR as a novel prognostic marker in breast cancer and indicate that blocking the TIGIT-PVR axis might represent a novel therapeutic option for the treatment of breast cancer patients.

3.
Cancers (Basel) ; 11(11)2019 Oct 26.
Article in English | MEDLINE | ID: mdl-31717802

ABSTRACT

Leukemia-initiating cells reside within the bone marrow in specialized niches where they undergo complex interactions with their surrounding stromal cells. We have identified the actin-binding protein Plastin-3 (PLS3) as potential player within the leukemic bone marrow niche and investigated its functional role in acute myeloid leukemia. High expression of PLS3 was associated with a poor overall and event-free survival for AML patients. These findings were supported by functional in vitro and in vivo experiments. AML cells with a PLS3 knockdown showed significantly reduced colony numbers in vitro while the PLS3 overexpression variants resulted in significantly enhanced colony numbers compared to their respective controls. Furthermore, the survival of NSG mice transplanted with the PLS3 knockdown cells showed a significantly prolonged survival in comparison to mice transplanted with the control AML cells. Further studies should focus on the underlying leukemia-promoting mechanisms and investigate PLS3 as therapeutic target.

4.
Mamm Genome ; 29(11-12): 694-702, 2018 12.
Article in English | MEDLINE | ID: mdl-30132062

ABSTRACT

Avoiding immune surveillance and inducing a tumor-promoting inflammatory milieu found entry into the new generation of the hallmarks of cancer. Cancer cells hijack immune mechanisms which physiologically protect the body from the development of autoimmune diseases and excessive tissue damage during inflammation by downregulating immune responses. This is frequently achieved by upregulation of immune checkpoints. Therefore, the blocking of immune checkpoint ligand-receptor interactions can reinstall the immune systems capability to fight cancer cells as shown for CTLA4 and PD-1 inhibitors in a clinical setting. Newly described checkpoint antigens are currently under investigation in cancer immunotherapy. Preclinical data emphasize the immune checkpoint axis TIGIT-PVR/PVRL2 as very promising target. This axis includes additional receptors such as DNAM-1, CD96, and CD112R. In this review, we discuss the recent findings of the relevance of this complex receptor ligand system in hematologic and solid cancers. Emphasis is also laid on the discussion of potential combinations with other immunotherapeutic approaches.


Subject(s)
Nectins/immunology , Neoplasms/immunology , Receptors, Immunologic/genetics , Receptors, Virus/genetics , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Humans , Immunotherapy , Nectins/genetics , Neoplasms/genetics , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology , Receptors, Virus/immunology
5.
Oncogene ; 37(39): 5269-5280, 2018 09.
Article in English | MEDLINE | ID: mdl-29855615

ABSTRACT

Immune checkpoints are promising targets in cancer therapy. Recently, poliovirus receptor (PVR) and poliovirus receptor-related 2 (PVRL2) have been identified as novel immune checkpoints. In this investigation we show that acute myeloid leukemia (AML) cell lines and AML patient samples highly express the T-cell immunoreceptor with Ig and ITIM domains (TIGIT) ligands PVR and PVRL2. Using two independent patient cohorts, we could demonstrate that high PVR and PVRL2 expression correlates with poor outcome in AML. We show for the first time that antibody blockade of PVR or PVRL2 on AML cell lines or primary AML cells or TIGIT blockade on immune cells increases the anti-leukemic effects mediated by PBMCs or purified CD3+ cells in vitro. The cytolytic activity of the BiTE® antibody construct AMG 330 against leukemic cells could be further enhanced by blockade of the TIGIT-PVR/PVRL2 axis. This increased immune reactivity is paralleled by augmented secretion of Granzyme B by immune cells. Employing CRISPR/Cas9-mediated knockout of PVR and PVRL2 in MV4-11 cells, the cytotoxic effects of antibody blockade could be recapitulated in vitro. In NSG mice reconstituted with human T cells and transplanted with either MV4-11 PVR/PVRL2 knockout or wildtype cells, prolonged survival was observed for the knockout cells. This survival benefit could be further extended by treating the mice with AMG 330. Therefore, targeting the TIGIT-PVR/PVRL2 axis with blocking antibodies might represent a promising future therapeutic option in AML.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Nectins/metabolism , Receptors, Virus/metabolism , Animals , Antibodies, Bispecific/pharmacology , Biomarkers, Tumor/analysis , Heterografts , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Mice , Prognosis , Receptors, Immunologic/metabolism
6.
Oncotarget ; 8(17): 29187-29201, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28418873

ABSTRACT

Activation of the Hedgehog pathway has been implicated in the pathogenesis of several tumor types including myeloid leukemia. Previously we demonstrated that overexpression of Hedgehog downstream mediators GLI1/2 confers an adverse prognosis to patients with acute myeloid leukemia (AML) and is correlated with a FLT3 mutated status. To analyze a possible non-canonical activation of the Hedgehog pathway via FLT3 and PI3K, we performed blocking experiments utilizing inhibitors for FLT3 (sunitinib), PI3K (PF-04691502) and GLI1/2 (GANT61) in FLT3-mutated and FLT3 wildtype AML cell lines and primary blasts. Combination of all three compounds had stronger anti-leukemic effects in FLT3-mutated compared to FLT3 wildtype AML cells in vitro. Interestingly, the colony growth of normal CD34+ cells from healthy donors was not impeded by the triple inhibitor combination possibly opening a therapeutic window for the clinical use of inhibitor combinations. Besides, combined treatment with sunitinib, PF-04691502 and GANT61 significantly prolonged the survival of mice transplanted with FLT3-mutated MV4-11 cells compared to the single agent treatments. Furthermore, the inhibition of FLT3 and PI3K resulted in reduced GLI protein expression and promotor activity in FLT3-mutated but not in FLT3 wildtype AML cell lines in western blotting and GLI1/2 promoter assays supporting our hypothesis of non-canonical GLI activation via FLT3.In summary, FLT3-mutated in contrast to FLT3 wildtype cells or normal human hematopoietic progenitor cells are exquisitely sensitive to combined inhibition by FLT3, PI3K and GLI1/2 overcoming some of the limitations of current FLT3 directed therapy in AML. The development of GLI1/2 inhibitors is highly desirable.


Subject(s)
Hedgehog Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Zinc Finger Protein GLI1/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Apoptosis , Cell Proliferation , Disease Models, Animal , Female , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Signal Transduction
7.
Eur J Haematol ; 97(3): 239-44, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26613208

ABSTRACT

OBJECTIVES: Leukaemia initiating cells reside within specialised niches in the bone marrow where they undergo complex interactions with different stromal cell types. The bone marrow niche is characterised by a low oxygen content resulting in high expression of hypoxia-inducible factor 1 α in leukaemic cells conferring a negative prognosis to patients with acute myeloid leukaemia (AML). METHODS AND RESULTS: In the current study, we investigated the impact of hypoxic vs. normoxic conditions on the sensitivity of AML cell lines and primary AML blasts to cytarabine. AML cells cultured under 6% oxygen were significantly more resistant against cytarabine compared to cells cultured under normoxic conditions in proliferation and colony-formation assays. Interestingly upon cultivation under hypoxia, the expression of the cytarabine-activating enzyme deoxycytidine kinase was downregulated in all analysed AML cell lines and primary AML samples representing a possible mechanism for resistance to chemotherapy. Furthermore, the downregulation of deoxycytidine kinase could be associated with hypoxia-inducible factor 1 α as treatment with its inhibitor BAY87-2243 hampered the downregulation of deoxycytidine kinase expression under hypoxic conditions. CONCLUSIONS: In conclusion, our data reveal that hypoxia-induced downregulation of deoxycytidine kinase represents one stroma-cell-independent mechanism of drug resistance to cytarabine in acute myeloid leukaemia.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Cytarabine/therapeutic use , Deoxycytidine Kinase/metabolism , Drug Resistance, Neoplasm , Hypoxia/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytarabine/pharmacology , Deoxycytidine Kinase/genetics , Down-Regulation , Gene Expression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism
8.
BMC Cancer ; 15: 896, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26560478

ABSTRACT

BACKGROUND: Disseminated tumor cells (DTCs) can be detected using ultrasensitive immunocytochemical assays and their presence in the bone marrow can predict the subsequent occurrence of overt metastasis formation and metastatic relapse. Using expression profiling on early stage primary breast tumors, low IRX2 expression was previously shown to be associated with the presence of DTCs in the bone marrow, suggesting a possible role of IRX2 in the early steps of metastasis formation. The purpose of this study is to gain insights into the significance of IRX2 protein function in the progression of breast cancer. METHODS: To assess the physiological relevance of IRX2 in breast cancer, we evaluated IRX2 expression in a large breast cancer cohort (n = 1992). Additionally, constitutive IRX2 over expression was established in BT-549 and Hs578T breast cancer cell lines. Subsequently we analyzed whether IRX2 overexpression effects chemokine secretion and cellular motility of these cells. RESULTS: Low IRX2 mRNA expression was found to correlate with high tumor grade, positive lymph node status, negative hormone receptor status, and basal type of primary breast tumors. Also in cell lines low IRX2 expression was associated with mainly basal breast cancer cell lines. The functional studies show that overexpression of the IRX2 transcription factor in basal cell lines suppressed secretion of the pro-metastatic chemokines and inhibited cellular motility but did not influence cell proliferation. CONCLUSION: Our results imply that the IRX2 transcription factor might represent a novel metastasis associated protein that acts as a negative regulator of cellular motility and as a repressor of chemokine expression. Loss of IRX2 expression could therefore contribute to early hematogenous dissemination of breast cancer by sustaining chemokine secretion and enabling mobilization of tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement/physiology , Chemokines/metabolism , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Transcription Factors/metabolism , Breast Neoplasms/physiopathology , Cell Line, Tumor , Disease Progression , Female , Gene Expression Profiling , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Humans , Neoplasm Metastasis , Neoplasm Recurrence, Local/pathology , Transcription Factors/genetics
9.
Lung ; 193(4): 567-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25926293

ABSTRACT

Gremlin-1, an intrinsic antagonist of bone morphogenetic protein (BMP) signaling, has been implicated in the pathophysiology of pulmonary arterial hypertension (PAH). However, it is unknown whether gremlin-1 can be detected in the circulation of PAH patients and whether it is associated with patients' functional status and outcome. With a mean level of 242 ± 24 ng/ml, gremlin-1 levels of 31 PAH patients were significantly elevated compared to 151 ± 18 ng/ml in 15 age- and gender-matched healthy subject (p = 0.016). In PAH patients, increasing gremlin-1 levels correlated with N-terminal prohormone of brain natriuretic peptide levels (r = 0.608, p < 0.001) and inversely with the 6-minute walking distance (r = -0.412, p = 0.029). Furthermore, gremlin-1 significantly stratified survival in PAH patients (p = 0.015). Gremlin-1 may represent a new biomarker for PAH which can be linked directly to the underlying pathomechanism. Elevated levels of gremlin-1 are associated with patients' functional status and survival, thus gremlin-1 neutralization could represent a potential therapeutic strategy to increase BMPR2 signaling.


Subject(s)
Hypertension, Pulmonary/blood , Intercellular Signaling Peptides and Proteins/blood , Aged , Biomarkers/blood , Bone Morphogenetic Proteins/metabolism , Case-Control Studies , Exercise Test , Female , Glomerular Filtration Rate , Humans , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Pulmonary Artery , Signal Transduction , Survival Rate , Walking/physiology
10.
Clin Cancer Res ; 21(10): 2388-98, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25745035

ABSTRACT

PURPOSE: The Hedgehog pathway plays an important role in stem-cell biology and malignant transformation. Therefore, we investigated the expression and prognostic impact of Hedgehog pathway members in acute myeloid leukemia (AML). EXPERIMENTAL DESIGN: Pretreatment samples from 104 newly diagnosed AML patients (AMLSG 07-04 trial) were analyzed by qPCR, and expression of Hedgehog family members was correlated with clinical outcome. Inhibition of GLI by GANT61 or shRNA was investigated in AML cells in vitro and in vivo. RESULTS: Expression of receptors Smoothened and Patched-1 and their downstream mediators, GLI1, GLI2, and GLI3, was found in AML patients in contrast to Hedgehog ligands. GLI2 expression had a significant negative influence on event-free survival (EFS), relapse-free survival (RFS), and overall survival (OS; P = 0.037, 0.026, and 0.013, respectively) and was correlated with FLT3 mutational status (P < 0.001). Analysis of a second, independent patient cohort confirmed the negative impact of GLI2 on EFS and OS (P = 0.007 and 0.003, respectively; n = 290). Within this cohort, GLI1 had a negative prognostic impact (P < 0.001 for both EFS and OS). Although AML cells did not express Hedgehog ligands by qPCR, AML patients had significantly increased Desert Hedgehog (DHH) plasma levels compared with healthy subjects (P = 0.002), in whom DHH was presumably provided by bone marrow niche cells. Moreover, the GLI inhibitor GANT61 or knockdown of GLI1/2 by shRNA caused antileukemic effects, including induction of apoptosis, reduced proliferation, and colony formation in AML cells, and a survival benefit in mice. CONCLUSIONS: GLI expression is a negative prognostic factor and might represent a novel druggable target in AML.


Subject(s)
Biomarkers, Tumor/metabolism , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/metabolism , Adolescent , Adult , Animals , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Line, Tumor , DNA Mutational Analysis , Disease-Free Survival , Female , Hedgehog Proteins/metabolism , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Prognosis , Proportional Hazards Models , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transcription Factors/genetics , Treatment Outcome , Young Adult , Zinc Finger Protein GLI1
11.
PLoS One ; 9(8): e104742, 2014.
Article in English | MEDLINE | ID: mdl-25116393

ABSTRACT

BACKGROUNDS: The Loeys-Dietz syndrome (LDS) is an inherited connective tissue disorder caused by mutations in the transforming growth factor ß (TGF-ß) receptors TGFBR1 or TGFBR2. Most patients with LDS develop severe aortic aneurysms resulting in early need of surgical intervention. In order to gain further insight into the pathophysiology of the disorder, we investigated circulating outgrowth endothelial cells (OEC) from the peripheral blood of LDS patients from a cohort of 23 patients including 6 patients with novel TGF-ß receptor mutations. METHODS AND RESULTS: We performed gene expression profiling of OECs using microarray analysis followed by quantitative PCR for verification of gene expression. Compared to OECs of age- and sex-matched healthy controls, OECs isolated from three LDS patients displayed altered expression of several genes belonging to the TGF-ß pathway, especially those affecting bone morphogenic protein (BMP) signalling including BMP2, BMP4 and BMPR1A. Gene expression of BMP antagonist Gremlin-1 (GREM1) showed the most prominent up-regulation. This increase was confirmed at the protein level by immunoblotting of LDS-OECs. In immunohistochemistry, abundant Gremlin-1 protein expression could be verified in endothelial cells as well as smooth muscle cells within the arterial media. Furthermore, Gremlin-1 plasma levels of LDS patients were significantly elevated compared to healthy control subjects. CONCLUSIONS: These findings open new avenues in the understanding of the pathogenesis of Loeys-Dietz syndrome and the development of new diagnostic serological methods for early disease detection.


Subject(s)
Endothelial Cells/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Loeys-Dietz Syndrome/physiopathology , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Adult , Aorta/metabolism , Aorta/pathology , Bone Morphogenetic Protein 2/biosynthesis , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors, Type I/biosynthesis , Bone Morphogenetic Protein Receptors, Type I/genetics , Cell Proliferation/genetics , Early Diagnosis , Female , Gene Expression Profiling , Humans , Latent TGF-beta Binding Proteins/biosynthesis , Loeys-Dietz Syndrome/diagnosis , Loeys-Dietz Syndrome/genetics , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/biosynthesis , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL