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










Publication year range
1.
Front Immunol ; 14: 1160116, 2023.
Article in English | MEDLINE | ID: mdl-37304285

ABSTRACT

Introduction: The ability to modulate and enhance the anti-tumor immune responses is critical in developing novel therapies in cancer. The Tumor Necrosis Factor (TNF) Receptor Super Family (TNFRSF) are potentially excellent targets for modulation which result in specific anti-tumor immune responses. CD40 is a member of the TNFRSF and several clinical therapies are under development. CD40 signaling plays a pivotal role in regulating the immune system from B cell responses to myeloid cell driven activation of T cells. The CD40 signaling axis is well characterized and here we compare next generation HERA-Ligands to conventional monoclonal antibody based immune modulation for the treatment of cancer. Methods & results: HERA-CD40L is a novel molecule that targets CD40 mediated signal transduction and demonstrates a clear mode of action in generating an activated receptor complex via recruitment of TRAFs, cIAP1, and HOIP, leading to TRAF2 phosphorylation and ultimately resulting in the enhanced activation of key inflammatory/survival pathway and transcription factors such asNFkB, AKT, p38, ERK1/2, JNK, and STAT1 in dendritic cells. Furthermore, HERA-CD40L demonstrated a strong modulation of the tumor microenvironment (TME) via the increase in intratumoral CD8+ T cells and the functional switch from pro-tumor macrophages (TAMs) to anti-tumor macrophages that together results in a significant reduction of tumor growth in a CT26 mouse model. Furthermore, radiotherapy which may have an immunosuppressive modulation of the TME, was shown to have an immunostimulatory effect in combination with HERA-CD40L. Radiotherapy in combination with HERA-CD40L treatment resulted in an increase in detected intratumoral CD4+/8+ T cells compared to RT alone and, additionally, the repolarization of TAMs was also observed, resulting in an inhibition of tumor growth in a TRAMP-C1 mouse model. Discussion: Taken together, HERA-CD40L resulted in activating signal transduction mechanisms in dendritic cells, resulting in an increase in intratumoral T cells and manipulation of the TME to be pro-inflammatory, repolarizing M2 macrophages to M1, enhancing tumor control.


Subject(s)
CD40 Ligand , Neoplasms , Animals , Mice , CD40 Antigens , Antigen-Presenting Cells , Macrophages , Neoplasms/radiotherapy , Disease Models, Animal , Tumor Microenvironment
2.
Cell Rep ; 29(8): 2295-2306.e6, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31747602

ABSTRACT

The death receptor CD95 is expressed in every cancer cell, thus providing a promising tool to target cancer. Activation of CD95 can, however, lead to apoptosis or proliferation. Yet the molecular determinants of CD95's mode of action remain unclear. Here, we identify an optimal distance between CD95Ligand molecules that enables specific clustering of receptor-ligand pairs, leading to efficient CD95 activation. Surprisingly, efficient CD95 activation leads to apoptosis in cancer cells in vitro and increased tumor growth in vivo. We show that allowing a 3D aggregation of cancer cells in vitro switches the apoptotic response to proliferation. Indeed, we demonstrate that the absence or presence of cell-cell contacts dictates the cell response to CD95. Cell contacts increase global levels of phosphorylated tyrosines, including CD95's tyrosine. A tyrosine-to-alanine CD95 mutant blocks proliferation in cells in contact. Our study sheds light into the regulatory mechanism of CD95 activation that can be further explored for anti-cancer therapies.


Subject(s)
Protein-Tyrosine Kinases/metabolism , fas Receptor/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Communication/genetics , Cell Communication/physiology , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Humans , Phosphorylation/genetics , Phosphorylation/physiology , Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Signal Transduction/physiology , fas Receptor/genetics
3.
J Immunother Cancer ; 7(1): 191, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324216

ABSTRACT

BACKGROUND: Glucocorticoid-induced TNFR-related protein (TNFRSF18, GITR, CD357), expressed by T cells, and its ligand (TNFSF18, GITRL), expressed by myeloid populations, provide co-stimulatory signals that boost T cell activity. Due to the important role that GITR plays in regulating immune functions, agonistic stimulation of GITR is a promising therapeutic concept. Multiple strategies to induce GITR signaling have been investigated. The limited clinical efficacy of antibody-based GITR agonists results from structural and functional characteristics of antibodies that are unsuitable for stimulating the well-defined trimeric members of the TNFRSF. METHODS: To overcome limitations of antibody-based TNFRSF agonists, we have developed HERA-GITRL, a fully human hexavalent TNF receptor agonist (HERA) targeting GITR and mimicking the natural signaling concept. HERA-GITRL is composed of a trivalent but single-chain GITRL-receptor-binding-domain (scGITRL-RBD) unit fused to an IgG1 derived silenced Fc-domain serving as dimerization scaffold. A specific mouse surrogate, mmHERA-GITRL, was also generated to examine in vivo activity in respective mouse tumor models. RESULTS: For functional characterization of HERA-GITRL in vitro, human immune cells were isolated from healthy-donor blood and stimulated with anti-CD3 antibody in the presence of HERA-GITRL. Consistently, HERA-GITRL increased the activity of T cells, including proliferation and differentiation, even in the presence of regulatory T cells. In line with these findings, mmHERA-GITRL enhanced antigen-specific clonal expansion of both CD4+ (OT-II) and CD8+ (OT-I) T cells in vivo while having no effect on non-specific T cells. In addition, mmHERA-GITRL showed single-agent anti-tumor activity in two subcutaneous syngeneic colon cancer models (CT26wt and MC38-CEA). Importantly, this activity is independent of its FcγR-binding functionality, as both mmHERA-GITRL with a functional Fc- and a silenced Fc-domain showed similar tumor growth inhibition. Finally, in a direct in vitro comparison to a bivalent clinical benchmark anti-GITR antibody and a trivalent GITRL, only the hexavalent HERA-GITRL showed full biological activity independent of additional crosslinking. CONCLUSION: In this manuscript, we describe the development of HERA-GITRL, a true GITR agonist with a clearly defined mechanism of action. By clustering six receptor chains in a spatially well-defined manner, HERA-GITRL induces potent agonistic activity without being dependent on additional FcγR-mediated crosslinking.


Subject(s)
Receptors, Tumor Necrosis Factor/agonists , Single-Chain Antibodies/administration & dosage , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factors/chemistry , Animals , Cell Line, Tumor , Humans , Immunoglobulin Fc Fragments/immunology , Lymphocyte Activation , Macaca fascicularis , Mice , Recombinant Fusion Proteins/immunology , Signal Transduction , Single-Chain Antibodies/immunology , Tumor Necrosis Factors/metabolism
4.
Pharmaceutics ; 11(4)2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30939793

ABSTRACT

The protein therapeutic and CD95L inhibitor asunercept is currently under clinical investigation for the treatment of glioblastoma and myelodysplastic syndrome. The purpose of this study was to predict the asunercept pharmacokinetics in children and to give dose recommendations for its first use in pediatric glioblastoma patients. A physiologically-based pharmacokinetic (PBPK) model of asunercept in healthy and diseased adults was successfully developed using the available clinical Phase I and Phase II study data. This model was then extrapolated to different pediatric populations, to predict the asunercept exposure in children and to find equivalent starting doses. Simulation of the asunercept serum concentration-time curves in children between 1⁻18 years of age shows that a dosing regimen based on body weight results in a similar asunercept steady-state exposure in all patients (pediatric or adult) above 12 years of age. For children between 1⁻12 years, higher doses per kg body weight are recommended, with the highest dose for the very young patients. Translational PBPK modeling is strongly encouraged by regulatory agencies to help with the initial dose selection for pediatric trials. To our knowledge, this is the first report of pediatric PBPK to support the dose selection of a therapeutic protein before its administration to children.

5.
Front Oncol ; 8: 387, 2018.
Article in English | MEDLINE | ID: mdl-30298117

ABSTRACT

Tumor necrosis factor receptor superfamily member 7 (TNFRSF7, CD27), expressed primarily by T cells, and its ligand CD27L (TNFSF7, CD70) provide co-stimulatory signals that boost T cell activation, differentiation, and survival. Agonistic stimulation of CD27 is therefore a promising therapeutic concept in immuno-oncology intended to boost and sustain T cell driven anti-tumor responses. Endogenous TNFSF/TNFRSF-based signal transmission is a structurally well-defined event that takes place during cell-to-cell-based contacts. It is well-established that the trimeric-trivalent TNFSF-receptor binding domain (TNFSF-RBD) exposed by the conducting cell and the resulting multi-trimer-based receptor clustering on the receiving cell are essential for agonistic signaling. Therefore, we have developed HERA-CD27L, a novel hexavalent TNF receptor agonist (HERA) targeting CD27 and mimicking the natural signaling concept. HERA-CD27L is composed of a trivalent but single-chain CD27L-receptor-binding-domain (scCD27L-RBD) fused to an IgG1 derived silenced Fc-domain serving as dimerization scaffold. The hexavalent agonist significantly boosted antigen-specific T cell responses while having no effect on non-specific T cells and was superior over stabilized recombinant trivalent CD27L. In addition, HERA-CD27L demonstrated potent single-agent anti-tumor efficacy in two different syngeneic tumor models, MC38-CEA and CT26wt. Furthermore, the combination of HERA-CD27L and an anti-PD-1 antibody showed additive anti-tumor effects highlighting the importance of both T cell activation and checkpoint inhibition in anti-tumor immunity. In this manuscript, we describe the development of HERA-CD27L, a true CD27 agonist with a clearly defined forward-signaling mechanism of action.

6.
J Immunother ; 41(9): 385-398, 2018.
Article in English | MEDLINE | ID: mdl-30273198

ABSTRACT

CD40 ligand (TNFSF5/CD154/CD40L), a member of the tumor necrosis factor (TNF) superfamily is a key regulator of the immune system. The cognate receptor CD40 (TNFRSF5) is expressed broadly on antigen-presenting cells and many tumor types, and has emerged as an attractive target for immunologic cancer treatment. Most of the CD40 targeting drugs in clinical development are antibodies which display some disadvantages: their activity typically depends on Fcγ receptor-mediated crosslinking, and depletion of CD40-expressing immune cells by antibody-dependent cellular cytotoxicity compromises an efficient antitumor response. To overcome the inadequacies of antibodies, we have developed the hexavalent receptor agonist (HERA) Technology. HERA compounds are fusion proteins composed of 3 receptor binding domains in a single chain arrangement, linked to an Fc-silenced human IgG1 thereby generating a hexavalent molecule. HERA-CD40L provides efficient receptor agonism on CD40-expressing cells and, importantly, does not require FcγR-mediated crosslinking. Strong activation of NFκB signaling was observed upon treatment of B cells with HERA-CD40L. Monocyte treatment with HERA-CD40L promoted differentiation towards the M1 spectrum and repolarization of M2 spectrum macrophages towards the M1 spectrum phenotype. Treatment of in vitro co-cultures of T and B cells with HERA-CD40L-triggered robust antitumor activation of T cells, which depended upon direct interaction with B cells. In contrast, bivalent anti-CD40 antibodies and trivalent soluble CD40L displayed weak activity which critically depended on crosslinking. In vivo, a murine surrogate of HERA-CD40L-stimulated clonal expansion of OT-I-specific murine CD8 T cells and showed single agent antitumor activity in the CD40 syngeneic MC38-CEA mouse model of colorectal cancer, suggesting an involvement of the immune system in controlling tumor growth. We conclude that HERA-CD40L is able to establish robust antitumor immune responses both in vitro and in vivo.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CD40 Antigens/antagonists & inhibitors , CD40 Ligand/pharmacology , Immunoglobulin G/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Macrophages/cytology , Macrophages/drug effects , Mice, Inbred C57BL , NF-kappa B/immunology
7.
Acta Neuropathol ; 132(3): 391-411, 2016 09.
Article in English | MEDLINE | ID: mdl-26910103

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating progressive neurodegenerative disease affecting primarily the upper and lower motor neurons. A common feature of all ALS cases is a well-characterized neuroinflammatory reaction within the central nervous system (CNS). However, much less is known about the role of the peripheral immune system and its interplay with CNS resident immune cells in motor neuron degeneration. Here, we characterized peripheral monocytes in both temporal and spatial dimensions of ALS pathogenesis. We found the circulating monocytes to be deregulated in ALS regarding subtype constitution, function and gene expression. Moreover, we show that CNS infiltration of peripheral monocytes correlates with improved motor neuron survival in a genetic ALS mouse model. Furthermore, application of human immunoglobulins or fusion proteins containing only the human Fc, but not the Fab antibody fragment, increased CNS invasion of peripheral monocytes and delayed the disease onset. Our results underline the importance of peripheral monocytes in ALS pathogenesis and are in agreement with a protective role of monocytes in the early phase of the disease. The possibility to boost this beneficial function of peripheral monocytes by application of human immunoglobulins should be evaluated in clinical trials.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Central Nervous System/metabolism , Leukocytes, Mononuclear/metabolism , Monocytes/metabolism , Mononuclear Phagocyte System/metabolism , Motor Neurons/pathology , Spinal Cord/pathology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Humans , Mice, Inbred C57BL , Spinal Cord/metabolism
8.
J Exp Med ; 212(4): 469-80, 2015 Apr 06.
Article in English | MEDLINE | ID: mdl-25779632

ABSTRACT

Neuroinflammation is increasingly recognized as a hallmark of neurodegeneration. Activated central nervous system-resident microglia and infiltrating immune cells contribute to the degeneration of dopaminergic neurons (DNs). However, how the inflammatory process leads to neuron loss and whether blocking this response would be beneficial to disease progression remains largely unknown. CD95 is a mediator of inflammation that has also been proposed as an apoptosis inducer in DNs, but previous studies using ubiquitous deletion of CD95 or CD95L in mouse models of neurodegeneration have generated conflicting results. Here we examine the role of CD95 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)-induced neurodegeneration using tissue-specific deletion of CD95 or CD95L. We show that DN death is not mediated by CD95-induced apoptosis because deletion of CD95 in DNs does not influence MPTP-induced neurodegeneration. In contrast, deletion of CD95L in peripheral myeloid cells significantly protects against MPTP neurotoxicity and preserves striatal dopamine levels. Systemic pharmacological inhibition of CD95L dampens the peripheral innate response, reduces the accumulation of infiltrating myeloid cells, and efficiently prevents MPTP-induced DN death. Altogether, this study emphasizes the role of the peripheral innate immune response in neurodegeneration and identifies CD95 as potential pharmacological target for neurodegenerative disease.


Subject(s)
Apoptosis/immunology , Dopaminergic Neurons/immunology , Fas Ligand Protein/immunology , Immunity, Innate , Myeloid Cells/immunology , Parkinsonian Disorders/immunology , Animals , Apoptosis/genetics , Corpus Striatum/immunology , Corpus Striatum/pathology , Dopamine/genetics , Dopamine/immunology , Dopaminergic Neurons/pathology , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/genetics , Inflammation , Mice , Mice, Knockout , Myeloid Cells/pathology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , fas Receptor/immunology
9.
Mol Cancer Ther ; 12(12): 2735-47, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24101228

ABSTRACT

Cancer cells can be specifically driven into apoptosis by activating Death-receptor-4 (DR4; TRAIL-R1) and/or Death-receptor-5 (DR5; TRAIL-R2). Albeit showing promising preclinical efficacy, first-generation protein therapeutics addressing this pathway, especially agonistic anti-DR4/DR5-monoclonal antibodies, have not been clinically successful to date. Due to their bivalent binding mode, effective apoptosis induction by agonistic TRAIL-R antibodies is achieved only upon additional events leading to antibody-multimer formation. The binding of these multimers to their target subsequently leads to effective receptor-clustering on cancer cells. The research results presented here report on a new class of TRAIL-receptor agonists overcoming this intrinsic limitation observed for antibodies in general. The main feature of these agonists is a TRAIL-mimic consisting of three TRAIL-protomer subsequences combined in one polypeptide chain, termed the single-chain TRAIL-receptor-binding domain (scTRAIL-RBD). In the active compounds, two scTRAIL-RBDs with three receptor binding sites each are brought molecularly in close proximity resulting in a fusion protein with a hexavalent binding mode. In the case of APG350-the prototype of this engineering concept-this is achieved by fusing the Fc-part of a human immunoglobulin G1 (IgG1)-mutein C-terminally to the scTRAIL-RBD polypeptide, thereby creating six receptor binding sites per drug molecule. In vitro, APG350 is a potent inducer of apoptosis on human tumor cell lines and primary tumor cells. In vivo, treatment of mice bearing Colo205-xenograft tumors with APG350 showed a dose-dependent antitumor efficacy. By dedicated muteins, we confirmed that the observed in vivo efficacy of the hexavalent scTRAIL-RBD fusion proteins is-in contrast to agonistic antibodies-independent of FcγR-based cross-linking events.


Subject(s)
Antineoplastic Agents/pharmacology , Peptide Fragments/pharmacology , Receptors, IgG/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Mice , Models, Biological , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Xenograft Model Antitumor Assays
10.
Int Immunopharmacol ; 13(1): 93-100, 2012 May.
Article in English | MEDLINE | ID: mdl-22446296

ABSTRACT

APG101 is a glycosylated fusion protein consisting of the extracellular domain of human CD95 (APO-1/Fas) and the Fc domain of human IgG1. Administration of APG101 blocks the interaction between CD95 and its cognate ligand CD95L, thereby inhibiting various pathways involved in e.g. proliferation, migration, differentiation and apoptosis induction. The safety and tolerability of ascending single doses of intravenously applied APG101 was examined in a randomized, double-blind, placebo-controlled, mono-centre "first in man" dose escalation study in 34 healthy male volunteers. Pharmacokinetics and pharmacodynamics were also assessed. The maximum serum concentration of 460 µg/ml was achieved following 1h infusion of the highest dose of 20 mg/kg. The systemic clearance was low (0.4 to 0.5 ml/hkg). Mean terminal elimination half-life was 12 to 15 days. Two patients suffering from malignant glioma received APG101 intravenously under compassionate use conditions. They received doses ranging from 5mg to 600 mg APG101. No adverse events and no clinical significant changes in laboratory parameters related to APG101 were reported. The presence of anti-drug-antibodies (ADA) was investigated and revealed no detectable levels of ADA. Overall, single ascending doses of APG101 up to 20 mg/kgbody weight (bw) administered as infusion over 1h were considered as safe and well tolerated in healthy volunteers. After the application of multiple doses of 400 mg in two glioma patients, steady state for APG101 seemed to be reached. These results support further clinical evaluation of APG101 at a dose of 400 mg per week in glioblastoma patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Immunoglobulin Fc Fragments/therapeutic use , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , fas Receptor/therapeutic use , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Apoptosis/drug effects , Brain Neoplasms/blood , Brain Neoplasms/pathology , Compassionate Use Trials , Dose-Response Relationship, Drug , Double-Blind Method , Glioma/blood , Glioma/pathology , Humans , Immunoglobulin Fc Fragments/adverse effects , Immunoglobulin Fc Fragments/blood , Immunoglobulin G/adverse effects , Immunoglobulin G/blood , Infusions, Intravenous , Magnetic Resonance Imaging , Male , Middle Aged , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/blood , Young Adult , fas Receptor/adverse effects , fas Receptor/blood
11.
Immunity ; 32(2): 240-52, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20153221

ABSTRACT

Injury to the central nervous system initiates an uncontrolled inflammatory response that results in both tissue repair and destruction. Here, we showed that, in rodents and humans, injury to the spinal cord triggered surface expression of CD95 ligand (CD95L, FasL) on peripheral blood myeloid cells. CD95L stimulation of CD95 on these cells activated phosphoinositide 3-kinase (PI3K) and metalloproteinase-9 (MMP-9) via recruitment and activation of Syk kinase, ultimately leading to increased migration. Exclusive CD95L deletion in myeloid cells greatly decreased the number of neutrophils and macrophages infiltrating the injured spinal cord or the inflamed peritoneum after thioglycollate injection. Importantly, deletion of myeloid CD95L, but not of CD95 on neural cells, led to functional recovery of spinal injured animals. Our results indicate that CD95L acts on peripheral myeloid cells to induce tissue damage. Thus, neutralization of CD95L should be considered as a means to create a controlled beneficial inflammatory response.


Subject(s)
Cell Movement , Fas Ligand Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid Cells/metabolism , Peritonitis/immunology , Protein-Tyrosine Kinases/metabolism , Animals , Cells, Cultured , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Humans , Inflammation , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/pathology , Peritoneum/immunology , Peritoneum/pathology , Peritonitis/chemically induced , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Spinal Cord/immunology , Spinal Cord/pathology , Syk Kinase , Thioglycolates/administration & dosage
13.
Cell Stem Cell ; 5(2): 178-90, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19664992

ABSTRACT

Adult neurogenesis persists in the subventricular zone and the dentate gyrus and can be induced upon central nervous system injury. However, the final contribution of newborn neurons to neuronal networks is limited. Here we show that in neural stem cells, stimulation of the "death receptor" CD95 does not trigger apoptosis but unexpectedly leads to increased stem cell survival and neuronal specification. These effects are mediated via activation of the Src/PI3K/AKT/mTOR signaling pathway, ultimately leading to a global increase in protein translation. Induction of neurogenesis by CD95 was further confirmed in the ischemic CA1 region, in the naive dentate gyrus, and after forced expression of CD95L in the adult subventricular zone. Lack of hippocampal CD95 resulted in a reduction in neurogenesis and working memory deficits. Following global ischemia, CD95-mediated brain repair rescued behavioral impairment. Thus, we identify the CD95/CD95L system as an instructive signal for ongoing and injury-induced neurogenesis.


Subject(s)
Adult Stem Cells/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Fas Ligand Protein/metabolism , Neurogenesis/physiology , fas Receptor/metabolism , Adult Stem Cells/transplantation , Animals , Brain Ischemia/therapy , Female , Gene Expression/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Protein Kinases/metabolism , Signal Transduction/physiology , Stem Cell Transplantation , TOR Serine-Threonine Kinases
15.
Bioorg Med Chem Lett ; 19(15): 4201-3, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19515557

ABSTRACT

Modifications of DPP-4 inhibitor 5, that was discovered by structure based design, are described and structure-activity relationships discussed. With analogue 7k one of the most potent non-covalent inhibitors of DPP-4 reported to date (IC(50)=0.38nM) was discovered. X-ray structure of inhibitor 7k bound to DPP-4 revealed a hydrogen bonding interaction with Q553. First successful efforts in balancing overall properties, as demonstrated by improved metabolic stability, highlight the potential of this series.


Subject(s)
Amides/chemical synthesis , Aminobutyrates/chemistry , Dipeptidyl-Peptidase IV Inhibitors , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Microsomes, Liver/drug effects , Sulfonamides/chemical synthesis , Amides/pharmacology , Animals , Chemistry, Pharmaceutical/methods , Crystallography, X-Ray/methods , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Design , Glucagon-Like Peptide 1/antagonists & inhibitors , Hydrogen Bonding , Inhibitory Concentration 50 , Microsomes, Liver/metabolism , Rats , Structure-Activity Relationship , Sulfonamides/pharmacology
16.
Cancer Cell ; 13(3): 235-48, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328427

ABSTRACT

Invasion of surrounding brain tissue by isolated tumor cells represents one of the main obstacles to a curative therapy of glioblastoma multiforme. Here we unravel a mechanism regulating glioma infiltration. Tumor interaction with the surrounding brain tissue induces CD95 Ligand expression. Binding of CD95 Ligand to CD95 on glioblastoma cells recruits the Src family member Yes and the p85 subunit of phosphatidylinositol 3-kinase to CD95, which signal invasion via the glycogen synthase kinase 3-beta pathway and subsequent expression of matrix metalloproteinases. In a murine syngeneic model of intracranial GBM, neutralization of CD95 activity dramatically reduced the number of invading cells. Our results uncover CD95 as an activator of PI3K and, most importantly, as a crucial trigger of basal invasion of glioblastoma in vivo.


Subject(s)
Brain Neoplasms/metabolism , Fas Ligand Protein/metabolism , Glioblastoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Signal Transduction , fas Receptor/metabolism , Animals , Apoptosis , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Proto-Oncogene Proteins c-yes/genetics , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , Transplantation, Isogeneic , Tumor Cells, Cultured , src-Family Kinases/metabolism
17.
Bioorg Med Chem Lett ; 16(6): 1744-8, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16376544

ABSTRACT

The co-crystal structure of beta-phenethylamine fragment inhibitor 5 bound to DPP-IV revealed that the phenyl ring occupied the proline pocket of the enzyme. This finding provided the basis for a general hypothesis of a reverse binding mode for beta-phenethylamine-based DPP-IV inhibitors. Novel inhibitor design concepts that obviate substrate-like structure-activity relationships (SAR) were thereby enabled, and novel, potent inhibitors were discovered.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Enzyme Inhibitors/chemistry , Phenethylamines , Animals , Binding Sites , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Molecular Structure , Phenethylamines/chemistry , Phenethylamines/metabolism , Proline/chemistry , Protein Binding , Structure-Activity Relationship , Swine
18.
Bioorg Med Chem Lett ; 16(5): 1405-9, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16321524

ABSTRACT

Dipeptidyl peptidase IV is a clinically validated target for type-2 diabetes and belongs to a family of peptidases with a quite unique post-proline cleavage specificity. Known inhibitors contain a limited number of molecular anchors occupying the small prototypical S1 pocket. A virtual screening approach for such S1-binding fragments was carried out using FlexX docking to evaluate its potential to confirm known and find novel compounds. Several low molecular weight inhibitors exhibiting activities in the micromolar range could be identified as starting points for structure-based design.


Subject(s)
Computational Biology , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Drug Design , Drug Evaluation, Preclinical/methods , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Binding Sites , Crystallography, X-Ray , Inhibitory Concentration 50 , Models, Molecular , Molecular Weight , Protease Inhibitors/pharmacology , Protein Structure, Tertiary , Structure-Activity Relationship
19.
Microsc Res Tech ; 61(2): 171-8, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12740823

ABSTRACT

Recent in vivo observations have revealed that peroxisomes are more dynamic and interactive than previously assumed. The growing recognition of the tubular and reticular morphology of peroxisomes in living cells, their association with microtubules, and the dynamic movements of peroxisomes in vivo and in vitro have inspired the query into the investigation of the cellular machinery that mediates such a complex behaviour. The characterisation of the underlying molecular components of this machinery is providing insight into the mechanisms regulating peroxisomal morphology and intracellular distribution.


Subject(s)
Microtubules/metabolism , Peroxisomes/physiology , Animals , CHO Cells , COS Cells , Cattle , Cricetinae , Gene Expression Regulation , Microtubules/ultrastructure , Peroxisomes/metabolism , Peroxisomes/ultrastructure , Rats
20.
J Biol Chem ; 278(10): 8597-605, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12499366

ABSTRACT

The mammalian dynamin-like protein 1 (DLP1), a member of the dynamin family of large GTPases, possesses mechanochemical properties known to constrict and tubulate membranes. In this study, we have combined two experimental approaches, induction of peroxisome proliferation by Pex11pbeta and expression of dominant-negative mutants, to test whether DLP1 plays a role in peroxisomal growth and division. We were able to localize DLP1 in spots on tubular peroxisomes in HepG2 cells. In addition, immunoblot analysis revealed the presence of DLP1 in highly purified peroxisomal fractions from rat liver and an increase of DLP1 after treatment of rats with the peroxisome proliferator bezafibrate. Expression of a dominant negative DLP1 mutant deficient in GTP hydrolysis (K38A) either alone or in combination with Pex11pbeta caused the appearance of tubular peroxisomes but had no influence on their intracellular distribution. In co-expressing cells, the formation of tubulo-reticular networks of peroxisomes was promoted, and peroxisomal division was completely inhibited. These findings were confirmed by silencing of DLP1 using siRNA. We propose a direct role for the dynamin-like protein DLP1 in peroxisomal fission and in the maintenance of peroxisomal morphology in mammalian cells.


Subject(s)
GTP Phosphohydrolases/physiology , Microtubule-Associated Proteins , Peroxisomes/physiology , Proteins/physiology , Animals , Cell Line , Dynamins , GTP Phosphohydrolases/genetics , Gene Silencing , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Mitochondrial Proteins , Proteins/genetics , RNA, Small Interfering/physiology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...