15 research outputs found

    Molekulare Analysen zum Einfluss der kombinierten "targeted"-Therapie beim Prostatakarzinom in-vitro und in-vivo

    Get PDF
    Das Prostatakarzinom (PCa) ist in Deutschland die häufigste bösartige Malignität beim Mann; jährlich erkranken etwa 58.000 Männer am PCa. Mit 11.000 Todesfällen pro Jahr liegt PCa in der Mortalitätsstatistik an dritter Stelle, hinter dem Bronchialkarzinom und dem Dickdarmkarzinom. Solange PCa auf die Vorsteherdrüse begrenzt ist, besteht die Möglichkeit einer Heilung über die Prostatektomie oder Bestrahlung. Bei etwa 15% der Neuerkrankungen tritt ein metastasierender PCa auf. Bei etwa 20% der Patienten mit organbegrenztem Tumor zeigen lokale Therapieformen keine Wirkung, so dass ebenfalls einer invasiven Ausbreitung Vorschub geleistet wird. Weitere Therapieformen bilden die Androgensuppression und die Chemotherapie. Bei der Androgensuppression kommt es sehr häufig zu einem hormonrefraktären Stadium, das über einen PSA-Anstieg definiert wird. Ist das hormonrefraktäre Stadium erreicht, stehen nur begrenzte Therapieoptionen zur Verfügung. In der jüngeren Vergangenheit wurden neue molekulare Substanzen entwickelt, die gezielt in den Metabolismus der Karzinomzellen eingreifen, die targeted therapy molecules. PCa ist eine sehr heterogene Karzinomentität. Das Verständnis des Zusammenspiels von parallelen und interagierenden mitogenen Signaltransduktionswegen beim PCa, die sein Wachstum, seine Differenzierung und seine Zellmotiliät regulieren, ist von enormer Bedeutung, um neue bzw. schon bestehende Inhibitoren gegen PCa erfolgreich zu entwickeln oder als Mono- bzw. Kombinationstherapie anzuwenden. Zu den beim PCa häufig überexprimierten Signalproteinen gehören der mammalian target of rapamycin (mTOR), die Rezeptor-Tyrosinkinasen (RTKs) und die Histondeacetylasen (HDACs). Ziel dieser Arbeit war es, die Effektivität von Inhibitoren auf diese Signalproteine als Einzel- und Kombinationstherapie im Hinblick auf das Zellwachstum und die Adhäsionseigenschaft der PCa-Zellen zu untersuchen. Zur Anwendung kamen RAD001, ein mTOR-Inhibitor, AEE788, ein Multikinaseinhibitor gegen den VEGF- und EGF-Rezeptor, die Valproinsäure (VPA), ein HDAC-Inhibitor und das Interferon-alpha-2a (IFNalpha2a), ein körpereigenes Zytokin. Die Untersuchungen basierten auf zellbiologischen und biochemischen Analysen in-vitro unter Verwendung der PCa-Zelllinien DU145, PC-3 und LNCaP und in-vivo mittels eines xenogenen Nacktmausmodells. Im Rahmen funktioneller Untersuchungen wurden unter den entsprechenden Therapien das Zellwachstum mit dem MTT-Test ermittelt, das Expressionsmuster der zellzyklusregulierenden Proteine durch die Western-Blot-Hybridisierung und die Progression des Zellzyklus mittels der Durchflusszytometrie untersucht. Im weiteren Verlauf wurden die Adhäsionsprozesse an Matrix und an Endothel analysiert und die Modulation der Integrinsubtypen mit Hilfe der fluorimetrischen und molekularbiologischen Methoden sowie deren Genaktivität evaluiert. Die Synthese der gebundenen und freien Form von PSA wurde mit dem ELISA-Verfahren gemessen. Zur detaillierten Aufklärung der Wirkmechanismen der Medikamente diente die nähere Untersuchung intrazellulärer Signalwege anhand molekularbiologischer Studien. AEE788, RAD001, VPA, nicht jedoch IFNalpha2a erzielten eine deutliche Reduktion im Zellwachstum und der Adhäsion der PCa-Zellen. Dabei war jede Substanz durch ein eigenes Wirkprofil charakterisiert und zeigte eine zelltypabhängige molekulare Aktivität. Während AEE788 und RAD001 eine direkte Wirkung auslösten, war der Effekt von VPA zeitversetzt. Im Gegensatz zu VPA und RAD001 beeinflusste AEE788 vor allem androgenresistente Zellen. Eine kombinierte Behandlung erwies sich nicht in allen Fällen als gleich effektiv. Letztendlich zeigten vor allem AEE788/RAD001, VPA/RAD001 und VPA/IFNalpha2a deutliche antitumorale Effekte. Dabei demonstrierten die Untersuchungen eine Verringerung des Zellwachstums, einhergehend mit einer deutlichen Modulation der relevanten regulatorischen Zellzyklusproteine, einer Zunahme der Tumorsuppressoren und einer deutlichen Verlangsamung der Zellzyklusprogression. AEE788/RAD001 und VPA/IFNalpha2a erzielten dabei eine effektive Reduktion von Wachstum und Adhäsion. In Analysen der Adhäsionsprozesse konnte die Modulation der Integrinsubtypen und der integrinassoziierenden Kinasen aufgrund der Substanzen demonstriert werden und zeigte bei androgenresistenen und androgensensitiven Zellen einen unterschiedlichen Wirkungseinfluss. Nur die Kombination von VPA/IFNalpha2a verringerte signifikant die PSA-Synthese und zeigte bei der Evaluation der intrazellulären Signalwege einen deutlichen Verstärkereffekt in der Regulation der Proteinexpression und der Aktivität von EGFR, ERK1/2 und P70S6K. In der darauffolgenden in-vivo-Untersuchung konnte der Verstärkereffekt von VPA/IFNalpha2a in einer effektiven Reduktion des Tumorvolumens demonstriert werden. Da insbesondere PCa einen sehr heterogenen Phänotyp aufweist, bietet vor allem die kombinierte targeted Therapie neue Hoffnung und vielversprechende Therapiemöglichkeiten. Die eigenen Daten demonstrieren, dass nur bestimmte Medikamenten-Kombinationen eine effektive Verstärkung der antikarzinogenen Effekte erzielen und eine wirksame Therapie des PCa ermöglichen. Bei der klinischen Anwendung ist zu beachten, dass abhängig von Geno- und Phänotyp individuelle Therapiekonzepte zu berücksichtigen sind

    New histone deacetylase inhibitors as potential therapeutic tools for advanced prostate carcinoma

    Get PDF
    The anti-epileptic drug valproic acid is also under trial as an anti-cancer agent due to its histone deacetylase (HDAC) inhibitory properties. However, the effects of valproic acid (VPA) are limited and concentrations required for exerting anti-neoplastic effects in vitro may not be reached in tumour patients. In this study, we tested in vitro and in vivo effects of two VPA-derivatives (ACS2, ACS33) on pre-clinical prostate cancer models. PC3 and DU-145 prostate tumour cell lines were treated with various concentrations of ACS2 or ACS33 to perform in vitro cell proliferation 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and to evaluate tumour cell adhesion to endothelial cell monolayers. Analysis of acetylated histones H3 and H4 protein expression was performed by western blotting. In vivo tumour growth was conducted in subcutaneous xenograft mouse models. Tumour sections were assessed by immunohistochemistry for histone H3 acetylation and proliferation. ACS2 and ACS33 significantly up-regulated histone H3 and H4 acetylation in prostate cancer cell lines. In micromolar concentrations both compounds exerted growth arrest in PC3 and DU-145 cells and prevented tumour cell attachment to endothelium. In vivo, ACS33 inhibited the growth of PC3 in subcutaneous xenografts. Immunohistochemistry and western blotting confirmed increased histone H3 acetylation and reduced proliferation. ACS2 and ACS33 represent novel VPA derivatives with superior anti-tumoural activities, compared to the mother compound. This investigation lends support to the clinical testing of ACS2 or ACS33 for the treatment of prostate cancer

    The histone deacetylase inhibitor valproic acid alters growth properties of renal cell carcinoma in vitro and in vivo

    Get PDF
    Histone deacetylase (HDAC) inhibitors represent a promising class of antineoplastic agents which affect tumour growth, differentiation and invasion. The effects of the HDAC inhibitor valproic acid (VPA) were tested in vitro and in vivo on pre-clinical renal cell carcinoma (RCC) models. Caki-1, KTC-26 or A498 cells were treated with various concentrations of VPA during in vitro cell proliferation 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays and to evaluate cell cycle manipulation. In vivo tumour growth was conducted in subcutaneous xenograft mouse models. The anti-tumoural potential of VPA combined with low-dosed interferon-α (IFN-α) was also investigated. VPA significantly and dose-dependently up-regulated histones H3 and H4 acetylation and caused growth arrest in RCC cells. VPA altered cell cycle regulating proteins, in particular CDK2, cyclin B, cyclin D3, p21 and Rb. In vivo, VPA significantly inhibited the growth of Caki-1 in subcutaneous xenografts, accompanied by a strong accumulation of p21 and bax in tissue specimens of VPA-treated animals. VPA–IFN-α combination markedly enhanced the effects of VPA monotherapy on RCC proliferation in vitro, but did not further enhance the anti-tumoural potential of VPA in vivo. VPA was found to have profound effects on RCC cell growth, lending support to the initiation of clinical testing of VPA for treating advanced RCC

    Combining the receptor tyrosine kinase inhibitor AEE788 and the mammalian target of rapamycin (mTOR) inhibitor RAD001 strongly inhibits adhesion and growth of renal cell carcinoma cells

    Get PDF
    Background Treatment options for metastatic renal cell carcinoma (RCC) are limited due to resistance to chemo- and radiotherapy. The development of small-molecule multikinase inhibitors have now opened novel treatment options. The influence of the receptor tyrosine kinase inhibitor AEE788, applied alone or combined with the mammalian target of rapamycin (mTOR) inhibitor RAD001, on RCC cell adhesion and proliferation in vitro has been evaluated. Methods RCC cell lines Caki-1, KTC-26 or A498 were treated with various concentrations of RAD001 or AEE788 and tumor cell proliferation, tumor cell adhesion to vascular endothelial cells or to immobilized extracellular matrix proteins (laminin, collagen, fibronectin) evaluated. The anti-tumoral potential of RAD001 combined with AEE788 was also investigated. Both, asynchronous and synchronized cell cultures were used to subsequently analyze drug induced cell cycle manipulation. Analysis of cell cycle regulating proteins was done by western blotting. Results RAD001 or AEE788 reduced adhesion of RCC cell lines to vascular endothelium and diminished RCC cell binding to immobilized laminin or collagen. Both drugs blocked RCC cell growth, impaired cell cycle progression and altered the expression level of the cell cycle regulating proteins cdk2, cdk4, cyclin D1, cyclin E and p27. The combination of AEE788 and RAD001 resulted in more pronounced RCC growth inhibition, greater rates of G0/G1 cells and lower rates of S-phase cells than either agent alone. Cell cycle proteins were much more strongly altered when both drugs were used in combination than with single drug application. The synergistic effects were observed in an asynchronous cell culture model, but were more pronounced in synchronous RCC cell cultures. Conclusions Potent anti-tumoral activitites of the multikinase inhibitors AEE788 or RAD001 have been demonstrated. Most importantly, the simultaneous use of both AEE788 and RAD001 offered a distinct combinatorial benefit and thus may provide a therapeutic advantage over either agent employed as a monotherapy for RCC treatment

    Molecular targeting of prostate cancer cells by a triple drug combination down-regulates integrin driven adhesion processes, delays cell cycle progression and interferes with the cdk-cyclin axis

    Get PDF
    Background: Single drug use has not achieved satisfactory results in the treatment of prostate cancer, despite application of increasingly widespread targeted therapeutics. In the present study, the combined impact of the mammalian target of rapamycin (mTOR)-inhibitor RAD001, the dual EGFr and VGEFr tyrosine kinase inhibitor AEE788 and the histone deacetylase (HDAC)-inhibitor valproic acid (VPA) on prostate cancer growth and adhesion in vitro was investigated. Methods: PC-3, DU-145 and LNCaP cells were treated with RAD001, AEE788 or VPA or with a RAD-AEE-VPA combination. Tumor cell growth, cell cycle progression and cell cycle regulating proteins were then investigated by MTT-assay, flow cytometry and western blotting, respectively. Furthermore, tumor cell adhesion to vascular endothelium or to immobilized extracellular matrix proteins as well as migratory properties of the cells was evaluated, and integrin alpha and beta subtypes were analyzed. Finally, effects of drug treatment on cell signaling pathways were determined. Results: All drugs, separately applied, reduced tumor cell adhesion, migration and growth. A much stronger anti-cancer effect was evoked by the triple drug combination. Particularly, cdk1, 2 and 4 and cyclin B were reduced, whereas p27 was elevated. In addition, simultaneous application of RAD001, AEE788 and VPA altered the membranous, cytoplasmic and gene expression pattern of various integrin alpha and beta subtypes, reduced integrin-linked kinase (ILK) and deactivated focal adhesion kinase (FAK). Signaling analysis revealed that EGFr and the downstream target Akt, as well as p70S6k was distinctly modified in the presence of the drug combination. Conclusions: Simultaneous targeting of several key proteins in prostate cancer cells provides an advantage over targeting a single pathway. Since strong anti-tumor properties became evident with respect to cell growth and adhesion dynamics, the triple drug combination might provide progress in the treatment of advanced prostate cancer

    Effects of electrical stimulation on rat limb regeneration, a new look at an old model

    No full text
    Limb loss is a devastating disability and while current treatments provide aesthetic and functional restoration, they are associated with complications and risks. The optimal solution would be to harness the body's regenerative capabilities to regrow new limbs. Several methods have been tried to regrow limbs in mammals, but none have succeeded. One such attempt, in the early 1970s, used electrical stimulation and demonstrated partial limb regeneration. Several researchers reproduced these findings, applying low voltage DC electrical stimulation to the stumps of amputated rat forelimbs reporting "blastema, and new bone, bone marrow, cartilage, nerve, skin, muscle and epiphyseal plate formation". In spite of these encouraging results this research was discontinued. Recently there has been renewed interest in studying electrical stimulation, primarily at a cellular and subcellular level, and studies have demonstrated changes in stem cell behavior with increased proliferation, differentiation, matrix formation and migration, all important in tissue regeneration. We applied electrical stimulation, in vivo, to the stumps of amputated rat limbs and observed significant new bone, cartilage and vessel formation and prevention of neuroma formation. These findings demonstrate that electricity stimulates tissue regeneration and form the basis for further research leading to possible new treatments for regenerating limbs

    Tumor-Endothelium Cross Talk Blocks Recruitment of Neutrophils to Endothelial Cells: A Novel Mechanism of Endothelial Cell Anergy1

    Get PDF
    Tumor cells have evolved effective strategies to escape the host immune response. The objective of this study was to determine whether tumor cells can condition endothelial cells in a specific manner to prevent subsequent adhesion of polymorphonuclear neutrophils (PMNs) and/or peripheral blood lymphocytes (PBLs). Human umbilical vein endothelial cells (HUVECs) and UKF-NB-4 neuroblastoma tumor cells were established in coculture on opposite sides of porous transwell filters. After 24 hours with and without HUVEC conditioning, PMNs or PBLs were added to the HUVEC monolayer. Adhesion to conditioned HUVEC versus adhesion to nonconditioned HUVEC was compared. Effects on endothelial CD44v4, CD44v5, CD44v7, intercellular adhesion molecule 1 (ICAM-1), E-selectin, and vascular cell adhesion molecule 1 (VCAM-1) adhesion receptor expression were analyzed by flow cytometry, intracellular signaling proteins of the mitogen-activated protein kinase pathway and protein kinase C (PKC) subtypes quantified by Western blot analysis. Endothelial conditioning led to a distinct reduction in PMN but not in PBL adhesion to HUVEC. CD44 was significantly reduced, whereas ICAM-1, E-selectin, and VCAM-1 were not altered during HUVEC conditioning. Antibody blockade against CD44v4, CD44v5, and CD44v7 inhibited PMN but not PBL binding. The observed effects were caused by direct tumor cell-HUVEC contact because addition of isolated tumor cell membrane fragments but not of soluble cell culture supernatant to HUVEC induced the CD44 receptor loss. PKCα activity was strongly enhanced in conditioned HUVEC. Blocking PKC prevented the reduction in PMN binding, indicating that this protein is involved in PMN adhesion regulation. A novel tumor escape strategy is presented here. Cell contact-dependent adhesion of tumor cells to the vascular wall promotes down-regulation of endothelial CD44 receptor expression, impairing an effective neutrophil attack

    Tumor-endothelium cross talk blocks recruitment of neutrophils to endothelial cells: a novel mechanism of endothelial cell anergy

    No full text
    The mode of the antitumoral activity of multimutated oncolytic herpes simplex virus type 1 G207 has not been fully elucidated yet. Because the antitumoral activity of many drugs involves the inhibition of tumor blood vessel formation, we determined if G207 had an influence on angiogenesis. Monolayers of human umbilical vein endothelial cells and human dermal microvascular endothelial cells, but not human dermal fibroblasts, bronchial epithelial cells, and retinal glial cells, were highly sensitive to the replicative and cytotoxic effects of G207. Moreover, G207 infection caused the destruction of endothelial cell tubes in vitro. In the in vivo Matrigel plug assay in mice, G207 suppressed the formation of perfused vessels. Intratumoral treatment of established human rhabdomyosarcoma xenografts with G207 led to the destruction of tumor vessels and tumor regression. Ultrastructural investigations revealed the presence of viral particles in both tumor and endothelial cells of G207-treated xenografts, but not in adjacent normal tissues. These findings show that G207 may suppress tumor growth, in part, due to inhibition of angiogenesis
    corecore