971 resultados para tumor model


Relevância:

60.00% 60.00%

Publicador:

Resumo:

TGF-beta ist ein Schlüsselmolekül zellvermittelter Immuntoleranz. So spielt es neben seiner pleiotropen Rolle in Immunzellen auch bei der Tumorentwicklung eine große Rolle. Das TGF-beta hat bei der Tumorentwicklung eine duale Rolle. So dient es in frühen Phasen als Tumorsuppressor, währenddessen es in späten Phasen der Entwicklung als Tumorpromotor wirkt. Eine strikte Regulation des TGF-beta Signalweges ist daher für ein funktionierendes Immunsystem von essentieller Bedeutung. Die Ubiquitin Ligase Smurf2 ist dabei ein wichtiger negativ Regulator des TGF-beta Signalweges.In der vorliegenden Arbeit konnte eine neue Spleißform des Smurf2 (dE2Smurf2) aus murinen CD4+ T-Zellen isoliert werden, deren Funktion in vitro und in vivo in T-Lymphozyten untersucht worden ist. Für diese Spleißform konnte zudem eine humane Relevanz nachgewiesen werden. Mit Hilfe von Überexpressionen in Cos7 Zellen konnte eine veränderte Lokalisation der Smurf2 Spleißformen (WT und dE2) festgestellt werden. Dabei konnten lysosomale und endosomale Kompartimente bei der Kolokalisation mit dem dE2Smurf2 Konstrukt beobachtet werden. Das Spleißen des Exons2 führte dabei zu Änderungen der Topologie der N-terminalen C2-Domäne, wodurch sich eine veränderte Lokalisation in der Zelle beschreiben ließ. Mit der veränderten intrazellulären Verteilung erfuhr auch die Funktion der dE2Smurf2 Ubiquitin Ligase eine Änderung. So konnte überraschenderweise eine positive Signalinduktion des TGF-beta Signalweges beobachtet werden, was im Gegensatz zum beschriebenen WTSmurf2 stand. Durch eine Überexpression des dE2Smurf2 Proteins in T-Lymphozyten wurde der TGF-beta Signalweg in CD4+ und CD8+ Zellen positiv reguliert, dabei wurde der TGFbetaRII vermehrt exprimiert und gleichzeitig fand eine verstärkte Phosphorylierung der Transkriptionsfaktoren Smad2 und Smad3 nach TGF-beta Stimulation statt. Die transgenen T-Lymphozyten waren somit sensitiver gegenüber TGF-beta. Dies führte zur Hypothese, die durch Western Blot Analyse bestätigt werden konnte, daß das dE2Smurf2 nach Überexpression seine WT-Form bindet und dadurch degradiert. Die Degradation der Ubiquitin Ligase war dabei Smad7 abhängig. Zur Analyse des Einflusses der Ubiquitin Ligase dE2Smurf2 auf die Differenzierung von CD4+ T-Zellen, sowie ihre Rolle bei der T-Zell Proliferation, konnte gezeigt werden, daß durch die höhere Sensitivität gegenüber TGF-beta naive T-Zellen unter Einfluß von TGF-beta und IL6 vermehrt in TH17 Zellen differenzierten. Zudem konnte gezeigt werden, daß die Proliferationsrate transgener naiver CD4+ T-Zellen bei geringen Mengen von TGF-beta starkt vermindert war. Weiterhin konnte gezeigt werden, daß bei einer Differenzierung der naiven CD4+ T-Zellen in TH1 Zellen, diese signifkant weniger das proinflammatorische Zytokin INFγ produzierten.So zeigten in vivo Versuche, daß die transgenen Tiere in der Entwicklung von Kolorektalen Karzinomen protektiert waren. Sowohl im kolitisassiziierten Tumor Modell als auch bei der spontanen Entwicklung von Tumoren im APCmin Modell. Dies konnte zum einen auf eine deutlich verminderte Entzündung (geringere Produktion an Zytokinen durch verminderte Proliferation) des Darms und zum anderen durch eine stärkere Produktion an zytotoxischen Genen, wie Perforin, INFγ und Granzym B erklärt werden. Interessanterweise konnte jedoch im Transfer Kolitis Modell eher eine proinflammatorische Wirkung des dE2Smurf2 Proteins nachgewiesen werden. So wiesen die immundefizienten Mäuse, in denen die transgenen T-Zellen injiziert wurden, eine signifikant stärkere Kolitis auf als die Kontrollen. Dies konnte mit einer Überproduktion an IL17 sezernierenden T-Zellen erklärt werden. Klonierungsexperimente führten zudem zur Identifikation einer bisher nicht beschriebenen nicht kodierenden RNA. Diese zeigte in Kombination mit dem dE2Smurf2 Protein in einer Reportergen Analyse eine Hyperaktivierung des Smad3 Promotors. Diese Daten liefern zum einen ein genaueres Modell über die Regulation des TGF-beta Signalweges sowie wichtige Erkenntnisse zur Pathophysiologie chronisch entzündlicher Darmerkrankung und daraus resultierende Tumorerkrankungen. So entwickelt sich das dE2Smurf2, Teil des TGF-beta Signalweges, als attraktives Zielprotein für die Modulation von chronisch entzündlichen Darmerkrankungen und (kolitisassoziierte) Kolonkarzinomen.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Ein neuer Ansatz der immunologischen Krebstherapie ist die Verwendung der bispezifischen, trifunktionalen Antikörper catumaxomab (anti-EpCAM x anti-CD3) und ertumaxomab (anti-Her2/neu x anti-CD3). Die Bispezifität besteht in der Bindung eines Tumor-assoziierten Antigens (EpCAM bzw. Her2/neu) und des CD3 Moleküls auf der Oberfläche von T-Zellen. Darüber hinaus stellt die Interaktion des Fc-Teils mit FcγRI/IIa/III positiven akzessorischen Immunzellen die dritte Funktion der Antikörper dar. Diese einzigartige Kombination ermöglicht theoretisch die Ausbildung eines Tri-Zell-Komplexes. In klinischen Studien konnte bereits die Wirksamkeit beider Antikörper nachgewiesen werden. Die eigentlichen Wirkmechanismen der trifunktionalen Antikörper jedoch sind noch nicht ausreichend bekannt. Um die Wechselwirkung zwischen den stark EpCAM- und schwach Her2/neu-positive FaDu- sowie den stabil mit humanem Her2/neu transfizierten FaDu E593-Tumorzellen, peripheren Blutmonozyten (PBMC) und trifunktionalen Antikörpern systematisch zu untersuchen wurde ein 3D-Tumormodell, die so genannten multizellulären Tumorsphäroide (MCTS), angewandt. Als Endpunkte zur Beurteilung der Therapieeffizienz dienten das Volumenwachstum der Sphäroide, sowie die Klonogenität und die Zellvitalität. Zur Beurteilung der PBMC-Penetration in die Sphäroide erfolgten immunhistochemische Färbungen und molekularbiologische Nachweise der Abwehrzellantigene. Entsprechend wurden in den Sphäroiden die Proliferationsrate über eine Ki67-Färbung sowie die Apoptoserate über eine FragEL-Markierung identifiziert. Die Aktivität der PBMC wurde durch die Bestimmung ausgewählter Zytokine (ELISA) und der Zellzahl aus den Medienüberständen charakterisiert. Die an den FaDu- und E593-Sphäroiden erzielten Ergebnisse zeigten, dass catumaxomab und ertumaxomab eine konzentrations- und zeitabhängige Abnahme des Sphäroidvolumens bewirkten. Die Schrumpfung der Tumorsphäroide ging mit einer Reduktion des proliferativen und mit einer Steigerung des apoptotischen Tumorzellanteils einher. Die histologischen Befunde weisen darauf hin, dass die Volumenreduktion durch eine gesteigerte Anzahl infiltrierender Leukozyten bedingt ist. Auf verschiedenen Methoden basierende Analysen der Immunzellsubtypen zeigten eine dominierende Infiltration von zytotoxischen T-Zellen in die Tumorsphäroide. Der Aktivitätsnachweis der T-Zellen wurde über die Detektion der IL-2 mRNA und des sekretierten Zytokins erbracht. Einen zusätzlichen Hinweis auf eine zelluläre Immunantwort liefert das Zytokinmuster mit hohen Konzentrationen an IFN-γ. Der direkte Vergleich beider Antikörper zeigte, dass der anti-tumorale Effekt abhängig von der Antigenexpression auf den Tumorzellen war. Die Analyse von Medienüberständen wies auf eine mehrheitlich höhere Zytokinausschüttung in Gegenwart des Tumorantigens hin. Sphäroid-Kokulturen, die mit dem parentalen anti-EpCAM Antikörper behandelt wurden, zeigten keine Volumenreduktion. Im Gegensatz dazu führte der parentale CD3-Antikörper, das CD3- und Tumorzell-bindende catumaxomab F(ab')2 Fragment oder eine Kombination beider parentaler Antikörper zu einer anti-tumoralen Wirkung, die jedoch nicht so stark war wie die des trifunktionalen Antikörpers catumaxomab. Demnach ist für catumaxomab gezeigt, dass für die Effektivität des Antikörpers die Trifunktionalität unabdingbar ist. Daraus leitet sich ab, dass die Aktivierung der Abwehrzellen durch kostimulatorische Signale notwendig ist und über die Tumorantigenbindung Mechanismen wie ADCC (antibody-dependent cellular cytotoxicity) zum Tragen kommen. Die Experimente mit gleichzeitiger Gabe von trifunktionalen Antikörpern und Immunsuppressiva haben gezeigt, dass eine Kombination beider Agenzien möglich ist. Die Konzentrationen sind jedoch sorgfältig derart zu wählen, dass die Zytokinausschüttung und die damit verbundenen Nebenwirkungen reduziert sind, ohne dass die anti-tumorale Wirkung der Antikörper maßgeblich beeinflusst wird. T-Zellen bedienen sich nach Aktivierung für die rasche Proliferation einer gesteigerten aeroben Glykolyse. Unter Behandlung der Kokulturen mit catumaxomab konnte im Vergleich zu anderen immunstimulatorischen Agenzien die größte Steigerung der Laktatproduktion bzw. der Azidifizierungs- und Sauerstoffverbrauchsrate detektiert werden. Diese Effekte weisen auf eine metabolische Aktivierung der PBMC durch catumaxomab hin. Das von den Tumorzellen abgegebene Laktat kann die Immunzellen jedoch inhibieren. Daher wäre die Kombination mit Glykolyseinhibitoren ein möglicher Ansatz, um die Therapieeffizienz weiter zu steigern. Darüber hinaus konnte gezeigt werden, dass eine Komedikation der trifunktionalen Antikörper mit Chemotherapeutika zu einer gesteigerter Wirkung führte. Insgesamt liegt die Zukunft der Immuntherapien wohl in der Kombination mit anderen Wirkstoffklassen, die anti-tumorale Effekte verstärken oder immunsupprimierende Mechanismen inhibieren.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

In dieser Arbeit wurde ein etabliertes Immunisierungsmodell auf der Basis eines synthetischen TLR 7-Liganden zur Aufhebung der suppressiven Eigenschaften von UV B-Strahlung verwendet. Es konnte gezeigt werden, dass nach Ermittlung eines geeigneten Immunisierungsprotokolls mit UV B die durch TCI vermittelte ausschließlich primäre Immunantwort verstärkt werden konnte, so dass eine Gedächtnisantwort induziert werden konnte. Durch die Immunisierung verschiedenster knockout sowie transgener Stämme konnte bewiesen werden, dass die Reaktion auch nach zusätzlicher UV B-Bestrahlung abhängig von TLR 7 und dem Adaptormolekül MyD88 ist. Bei der Aufklärung der zellulären Mechanismen, die dieser Immunisierungsmethode zu Grunde liegen, konnten dermale DCs, sowie GR1+-Zellen als wichtige Mediatoren identifiziert werden. Eine wesentliche Funktion von epidermalen Langerhans Zellen konnte in diesem Zusammenhang ausgeschlossen werden. Des Weiteren konnte gezeigt werden, dass auf Zytokinebene Typ I Interferone eine tragende Rolle spielen und die Produktion von IL-12p35 in den Haut-drainierenden Lymphknoten angeregt wird. Supprimiert wird die Immunantwort durch regulatorische T-Zellen, sowie durch die Freisetzung von IL-10, welches nicht ausschließlich von regulatorischen T-Zellen produziert wird. Die Applikation eines blockierenden IL-10-Rezeptor Antikörpers verhinderte die IL-10-vermittelte Suppression und führte zu einer weiteren Verstärkung der Immunantwort. Durch die induzierte Gedächtnisantwort nach UV B/TCI konnte das Immunisierungs-Modell in einem therapeutischen Tumormodell angewandt werden und führt hier zu einer verstärkten Abstoßung von Tumoren, verglichen mit TCI alleine, was die Basis für zukünftige nadelfreie Vakzinierungen für die Behandlung von Krebs oder persistierender Infektionen darstellen könnte.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Nuclear medicine imaging techniques such as PET are of increasing relevance in pharmaceutical research being valuable (pre)clinical tools to non-invasively assess drug performance in vivo. Therapeutic drugs, e.g. chemotherapeutics, often suffer from a poor balance between their efficacy and toxicity. Here, polymer based drug delivery systems can modulate the pharmacokinetics of low Mw therapeutics (prolonging blood circulation time, reducing toxic side effects, increasing target site accumulation) and therefore leading to a more efficient therapy. In this regard, poly-N-(2-hydroxypropyl)-methacrylamide (HPMA) constitutes a promising biocompatible polymer. Towards the further development of these structures, non-invasive PET imaging allows insight into structure-property relationships in vivo. This performant tool can guide design optimization towards more effective drug delivery. Hence, versatile radiolabeling strategies need to be developed and establishing 18F- as well as 131I-labeling of diverse HPMA architectures forms the basis for short- as well as long-term in vivo evaluations. By means of the prosthetic group [18F]FETos, 18F-labeling of distinct HPMA polymer architectures (homopolymers, amphiphilic copolymers as well as block copolymers) was successfully accomplished enabling their systematic evaluation in tumor bearing rats. These investigations revealed pronounced differences depending on individual polymer characteristics (molecular weight, amphiphilicity due to incorporated hydrophobic laurylmethacrylate (LMA) segments, architecture) as well as on the studied tumor model. Polymers showed higher uptake for up to 4 h p.i. into Walker 256 tumors vs. AT1 tumors (correlating to a higher cellular uptake in vitro). Highest tumor concentrations were found for amphiphilic HPMA-ran-LMA copolymers in comparison to homopolymers and block copolymers. Notably, the random LMA copolymer P4* (Mw=55 kDa, 25% LMA) exhibited most promising in vivo behavior such as highest blood retention as well as tumor uptake. Further studies concentrated on the influence of PEGylation (‘stealth effect’) in terms of improving drug delivery properties of defined polymeric micelles. Here, [18F]fluoroethylation of distinct PEGylated block copolymers (0%, 1%, 5%, 7%, 11% of incorporated PEG2kDa) enabled to systematically study the impact of PEG incorporation ratio and respective architecture on the in vivo performance. Most strikingly, higher PEG content caused prolonged blood circulation as well as a linear increase in tumor uptake (Walker 256 carcinoma). Due to the structural diversity of potential polymeric carrier systems, further versatile 18F-labeling strategies are needed. Therefore, a prosthetic 18F-labeling approach based on the Cu(I)-catalyzed click reaction was established for HPMA-based polymers, providing incorporation of fluorine-18 under mild conditions and in high yields. On this basis, a preliminary µPET study of a HPMA-based polymer – radiolabeled via the prosthetic group [18F]F-PEG3-N3 – was successfully accomplished. By revealing early pharmacokinetics, 18F-labeling enables to time-efficiently assess the potential of HPMA polymers for efficient drug delivery. Yet, investigating the long-term fate is essential, especially regarding prolonged circulation properties and passive tumor accumulation (EPR effect). Therefore, radiolabeling of diverse HPMA copolymers with the longer-lived isotope iodine-131 was accomplished enabling in vivo evaluation of copolymer P4* over several days. In this study, tumor retention of 131I-P4* could be demonstrated at least over 48h with concurrent blood clearance thereby confirming promising tumor targeting properties of amphiphilic HPMA copolymer systems based on the EPR effect.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

T helper (Th) 9 cells are an important subpopulation of the CD4+ T helper cells. Due to their ability to secrete Interleukin-(IL-)9, Th9 cells essentially contribute to the expulsion of parasitic helminths from the intestinal tract but they play also an immunopathological role in the course of asthma. Recently, a beneficial function of Th9 cells in anti-tumor immune responses was published. In a murine melanoma tumor model Th9 cells were shown to enhance the anti-melanoma immune response via the recruitment of CD8+ T cells, dendritic cells and mast cells. In contrast to Th9 effector cells regulatory T cells (Tregs) are able to control an immune response with the aid of different suppressive mechanisms. Based on their ability to suppress an immune response Tregs are believed to be beneficial in asthma by diminishing excessive allergic reactions. However, concerning cancer they can have a detrimental function because Tregs inhibit an effective anti-tumor immune reaction. Thus, the analysis of Th9 suppression by Tregs is of central importance concerning the development of therapeutic strategies for the treatment of cancer and allergic diseases and was therefore the main objective of this PhD thesis.rnIn general it could be demonstrated that the development of Th9 cells can be inhibited by Tregs in vitro. The production of the lineage-specific cytokine IL-9 by developing Th9 cells was completely suppressed at a Treg/Th9 ratio of 1:1 on the transcriptional (qRT-PCR) as well as on the translational level (ELISA). In contrast, the expression of IRF4 that was found to strongly promote Th9 development was not reduced in the presence of Tregs, suggesting that IRF4 requires additional transcription factors to induce the differentiation of Th9 cells. In order to identify such factors, which regulate Th9 development and therefore represent potential targets for Treg-mediated suppressive mechanisms, a transcriptome analysis using “next-generation sequencing” was performed. The expression of some genes which were found to be up- or downregulated in Th9 cells in the presence of Tregs was validated with qRT-PCR. Time limitations prevented a detailed functional analysis of these candidate genes. Nevertheless, the analysis of the suppressive mechanisms revealed that Tregs probably suppress Th9 cells via the increase of the intracellular cAMP concentration. In contrast, IL-9 production by differentiated Th9 cells was only marginally affected by Tregs in vitro and in vivo analysis (asthma, melanoma model). Hence, Tregs represent very effective inhibitors of Th9 development whereas they have only a minimal suppressive influence on differentiated Th9 cells.rn

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Adoptive T cell therapy using antigen-specific T lymphocytes is a powerful immunotherapeutic approach against cancer. Nevertheless, many T cells against tumor-antigens exhibit only weak anti-tumoral response. To overcome this barrier it is necessary to improve the potency and anti-tumoral efficacy of these T cells. Activation and activity of T cells are tightly controlled to inhibit unwanted T cell responses and to reduce the risk of autoimmunity. Both are regulated by extrinsic signals and intrinsic mechanisms which suppress T cell activation. The intrinsic mechanisms include the expression of phosphatases that counteract the activation-inducing kinases. Modifying the expression of these phosphatases allows the targeted modulation of T cell reactivity. MicroRNAs (miRNAs) are regulatory small noncoding RNA molecules that control gene expression by targeting messenger RNAs in a sequence specific manner. Gene-specific silencing plays a key role in diverse biological processes, such as development, differentiation, and functionality. miR181a has been shown to be highly expressed in immature T cells that recognize low-affinity antigens.rnThe present study successfully shows that ectopic expression of miR181a is able to enhance the sensitivity of both murine and human T cells. In CD4+ T helper cells as well as in CD8+ cytotoxic T cells the overexpression of miR181a leads to downregulation of multiple phosphatases involved in the T cell receptor signaling pathway. Overexpression of miR181a in human T cells achieves a co-stimulatory independent activation and has an anti-apoptotic effect on CD4+ T helper cells. Additionally, increasing the amount of miR181a enhances the cytolytic activity of murine CD8+ TCRtg T cells in an antigen-specific manner.rnTo test miR181a overexpressing T cells in vivo, a mouse tumor model using a B cell lymphoma cell line (A20-HA) expressing the Influenza hemagglutinin (Infl.-HA) antigen was established. The expression of model antigens in tumor cell lines enables targeted elimination of tumors using TCRtg T cells. The transfer of miR181a overexpressing Infl.-HA TCRtg CD8+ T cells alone has no positive effect neither on tumor control nor on survival of A20-HA tumor-bearing mice. In contrast, the co-transfer of miR181a overexpressing Infl.-HA TCRtg CD8+ and CD4+ T cells leads to improved tumor control and prolongs survival of A20-HA tumor-bearing mice. This effect is characterized by higher amounts of effector T cells and the expansion of Infl.-HA TCRtg CD8+ T cells.rnAll effects were achieved by changes in expression of several genes including molecules involved in T cell differentiation, activation, and regulation, cytotoxic effector molecules, and receptors important for the homing process of T cells in miR181a overexpressing T cells. The present study demonstrates that miR181a is able to enhance the anti-tumoral response of antigen-specific T cells and is a promising candidate for improving adoptive cell therapy.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Somatostatin receptor targeting of neuroendocrine tumors using radiolabeled somatostatin agonists is today an established method to image and treat cancer patients. However, in a study using an animal tumor model, somatostatin receptor antagonists were shown to label sst(2)- and sst(3)-expressing tumors in vivo better than agonists, with comparable affinity even though they are not internalized into the tumor cell. In the present study, we evaluated the in vitro binding of the antagonist (177)Lu-DOTA-pNO(2)-Phe-c (DCys-Tyr-DTrp-Lys-Thr-Cys) DTyrNH(2) ((177)Lu-DOTA-BASS) or the (177)Lu-DOTATATE agonist to sst(2)-expressing human tumor samples.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

High levels of glucagon-like peptide-1 (GLP-1) receptor expression in human insulinomas and gastrinomas provide an attractive target for imaging, therapy, and intraoperative tumor localization, using receptor-avid radioligands. The goal of this study was to establish a tumor model for GLP-1 receptor targeting and to use a newly designed exendin-4-DTPA (DTPA is diethylenetriaminepentaacetic acid) conjugate for GLP-1 receptor targeting. METHODS: Exendin-4 was modified C-terminally with Lys(40)-NH(2), whereby the lysine side chain was conjugated with Ahx-DTPA (Ahx is aminohexanoic acid). The GLP-1 receptor affinity (50% inhibitory concentration [IC(50)] value) of [Lys(40)(Ahx-DTPA)NH(2)]exendin-4 as well as the GLP-1 receptor density in tumors and different organs of Rip1Tag2 mice were determined. Rip1Tag2 mice are transgenic mice that develop insulinomas in a well-defined multistage tumorigenesis pathway. This animal model was used for biodistribution studies, pinhole SPECT/MRI, and SPECT/CT. Peptide stability, internalization, and efflux studies were performed in cultured beta-tumor cells established from tumors of Rip1Tag2 mice. RESULTS: The GLP-1 receptor affinity of [Lys(40)(Ahx-DTPA)NH(2)]exendin-4 was found to be 2.1 +/- 1.1 nmol/L (mean +/- SEM). Because the GLP-1 receptor density in tumors of Rip1Tag2 mice was very high, a remarkably high tumor uptake of 287 +/- 62 %IA/g (% injected activity per gram tissue) was found 4 h after injection. This resulted in excellent tumor visualization by pinhole SPECT/MRI and SPECT/CT. In accordance with in vitro data, [Lys(40)(Ahx-DTPA-(111)In)NH(2)]exendin-4 uptake in Rip1Tag2 mice was also found in nonneoplastic tissues such as pancreas and lung. However, lung and pancreas uptake was distinctly lower compared with that of tumors, resulting in a tumor-to-pancreas ratio of 13.6 and in a tumor-to-lung ratio of 4.4 at 4 h after injection. Furthermore, in vitro studies in cultured beta-tumor cells demonstrated a specific internalization of [Lys(40)(Ahx-DTPA-(111)In)NH(2)]exendin-4, whereas peptide stability studies indicated a high metabolic stability of the radiopeptide in beta-tumor cells and human blood serum. CONCLUSION: The high density of GLP-1 receptors in insulinomas as well as the high specific uptake of [Lys(40)(Ahx-DTPA-(111)In)NH(2)]exendin-4 in the tumor of Rip1Tag2 mice indicate that targeting of GLP-1 receptors in insulinomas may become a useful imaging method to localize insulinomas in patients, either preoperatively or intraoperatively. In addition, Rip1Tag2 transgenic mice represent a suitable animal tumor model for GLP-1 receptor targeting.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

AIM: Peptide receptor radionuclide therapy using the somatostatin analogue [(177)Lu-DOTA(0),Tyr(3)]octreotate is a convincing treatment modality for metastasized neuroendocrine tumors. Therapeutic doses are administered in 4 cycles with 6-10 week intervals. A high somatostatin receptor density on tumor cells is a prerequisite at every administration to enable effective therapy. In this study, the density of the somatostatin receptor subtype 2 (sst2) was investigated in the rat CA20948 pancreatic tumor model after low dose [(177)Lu-DOTA(0), Tyr(3)]octreotate administration resulting in approximately 20 Gy tumor radiation absorbed dose, whereas 60 Gy is needed to induce complete tumor regression in these and the majority of tumors. METHODS: Sixteen days after inoculation of the CA20948 tumor, male Lewis rats were injected with 185 MBq [(177)Lu-DOTA(0),Tyr(3)]octreotate to initiate a decline in tumor size. Approximately 40 days after injection, tumors re-grew progressively after initial response. Quantification of sst2 expression was performed using in vitro autoradiography on frozen sections of three groups: control (not-treated) tumors, tumors in regression and tumors in re-growth. Histology and proliferation were determined using HE- and anti-Ki-67-staining. RESULTS: The sst2 expression on CA20948 tumor cells decreased significantly after therapy to 5% of control level. However, tumors escaping from therapy showed an up-regulated sst2 level of 2-5 times higher sst2 density compared to control tumors. CONCLUSION: After a suboptimal therapeutic dose of [(177)Lu-DOTA(0),Tyr(3)]octreotate, escape of tumors is likely to occur. Since these cells show an up-regulated sst2 receptor density, a next therapeutic administration of radiolabelled sst2 analogue can be expected to be highly effective.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

The receptor tyrosine kinase MET is a prime target in clinical oncology due to its aberrant activation and involvement in the pathogenesis of a broad spectrum of malignancies. Similar to other targeted kinases, primary and secondary mutations seem to represent an important resistance mechanism to MET inhibitors. Here, we report the biologic activity of a novel MET inhibitor, EMD1214063, on cells that ectopically express the mutated MET variants M1268T, Y1248H, H1112Y, L1213V, H1112L, V1110I, V1206L, and V1238I. Our results demonstrate a dose-dependent decrease in MET autophosphorylation in response to EMD1214063 in five out of the eight cell lines (IC50 2-43nM). Blockade of MET by EMD1214063 was accompanied by a reduced activation of downstream effectors in cells expressing EMD1214063-sensitive mutants. In all sensitive mutant-expressing lines, EMD1214063 altered cell cycle distribution, primarily with an increase in G1 phase. EMD1214063 strongly influenced MET-driven biological functions, such as cellular morphology, MET-dependent cell motility and anchorage-independent growth. To assess the in vivo efficacy of EMD1214063, we used a xenograft tumor model in immunocompromised mice bearing NIH3T3 cells expressing sensitive and resistant MET mutated variants. Animals were randomized for the treatment with EMD1214063 (50mg/kg/day) or vehicle only. Remarkably, five days of EMD1214063 treatment resulted in a complete regression of the sensitive H1112L-derived tumors, while tumor growth remained unaffected in mice with L1213V tumors and in vehicle-treated animals. Collectively, the current data identifies EMD1214063 as a potent MET small molecule inhibitor with selective activity towards mutated MET variants.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

EphB4 receptors, a member of the largest family of receptor tyrosine kinases, are found over-expressed in a variety of tumors cells including glioma cells as well as angiogenic blood vessels. Noninvasive imaging of EphB4 could potentially increase early detection rates, monitor response to therapy directed against EphB4, and improve patient outcomes. Targeted delivery of EphB4 receptor specific peptide conjugated hollow gold nanoshells (HAuNS) into tumors has great potential in cancer imaging and photothermal therapy. In this study, we developed an EphB4 specific peptide named TNYL-RAW and labeled with radioisotope 64Cu and Cy5.5 dye. We also conjugate this specific peptide with hollow gold nanoshells (HAuNS) to evaluate targeted photothermal therapy of cancers. In vitro, 64Cu-DOTA-TNYL- RAW specifically bind to CT26 and PC-3M cells but not to A549 cells. In vivo, Small-animal PET/CT clearly showed the significant uptake of 64Cu-DOTA-TNYL-RAW in CT26 and PC-3M tumors but not in A549 tumors. Furthermore, µPET/CT and near-infrared optical imaging clearly showed the uptake of the dual labeled TNYL-RAW peptide in both U251 and U87 tumors in the brains of nude mice. In U251 tumors, Cy5.5-labeled peptide can bind to EphB4-expressing tumor blood vessels and tumors cells. But in U87 models, dual labeled peptide only could bind to tumor associated blood vessels. Also, Irradiation of PC-3M and CT-26 cell treated with TNYL-PEG-HAuNS nanopatilces with near-infrared (NIR) laser resulted in selective destruction of these cells in vitro. EphB4 targeted TNYL-PEG-HAuNS showed more photothermal killing effect on CT26 tumor model than PEG-HAuNS did. In summary, tumors with overexpression of EphB4 receptors can be noninvasively visualized by micro PET/CT with 64Cu labeled or dual labeled TNYL-RAW peptide. Targeted delivery of TNYL-RAW conjugated HAuNS into tumors can greatly improve the treatment effect of photothermal therapy. The information acquired with this study should be advantageous in improving diagnostics and future applications in photothermal ablation therapy in clinical.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Vasculogenesis is the process by which Endothelial Precursor Cells (EPCs) form a vasculature. This process has been traditionally regarded as an embryological process of vessel formation. However, as early as in the 60's the concept of postnatal vasculogenesis was introduced, with a strong resurface of this idea in recent years. Similarly, previous work on a mouse skin tumor model provided us with the grounds to consider the role of vasculogenesis during tumor formation. ^ We examined the contribution of donor bone marrow (BM)-derived cells to neovascularization in recipient nude mice with Ewing's sarcoma. Ewing's sarcoma is a primitive neuroectodermal tumor that most often affects children and young adults between 5 and 30 years of age. Despite multiple attempts to improve the efficacy of chemotherapy for the disease, the 2-year metastases-free survival rate for patients with Ewing's sarcoma has not improved over the past 15 years. New therapeutic approaches are therefore needed to reduce the mortality rate. ^ The contribution of BM endothelial precursor cells in the development of Ewing's sarcoma was examined using different strategies to track the donor-derived cells. Using a BMT model that takes advantage of MHC differences between donor and recipient mice, we have found that donor BM cells were involved in the formation of Ewing's sarcoma vasculature. ^ Cells responsible for this vasculogenesis activity may be located within the stem cell population of the murine BM. These stem cells would not only generate the hematopoietic lineage but they would also generate ECs. Bone marrow SP (Side Population) cells pertain to a subpopulation that can be identified using flow cytometric analysis of Hoechst 33342-stained BM. This population of cells has HSC activity. We have tested the ability of BM SP cells to contribute to vasculogenesis in Ewing's sarcoma using our MHC mismatched transplant model. Mice transplanted with SP cells developed tumor neovessels that were derived from the donor SP cells. Thus, SP cells not only replenished the hematopoietic system of the lethally irradiated mice, but also differentiated into a non-hematopoietic cell lineage and contributed to the formation of the tumor vasculature. ^ In summary, we have demonstrated that BM-derived cells are involved in the generation of the new vasculature during the growth of Ewing's sarcoma. The finding that vasculogenesis plays a role in Ewing's sarcoma development opens the possibility of using genetically modified BM-derived cells for the treatment of Ewing's sarcomas. ^

Relevância:

60.00% 60.00%

Publicador:

Resumo:

The presentation of MHC class I (MHC-I)/peptide complexes by dendritic cells (DCs) is critical for the maintenance of central tolerance to self and for the regulation of cytotoxic T lymphocytes (CTL)-mediated adaptive immune responses against pathogens and cancer cells. Interestingly, several findings have suggested that the cytoplasmic tail of MHC class I plays a functional role in the regulation of CTL immune responses. For example, our previous studies demonstrated that exon 7-deleted MHC-I molecules not only showed extended DC cell surface half-lives but also induced significantly increased CTL responses to viral challange invivo. Although exon 7-deleted variant of MHC-I does not occur naturally in humans, the animal studies prompted us to examine whether exon 7-deleted MHC-I molecules could generate augmented CTL responses in a therapeutic DC-based vaccine setting. To examine the stimulatory capacity of exon 7-deleted MHC-I molecules, we generated a lentivirus-mediated gene transfer system to induce the expression of different MHC-I cytoplasmic tail isoforms in both mouse and human DCs. These DCs were then used as vaccines in a melanoma mouse tumor model and in a human invitro co-culture system. In this thesis, we show that DCs expressing exon 7-deleted MHC-I molecules, stimulated remarkably higher levels of T-cell cytokine production and significantly increased the proliferation of meanoma-specific (Pmel-1) T cells compared with DCs expressing wild type MHC-I. We also demonstrate that, in combination with adoptive transfer of Pmel-1 T-cell, DCs expressing exon 7-deleted Db molecules induced greater anti-tumor responses against established B16 melanoma tumors, significantly extending mouse survival as compared to DCs expressing wild-type Db molecules. Moreover, we also observed that human DCs expressing exon 7-deleted HLA-A2 molecules showed similarly augmented CTL stimulatory ability. Mechanistic studies suggest that exon 7-deleted MHC-I molecules showed impaired lateral membrane movement and extended cell surface half-lives within the DC/T-cell interface, leading to increased spatial availability of MHC-I/peptide complexes for recognition by CD8+ T cells. Collectively, these results suggesr that targeting exon 7 within the cytoplasmic tail of MHC-I molecules in DC vaccines has the potential to enhance CD8+ T cell stimulatory capacity and improve clinical outcomes in patients with cancer or viral infections.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Tumor growth often outpaces its vascularization, leading to development of a hypoxic tumor microenvironment. In response, an intracellular hypoxia survival pathway is initiated by heterodimerization of hypoxia-inducible factor (HIF)-1α and HIF-1β, which subsequently upregulates the expression of several hypoxia-inducible genes, promotes cell survival and stimulates angiogenesis in the oxygen-deprived environment. Hypoxic tumor regions are often associated with resistance to various classes of radio- or chemotherapeutic agents. Therefore, development of HIF-1α/β heterodimerization inhibitors may provide a novel approach to anti-cancer therapy. To this end, a novel approach for imaging HIF-1α/β heterodimerization in vitro and in vivo was developed in this study. Using this screening platform, we identified a promising lead candidate and further chemically derivatized the lead candidate to assess the structure-activity relationship (SAR). The most effective first generation drug inhibitors were selected and their pharmacodynamics and anti-tumor efficacy in vivo were verified by bioluminescence imaging (BLI) of HIF-1α/β heterodimerization in the xenograft tumor model. Furthermore, the first generation drug inhibitors, M-TMCP and D-TMCP, demonstrated efficacy as monotherapies, resulting in tumor growth inhibition via disruption of HIF-1 signaling-mediated tumor stromal neoangiogenesis.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

α-Melanocyte stimulating hormone (α-MSH) analogs, cyclized through site-specific rhenium (Re) and technetium (Tc) metal coordination, were structurally characterized and analyzed for their abilities to bind α-MSH receptors present on melanoma cells and in tumor-bearing mice. Results from receptor-binding assays conducted with B16 F1 murine melanoma cells indicated that receptor-binding affinity was reduced to approximately 1% of its original levels after Re incorporation into the cyclic Cys4,10, d-Phe7–α-MSH4-13 analog. Structural analysis of the Re–peptide complex showed that the disulfide bond of the original peptide was replaced by thiolate–metal–thiolate cyclization. A comparison of the metal-bound and metal-free structures indicated that metal complexation dramatically altered the structure of the receptor-binding core sequence. Redesign of the metal binding site resulted in a second-generation Re–peptide complex (ReCCMSH) that displayed a receptor-binding affinity of 2.9 nM, 25-fold higher than the initial Re–α-MSH analog. Characterization of the second-generation Re–peptide complex indicated that the peptide was still cyclized through Re coordination, but the structure of the receptor-binding sequence was no longer constrained. The corresponding 99mTc- and 188ReCCMSH complexes were synthesized and shown to be stable in phosphate-buffered saline and to challenges from diethylenetriaminepentaacetic acid (DTPA) and free cysteine. In vivo, the 99mTcCCMSH complex exhibited significant tumor uptake and retention and was effective in imaging melanoma in a murine-tumor model system. Cyclization of α-MSH analogs via 99mTc and 188Re yields chemically stable and biologically active molecules with potential melanoma-imaging and therapeutic properties.