• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 123
  • 31
  • 21
  • 15
  • 12
  • 9
  • 9
  • 7
  • 3
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 282
  • 282
  • 33
  • 29
  • 28
  • 24
  • 24
  • 23
  • 23
  • 20
  • 18
  • 18
  • 17
  • 16
  • 16
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
231

Évaluation des propriétés biologiques de sels de biguanidium : perturbation membranaire et applications dans le traitement du cancer du pancréas

Hébert, Audrey 08 1900 (has links)
La metformine, un médicament couramment utilisé pour le traitement du diabète de type II, fut récemment identifiée comme un composé ayant des propriétés anticancéreuses très intéressantes, notamment pour le cancer du pancréas. Toutefois, malgré les nombreuses expériences in vitro et sur des modèles murins qui ont confirmé cet effet, les essais cliniques sur les humains sont restés infructueux. Un des facteurs mis en cause pour expliquer ces résultats est la grande hydrophilie de la metformine, qui diminue sa biodisponibilité et limite son transport au travers des membranes cellulaires. Nous nous sommes donc intéressés à la synthèse de sels de biguanidium amphiphiles inspirés de la metformine qui se partitionnent plus facilement dans les bicouches phospholipidiques et qui possèdent de meilleures activités anticancéreuses. Pour ce faire, nous avons tout d’abord étudié la perturbation des membranes par de simples alkylbiguanidium. Nous avons démontré que ces composés peuvent transporter des ions H+/OH- et dépolariser les membranes bactériennes, ce qui leur confère des propriétés antibactériennes et antifongiques. Afin de limiter l’hémolyse associée à ces composés, des sels de biguanidium substitués par le groupement phényléthynylbenzyle ont par la suite été synthétisés. La structure de ceux-ci leur permet de mieux se partitionner dans les membranes et s’accumuler dans les mitochondries, tout en diminuant la toxicité associée à une perturbation membranaire trop forte. Leur activité sur les cellules cancéreuses du pancréas est ainsi beaucoup plus importante que celle de la metformine, de même que leur capacité à inhiber la croissance de xénogreffes chez les souris. Ces résultats nous ont ensuite amené vers la synthèse d’une petite librairie de sels de biguanidium et l’étude de leurs activités anticancéreuses et antibactériennes. Les modifications structurales et les changements de contre-ion apportés à cette librairie ont permis d’obtenir des composés encore plus efficaces et surtout beaucoup plus sélectifs envers les cellules saines, ouvrant ainsi la porte à une nouvelle classe de médicaments anticancéreux à base de sels de biguanidium. / Metformin, a common drug used for the treatment of type II diabetes, has recently been linked to interesting anticancer properties, notably on pancreatic cancer. Although there have been many experiments in vitro and on murine models that have confirmed this effect, human clinical trials featuring metformin have been unsuccessful. One of the reasons brought forward to explain these results is the high hydrophilicity of metformin, which limits its bioavailability and transport through cellular membranes. For this reason, we have been interested in the synthesis of amphiphilic biguanidium salts inspired from metformin that can partition more easily in phospholipid membranes and thus have better anticancer properties. We first studied the membrane perturbation properties of simple alkylbiguanidium salts and showed that these compounds can transport H+/OH- ions and depolarize bacterial membranes, which in turn gives them antibacterial and antifungal properties. To limit the hemolytic activity associated with these compounds, biguanidium salts substituted by the phenylethynylbenzyl moiety were then synthesised. Their structure allows them to partition more easily in membranes and accumulate in mitochondria, while lowering the toxicity associated with high membrane perturbation. For those reasons, their activity on pancreatic cancer cells is much higher than metformin, as is their inhibition of xenograft growth in mice. These results encouraged us to synthesise a small library of biguanidium salts and study their anticancer activity. The structural modifications and counter-anion variations brought to this library have improved the efficiency of these compounds as well as their selectivity towards healthy cells, thus opening the door to a new class of anticancer drugs based on biguanidium salts.
232

Einfluss des Histondeacetylase-Inhibitors 4-Phenylbutyrat auf das Wachstum des experimentell-induzierten Pankreaskarzinoms

Friske, Alexandra 14 April 2015 (has links)
Das Pankreaskarzinom bleibt trotz verbesserter Diagnose- und Therapiemöglichkeiten weiterhin eine Krankheit mit einer sehr schlechten Prognose und Lebenserwartung nach Diagnosestellung. Eine innovative Therapiemöglichkeit stellt eine Gruppe von Histondeacetylase-Inhibitoren dar, die einen direkten Einfluss auf die Regulation der Genexpression in Tumorzellen haben. Das Ziel der vorliegenden Arbeit bestand darin, die Wirkung des HDAC-Inhibitors 4-Phenylbutyrat auf Pankreaskarzinomzellen in-vitro und vor allem in-vivo zu untersuchen. Neben dem Einfluss auf die Zellproliferation in-vitro und in-vivo wurde in-vivo im subkutanen und orthotopen Tumormodell der Einfluss auf Tumorwachstum, Zellproliferation, Nekroseausbreitung, Regulation des Connexin 43 und Histonacetylierung im Tumorgewebe untersucht. Die Untersuchungen zeigen, dass 4-PB durch seinen hemmenden Effekt auf das Wachstum von Xenografttumoren und auf die Proliferation von Pankreastumorzellen sowie durch seine fördernde Wirkung auf die Expression von Connexin 43, Acetylierung von H4 und Bildung eine Pseudokapsel, ein potentiell wirksames Medikament bei der experimentellen Behandlung des Pankreaskarzinoms ist.
233

Die Dysregulation der Apoptose im Pankreaskarzinom und ein darauf basierendes multimodales Therapiekonzept

Werner, Kristin 03 June 2016 (has links)
Weltweit hat das duktale Adenokarzinom des Pankreas (PDAC) unter den Krebserkrankungen die schlechteste Prognose. Aufgrund der unspezifischen Symptome wird die Erkrankung meist erst in einem lokal fortgeschrittenem Stadium diagnostiziert, wenn eine kurative Tumorresektion nicht mehr möglich ist und es bereits zur Metastasierung gekommen ist. Trotz intensiver klinischer Forschung wird mit bisherigen Therapieansätzen wie zum Beispiel Gemcitabin (auch in Kombination mit nab-Paclitaxel) nur eine geringfügige Verbesserung des medianen Überlebens erzielt. Diese Therapieresistenz gegenüber Gemcitabin wurde in in vitro Versuchen mit verschiedenen humanen PDAC-Zelllinien bestätigt. Die zusätzlich behandelte nichttumorigene Pankreasdukt-Zelllinie HDPE-E6E7 zeigte hingegen eine starke Sensitivität gegenüber dem Chemotherapeutikum. Ursächlich beteiligt an der generellen Resistenz dieser Tumore gegenüber Chemo- und Strahlentherapie sind verschiedene Apoptose-Evasionsmechanismen. Diese sind vor allem durch ein Ungleichgewicht pro- und antiapoptotischer Faktoren bedingt (Lowe 2004). Zusätzlich fördern KRAS-Mutationen, die nahezu universell in allen PDACs vorliegen, die Proliferation der Tumorzellen und unterstützen die Apoptose-Dysregulation durch eine verstärkte Expression antiapoptotischer Proteine. Nach umfangreichen Literaturrecherchen (Werner 2011a, Werner 2011b) wurden verschiedenste PDAC-Zelllinien hinsichtlich ihrer Expression von Apoptose-assoziierten Genen analysiert. Untersucht wurden diesbezüglich verschiedene humane PDAC-Zelllinien, aber auch Primärzellkulturen (PaCaDD-Zellen), die in unserem Labor per Outgrowth-Methode aus Patiententumorgewebe etabliert wurden. Ergänzend wurden murine Zelllinien analysiert, die aus einem genetischen PDAC-Mausmodell (KRASG12D; P53R172H; PDX1 CRE) stammen. Normiert bezüglich der Expression der HDPE-E6E7-Zellen wurde eine vielfältige, sehr heterogene Dysregulation von verschiedenen, mit der Apoptose assoziierten Proteinen festgestellt. Per siRNA-basiertem Knockdown wurden verschiedene Kandidatengene hinsichtlich ihrer funktionellen Bedeutung für die Zellproliferation und Apoptose-Induktion näher charakterisiert. In einem dynamischen Prozess wurde eine multimodale Therapie entwickelt, bei der fünf antiapoptotische Zielgene (BCLXL, FLIP, MCL1L, SURVIVIN und XIAP) kombiniert mit KRAS als zentralem Onkogen simultan in ihrer Expression inhibiert wurden. Ziel war es, hierdurch sowohl die extrinsische als auch intrinsische Apoptose zu normalisieren und eine möglicherweise durch Caspase-Inhibitoren blockierte Signaltransduktion zu ermöglichen. Ein zusätzlicher Knockdown von KRAS sollte einer Gegenregulierung dieser Therapie vorbeugen und die gesteigerte Proliferation der Tumorzellen unterbinden. Dieser so genannte SGS6-Therapieansatz zeigte in vitro in allen fünf getesteten humanen sowie in zwei murinen Zelllinien eine starke Apoptose-Induktion und Verminderung der Zellzahl. Durch Zweit-siRNAs wurde die spezifische Wirksamkeit der Knockdowns bestätigt und zelluläre off-target-Effekte wurden ausgeschlossen. Auch in vivo wurde in einem subkutanen Allograftmodell mit dem SGS6-Therapiekonzept eine starke Tumorreduktion erzielt. Die analoge Untersuchung der nichttumorösen Pankreasdukt-Zelllinie HDPE-E6E7 zeigte, dass die SGS6-Therapie deutlich spezifischer für die Krebszellen war verglichen zur Behandlung mit Gemcitabin.
234

Auswirkung der portalvenösen Infiltration nach kurativer Resektion duktaler Adenokarzinome des Pankreas auf das Metastasierungsmuster und das progressionsfreie Überleben: Eine retrospektive Kohortenstudie

Mierke, Franz 05 December 2017 (has links)
Hintergrund: Ziel der Studie war der Vergleich von Patienten mit duktalem Pankreaskarzinom (PDAC) im progressionsfreien und Gesamtüberleben sowie im Rezidivmuster in Abhängigkeit einer Resektion der Vena portae oder der Vena mesenterica superior (PV/SMV). Methoden: Es wurde eine retrospektive Analyse durchgeführt. Hierbei wurden Patienten betrachtet, die zwischen 2005 und 2015 eine pyloruserhaltende partielle Pankreatoduodenektomie (PPPD), eine klassische Pankreatoduodenektomie (kPD) oder eine totale Pankreatektomie (TP) erhielten. Diese wurden in drei Gruppen eingeteilt. Die P+I+- Gruppe bestand aus Patienten mit Venenresektion (P+), bei denen eine pathohistologische Infiltration der PV oder SMV vorlag (I+). Fand sich bei durchgeführter Venenresektion keine portalvenöse Infiltration (I-), wurden die Patienten der P+I--Gruppe zugeordnet. Als Kontrollgruppe galten Patienten ohne Venenresektion (P-I-), welche zu denen der P+I+- Gruppe gematcht wurden. Die statistischen Analysen wurden mit dem R Softwarepaket durchgeführt. Das Signifikanzlevel wurde für alle Berechnungen auf α = 0,05 festgelegt. Ergebnisse: Insgesamt wurden 179 Patienten eingeschlossen. 113 erhielten eine portalvenöse Resektion. Davon hatten 36 (31,9%) eine pathohistologische Lumeninfiltration (P+I+), bei 77 (68,1%) lag dagegen keine Infiltration vor (P+I-). 66 Patienten ohne Venenresektion wurden zu den Patienten der P+I+-Gruppe gematcht (P-I-). Zwischen den drei Gruppen waren die meisten pathohistologischen Parameter vergleichbar. 17 Patienten (9,5%) wurden neoadjuvant therapiert, davon erhielten 16 eine Venenresektion (P+). Für das Gesamtüberleben konnten signifikante Unterschiede nachgewiesen werden (11,9 Monate [P+I+] vs. 16,1 Monate [P+I-] vs. 20,1 Monate [P-I-]; p=0,01). In der univariaten Überlebensanalyse konnte für den erhöhten präoperativen CA19-9 Wert, den Resektionsstatus (R), den Lymphknotenstatus (N), das Lymphknotenverhältnis (LNR), die mikroskopische Veneninvasion (V) sowie die pathohistologisch gesicherte Infiltration der PV/SMV ein negativer Einfluss nachgewiesen werden. In der multivariaten Analyse blieb die wahre Infiltration der PV/SMV als einziger signifikanter negativer Einflussfaktor auf das Gesamtüberleben erhalten (p=0,014). Die Inzidenz an Fernmetastasen war in der P+I+- Gruppe signifikant erhöht (75% [P+I+] vs. 45,8% [P+I-] vs. 54,7% [P-I-], p=0,01). Für ein Lokalrezidiv fanden sich dagegen keine Häufigkeitsunterschiede zwischen den Gruppen (p=0,96). Das mediane progressionsfreie Überleben war für Patienten der P+I+-Gruppe signifikant verkürzt (7,4 Monate [P+I+] vs. 10,9 Monate [P+I-] vs. 11,6 Monate [P-I-]; p=0,02). Die Lumeninfiltration der PV/SMV, die mikroskopische Veneninvasion (V), der präoperative CA19-9 Wert sowie der Differenzierungsgrad (G) waren negative Einflussfaktoren auf das progressionsfreie Überleben. In der multivariaten Analyse blieben die pathohistologisch gesicherte Infiltration sowie das Grading als negative unabhängige Einflussfaktoren nachweisbar. In 25% der Fälle manifestierte sich das Rezidiv initial in der Leber. Schlussfolgerung: Die pathohistologisch gesicherte Infiltration der PV/SMV ist ein unabhängiger Risikofaktor für das progressionsfreie und das Gesamtüberleben. Die Inzidenz an Fernmetastasen ist für die Patienten der P+I+-Gruppe erhöht. Eine potentiell kurative venöse Resektion kann den Einfluss der aggressiven Tumorbiologie und des fortgeschrittenen Krankheitsbildes nicht vollständig kompensieren. / Background. The present study aims to evaluate the longterm outcome and metastatatic pattern of patients who underwent an operation for pancreatic ductal adenocarcinoma (PDAC) with portal or superior mesenteric vein (PV/SMV) resection. Methods. Patients who underwent a pylorus preserving pancreaticoduodenectomy (PPPD), Whipple procedure (kPD) or total pancreatoduodenectomy (TP) between 2005 and 2015 were retrospectively analyzed. The patients were categorized in three subgroups. Those whom received a vein resection with pathohistological tumor invasion of the PV/SMV (P+I+) those at whom underwent vein resection but without pathohistological tumor invasion (P+I-) and lastly a third group (P-I-) matched to the P+I+ included patients without vein resection. Statistical analysis was performed using the R software package. The significance level for all calculations was set at α = 0.05. Results. The study cohort included 179 patients, 113 of whom underwent simultaneous PV/SMV resection. 36 patients (31,9%) had pathohistological tumor infiltration (P+I+), 77 (68,1%) did not (P+I-). 66 patients without vein resection (P-I-) were balanced by the P+I+ group. Most of pathohistological tumor characteristics were comparable between groups. 17 patients (9.5%) received neoadjuvant therapy, 16 of them were in vein resection group (P+). The study revealed differences in overall median survival (11.9 months [P+I+] vs. 16.1 months [P+I-] vs. 20.1 months [P-I-]; p=0.01). Univariate survival analysis shown negative consequences for CA19-9, resection margin (R), status of nodal metastasis (N), lymph node ratio (LNR), microvascular vein invasion (V) and true invasion of the PV/SMV. Multivariate survival analysis identified true invasion of the PV/SMV as the only significant, negative prognostic factor (p= 0.01). Whereas the incidence of local tumor recurrence was comparable (p=0.96), the proportion of patients with distant metastasis showed significant differences (75% [P+I+] vs. 45.8% [P+I-] vs 54.7% [P-I-]; p=0.01). The median time to progression were significantly shorter if the PV/SMV was infiltrated (7,4 months [P+I+] vs. 10,9 months [P+I-] vs. 11,6 months [P-I-]; p=0.02). Univariate progression analysis revealed significances for true invasion of the PV/SMV, microvascular vein invasion (V), CA19-9 and histologic classification (G). Multivariate progression analysis detected pathohistological invasion of the PV/SMV and histologic classification (G) as independent factors. Initial liver metastasis occurred in 25% of the patients. Conclusions. Pathohistological invasion of the PV/SMV is an independent risk factor for overall and progression free survival. Patients of P+I+-group had a higher incidence of distant metastasis, local progression is comparable. Even radical and complete resection cannot completely compensate for aggressive tumor biology and advanced disease. Modifiziert nach Mierke et al., 2016
235

From cancer gene expression to protein interaction: Interaction prediction, network reasoning and applications in pancreatic cancer

Daw Elbait, Gihan Elsir Ahmed 16 June 2009 (has links)
Microarray technologies enable scientists to identify co-expressed genes at large scale. However, the gene expression analysis does not show functional relationships between co-expressed genes. There is a demand for effective approaches to analyse gene expression data to enable biological discoveries that can lead to identification of markers or therapeutic targets of many diseases. In cancer research, a number of gene expression screens have been carried out to identify genes differentially expressed in cancerous tissue such as Pancreatic Ductal Adenocarcinoma (PDAC). PDAC carries very poor prognosis, it eludes early detection and is characterised by its aggressiveness and resistance to currently available therapies. To identify molecular markers and suitable targets, there exist a research effort that maps differentially expressed genes to protein interactions to gain an understanding at systems level. Such interaction networks have a complex interconnected structure, whose the understanding of which is not a trivial task. Several formal approaches use simulation to support the investigation of such networks. These approaches suffer from the missing knowledge concerning biological systems. Reasoning in the other hand has the advantage of dealing with incomplete and partial information of the network knowledge. The initial approach adopted was to provide an algorithm that utilises a network-centric approach to pancreatic cancer, by re-constructing networks from known interactions and predicting novel protein interactions from structural templates. This method was applied to a data set of co-expressed PDAC genes. To this end, structural domains for the gene products are identified by using threading which is a 3D structure prediction technique. Next, the Protein Structure Interaction Database (SCOPPI), a database that classifies and annotates domain interactions derived from all known protein structures, is used to find templates of structurally interacting domains. Moreover, a network of related biological pathways for the PDAC data was constructed. In order to reason over molecular networks that are affected by dysregulation of gene expression, BioRevise was implemented. It is a belief revision system where the inhibition behaviour of reactions is modelled using extended logic programming. The system computes a minimal set of enzymes whose malfunction explains the abnormal expression levels of observed metabolites or enzymes. As a result of this research, two complementary approaches for the analysis of pancreatic cancer gene expression data are presented. Using the first approach, the pathways found to be largely affected in pancreatic cancer are signal transduction, actin cytoskeleton regulation, cell growth and cell communication. The analysis indicates that the alteration of the calcium pathway plays an important role in pancreas specific tumorigenesis. Furthermore, the structural prediction method reveals ~ 700 potential protein-protein interactions from the PDAC microarray data, among them, 81 novel interactions such as: serine/threonine kinase CDC2L1 interacting with cyclin-dependent kinase inhibitor CDKN3 and the tissue factor pathway inhibitor 2 (TFPI2) interacting with the transmembrane protease serine 4 (TMPRSS4). These resulting genes were further investigated and some were found to be potential therapeutic markers for PDAC. Since TMPRSS4 is involved in metastasis formation, it is hypothesised that the upregulation of TMPRSS4 and the downregulation of its predicted inhibitor TFPI2 plays an important role in this process. The predicted protein-protein network inspired the analysis of the data from two other perspectives. The resulting protein-protein interaction network highlighted the importance of the co-expression of KLK6 and KLK10 as prognostic factors for survival in PDAC as well as the construction of a PDAC specific apoptosis pathway to study different effects of multiple gene silencing in order to reactivate apoptosis in PDAC. Using the second approach, the behaviour of biological interaction networks using computational logic formalism was modelled, reasoning over the networks is enabled and the abnormal behaviour of its components is explained. The usability of the BioRevise system is demonstrated through two examples, a metabolic disorder disease and a deficiency in a pancreatic cancer associated pathway. The system successfully identified the inhibition of the enzyme glucose-6-phosphatase as responsible for the Glycogen storage disease type I, which according to literature is known to be the main reason for this disease. Furthermore, BioRevise was used to model reaction inhibition in the Glycolysis pathway which is known to be affected by Pancreatic cancer.
236

γ-Tocotrienol Induces Apoptosis in Pancreatic Cancer Cells by Upregulation of Ceramide Synthesis and Modulation of Sphingolipid Transport

Palau, Victoria E., Chakraborty, Kanishka, Wann, Daniel, Lightner, Janet, Hilton, Keely, Brannon, Marianne, Stone, William, Krishnan, Koyamangalath 16 May 2018 (has links)
Background: Ceramide synthesis and metabolism is a promising target in cancer drug development. γ-tocotrienol (GT3), a member of the vitamin E family, orchestrates multiple effects that ensure the induction of apoptosis in both, wild-type and RAS-mutated pancreatic cancer cells. Here, we investigated whether these effects involve changes in ceramide synthesis and transport. Methods: The effects of GT3 on the synthesis of ceramide via the de novo pathway, and the hydrolysis of sphingomyelin were analyzed by the expression levels of the enzymes serine palmitoyl transferase, ceramide synthase-6, and dihydroceramide desaturase, and acid sphingomyelinase in wild-type RAS BxPC3, and RAS-mutated MIA PaCa-2 and Panc 1 pancreatic cancer cells. Quantitative changes in ceramides, dihydroceramides, and sphingomyelin at the cell membrane were detected by LCMS. Modulation of ceramide transport by GT3 was studied by immunochemistry of CERT and ARV-1, and the subsequent effects at the cell membrane was analyzed via immunofluorescence of ceramide, caveolin, and DR5. Results: GT3 favors the upregulation of ceramide by stimulating synthesis at the ER and the plasma membrane. Additionally, the conversion of newly synthesized ceramide to sphingomyelin and glucosylceramide at the Golgi is prevented by the inhibition of CERT. Modulation ARV1 and previously observed inhibition of the HMG-CoA pathway, contribute to changes in membrane structure and signaling functions, allows the clustering of DR5, effectively initiating apoptosis. Conclusions: Our results suggest that GT3 targets ceramide synthesis and transport, and that the upregulation of ceramide and modulation of transporters CERT and ARV1 are important contributors to the apoptotic properties demonstrated by GT3 in pancreatic cancer cells.
237

NONINVASIVE CHARACTERIZATION AND DEVELOPMENT OF IN SITU FORMING IMPLANTS FOR USE AS A LOCAL PANCREATIC CANCER THERAPY

Kelsey A Hopkins (12468513) 27 April 2022 (has links)
<p>Pancreatic ductal adenocarcinoma is an especially deadly disease having the lowest 5-year survival rate of any major cancer at just 11%. As in many cancers, systemically-delivered chemotherapy forms the backbone of clinical treatment. However, limitations of systemic delivery exacerbated by the unique desmoplastic and avascular microenvironment surrounding the pancreatic tumor cells result in the failed efficacy of current treatments. The high stromal content in the microenvironment, which is especially overabundant in hyaluronic acid, is thought to physically impede drug perfusion into the tissue. Thus, there is clearly a <strong>critical need</strong> to develop novel treatments for pancreatic ductal adenocarcinoma that can overcome these drug delivery barriers. Long-acting injectable implants offer an attractive drug delivery method that can provide <em><strong>sustained</strong></em> drug release directly at the <em><strong>local</strong></em> targeted site, rather than transient, systemic release. Here we use in situ forming implants (ISFIs), which are a low-viscosity solution outside of the body but transition into a solid drug-eluting depot after injection into an aqueous environment. Our <strong>objective</strong> is to develop and characterize an ISFI that can provide sustained release of bioactive hyaluronidase for use as an intratumoral injection to degrade hyaluronic acid in pancreatic tumors. This work was accomplished in four aims. First, a method was developed using diffusion-weighted MRI for noninvasive characterization of the implants. Second, because hyaluronidase is a protein drug, we studied factors affecting protein release from ISFIs, focusing on external factors of the injection site. Third, we showed that basic salt additives can be used to neutralize the acidic environment created by the implants which may improve protein stability. Finally, we formulated an implant to provide sustained release of hyaluronidase and demonstrated retention of its bioactivity both <em>in vitro</em> and <em>ex vivo</em>.</p>
238

Nouvelle approche d'imagerie pour l’étude de la biodistribution de nanomédicaments / New imaging approach to the biodistribution study of innovative nanomedicine

Epaule, Céline 04 December 2017 (has links)
La distribution in vivo des médicaments est étudiée par des techniques quantitatives faiblement ou non résolues spatialement. Avec l'apparition des thérapies personnalisées, des études plus approfondies sont nécessaires pour connaître précisément le comportement des molécules vectorisées sous la forme de nanoparticules (NPs). Dans le cadre du programme européen Ternanomed, ce projet de recherche a pour objectif d’évaluer la capacité de deux techniques d’imagerie appliquées à l’étude de la distribution de nanomédicaments à base de squalène et de Cis platine (Cis-Pt). Ces deux techniques ont été sélectionnées pour leur apport d’informations complémentaires à l’échelle des organes et des tissus : i) l’imagerie par résonnance magnétique (IRM) pour suivre in vivo la biodistribution de NPs modèles à base de Cis-Pt et BiSqualène (BiSQ), marquées par des agents de contraste type oxyde de fer (USPIO), ii) l’imagerie de microfluorescence X, couplée au rayonnement synchrotron (SR-μXRF), qui ne nécessite pas de marquage préalable des nanomédicaments, pour le suivi tissulaire du Cis-Pt.Concernant l’approche par IRM, nous avons encapsulé avec succès nos USPIO synthétisées au sein des NPs de Cis-Pt BiSQ (210nm, polydispersité 0,1), tout en leur conférant un pouvoir contrastant à 7 tesla (r2=404 ms.mol-1 et r1=3 ms.mol-1). Ces NPs nouvellement préparées sont également traçables chez notre modèle murin Nudes. Les résultats de biodistribution montrent une arrivée rapide du contraste dans les organes épurateurs : le foie et la rate (5 minutes après l’injection). Au final, l’analyse par IRM a permis d’obtenir les données de biodistribution en temps réel des NPs à base de Cis Pt BiSQ, grâce au suivi du contraste apporté par les USPIO encapsulés. Concernant l’imagerie par SR-μXRF, nous avons démontré que cette technique est suffisamment sensible pour détecter et cartographier le Cis-Pt, vectorisé par nos NPs modèles. La distribution du Cis Pt a été quantifiée localement à partir d’une référence interne de concentration connue, le Zinc, à partir de notre méthode validée par le dosage globale du Platine par spectrométrie d’absorption atomique. Lorsque notre référence tissulaire n’est pas distribuée de façon homogène, une méthode semi-quantitative a été mise au point pour comparer la distribution à 2h, 8h et 24h, tel qu’au niveau des coupes de tumeurs PANC-1.Au final, ces travaux ont permis de démontrer, que la SR-μXRF et l’IRM sont des approches de choix pour l’étude pharmacocinétique et pharmacodynamique de nanomédicaments tels que les NPs à base de Cis-Pt. La technique de microfluorescence X contribue au caractère original et pionnier de ce travail de recherche, apportant des nouveaux résultats de détection et quantification important dans le domaine des nanomédecines. / Nowadays, the in vivo distribution of drugs is studied by non-spatial or partially spatial quantitative techniques. With the development of personalized therapies, many studies are required to know the in vivo behaviour of these innovative treatments, which target drugs, such as nanoparticles (NPs). Into the European funded program Ternanomed, the aim of this multidisciplinary research project was to evaluate two complementary imaging methods to study the distribution of squalene and Cis platinum (Cis Pt) NPs. The 2 imaging methods were selected to provide complementary data at the scale of organs and tissues: i) Magnetic resonance imaging (MRI) to monitor the in vivo biodistribution of NPs models based on Cis-Pt and BiSqualene (BiSQ), labelled with "UltraSmall Iron Oxide Particle" (USPIO) contrast agents, ii) X-ray microfluorescence imaging, coupled with synchrotron radiation (SR-μXRF) without any labelling of these nanomedicines, by following the Cis-Pt drug distribution into tissues.Regarding the MRI approach, we first successfully prepared Cis-Pt BiSQ NPs loading with USPIO (210nm, polydispersity 0,1). These NPs were given a contrast at 7 Tesla (r2 = 404 ms.mol-1 and r1 = 3 ms.mol-1). These newly prepared and characterized NPs were also trackable into our Nude murine model. The results show a rapid arrival of contrast in the liver and spleen scavengers (5 minutes after injection). Ultimately, MRI analysis yielded real-time biodistribution data for Cis-Pt BiSQ-based NPs by monitoring the contrast provided by encapsulated USPIO. Regarding the SR-μXRF imaging analysis, we demonstrated that this technique is very sensitive to detect and map the Cis-Pt distribution, the drug vectorized by our squalene NPs models. Additionally, a local quantitative analysis is feasible when a microelement present in the tissue is used as a reference, in our study the Zinc element. The distribution of Cis-Pt was quantified in the hepatic, renal and fat tissues, after 2h and 24h, with our method validated by the global Platinum microanalyse using atomic absorption spectrometry. When the tissue reference appears not homogenously distributed, a semi-quantitative analysis method is possible to compare the distribution such as into PANC-1 tumour sections.Finally, these two complementary approaches illustrate the use of SR-μXRF and lay the optimized bases of MRI to study the pharmacokinetics and pharmacodynamics of two new types of Cis-Pt/squalene NPs. The SR-μXRF technique, newly used in pharmaceutical field, had an effective contribution to these original and pioneering research studies with an original way of in vivo assessment of the distribution of drug embedded into nanomedicine system. The issue of detecting correct and measurable distribution of the drugs is extremely important, timely and relevant to improve current knowledge in the state of the art. This research study brings new data which can produce significant impact to the overall area of nanomedicine.
239

Tumor-stroma interaction mediated by tissue transglutaminase in pancreatic cancer

Lee, Jiyoon 08 July 2015 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Pancreatic ductal adenocarcinoma (PDA) is a deadly disease due to early metastasis and resistance to chemotherapy. PDA is commonly associated with a dense desmoplastic stroma, which forms a protective niche for cancer cells. Tissue transglutaminase (TG2), a Ca2+-dependent enzyme, is abundantly expressed in pancreatic cancer cells and crosslinks proteins through acyl-transfer transamidation between glutamine and lysine residues. The objective of the study was to determine the functions of TG2 in the pancreatic stroma. Orthotopic pancreatic xenografts and co-culture systems tested the mechanisms by which the enzyme modulates tumor-stroma interactions. We showed that TG2 secreted by cancer cells is enzymatically active and renders the stroma denser by crosslinking collagen, which in turn activates fibroblasts and stimulates their proliferation. Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are transcription factors involved in mechanotransduction. The TG2-mediated fibrosis-rich, stiff microenvironment conveys mechanical cues to cancer cells leading to activation of YAP and TAZ, promoting cell proliferation and tumor growth. Stable knockdown of TG2 in pancreatic cancer cells led to decreased size of pancreatic xenografts and increased sensitivity of xenografts to gemcitabine. Taken together, our results demonstrate that TG2 secreted in the tumor microenvironment orchestrates the crosstalk between cancer cells and the stroma, fundamentally impacting tumor growth and response to chemotherapy. Our study supports TG2 inhibition in the pancreatic stroma as a novel strategy to block pancreatic cancer progression.
240

Angiogenic gene signature in human pancreatic cancer correlates with TGF-beta and inflammatory transcriptomes

Craven, Kelly E. 11 April 2016 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Pancreatic ductal adenocarcinoma (PDAC), which comprises 85% of pancreatic cancers, is the 4th leading cause of cancer death in the United States with a 5-year survival rate of 8%. While human PDACs (hPDACs) are hypovascular, they also overexpress a number of angiogenic growth factors and receptors. Additionally, the use of anti-angiogenic agents in murine models of PDAC leads to reduced tumor volume, tumor spread, and microvessel density (MVD), and improved survival. Nonetheless, clinical trials using anti-angiogenic therapy have been overwhelmingly unsuccessful in hPDAC. On the other hand, pancreatic neuroendocrine tumors (PNETs) account for only 2% of pancreatic tumors, yet they are very vascular and classically angiogenic, respond to anti-angiogenic therapy, and confer a better prognosis than PDAC even in the metastatic setting. In an effort to compare and contrast the angiogenic transcriptomes of these two tumor types, we analyzed RNA-Sequencing (RNA-Seq) data from The Cancer Genome Atlas (TCGA) and found that a pro-angiogenic gene signature is present in 35% of PDACs and that it is mostly distinct from the angiogenic signature present in PNETs. The pro-angiogenic PDAC subgroup also exhibits a transcriptome that reflects active TGF-β signaling, less frequent SMAD4 inactivation than PDACs without the signature, and up-regulation of several pro-inflammatory genes, including members of JAK signaling pathways. Consequently, targeting the TGF-β receptor type-1 kinase with SB505124 and JAK1/2 with ruxolitinib blocks proliferative crosstalk between human pancreatic cancer cells (PCCs) and human endothelial cells (ECs). Additionally, treatment of the KRC (oncogenic Kras, homozygous deletion of Rb1) and KPC (oncogenic Kras, mutated Trp53) genetically engineered PDAC mouse models with ruxolitinib suppresses murine PDAC (mPDAC) progression only in the KRC model, which shows superior enrichment and differential expression of the human pro-angiogenic gene signature as compared to KPC tumors. These findings suggest that targeting both TGF-β and JAK signaling in the 35% of PDAC patients whose cancers exhibit an pro-angiogenic gene signature should be explored in a clinical trial.

Page generated in 0.2973 seconds