• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 3
  • 2
  • Tagged with
  • 17
  • 17
  • 7
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 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.
1

Carfilzomib demonstrates broad anti-tumor activity in pre-clinical non-small cell and small cell lung cancer models

Baker, Amanda F., Hanke, Neale T., Sands, Barbara J., Carbajal, Liliana, Anderl, Janet L., Garland, Linda L. January 2014 (has links)
BACKGROUND: Carfilzomib (CFZ) is a proteasome inhibitor that selectively and irreversibly binds to its target and has been approved in the US for treatment of relapsed and refractory multiple myeloma. Phase 1B studies of CFZ reported signals of clinical activity in solid tumors, including small cell lung cancer (SCLC). The aim of this study was to investigate the activity of CFZ in lung cancer models. METHODS: A diverse panel of human lung cancer cell lines and a SHP77 small cell lung cancer xenograft model were used to investigate the anti-tumor activity of CFZ. RESULTS: CFZ treatment inhibited both the constitutive proteasome and the immunoproteasome in lung cancer cell lines. CFZ had marked anti-proliferative activity in A549, H1993, H520, H460, and H1299 non-small cell lung cancer (NSCLC) cell lines, with IC₅₀ values after 96 hour exposure from <1.0 nM to 36 nM. CFZ had more variable effects in the SHP77 and DMS114 SCLC cell lines, with IC₅₀ values at 96 hours from <1 nM to 203 nM. Western blot analysis of CFZ-treated H1993 and SHP77 cells showed cleavage of poly ADP ribose polymerase (PARP) and caspase-3, indicative of apoptosis, and induction of microtubule-associated protein-1 light chain-3B (LC3B), indicative of autophagy. In SHP77 flank xenograft tumors, CFZ monotherapy inhibited tumor growth and prolonged survival, while no additive or synergistic anti-tumor efficacy was observed for CFZ + cisplatin (CDDP). CONCLUSIONS: CFZ demonstrated anti-proliferative activity in lung cancer cell lines in vitro and resulted in a significant survival advantage in mice with SHP77 SCLC xenografts, supporting further pre-clinical and clinical investigations of CFZ in NSCLC and SCLC.
2

THE DEVELOPMENT OF NOVEL PROTEASOME INHIBITORS FOR THE TREATMENT OF MULTIPLE MYELOMA AND ALZHEIMER’S DISEASE

Lee, Min Jae 01 January 2019 (has links)
Over a decade, proteasome inhibitors (PIs), bortezomib, carfilzomib (Cfz) and ixazomib, have contributed to a significant improvement in the overall survival for multiple myeloma (MM) patients. However, the response rate of PI was fairly low, leaving a huge gap in MM patient care. Given this, mechanistic understanding of PI resistance is crucial towards developing new therapeutic strategies for refractory/relapsed MM patients. In this dissertation work, we found H727 human bronchial carcinoid cells are inherently resistant to Cfz, yet susceptible to other PIs and inhibitors targeting upstream components of the ubiquitin-proteasome system (UPS). It indicated H727 cells may serve as a cell line model for de novo Cfz resistance and remains UPS dependent for survival. To examine the potential link between proteasome catalytic subunit composition and cellular response to Cfz, we altered the composition of proteasome catalytic subunits via interferon-γ treatment or siRNA knockdown in H727 cells. Our results showed alteration in composition of proteasome catalytic subunits results in sensitization of H727 cells to Cfz. It supported that proteasome inhibition by alternative PIs may still be a valid therapeutic strategy for patients with relapsed MM after having received treatment with Cfz. With this in mind, we designed and synthesized a small library of epoxyketone-based PIs by structural modifications at the P1′ site. We observed that a Cfz analog, harboring a hydroxyl substituent at its P1′ position was cytotoxic against cancer cell lines with de novo or acquired resistance to Cfz. These results suggested that peptide epoxyketones incorporating P1′-targeting moieties may have the potential to overcome Cfz resistance mechanisms in cells. The immunoproteasome (IP), an inducible proteasome variant which is harboring distinct catalytic subunits, LMP2, MECL1 and LMP7 of the proteasome typically expressed in cells of hematopoietic origin, plays a role in immune response and is closely linked to inflammatory diseases. It has been reported that the IP is upregulated in reactive glial cells surrounding amyloid β (Aβ) deposits in brains of Alzheimer’s disease (AD) patients and AD animal models. To investigate whether the IP is involved in the pathogenesis of AD, we examined the impact of IP inhibition on cognitive function in AD mouse models. We observed that YU102, an epoxyketone peptide targeting the IP catalytic subunit LMP2, improved cognitive dysfunction in AD mice without clearance of Aβ deposition or tau aggregation. Our cell line model study also showed a potential mode of action of YU102 which is suppressing pro-inflammatory cytokine production in microglial cells. It suggested that LMP2 contributes to microglia-mediated inflammatory response. These findings supported that LMP2 may offers a valuable therapeutic target for treatment of Alzheimer’s disease, expanding the therapeutic potential of the LMP2-targeting strategy.
3

Pre-Clinical Assessment of the Proteasomal Inhibitor Bortezomib as a Generalized Therapeutic Approach for Recessively Inherited Disorders

Jary, Calvin January 2017 (has links)
The number of known monogenic rare diseases (~7000) exceeds the number of effective treatments (~500) by more than an order of magnitude underlining the pressing need for generalizable therapeutic approaches for this class of conditions. In this regard, the majority of recessive and x-linked recessive disorders are caused by missense mutations encoding proteins that frequently have residual function but are rapidly degraded by the 26S proteasome. Bortezomib is a small molecule that inhibits the 26S proteasome and has been approved for use in patients for an unrelated condition; multiple myeloma. Previous work has shown that, for a small number of disorders, bortezomib can inhibit the degradation of the mutant protein, thereby increasing the protein level and activity, holding out the promise of a beneficial therapeutic effect by the repurposing of this agent. We present here a high level western blot based survey of nine recessive disorders to characterize the general effectiveness of such an approach. Thirteen patient fibroblast cell lines comprising 9 different diseases with 19 known mutations were selected on the basis of missense mutations protein expression data when available. The cell lines were incubated with bortezomib (10 nM and 50 nM; 24 hrs) and levels of the mutated protein were quantified by western blot. Unfortunately, no consistent, appreciable increase was observed for any of the conditions tested. The general therapeutic value of re-purposing bortezomib for recessive and x-linked diseases appears limited at best. The few reported cases of bortezomib successfully working in increasing mutated protein levels appear to be the exceptions and not the norm. Moreover successes are more often limited to cell lines carrying a transgene expressing the mutated protein rather than endogenous mutated protein in patient cell lines.
4

TARGET VALIDATION OF UK-101 AND FUNCTIONAL STUDIES OF β1i

Wehenkel, Marie V. 01 January 2011 (has links)
β1i is a major catalytic subunit of the immunoproteasome, an alternative form of the constitutive proteasome, and its upregulation has been demonstrated in a variety of disease states including cancer. Our lab has developed a small molecule inhibitor of β1i, dubbed UK-101. While UK-101 causes apoptosis in cancer cell lines, it was not clear whether this apoptotic effect was directly mediated by its irreversible inhibition of β1i. Since off-target effects are major roadblocks for the development of new and effective pharmaceuticals, target validation studies in this system would assist in the further progression of β1i inhibitors towards preclinical trials. Our hypothesis was that the expression and catalytic activity of β1i is important for the growth and proliferation of the PC-3 prostate cancer cell line, therefore the apoptotic effect seen upon treatment of PC-3 cells with UK-101 was due solely to its covalent inhibition of β1i. To test this hypothesis, a number of complementary approaches were used. The expression of β1i in PC-3 cells was increased by the treatment of these cells with interferon-gamma or tumor necrosis factor-alpha, natural inducers of the immunoproteasome. The expression of β1i in PC-3 cells was decreased using small interfering RNA or short hairpin RNA, in a transient or stable manner, respectively. All of these cells were then treated with UK-101. The efficacy of UK-101 decreased in the interferon-gamma treated cells but did not change in any other the other cell lines, suggesting that UK-101 was not specific for β1i. This was confirmed using a molecular probe of the proteasome and demonstrated that UK-101 bound to other proteasome catalytic subunits. Additional experiments were performed to determine the effect of β1i on the proliferation of PC-3 cells. Simply removing the β1i using small interfering RNA reduces the viability of these cells. Other studies demonstrated that a mutation of β1i which inhibited its catalytic activity reduced the viability of cells when compared to those containing the wild type protein. Overall, our data indicate that β1i is a potential therapeutic target in prostate cancer. Further medicinal chemistry efforts will be required develop UK-101 into a truly selective proteasome inhibitor.
5

PROTEASOME REGULATION OF CASPASE-8: SIGNIFICANCE IN CANCER

Fiandalo, Michael Vincent 01 January 2012 (has links)
Anti-tumor therapeutic strategies based on combinations of chemotherapeutic agents with a death inducing ligand such as TNF-α Related Apoptosis Inducing Ligand (TRAIL), are directed towards selective and effective cancer cell apoptosis and enhanced therapeutic response. We previously demonstrated that proteasome inhibition sensitizes TRAIL resistant prostate cancer cells to TRAIL-mediated apoptosis via stabilization of the active p18 subunit of initiator caspase-8. The present study investigated the functional link between caspase-8 and the proteasome, by analyzing the impact of caspase-8 ubiquitination and proteasomal degradation on the outcomes of the extrinsic apoptosis pathway in cancer cells. Caspase-8 ubiquitination status was assessed by polyubiquitin immunoprecipitation (IP) and fluorescent microscopy. Apoptosis induction in response to death receptor stimuli or proteasome inhibitor was evaluated using the Annexin V/Propidium iodide staining (PI). To determine the consequences of proteasome inhibition on caspase-8 stability, trafficking, and activity following death receptor activation, we used the TRAIL-resistant human prostate cancer LNCaP cells, and the caspase-8 deficient Neuroblastoma 7 (NB7) cells, as cellular models for reconstituting the non-cleavable mutant forms of caspase-8. Our findings demonstrate that the non-cleavable forms of caspase-8 are capable of inducing apoptosis comparably to wild-type caspase-8 upon treatment with proteasome inhibitor and GST-TRAIL. Furthermore, caspase-8 processing into its active subunits preceded caspase-8 polyubiquitination, implicating caspase-8 processing as a potential regulatory mechanism, rather than a requirement for caspase-8 activation in apoptosis induction. The mechanistic control of caspase-8 by ubiquitination in cancer cells may have significant significance in bypassing mechanisms of therapeutic resistance in human tumors and optimization of anti-cancer treatment strategies in human tumors and optimization of anti-cancer treatment strategies.
6

Studies in Rhodium Catalyzed Intramolecular C-H Insertion of Amino Acid Derived α-Diazo-α-(substituted)acetamides and its Application to the Total Synthesis of <em>clasto</em>-Lactacystin β-Lactone

Flanigan, David L, Jr. 24 May 2004 (has links)
Lactacystin is a microbial metabolite isolated by Omura that exhibits neurotrophic activity in neuroblastoma cell lines. Lactacystin and especially its β-lactone analog are the first examples of non-polypeptide small molecules capable of specifically inhibiting the 20S proteasome. Various asymmetric total syntheses of lactacystin and its analogs have been reported. The total synthesis of clasto -lactacystin β-lactone is achieved using L-serine methyl ester as the starting material and the sole source of stereochemical induction. The success of this synthesis hinges on two featured transformations. The first key step involves formation of the γ -lactam core via rhodium (II) catalyzed intramolecular C-H insertion of the α-diazo-α-(phenylsulfonyl)acetamide intermediate. The methodology for this transformation has been developed and applied to the synthesis of highly functionalized stereogenic γ-lactams from natural α-amino acids. Three control elements that govern γ-lactam formation are described. This step is highlighted by the xvi simultaneous creation of two stereogenic centers of the γ-lactam core. The second key step involves the late stage aldol coupling for quaternary carbon formation and installation of the hydroxyisobutyl group. In all previously reported syntheses, this is the very first aspect which is addressed. The stereochemical outcome of this step is directed by the chiral environment of the enolate itself. Various attempts to achieve selectivity are explored and reported. Completion of the synthesis of clasto-lactacystin β-lactone requires 17 steps with an overall yield of 10%. Some general attempts for optimizing the synthetic scheme are discussed as well as the future direction of this research.
7

STRUCTURAL AND FUNCTIONAL STUDIES OF F-BOX-ONLY PROTEIN FBXO7 AND ITS INTERACTIONS WITH PROTEASOME INHIBITOR PI31

Shang, Jinsai 01 August 2015 (has links)
F-box only protein 7 (Fbxo7), a member of the F-box-only subfamily of FBPs, is a biologically and pathophysiologically important human protein that assumes many critical functions. The different functions of Fbxo7 depend on the formation of various multi-protein complexes. Possible interplay between different Fbxo7 functions further complicate the protein-protein interaction networks involved in Fbxo7 biology. Although significant progresses have been made to understand the functions, regulation, specificity, and protein interaction network of Fbxo7, a myriad of questions remain to be answered. The objectives of the work presented in this dissertation are to elucidate the molecular structures underlying the functions of Fbxo7 and the interaction with its protein partners, such as proteasome inhibitor PI31. The best known biological function of Fbxo7 is its role as the substrate-recognition subunit of the SCFFbxo7 (Skp1-Cul1-F-box protein) E3 ubiquitin ligase that catalyzes the ubiquitination of hepatoma up-regulated protein (HURP) and inhibitor of apoptosis protein (IAP). Fbxo7 also assumes various SCF-independent functions through interact with its protein partners that are not the substrates of the ubiquitin proteasome system, such as PI31, Cdk6, p27, PINK1 (PTEN-induced kinase 1), and Parkin. PI31 is a known proteasome regulator which was initially characterized as a proteasome inhibitor in vitro. The binding affinity between Fbxo7 and PI31 is very strong, and The Fbxo7-PI31 interaction is mediated by heterodimerization of the FP domains of the two proteins. This work is focus on study the protein structure of the two FP domains in Fbxo7 and PI3. Chapter 1 reviewed the F-box-only protein Fbxo7 biology including the function of Fbxo7 protein in ubiquitination proteasome pathway and some SCF-independent functions which are relate to human disease. Chapter 2 discussed the function of proteasome inhibitor PI31. With the many important biological functions, Fbxo7 is clearly an extraordinary important protein, but the lack of structural knowledge has hampered efforts to achieve a better understanding of Fbxo7 biology. In this work, we have determined the crystal structure of Fbxo7 FP domain (residues 181-335) and the crystal structure of the PI31 FP domain (residues 1-161) using a longer protein construct both at 2.0Å resolution. The Fbxo7 FP domain adopts an α/β-fold similar to that of the PI31 FP domain and the secondary structure elements of the two FP domains are comparable including the C-terminal helix, indicating that the two FP domains share the same overall global fold. However, an α helix and three β strands in the Fbxo7 are longer than their counterparts in the PI31 FP domain. The two FP domains also differ substantially in the length and conformation of the longest connecting loop. More importantly, structural differences between the two FP domains lead to drastically different modes of inter-domain protein–protein interaction: the PI31 FP domain utilizes either an α interface or β interface for homodimeric interaction, whereas the Fbxo7 FP domain utilizes an αβ interface. We have note that the inter-domain interaction of the Fbxo7 FP domain is much more extensive, featuring a larger contact surface area, better shape complementarity and more hydrophobic and hydrogen-bonding interactions. The results of this structural study provide critical insights into how Fbxo7 may dimerize (or multimerize) and interact with PI31 via the FP domain. Chapter 4 and Chapter 5 discussed the structure determinations, structure features and detail of protein-protein interactions of Fbxo7 and PI31 FP domains. Chapter 2 reviewed the corresponding fundamental biochemical techniques that been used in this study. Chapter 3 discussed protein structure determination by X-ray crystallography in structural biology studies. It was believed that the FP domains of Fbxo7 and PI31 mediate homodimerization and heterodimerization of the proteins and the FP domain is not present in other human proteins. In order to study the Fbxo7-PI31 heterodimerization protein-protein interactions, we performed modeling studies. Chapter 6 discussed the model building and binding studies. Based on the result of model building studies, we propose that an interaction between the two FP domains of Fbxo7 and PI31 should be mediated by a αβ interface using the α-helical surface of the Fbxo7 FP domain and the β-sheet surface of the PI31 FP domain. According to the result of pull down assay, the PI31 FP domain may complete with Skp1 for the binding with Fbxo7. It is possible that the formation of heterodimer between the Fbxo7 and PI31 mediate by FP domains may lead to the Fbxo7 dissociation from SCFFbxo7 complex which might reveal a new regulation mechanism.
8

Elucidating Proteasome Catalytic Subunit Composition and Its Role in Proteasome Inhibitor Resistance

Carmony, Kimberly C. 01 January 2016 (has links)
Proteasome inhibitors bortezomib and carfilzomib are FDA-approved anticancer agents that have contributed to significant improvements in treatment outcomes. However, the eventual onset of acquired resistance continues to limit their clinical utility, yet a clear consensus regarding the underlying mechanisms has not been reached. Bortezomib and carfilzomib are known to target both the constitutive proteasome and the immunoproteasome, two conventional proteasome subtypes comprising distinctive sets of catalytic subunits. While it has become increasingly evident that additional, ‘intermediate’ proteasome subtypes, which harbor non-standard mixtures of constitutive proteasome and immunoproteasome catalytic subunits, represent a considerable proportion of the proteasome population in many cell types, less is known regarding their contribution to cellular responses to proteasome inhibitors. Importantly, previous studies in murine models have shown that individual proteasome subtypes differ in sensitivity to specific proteasome inhibitors. Furthermore, research efforts in our laboratory and others have revealed that proteasome catalytic subunit expression levels and activity profiles are altered when human cancer cells acquire resistance to proteasome inhibitors. We therefore hypothesized that changes in the relative abundances of individual proteasome subtypes contribute to the acquired resistance of cancer cells to bortezomib and carfilzomib. A major obstacle in testing our hypothesis was a lack of chemical probes suitable for use in identifying distinct proteasome subtypes. We addressed this by developing a series of bifunctional proteasome probes capable of crosslinking specific pairs of catalytic subunits colocalized within individual proteasome complexes and compatible with immunoblotting-based detection of the crosslinked subunit pairs. We confirmed the utility of these probes in discerning the identities of individual proteasome subtypes in a multiple myeloma cell line that abundantly expresses catalytic subunits of both the constitutive proteasome and immunoproteasome. Our findings indicate that constitutive proteasomes, immunoproteasomes, and intermediate proteasomes co-exist within these cells and support conclusions drawn from previous studies in other cell types. We also established non-small cell lung cancer cell line models of acquired bortezomib and carfilzomib resistance in which to test our hypothesis. Using immunoblotting and proteasome activity assays, we discovered that changes in the expression levels and activities of individual catalytic proteasome subunits were associated with the emergence of acquired resistance to bortezomib or carfilzomib. These changes were inhibitor-dependent and persisted after the selective pressure of the inhibitor was removed. Finally, results obtained using our bifunctional proteasome probes suggest that the altered abundance of an intermediate proteasome subtype is associated with acquired proteasome inhibitor resistance. Collectively, our results provide evidence linking changes proteasome composition with acquired proteasome inhibitor resistance and support the hypothesis that such changes are involved in resistance mechanisms to these inhibitors.
9

Inhibitory effect on the proteasome regulatory subunit, RPN11/POH1, with the use of Capzimin-PROTAC to trigger apoptosis in cancer cells

Holmqvist, Andreas January 2020 (has links)
Most patients diagnosed with cancer will receive systematic chemotherapy at some point during their illness, which almost always cause severe side effects for the patients such as, anemia, nausea and vomiting. The problems with today’s chemotherapy is not only that it cause severe side effects, but also that the cancer may develop resistance to the therapy, which is why the development of a new type of therapeutic agent is in dire need. The ubiquitin proteasome system (UPS) is a vital machinery for the cancer cells to maintain protein homeostasis, which also make them vulnerable to any disruption of this system. In recent years, a new technology has been developed that utilize the UPS by chemically bringing an E3 ubiquitin ligase into close proximity of a protein of choice and tagging the protein with ubiquitin for degradation. This technology is called proteolysis targeting chimera (PROTAC). In this project, we managed to theoretically develop a new type of cancer therapeutic agent, that utilize the PROTAC system together with the first-in-class proteasome regulatory subunit, POH1, inhibitor Capzimin as a warhead. By using Capzimin as a warhead it should be possible to polyubiquitinate POH1, and thus induce proteotoxic stress in the cancer cells to trigger apoptosis. This theoretically developed drug is therefore called Capzimin-PROTAC, which should be able to trigger apoptosis in cancer cells, and at the same time being relatively safe to normal healthy cells.
10

Untersuchungen zur Pathogenese und Therapie der Notch1-abhängigen T-ALL in einem transgenen Rattenmodell. / Investigation of pathogenesis and therapy of Notch1 dependent T-ALL in a transgenic rat model

Helbron, Kai-Torben 18 October 2016 (has links)
Die T-Zell akute lymphoblastische Leukämie (T-ALL) ist eine aggressive Krebserkrankung mit hoher Rezidivwahrscheinlichkeit und insgesamt schlechter Prognose, die häufig durch ein fehlreguliertes, meist konstitutiv aktives Notch1 gekennzeichnet ist. Ungeachtet der wichtigen Rolle von Notch1 in der T-Zell-Entwicklung sind die genauen Mechanismen, mit denen ein fehlreguliertes Notch1-Signal zur Karzinogenese führt, bislang unverstanden. Diese Fragen können mithilfe eines Rattenmodells (NICA-Ratten), das durch die transgene Expression von konstitutiv aktivem Notch1IC in Thymozyten charakterisiert ist, untersucht werden. Diese Tiere entwickeln mit hoher Inzidenz thymische Lymphome, welche sich durch unerwartete phänotypische Eigenschaften und eine rasche leukämische Aussaat auszeichnen. Im Rahmen dieser Arbeit wurde ein Rattenmodell der T-ALL verwendet, beim dem es durch adoptiven Transfer von Lymphomzellen aus NICA-Ratten in syngene Wirte kurzfristig zu neuem und aggressivem Tumorwachstum ähnlich der humanen Variante kommt. Histologische und durchflusszytometrische Analysen offenbarten die initiale Infiltration in Lymphknoten und Knochenmark, bevor im Zuge der anschließenden leukämischen Phase nicht-lymphatische Organe kleinherdig durchsetzt wurden. Die aus den Wirten extrahierten Lymphomzelllinien zeigten einen heterogenen Phänotyp und eine ausgeprägte Neigung zur Spontanapoptose ex vivo. Die Abwesenheit zytotoxischer T-Zellen im Wirt hatte keinen Einfluss auf die Lymphometablierung und -progression, was möglicherweise auf die geringe Expression von MHC-I Molekülen zurückzuführen ist. Im weiteren Verlauf der Arbeit wurden neue Therapieansätze in der Behandlung der Notch1-abhängigen T-ALL getestet. Da der routinemäßige Einsatz von Glukokortikoiden (GC) durch das Auftreten schwerer Nebenwirkungen und Resistenzen gekennzeichnet ist, wurde als Ansatz zur ihrer Optimierung liposomal verpacktes Prednisolon mit freiem Dexamethason in der Therapie von T-ALL in vivo im Tiermodell verglichen. Die alternative Darreichungsfom war durch ein auffallendes Wirkdefizit im lymphatischen System und ein schlechteres Gesamtüberleben gekennzeichnet. Als potentielle Ursache der rasch einsetzenden GC-Resistenz im Zuge der repetitiven Dex-Behandlung konnte eine Herunterregulierung des GC-Rezeptors nachgewiesen werden. Weiterhin wurde die Frage adressiert, inwieweit eine medikamentöse Hemmung von NF-κB durch die Verwendung des Proteasomen-Inhibitors Bortezomib eine therapeutische Wirkung auf die T-ALL im Rattenmodell entfalten würde. Hierzu wurde Bortezomib im direkten Vergleich mit Dexamethason in vivo evaluiert. Die Monotherapie mit Bortezomib zeigte eine dem Dexamethason vergleichbare Wirkung in den Lymphknoten, andererseits korrelierte ein signifikantes Wirkdefizit in Milz, Knochenmark und Blut mit einem insgesamt schlechteren Gesamtansprechen. Ferner legten die gewonnenen Daten nahe, dass Bortezomib eine GC-Resistenz der T-ALL mit konstitutiv aktivem Notch1 in vivo nicht zu überwinden vermag. Zusammengefasst wurden im Rahmen dieser Arbeit neue Erkenntnisse zur Pathogenese der Notch1-abhängigen T-ALL gewonnen und darüber hinaus limitierte Therapieeffizienzen neuartiger Behandlungsansätze mit liposomalem Prednisolon und dem Proteasominhibitor Bortezomib im Vergleich zu freiem Dex in vivo aufgedeckt. Im Hinblick auf die Erfassung der Pathomechanismen sowie auf die Entwicklung neuer Therapieansätze bietet die Möglichkeit des adoptiven Lymphomtransfers und der Generierung von neuem aggressivem Wachstum im Wirt mit hoher Analogie zur humanen Erkrankung ideale Voraussetzungen, um die Rolle von Notch1 in der der T-ALL zu untersuchen. Die Ergebnisse dieser Arbeit könnten somit zu einer Verbesserung von spezifischer Risikostratifizierung, Behandlungs-möglichkeit und Prognose der an T-ALL erkrankten Patienten beitragen.

Page generated in 0.0727 seconds