@phdthesis{Gnamlin2015, author = {Gnamlin, Prisca}, title = {Use of Tumor Vasculature for Successful Treatment of Carcinomas by Oncolytic Vaccinia Virus}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119019}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Tumor-induced angiogenesis is of major interest for oncology research. Vascular endothelial growth factor (VEGF) is the most potent angiogenic factor characterized so far. VEGF blockade was shown to be sufficient for angiogenesis inhibition and subsequent tumor regression in several preclinical tumor models. Bevacizumab was the first treatment targeting specifically tumor-induced angiogenesis through VEGF blockade to be approved by the Food and Drugs Administration (FDA) for cancer treatment. However, after very promising results in preclinical evaluations, VEGF blockade did not show the expected success in patients. Some tumors became resistant to VEGF blockade. Several factors have been accounted responsible, the over-expression of other angiogenic factors, the noxious influence of VEFG blockade on normal tissues, the selection of hypoxia resistant neoplastic cells, the recruitment of hematopoietic progenitor cells and finally the transient nature of angiogenesis inhibition by VEGF blockade. The development of blocking agents against other angiogenic factors like placental growth factor (PlGF) and Angiopoietin-2 (Ang-2) allows the development of an anti-angiogenesis strategy adapted to the profile of the tumor. Oncolytic virotherapy uses the natural propensity of viruses to colonize tumors to treat cancer. The recombinant vaccinia virus GLV-1h68 was shown to infect, colonize and lyse several tumor types. Its descendant GLV-1h108, expressing an anti-VEGF antibody, was proved in previous studies to inhibit efficiently tumor induced angiogenesis. Additional VACVs expressing single chain antibodies (scAb) antibodies against PlGF and Ang-2 alone or in combination with anti VEGF scAb were designed. In this study, VACV-mediated anti-angiogenesis treatments have been evaluated in several preclinical tumor models. The efficiency of PlGF blockade, alone or in combination with VEGF, mediated by VACV has been established and confirmed. PlGF inhibition alone or with VEGF reduced tumor burden 5- and 2-folds more efficiently than the control virus, respectively. Ang-2 blockade efficiency for cancer treatment gave controversial results when tested in different laboratories. Here we demonstrated that unlike VEGF, the success of Ang-2 blockade is not only correlated to the strength of the blockade. A particular balance between Ang-2, VEGF and Ang-1 needs to be induced by the treatment to see a regression of the tumor and an improved survival. We saw that Ang-2 inhibition delayed tumor growth up to 3-folds compared to the control virus. These same viruses induced statistically significant tumor growth delays. This study unveiled the need to establish an angiogenic profile of the tumor to be treated as well as the necessity to better understand the synergic effects of VEGF and Ang-2. In addition angiogenesis inhibition by VACV-mediated PlGF and Ang-2 blockade was able to reduce the number of metastases and migrating tumor cells (even more efficiently than VEGF blockade). VACV colonization of tumor cells, in vitro, was limited by VEGF, when the use of the anti-VEGF VACV GLV-1h108 drastically improved the colonization efficiency up to 2-fold, 72 hours post-infection. These in vitro data were confirmed by in vivo analysis of tumors. Fourteen days post-treatment, the anti-VEGF virus GLV-1h108 was colonizing 78.8\% of the tumors when GLV-1h68 colonization rate was 49.6\%. These data confirmed the synergistic effect of VEGF blockade and VACV replication for tumor regression. Three of the tumor cell lines used to assess VACV-mediated angiogenesis inhibition were found, in certain conditions, to mimic either endothelial cell or pericyte functions, and participate directly to the vascular structure. The expression by these tumor cells of e-selectin, p-selectin, ICAM-1 and VCAM-1, normally expressed on activated endothelial cells, corroborates our findings. These proteins play an important role in immune cell recruitment, and there amount vary in presence of VEGF, PlGF and Ang-2, confirming the involvement of angiogenic factors in the immuno-modulatory abilities of tumors. In this study VACV-mediated angiogenesis blockade proved its potential as a therapeutic agent able to treat different tumor types and prevent resistance observed during bevacizumab treatment by acting on different factors. First, the expression of several antibodies by VACV would prevent another angiogenic factor to take over VEGF and stimulate angiogenesis. Then, the ability of VACV to infect tumor cells would prevent them to form blood vessel-like structures to sustain tumor growth, and the localized delivery of the antibody would decrease the risk of adverse effects. Next, the blockade of angiogenic factors would improve VACV replication and decrease the immune-modulatory effect of tumors. Finally the fact that angiogenesis blockade lasts until total regression of the tumor would prevent the recovery of the tumor-associated vasculature and the relapse of patients.}, subject = {Vaccinia-Virus}, language = {en} } @article{SchneiderSchauliesvonBrunnSchachner1990, author = {Schneider-Schaulies, J{\"u}rgen and von Brunn, A. and Schachner, M.}, title = {Recombinant peripheral myelin protein P\(_o\) confers both adhesion and neurite outgrowth promoting properties}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-54841}, year = {1990}, abstract = {To probe into the functional properties of the major peripheral myelin cell surface glycoprotein P 0 , its ability to confer adhesion and neurite outgrowth-promoting properfies was studied in cell culture. Tothis aim, Po was expressed as integral membrane glycoprotein at the surface of CV -1 cells with the help of a recombinant vaccinia virus expression system. Furthermore, the immunoglobulin-like extracellular domain of P0 (P0 -ED) was expressed as soluble profein in a bacterial expression system and used as substrafe coated to plastic dishes or as competitor in cell adhesion and neurite outgrowth-promoting assays. The adhesion of P0 -expressing CV-1 cells to P0 -ED substrafe was specifically inhibitable by polyclonal Po antibodies (54\% :t 6\% ). In addition, the specific interaction between Po molecules could be reduced ( 49\% ± 8\%) by adding soluble P0 -ED to the culture medium, demonstrating that the homophilic inter~ction between recombinant Po molecules can be mediated, at least on one partner of interacting molecules, by the unglycosylated Ig-like domain. Substrate-coated p -ED also conferred adhesion and neurite outgrowth ability to dorsal root ganglion neurons with neurites of a mean length of about 150 ,_..m. This neurite outgrowth was specifically inhibitable by soluble P" (74\% ± 14\%) and P 0 antibodies (65\% ± 9\% ). These observations indicate that Po is capable of displaying two different types of functional roles in the myelination process of . peripheral nerves: The heterophilic interaction with neurons may be responsible for the recognition between axon and myelinating Schwann cell at the onset of myelination, whereas the homophilic interacton may indicate its roJe in the selfrecognition of the apposing loops of Schwann cell surface membranes during the myelination process and in the mature compact myelin sheath.}, subject = {Immunologie}, language = {en} } @phdthesis{Adelfinger2012, author = {Adelfinger, Marion}, title = {Pr{\"a}klinische Verwendung verschiedener onkolytischer Vaccinia-Viren zur Therapie von Human- und Hundetumoren}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-70688}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Nach Einsch{\"a}tzung der Weltgesundheitsorganisation WHO wird Krebs im Jahr 2013 die weltweit h{\"a}ufigste Todesursache bei Menschen und Haustieren sein. Diese Situation erfordert die Entwicklung neuer therapeutischer Ans{\"a}tze. Hauptziel einer Tumortherapie ist es, sowohl den Prim{\"a}rtumor als auch die Metastasen m{\"o}glichst vollst{\"a}ndig zu entfernen. Dabei wird nach Methoden gesucht, die im Gegensatz zu den meisten gegenw{\"a}rtigen therapeutischen Eins{\"a}tzen, wie der chirurgischen Entfernung b{\"o}sartiger Neubildungen, Chemotherapie und Strahlentherapie, selektiv die b{\"o}sartigen Zellen erkennen und zerst{\"o}ren k{\"o}nnen. Eine faszinierende M{\"o}glichkeit in dieser Hinsicht ist die Verwendung von onkolytischen Viren, die die F{\"a}higkeit besitzen, sich selektiv sowohl in Prim{\"a}rtumoren als auch in Metastasen anzusiedeln und die Krebszellen dort zu zerst{\"o}ren. Das Konzept, dass Viren n{\"u}tzlich f{\"u}r die Bek{\"a}mpfung von Krebs sein k{\"o}nnten, ist nicht neu. Allerdings konnte erst in den letzten Jahren durch zahlreiche Studien best{\"a}tigt werden, dass verschiedene Viren in der Lage sind, eine signifikante Antitumorwirkung in vivo auszu{\"u}ben. Zu den erfolgversprechenden onkolytischen Viren z{\"a}hlen insbesondere Adenovirus, Herpes simplex Virus, Reovirus und Vaccinia-Virus, die sich bereits in Phase III der klinischen Studien befinden oder kurz davor sind. Die therapeutische Nutzung von tumorspezifischen onkolytischen Viren beim Menschen hat bereits begonnen. Im Rahmen der vorliegenden Doktorarbeit wurden verschiedene Aspekte der Wirkungsweise von Vaccinia-Virus-St{\"a}mmen bei der Therapie verschiedener Tumore aus Mensch und Hund im Xenotransplantat-Mausmodell bearbeitet: die Onkolyse der Krebszellen und Inhibition des Tumorwachstums sowie die Effekte der Virusinfektion auf das Tumormikromilieu und die Mitwirkung des angeborenen Immunsystems bei der Virotherapie. Das Tumormikromilieu (Stroma) setzt sich aus einer Vielzahl verschiedener Zellen und Komponenten der extrazellul{\"a}ren Matrix zusammen. Die Krebszellen bilden unter anderem mit Endothelzellen des Blut- und Lymphsystems und verschiedenen Immunzellen eine komplexe Organ-{\"a}hnliche Struktur. Weitere wichtige Bestandteile des Stromas sind Wachstumsfaktoren, Chemokine und Zytokine und die Tumorvaskulatur. Diese ist durch zahlreiche strukturelle und funktionelle Abnormalit{\"a}ten charakterisiert, wodurch die Effektivit{\"a}t von Strahlen- und Chemotherapie herabgesetzt wird. Weiterhin ist das Tumormikromilieu durch seine {\"A}hnlichkeit mit einer chronischen Entz{\"u}ndungsreaktion gekennzeichnet und wirkt immunsupprimierend auf rekrutierte Leukozyten, die wiederum die Inflammation verst{\"a}rken und die Angiogenese und das Tumorwachstum weiter f{\"o}rdern. Aufgrund dieser vielen Komponenten ist die Zusammensetzung jedes Tumors einzigartig, weswegen Standardtherapien h{\"a}ufig nicht zu einer Heilung f{\"u}hren. Die Wirkung der Viren bei der Virotherapie beruht vermutlich auf 4 Mechanismen, die einzeln oder in Kombination auftreten k{\"o}nnen: die direkte Onkolyse der Krebszellen, die Zerst{\"o}rung des Tumorblutgef{\"a}ßsystems, die Aktivierung des Immunsystems des Wirts und die Suppression der microRNA-Expression des Wirtes. Zus{\"a}tzlich kann die Expression therapeutischer Gene die onkolytische Wirkung verst{\"a}rken. Zum Nachweis der Onkolyse der Krebszellen und Inhibition des Tumorwachstums wurde zuerst das Virus GLV-1h68 in einem autologen humanen Melanomzellpaar, 888-MEL und 1936-MEL, eingesetzt. Das GLV-1h68-Virus wurde auf Basis des Wildtyp Vaccinia-Virus LIVP durch die Insertion von 3 Expressionskassetten in den drei Genloci F14.5L, J2R und A56 genetisch konstruiert. 888-MEL, eine zu einem fr{\"u}hen Zeitpunkt der Krebserkrankung aus einer Metastase isolierte Zelllinie, zeigt nach Infektion mit GLV-1h68 im Mausmodell Tumornekrose („Responder"), w{\"a}hrend 1936-MEL aus einer sp{\"a}ten Metastasierungsphase kaum mit Onkolyse auf eine Virusinfektion reagiert („Poor-Responder"). Die onkolytische Wirkung konnte mittels Durchflusszytometrie in Tumoren beider Zelllinien zu einem fr{\"u}hen Zeitpunkt nach Virusinfektion nachgewiesen werden. In 888-MEL-Tumoren wurde hierbei eine große Zahl infizierter und toter Zellen nach Virusinfektion gefunden. Gleichzeitig wurde eine hohe Zahl an Immunzellen detektiert, die nach Virusinfektion reduziert war. In den schw{\"a}cher reagierenden 1936-MEL-Tumoren konnte eine Onkolyse bei Infektion mit h{\"o}herer Virusmenge und zu einem fr{\"u}heren Zeitpunkt demonstriert werden, wodurch mehr Zellen infiziert wurden. Zus{\"a}tzlich wurde eine Steigerung der nur in geringer Zahl vorhandenen Immunzellen nachgewiesen. Trotz des unterschiedlichen Tumormikromilieus konnte somit ein onkolytischer Effekt in beiden Tumormodellen erzielt werden. ...}, subject = {Vaccinia-Virus}, language = {de} } @phdthesis{Cook2012, author = {Cook, Vanessa Janine}, title = {Protection of healthy tissues from infection with systemically administered vaccinia virus strains}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-69654}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Oncolytic virotherapy using recombinant vaccinia virus strains is a promising approach for the treatment of cancer. To further improve the safety of oncolytic vaccinia viruses, the cellular microRNA machinery can be applied as the host's own security mechanism to avoid unwanted viral replication in healthy tissues. MicroRNAs are a class of small single-stranded RNAs which due to their ability to mediate post-transcriptional gene-silencing, play a crucial role in almost every regulatory process in cellular metabolism. Different cancers display unique microRNA expression patterns, showing significant up- or downregulation of endogenously expressed microRNAs. Furthermore, the behavior of cancer cells can be altered by either adding microRNAs known to inhibit cancer cell spread and proliferation or suppressing cancer promoting microRNAs (oncomirs) making microRNAs promising targets for cancer gene therapy. The cell's own RNAi machinery can also be utilized to control viral replication due to the virus dependence on the host cell replication machinery, a process controlled by microRNAs. GLV-1h68 is a replication-competent recombinant oncolytic vaccinia virus constructed and generated by Genelux Corp., San Diego, CA, USA which carries insertions of three reporter gene cassettes for detection and attenuation purposes and is currently being evaluated for cancer treatment in clinical trials. Though there are hardly any side effects found in GLV-1h68 mediated oncolytic therapy an increased tropism for replication exclusively in cancer cells is desirable. Therefore it was investigated whether or not further cancer cell specificity of a recombinant vaccinia virus strain could be obtained without compromising its oncolytic activity using microRNA interference. Let-7a is a well characterized microRNA known to be expressed in high levels in healthy tissues and strongly downregulated in most cancers. To control vaccinia virus replication rates, four copies of the mature human microRNA let-7a target sequence were cloned behind the stop codon in the 3'end of the vaccinia virus D4R gene, using a GLV-1h68 derivative, GLV-1h190, as parental strain yielding the new recombinant virus strain GLV-1h250. The D4R gene belongs to the group of early transcribed vaccinia genes and encodes an essential enzyme, uracil DNA glycosylase, which catalyzes the removal of uracil residues from double-stranded DNA. A defect in D4R prevents vaccinia virus from entering into the intermediate and late phase of replication, leading to an aborted virus replication. After expression of the microRNA target sequence from the vaccinia virus genome, the endogenously expressed microRNA-let-7a should recognize its target structure within the viral mRNA transcript, thereby binding and degrading the viral mRNA which should lead to a strong inhibition of the virus replication in healthy cells. GLV-1h250 replication rates in cancerous A549 lung adenocarcinoma cells, which show a strong down-regulation of microRNA let-7a, was comparable to the replication rates of its parental strain GLV-1h190 and the control strain GLV-1h68. In contrast, GLV-1h250 displayed a 10-fold decrease in viral replication in non-cancerous ERC cells when compared to GLV-1h190 and GLV-1h68. In A549 tumor bearing nude mice GLV-1h250 replicated exclusively in the tumorous tissue and resulted in efficient tumor regression without adverse effects leading to the conclusion that GLV-1h250 replicates preferentially in cancerous cells and tissues, which display low endogenous let-7a expression levels.}, subject = {Vaccinia-Virus}, language = {en} } @phdthesis{Krueger2012, author = {Kr{\"u}ger, Beate}, title = {Integration und Kombination bioinformatischer Methoden in Biotechnologie, synthetischer Biologie und Pharmaindustrie}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-70702}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Die Bioinformatik ist eine interdisziplin{\"a}re Wissenschaft, welche Probleme aus allen Lebenswissenschaften mit Hilfe computergest{\"u}tzter Methoden bearbeitet. Ihr Ziel ist es, die Verarbeitung und Interpretation großer Datenmengen zu erm{\"o}glichen. Zudem unterst{\"u}tzt sie den Designprozess von Experimenten in der Synthetischen Biologie. Die synthetische Biologie besch{\"a}ftigt sich mit der Generierung neuer Komponenten und deren Eigenschaften, welche durch die Behandlung und Manipulation lebender Organismen oder Teilen daraus entstehen. Ein besonders interessantes Themengebiet hierbei sind Zweikomponenten-Systeme (Two-Component System, TCS). TCS sind wichtige Signalkaskaden in Bakterien, welche in der Lage sind Informationen aus der Umgebung in eine Zelle zu {\"u}bertragen und darauf zu reagieren. Die vorliegende Dissertation besch{\"a}ftigt sich mit der Beurteilung, Nutzung und Weiterentwicklung von bioinformatischen Methoden zur Untersuchung von Proteininteraktionen und biologischen Systemen. Der wissenschaftliche Beitrag der vorliegenden Arbeit kann in drei Aspekte unterteilt werden: - Untersuchung und Beurteilung von bioinformatischen Methoden und Weiterf{\"u}hrung der Ergebnisse aus der vorhergehenden Diplomarbeit zum Thema Protein-Protein-Interaktionsvorhersagen. - Analyse genereller evolution{\"a}rer Modifikationsm{\"o}glichkeiten von TCS sowie deren Design und spezifische Unterschiede. - Abstraktion bzw. Transfer der gewonnenen Erkenntnisse auf technische und biologische Zusammenh{\"a}nge. Mit dem Ziel das Design neuer Experimente in der synthetischen Biologie zu vereinfachen und die Vergleichbarkeit von technischen und biologischen Prozessen sowie zwischen Organismen zu erm{\"o}glichen. Das Ergebnis der durchgef{\"u}hrten Studie zeigte, dass Zweikomponenten-Systeme in ihrem Aufbau sehr konserviert sind. Nichtsdestotrotz konnten viele spezifische Eigenschaften und drei generelle Modifikationsm{\"o}glichkeiten entdeckt werden. Die Untersuchungen erm{\"o}glichten die Identifikation neuer Promotorstellen, erlaubten aber auch die Beschreibung der Beschaffenheit unterschiedlicher Signalbindestellen. Zudem konnten bisher fehlende Komponenten aus TCS entdeckt werden, ebenso wie neue divergierte TCS-Dom{\"a}nen im Organismus Mycoplasma. Eine Kombination aus technischen Ans{\"a}tzen und synthetischer Biologie vereinfachte die gezielte Manipulation von TCS oder anderen modularen Systemen. Die Etablierung der vorgestellten zweistufigen Modul-Klassifikation erm{\"o}glichte eine effizientere Analyse modular aufgebauter Prozesse und erlaubte somit das molekulare Design synthetischer, biologischer Anwendungen. Zur einfachen Nutzung dieses Ansatzes wurde eine frei zug{\"a}ngliche Software GoSynthetic entwickelt. Konkrete Beispiele demonstrierten die praktische Anwendbarkeit dieser Analysesoftware. Die vorgestellte Klassifikation der synthetisch-biologischen und technischen Einheiten soll die Planung zuk{\"u}nftiger Designexperimente vereinfachen und neue Wege f{\"u}r sinnverwandte Bereiche aufzeigen. Es ist nicht die Hauptaufgabe der Bioinformatik, Experimente zu ersetzen, sondern resultierende große Datenmengen sinnvoll und effizient auszuwerten. Daraus sollen neue Ideen f{\"u}r weitere Analysen und alternative Anwendungen gewonnen werden, um fehlerhafte oder falsche Ans{\"a}tze fr{\"u}hzeitig zu erkennen. Die Bioinformatik bietet moderne, technische Verfahren, um vertraute, aber oft m{\"u}hsame experimentelle Wege durch neue, vielversprechende Ans{\"a}tze zur Datenstrukturierung und Auswertung großer Datenmengen zu erg{\"a}nzen. Neue Sichtweisen werden durch die Erleichterung des Testprozederes gef{\"o}rdert. Die resultierende Zeitersparnis f{\"u}hrt zudem zu einer Kostenreduktion.}, subject = {Biotechnologie}, language = {de} } @phdthesis{Tsoneva2017, author = {Tsoneva, Desislava}, title = {Humanized mouse model: a system to study the interactions of human immune system with vaccinia virus-infected human tumors in mice}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-118983}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {Ein vielversprechender neuer Ansatz zur Behandlung von Krebs beim Menschen ist die Verwendung von onkolytischen Viren, die einen Tumor-spezifischen Tropismus aufweisen. Einer der Top-Kandidaten in diesem Bereich ist das onkolytische Vaccinia Virus (VACV), das bereits vielversprechende Ergebnisse in Tierversuchen und in klinischen Studien gezeigt hat. Aber die von den in vivo in tierischen Modellen erhaltenen Resultate k{\"o}nnten ungenaue Informationen wegen der anatomischen und physiologischen Unterschiede zwischen den Spezies liefern. Andererseits sind Studien in Menschen aufgrund ethischer Erw{\"a}gungen und potenzieller Toxizit{\"a}t nur limitiert m{\"o}glich. Die zahlreichen Einschr{\"a}nkungen und Risiken, die mit den Humanstudien verbunden sind, k{\"o}nnten mit der Verwendung eines humanisierten Mausmodells vermieden werden. Die LIVP-1.1.1, GLV-2b372, GLV-1h68, GLV-1h375, GLV-1h376 and GLV-1h377 VACV St{\"a}mmen wurden von der Genelux Corporation zur Verf{\"u}gung gestellt. GLV-2b372 wurde durch Einf{\"u}gen der TurboFP635 Expressionskassette in den J2R Genlocus des parentalen LIVP-1.1.1-Stammes konstruiert. GLV-1h375, -1h376 and -1h377 kodiert das Gen f{\"u}r den menschlichen CTLA4-blockierenden Einzelketten-Antik{\"o}rper (CTLA4 scAb). Befunde aus Replikations- and Zytotoxizit{\"a}tsstudien zeigten, dass alle sechs Viren Tumorzellen infizieren, sich in ihnen replizieren und sie in Zellkultur schließlich ebenso dosis- und zeitabh{\"a}ngig effizient abt{\"o}ten konnten. CTLA4 scAb und β-Glucuronidase (GusA) Expression sowie Virus Titer in GLV-1h376-infizierten A549-Zellen wurde anhand von ELISA-, β-Glucuronidase- and Standard Plaque-Assays bestimmt. Hierbei zeigte sich eine ausgezeichnete Korrelation mit Korrelationskoeffizienten R2>0.9806. Der durch das GLV-1h376 kodierte CTLA4 scAb wurde erfolgreich aus {\"U}berst{\"a}nden von infizierten CV-1-Zellen gereinigt. CTLA4 scAb hat eine hohe in-vitro-Affinit{\"a}t zu seinem menschlichen CTLA4-Zielmolek{\"u}l sowie abwesende Kreuzreaktivit{\"a}t gegen{\"u}ber murine CTLA4 gezeigt. CTLA4 scAb Funktionalit{\"a}t wurde in Jurkat-Zellen best{\"a}tigt. LIVP-1.1.1, GLV-2b372, GLV-1h68 und GLV-1h376 wurden auch in nicht-tumor{\"o}sen und/oder tumortragenden humanisierten M{\"a}usen getestet. Zun{\"a}chst wurde gezeigt, dass die Injektion von menschlichen CD34+ Stammzellen in die Leber von vorkonditionierten neugeborenen NSG M{\"a}usen zu einer erfolgreichen systemische Rekonstitution mit menschlichen Immunzellen gef{\"u}hrt hat. CD19+-B-Zellen, CD4+- und CD8+-CD3+-T-Zellen, NKp46+CD56- und NKp46+CD56+-NK-Zellen sowie CD33+-myeloischen Zellen wurden detektiert. Die Mehrheit der nachgewisenen humanen h{\"a}matopoetischen Zellen im M{\"a}useblut in den ersten Wochen nach der Humanisierung waren CD19+-B-Zellen, und nur ein kleiner Teil waren CD3+-T-Zellen. Mit der Zeit wurde eine signifikante Ver{\"a}nderung in CD19+/CD3+-Verh{\"a}ltnis beobachtet, die parallel zur Abnahme der B-Zellen und einem Anstieg der T-Zellen kam. Die Implantation von A549-Zellen unter die Haut dieser M{\"a}use f{\"u}hrte zu einem progressiven Tumorwachstum. Bildgebende Verfahren zur Detektion von Virus-vermittelter TurboFP635- und GFP-Expression, Standard Plaque Assays sowie immunohistochemische Analysen best{\"a}tigten die erfolgreiche Invasion der Viren in die subkutanen Tumoren. Die humane CD45+-Zellpopulation in Tumoren wurde haupts{\"a}chlich durch NKp46+CD56bright-NK-Zellen und einen hohen Anteil von aktivierten CD4+- und zytotoxische CD8+-T-Zellen dargestellt. Es wurden jedoch keine signifikanten Unterschiede zwischen den Kontroll- und LIVP-1.1.1-infizierten Tumoren beobachtet, was darauf hindeutete, dass die Rekrutierung von NK- und aktivierten T-Zellen, mehr Tumorgewebe-spezifisch als Virus-abh{\"a}ngig waren. Die GLV-1h376-vermittelten CTLA4 scAb-Expression in den infizierten Tumoren war ebenfalls nicht in der Lage, die Aktivierung von Tumor-infiltrierenden T-Zellen im Vergleich zur Kontrolle und GLV-1h68-behandelten M{\"a}usen, signifikant zu erh{\"o}hen. ELISA-, β-Glucuronidase- and Standard Plaque-Assays zeigten eine eindeutige Korrelation mit den Korrelationskoeffizienten R2>0,9454 zwischen CTLA4 scAb- und GusA-Konzentrationen und Virus Titer in Tumorproben von GLV-1h376-behandelten M{\"a}usen. T-Zellen, die aus der Milz dieser Tumor-tragenden M{\"a}use isoliert wurden, waren funktionell und konnten erfolgreich mit Beads aktiviert werden. Mehr CD25+ und IFN-ɣ+ T-Zellen wurden in der GLV-1h376-Gruppe gefunden, wahrscheinlich aufgrund der CTLA4-Blockade durch die Virus-vermittelte CTLA4 scAb-Expression in den M{\"a}usen. Außerdem wurde eine h{\"o}here Konzentration von IL-2 in dem Kultur{\"u}berstand von diesen Splenozyten im Vergleich zu Kontrollproben nachgewiesen. Im Gegensatz zu der Aktivierung mit Beads konnten T-Zellen von allen drei Maus-Gruppen nicht durch A549 Tumorzellen ex vivo aktiviert werden. Unser Mausmodell hat den besonderen Vorteil, dass sich Tumoren unter der Haut der humanisierten M{\"a}use entwickeln, was eine genaue {\"U}berwachung des Tumorwachstums und Auswertung der onkolytischen Virotherapie erm{\"o}glicht.}, subject = {Vaccinia virus}, language = {en} } @article{EhrigKilincChenetal.2013, author = {Ehrig, Klaas and Kilinc, Mehmet O. and Chen, Nanhai G. and Stritzker, Jochen and Buckel, Lisa and Zhang, Qian and Szalay, Aladar A.}, title = {Growth inhibition of different human colorectal cancer xenografts after a single intravenous injection of oncolytic vaccinia virus GLV-1h68}, series = {Journal of Translational Medicine}, volume = {11}, journal = {Journal of Translational Medicine}, number = {79}, doi = {10.1186/1479-5876-11-79}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-129619}, year = {2013}, abstract = {Background: Despite availability of efficient treatment regimens for early stage colorectal cancer, treatment regimens for late stage colorectal cancer are generally not effective and thus need improvement. Oncolytic virotherapy using replication-competent vaccinia virus (VACV) strains is a promising new strategy for therapy of a variety of human cancers. Methods: Oncolytic efficacy of replication-competent vaccinia virus GLV-1h68 was analyzed in both, cell cultures and subcutaneous xenograft tumor models. Results: In this study we demonstrated for the first time that the replication-competent recombinant VACV GLV-1h68 efficiently infected, replicated in, and subsequently lysed various human colorectal cancer lines (Colo 205, HCT-15, HCT-116, HT-29, and SW-620) derived from patients at all four stages of disease. Additionally, in tumor xenograft models in athymic nude mice, a single injection of intravenously administered GLV-1h68 significantly inhibited tumor growth of two different human colorectal cell line tumors (Duke's type A-stage HCT-116 and Duke's type C-stage SW-620), significantly improving survival compared to untreated mice. Expression of the viral marker gene ruc-gfp allowed for real-time analysis of the virus infection in cell cultures and in mice. GLV-1h68 treatment was well-tolerated in all animals and viral replication was confined to the tumor. GLV-1h68 treatment elicited a significant up-regulation of murine immune-related antigens like IFN-γ, IP-10, MCP-1, MCP-3, MCP-5, RANTES and TNF-γ and a greater infiltration of macrophages and NK cells in tumors as compared to untreated controls. Conclusion: The anti-tumor activity observed against colorectal cancer cells in these studies was a result of direct viral oncolysis by GLV-1h68 and inflammation-mediated innate immune responses. The therapeutic effects occurred in tumors regardless of the stage of disease from which the cells were derived. Thus, the recombinant vaccinia virus GLV-1h68 has the potential to treat colorectal cancers independently of the stage of progression.}, language = {en} } @article{SturmHessWeibeletal.2012, author = {Sturm, Julia B. and Hess, Michael and Weibel, Stephanie and Chen, Nanhei G. and Yu, Yong A. and Zhang, Quian and Donat, Ulrike and Reiss, Cora and Gambaryan, Stepan and Krohne, Georg and Stritzker, Jochen and Szalay, Aladar A.}, title = {Functional hyper-IL-6 from vaccinia virus-colonized tumors triggers platelet formation and helps to alleviate toxicity of mitomycin C enhanced virus therapy}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-75224}, year = {2012}, abstract = {Background: Combination of oncolytic vaccinia virus therapy with conventional chemotherapy has shown promise for tumor therapy. However, side effects of chemotherapy including thrombocytopenia, still remain problematic. Methods: Here, we describe a novel approach to optimize combination therapy of oncolytic virus and chemotherapy utilizing virus-encoding hyper-IL-6, GLV-1h90, to reduce chemotherapy-associated side effects. Results: We showed that the hyper-IL-6 cytokine was successfully produced by GLV-1h90 and was functional both in cell culture as well as in tumor-bearing animals, in which the cytokine-producing vaccinia virus strain was well tolerated. When combined with the chemotherapeutic mitomycin C, the anti-tumor effect of the oncolytic virotherapy was significantly enhanced. Moreover, hyper-IL-6 expression greatly reduced the time interval during which the mice suffered from chemotherapy-induced thrombocytopenia. Conclusion: Therefore, future clinical application would benefit from careful investigation of additional cytokine treatment to reduce chemotherapy-induced side effects.}, subject = {Biologie}, language = {en} } @article{AdelfingerGentschevdeGuibertetal.2014, author = {Adelfinger, Marion and Gentschev, Ivaylo and de Guibert, Julio Grimm and Weibel, Stephanie and Langbein-Laugwitz, Johanna and H{\"a}rtl, Barbara and Escobar, Hugo Murua and Nolte, Ingo and Chen, Nanhai G. and Aguilar, Richard J. and Yu, Yong A. and Zhang, Qian and Frentzen, Alexa and Szalay, Aladar A.}, title = {Evaluation of a New Recombinant Oncolytic Vaccinia Virus Strain GLV-5b451 for Feline Mammary Carcinoma Therapy}, series = {PLoS ONE}, volume = {9}, journal = {PLoS ONE}, number = {8}, doi = {10.1371/journal.pone.0104337}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119387}, pages = {e104337}, year = {2014}, abstract = {Virotherapy on the basis of oncolytic vaccinia virus (VACV) infection is a promising approach for cancer therapy. In this study we describe the establishment of a new preclinical model of feline mammary carcinoma (FMC) using a recently established cancer cell line, DT09/06. In addition, we evaluated a recombinant vaccinia virus strain, GLV-5b451, expressing the anti-vascular endothelial growth factor (VEGF) single-chain antibody (scAb) GLAF-2 as an oncolytic agent against FMC. Cell culture data demonstrate that GLV-5b451 virus efficiently infected, replicated in and destroyed DT09/06 cancer cells. In the selected xenografts of FMC, a single systemic administration of GLV-5b451 led to significant inhibition of tumor growth in comparison to untreated tumor-bearing mice. Furthermore, tumor-specific virus infection led to overproduction of functional scAb GLAF-2, which caused drastic reduction of intratumoral VEGF levels and inhibition of angiogenesis. In summary, here we have shown, for the first time, that the vaccinia virus strains and especially GLV-5b451 have great potential for effective treatment of FMC in animal model.}, language = {en} } @phdthesis{Buckel2012, author = {Buckel, Lisa}, title = {Evaluating the combination of oncolytic vaccinia virus and ionizing radiation in therapy of preclinical glioma models}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-85309}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Glioblastoma multiforme (GBM) represents the most aggressive form of malignant brain tumors and remains a therapeutically challenge. Intense research in the field has lead to the testing of oncolytic viruses to improve tumor control. Currently, a variety of different oncolytic viruses are being evaluated for their ability to be used in anti-cancer therapy and a few have entered clinical trials. Vaccinia virus, is one of the viruses being studied. GLV-1h68, an oncolytic vaccinia virus engineered by Genelux Corporation, was constructed by insertion of three gene cassettes, RUC-GFP fusion, β-galactosidase and β- glucuronidase into the genome of the LIVP strain. Since focal tumor radiotherapy is a mainstay for cancer treatment, including glioma therapy, it is of clinical relevance to assess how systemically administered oncolytic vaccinia virus could be combined with targeted ionizing radiation for therapeutic gain. In this work we show how focal ionizing radiation (IR) can be combined with multiple systemically delivered oncolytic vaccinia virus strains in murine models of human U-87 glioma. After initial experiments which confirmed that ionizing radiation does not damage viral DNA or alter viral tropism, animal studies were carried out to analyze the interaction of vaccinia virus and ionizing radiation in the in vivo setting. We found that irradiation of the tumor target, prior to systemic administration of oncolytic vaccinia virus GLV-1h68, increased viral replication within the U-87 xenografts as measured by viral reporter gene expression and viral titers. Importantly, while GLV-1h68 alone had minimal effect on U-87 tumor growth delay, IR enhanced GLV-1h68 replication, which translated to increased tumor growth delay and mouse survival in subcutaneous and orthotopic U-87 glioma murine models compared to monotherapy with IR or GLV-1h68. The ability of IR to enhance vaccinia replication was not restricted to the multi-mutated GLV-1h68, but was also seen with the less attenuated oncolytic vaccinia, LIVP 1.1.1. We have demonstrated that in animals treated with combination of ionizing radiation and LIVP 1.1.1 a strong pro-inflammatory tissue response was induced. When IR was given in a more clinically relevant fractionated scheme, we found oncolytic vaccinia virus replication also increased. This indicates that vaccinia virus could be incorporated into either larger hypo-fraction or more conventionally fractionated radiotherapy schemes. The ability of focal IR to mediate selective replication of systemically injected oncolytic vaccinia was demonstrated in a bilateral glioma model. In mice with bilateral U-87 tumors in both hindlimbs, systemically administered oncolytic vaccinia replicated preferentially in the focally irradiated tumor compared to the shielded non- irradiated tumor in the same mouse We demonstrated that tumor control could be further improved when fractionated focal ionizing radiation was combined with a vaccinia virus caring an anti-angiogenic payload targeting vascular endothelial growth factor (VEGF). Our studies showed that following ionizing radiation expression of VEGF is upregulated in U-87 glioma cells in culture. We further showed a concentration dependent increase in radioresistance of human endothelial cells in presence of VEGF. Interestingly, we found effects of vascular endothelial growth factor on endothelial cells were reversible by adding purified GLAF-1 to the cells. GLAF-1 is a single- chain antibody targeting human and murine VEGF and is expressed by oncolytic vaccinia virus GLV-109. In U-87 glioma xenograft murine models the combination of fractionated ionizing radiation with GLV-1h164, a vaccinia virus also targeting VEGF, resulted in the best volumetric tumor response and a drastic decrease in vascular endothelial growth factor. Histological analysis of embedded tumor sections 14 days after viral administration confirmed that blocking VEGF translated into a decrease in vessel number to 30\% of vessel number found in control tumors in animals treated with GLV-164 and fractionated IR which was lower than for all other treatment groups. Our experiments with GLV-1h164 and fractionated radiotherapy have shown that in addition to ionizing radiation and viral induced tumor cell destruction we were able to effectively target the tumor vasculature. This was achieved by enhanced viral replication translating in increased levels of GLAF-2 disrupting tumor vessels as well as the radiosensitization of tumor vasculature to IR by blocking VEGF. Our preclinical results have important clinical implications of how focal radiotherapy can be combined with systemic oncolytic viral administration for highly aggressive, locally advanced tumors with the potential, by using a vaccinia virus targeting human vascular endothelial growth factor, to further increase tumor radiation sensitivity by engaging the vascular component in addition to cancer cells.}, subject = {Gliom}, language = {en} }