TY - JOUR A1 - Gentschev, Ivaylo A1 - Patil, Sadeep S. A1 - Petrov, Ivan A1 - Cappello, Joseph A1 - Adelfinger, Marion A1 - Szalay, Aladar A. T1 - Oncolytic Virotherapy of Canine and Feline Cancer JF - Viruses N2 - Cancer is the leading cause of disease-related death in companion animals such as dogs and cats. Despite recent progress in the diagnosis and treatment of advanced canine and feline cancer, overall patient treatment outcome has not been substantially improved. Virotherapy using oncolytic viruses is one promising new strategy for cancer therapy. Oncolytic viruses (OVs) preferentially infect and lyse cancer cells, without causing excessive damage to surrounding healthy tissue, and initiate tumor-specific immunity. The current review describes the use of different oncolytic viruses for cancer therapy and their application to canine and feline cancer. KW - oncolytic virus KW - oncolysis KW - cancer KW - canine and feline cancer therapy Y1 - 2014 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-119753 VL - 6 IS - 5 ER - TY - JOUR A1 - Draganov, Dobrin D. A1 - Santidrian, Antonio F. A1 - Minev, Ivelina A1 - Duong, Nguyen A1 - Kilinc, Mehmet Okyay A1 - Petrov, Ivan A1 - Vyalkova, Anna A1 - Lander, Elliot A1 - Berman, Mark A1 - Minev, Boris A1 - Szalay, Aladar A. T1 - Delivery of oncolytic vaccinia virus by matched allogeneic stem cells overcomes critical innate and adaptive immune barriers JF - Journal of Translational Medicine N2 - Background Previous studies have identified IFNγ as an important early barrier to oncolytic viruses including vaccinia. The existing innate and adaptive immune barriers restricting oncolytic virotherapy, however, can be overcome using autologous or allogeneic mesenchymal stem cells as carrier cells with unique immunosuppressive properties. Methods To test the ability of mesenchymal stem cells to overcome innate and adaptive immune barriers and to successfully deliver oncolytic vaccinia virus to tumor cells, we performed flow cytometry and virus plaque assay analysis of ex vivo co-cultures of stem cells infected with vaccinia virus in the presence of peripheral blood mononuclear cells from healthy donors. Comparative analysis was performed to establish statistically significant correlations and to evaluate the effect of stem cells on the activity of key immune cell populations. Results Here, we demonstrate that adipose-derived stem cells (ADSCs) have the potential to eradicate resistant tumor cells through a combination of potent virus amplification and sensitization of the tumor cells to virus infection. Moreover, the ADSCs demonstrate ability to function as a virus-amplifying Trojan horse in the presence of both autologous and allogeneic human PBMCs, which can be linked to the intrinsic immunosuppressive properties of stem cells and their unique potential to overcome innate and adaptive immune barriers. The clinical application of ready-to-use ex vivo expanded allogeneic stem cell lines, however, appears significantly restricted by patient-specific allogeneic differences associated with the induction of potent anti-stem cell cytotoxic and IFNγ responses. These allogeneic responses originate from both innate (NK)- and adaptive (T)- immune cells and might compromise therapeutic efficacy through direct elimination of the stem cells or the induction of an anti-viral state, which can block the potential of the Trojan horse to amplify and deliver vaccinia virus to the tumor. Conclusions Overall, our findings and data indicate the feasibility to establish simple and informative assays that capture critically important patient-specific differences in the immune responses to the virus and stem cells, which allows for proper patient-stem cell matching and enables the effective use of off-the-shelf allogeneic cell-based delivery platforms, thus providing a more practical and commercially viable alternative to the autologous stem cell approach. KW - vaccinia KW - cancer KW - stem Cells KW - oncolysis KW - oncolytic virus KW - virotherapy KW - immunity KW - immunotherapy Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-226312 SN - 100 VL - 17 ER - TY - JOUR A1 - Cecil, Alexander A1 - Gentschev, Ivaylo A1 - Adelfinger, Marion A1 - Dandekar, Thomas A1 - Szalay, Aladar A. T1 - Vaccinia virus injected human tumors: oncolytic virus efficiency predicted by antigen profiling analysis fitted boolean models JF - Bioengineered N2 - Virotherapy on the basis of oncolytic vaccinia virus (VACV) strains is a promising approach for cancer therapy. Recently, we showed that the oncolytic vaccinia virus GLV-1h68 has a therapeutic potential in treating human prostate and hepatocellular carcinomas in xenografted mice. In this study, we describe the use of dynamic boolean modeling for tumor growth prediction of vaccinia virus-injected human tumors. Antigen profiling data of vaccinia virus GLV-1h68-injected human xenografted mice were obtained, analyzed and used to calculate differences in the tumor growth signaling network by tumor type and gender. Our model combines networks for apoptosis, MAPK, p53, WNT, Hedgehog, the T-killer cell mediated cell death, Interferon and Interleukin signaling networks. The in silico findings conform very well with in vivo findings of tumor growth. Similar to a previously published analysis of vaccinia virus-injected canine tumors, we were able to confirm the suitability of our boolean modeling for prediction of human tumor growth after virus infection in the current study as well. In summary, these findings indicate that our boolean models could be a useful tool for testing of the efficacy of VACV-mediated cancer therapy already before its use in human patients. KW - boolean modeling KW - oncolytic virus KW - human xenografted mouse models KW - cancer therapy Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-200507 VL - 10 IS - 1 ER - TY - JOUR A1 - Adelfinger, Marion A1 - Bessler, Simon A1 - Cecil, Alexander A1 - Langbein-Laugwitz, Johanna A1 - Frentzen, Alexa A1 - Gentschev, Ivaylo A1 - Szalay, Aladar A. T1 - Preclinical Testing Oncolytic Vaccinia Virus Strain GLV-5b451 Expressing an Anti-VEGF Single-Chain Antibody for Canine Cancer Therapy JF - Viruses N2 - Virotherapy on the basis of oncolytic vaccinia virus (VACV) strains is a novel approach for canine cancer therapy. Here we describe, for the first time, the characterization and the use of VACV strain GLV-5b451 expressing the anti-vascular endothelial growth factor (VEGF) single-chain antibody (scAb) GLAF-2 as therapeutic agent against different canine cancers. Cell culture data demonstrated that GLV-5b451 efficiently infected and destroyed all four tested canine cancer cell lines including: mammary carcinoma (MTH52c), mammary adenoma (ZMTH3), prostate carcinoma (CT1258), and soft tissue sarcoma (STSA-1). The GLV-5b451 virus-mediated production of GLAF-2 antibody was observed in all four cancer cell lines. In addition, this antibody specifically recognized canine VEGF. Finally, in canine soft tissue sarcoma (CSTS) xenografted mice, a single systemic administration of GLV-5b451 was found to be safe and led to anti-tumor effects resulting in the significant reduction and substantial long-term inhibition of tumor growth. A CD31-based immuno-staining showed significantly decreased neo-angiogenesis in GLV-5b451-treated tumors compared to the controls. In summary, these findings indicate that GLV-5b451 has potential for use as a therapeutic agent in the treatment of CSTS. KW - canine cancer therapy KW - canine soft tissue sarcoma (CSTS) KW - oncolytic virus KW - cancer KW - canine cancer cell lines KW - antibody production KW - angiogenesis Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-125705 VL - 7 ER - TY - THES A1 - Adelfinger, Marion T1 - Präklinische Verwendung verschiedener onkolytischer Vaccinia-Viren zur Therapie von Human- und Hundetumoren T1 - Preclinical application of different oncolytic vaccinia viruses in human and canine tumor therapy N2 - Nach Einschätzung der Weltgesundheitsorganisation WHO wird Krebs im Jahr 2013 die weltweit häufigste Todesursache bei Menschen und Haustieren sein. Diese Situation erfordert die Entwicklung neuer therapeutischer Ansätze. Hauptziel einer Tumortherapie ist es, sowohl den Primärtumor als auch die Metastasen möglichst vollständig zu entfernen. Dabei wird nach Methoden gesucht, die im Gegensatz zu den meisten gegenwärtigen therapeutischen Einsätzen, wie der chirurgischen Entfernung bösartiger Neubildungen, Chemotherapie und Strahlentherapie, selektiv die bösartigen Zellen erkennen und zerstören können. Eine faszinierende Möglichkeit in dieser Hinsicht ist die Verwendung von onkolytischen Viren, die die Fähigkeit besitzen, sich selektiv sowohl in Primärtumoren als auch in Metastasen anzusiedeln und die Krebszellen dort zu zerstören. Das Konzept, dass Viren nützlich für die Bekämpfung von Krebs sein könnten, ist nicht neu. Allerdings konnte erst in den letzten Jahren durch zahlreiche Studien bestätigt werden, dass verschiedene Viren in der Lage sind, eine signifikante Antitumorwirkung in vivo auszuüben. Zu den erfolgversprechenden onkolytischen Viren zählen 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-Stämmen 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 extrazellulären Matrix zusammen. Die Krebszellen bilden unter anderem mit Endothelzellen des Blut- und Lymphsystems und verschiedenen Immunzellen eine komplexe Organ-ähnliche Struktur. Weitere wichtige Bestandteile des Stromas sind Wachstumsfaktoren, Chemokine und Zytokine und die Tumorvaskulatur. Diese ist durch zahlreiche strukturelle und funktionelle Abnormalitäten charakterisiert, wodurch die Effektivität von Strahlen- und Chemotherapie herabgesetzt wird. Weiterhin ist das Tumormikromilieu durch seine Ähnlichkeit mit einer chronischen Entzündungsreaktion gekennzeichnet und wirkt immunsupprimierend auf rekrutierte Leukozyten, die wiederum die Inflammation verstärken und die Angiogenese und das Tumorwachstum weiter fördern. Aufgrund dieser vielen Komponenten ist die Zusammensetzung jedes Tumors einzigartig, weswegen Standardtherapien häufig nicht zu einer Heilung führen. Die Wirkung der Viren bei der Virotherapie beruht vermutlich auf 4 Mechanismen, die einzeln oder in Kombination auftreten können: die direkte Onkolyse der Krebszellen, die Zerstörung des Tumorblutgefäßsystems, die Aktivierung des Immunsystems des Wirts und die Suppression der microRNA-Expression des Wirtes. Zusätzlich kann die Expression therapeutischer Gene die onkolytische Wirkung verstärken. 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 frühen Zeitpunkt der Krebserkrankung aus einer Metastase isolierte Zelllinie, zeigt nach Infektion mit GLV-1h68 im Mausmodell Tumornekrose („Responder“), während 1936-MEL aus einer späten Metastasierungsphase kaum mit Onkolyse auf eine Virusinfektion reagiert („Poor-Responder“). Die onkolytische Wirkung konnte mittels Durchflusszytometrie in Tumoren beider Zelllinien zu einem frühen 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 schwächer reagierenden 1936-MEL-Tumoren konnte eine Onkolyse bei Infektion mit höherer Virusmenge und zu einem früheren Zeitpunkt demonstriert werden, wodurch mehr Zellen infiziert wurden. Zusätzlich 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. ... N2 - The cancer mortality rate is increasing steadily in man and pet animals and it is estimated by the World Health Organisation (WHO) to be number one cause of deaths in 2013. This situation raises the need for developing new therapeutic approaches whereby the main goal of tumor therapy always is the complete elimination of primary tumor and metastases. In contrast to most current therapies, like surgical resection of malignant neoplasia, chemotherapy and radiation, the new cancer treatments should selectively recognize and destroy malignant cells. The application of oncolytic viruses offers fascinating opportunities, because of their ability to selectively colonize primary tumors and metastases and directly destroy cancer cells. The concept of fighting cancer with viruses is not new, but the significant anti-tumoral effect of different viruses in vivo was demonstrated by several studies in the last few years. Promising oncolytic viruses are adenovirus, Herpes simplex virus, reovirus and vaccinia virus. These viruses are currently in or entering phase III clinical trials but the therapeutic use of tumor specific oncolytic viruses for humans has already started. In this work, different aspects of the effects of vaccinia virus strains during therapy of canine and human tumors in mouse xenograft models were analyzed: oncolysis of cancer cells, inhibition of tumor growth, influence of virus infection on the tumor microenvironment and participation of the innate immunity in virotherapy. The tumor microenvironment (stroma) is composed of many different cells and components of the extracellular matrix. Cancer cells form structures similar to organs including endothelial cells of the blood and lymph system, different immune cells and many other cell types. Other important components of the stroma are growth factors, chemokines and cytokines as well as the tumor vasculature that is characterized by numerous structural and functional abnormalities which decrease the efficacy of radiation and chemotherapy. Another hallmark of the tumor microenvironment is its similarity to chronic inflammation and its immunosuppressive effect on recruited leukocytes which in turn increase the inflammation and angiogenesis and promote tumor growth. Due to the many different components the composition of each tumor is unique and that is why standard therapies often do not result in cure. The effects of the viruses in the virotherapy are propably based on four mechanisms working alone or in combination: direct oncolysis of cancer cells, destruction of tumor vasculature, activation of host immune system and suppression of host microRNA expression. Additionally the expression of therapeutic genes can increase the oncolytic effect of the viruses. To analyze the oncolysis of cancer cells and the inhibition of tumor growth first the GLV-1h68 virus was used in an autologous pair of human melanoma 888-MEL and 1936-MEL. GLV-1h68 was genetically constructed on the basis of wild-type vaccinia virus LIVP by insertion of 3 expression cassettes into F14.5L, J2R and A56 gene loci. 888-MEL was isolated from metastases early during cancer disease and exhibits tumor necrosis after infection with GLV-1h68 in mouse xenograft model (responder) whereas 1936-MEL which was isolated in the late phase of metastases formation hardly shows oncolysis following virus treatment (poor-responder). The oncolytic effect was demonstrated by flow cytometric analysis of tumors of both cell lines at an early time after virus infection. In 888-MEL tumors high numbers of infected and dead cells were found after virus infection. Simultaneously, a high number of immune cells were detected which was reduced after infection. In the lower responding 1936-MEL tumors oncolysis was only observed after infection with higher amount of virus at an earlier time whereby the number of infected cells increased. Additionally, the low number of immune cells increased after infection. Despite the different tumor microenvironment an oncolytic effect was achieved in both tumor models. ... KW - Vaccinia-Virus KW - Tumortherapie KW - Tumorimmunologie KW - Hundetumor KW - onkolytisches Virus KW - vaccinia virus KW - tumor therapy KW - tumor immunology KW - canine tumor KW - oncolytic virus KW - Tumor KW - Therapie KW - Onkolyse Y1 - 2012 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-70688 ER -