@article{SchmidtAltDeoghareetal.2022, author = {Schmidt, Sven and Alt, Yvonne and Deoghare, Nikita and Kr{\"u}ger, Sarah and Kern, Anna and Rockel, Anna Frederike and Wagner, Nicole and Erg{\"u}n, S{\"u}leyman and W{\"o}rsd{\"o}rfer, Philipp}, title = {A blood vessel organoid model recapitulating aspects of vasculogenesis, angiogenesis and vessel wall maturation}, series = {Organoids}, volume = {1}, journal = {Organoids}, number = {1}, issn = {2674-1172}, doi = {10.3390/organoids1010005}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-284043}, pages = {41 -- 53}, year = {2022}, abstract = {Blood vessel organoids are an important in vitro model to understand the underlying mechanisms of human blood vessel development and for toxicity testing or high throughput drug screening. Here we present a novel, cost-effective, and easy to manufacture vascular organoid model. To engineer the organoids, a defined number of human induced pluripotent stem cells are seeded in non-adhesive agarose coated wells of a 96-well plate and directed towards a lateral plate mesoderm fate by activation of Wnt and BMP4 signaling. We observe the formation of a circular layer of angioblasts around days 5-6. Induced by VEGF application, CD31\(^+\) vascular endothelial cells appear within this vasculogenic zone at approximately day 7 of organoid culture. These cells arrange to form a primitive vascular plexus from which angiogenic sprouting is observed after 10 days of culture. The differentiation outcome is highly reproducible, and the size of organoids is scalable depending on the number of starting cells. We observe that the initial vascular ring forms at the interface between two cell populations. The inner cellular compartment can be distinguished from the outer by the expression of GATA6, a marker of lateral plate mesoderm. Finally, 14-days-old organoids were transplanted on the chorioallantois membrane of chicken embryos resulting in a functional connection of the human vascular network to the chicken circulation. Perfusion of the vessels leads to vessel wall maturation and remodeling as indicated by the formation of a continuous layer of smooth muscle actin expressing cells enwrapping the endothelium. In summary, our organoid model recapitulates human vasculogenesis, angiogenesis as well as vessel wall maturation and therefore represents an easy and cost-effective tool to study all steps of blood vessel development and maturation directly in the human setting without animal experimentation.}, language = {en} } @phdthesis{Schleider2010, author = {Schleider, Elisa}, title = {Angiogenese-Modellsysteme zur Funktionsanalyse des humanen CCM3 Proteins}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-51504}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2010}, abstract = {Zerebrovaskul{\"a}re kavern{\"o}se Malformationen (CCM) sind Blutgef{\"a}ßfehlbildungen, welche haupts{\"a}chlich im Gehirn vorkommen. Sie sind gekennzeichnet durch stark dilatierte kapillar{\"a}hnliche Gef{\"a}ße mit niedriger Flussrate („slow-flow lesions"). Intervenierendes Gehirnparenchym fehlt ebenso wie Perizyten oder glatte Gef{\"a}ßmuskelzellen. Die klinischen Symptome reichen von starken Kopfschmerzen {\"u}ber Epilepsie bis hin zum Schlaganfall. Dennoch bleiben viele Kavernomtr{\"a}ger aufgrund unvollst{\"a}ndiger Penetranz ihr Leben lang asymptomatisch. Die Pr{\"a}valenz betr{\"a}gt ca. 0,5\% in der Gesamtbev{\"o}lkerung. Es gibt sowohl sporadische als auch dominant vererbte Krankheitsformen. In den letzten Jahren konnten 3 Gene urs{\"a}chlich mit der Krankheit in Verbindung gebracht werden. Mutationen in CCM1, CCM2 oder CCM3 f{\"u}hren zu einem nicht unterscheidbaren klinischen Ph{\"a}notyp. Alle drei Proteine bilden einen tern{\"a}ren Komplex in vitro, was eine Beteiligung an einem gemeinsamen molekularen Signalweg bekr{\"a}ftigt. W{\"a}hrend die Proteine CCM1 und CCM2 in den letzten Jahren umfangreich erforscht wurden, ist {\"u}ber das CCM3-Protein bis heute wenig bekannt. In dieser Arbeit konnte gezeigt werden, dass CCM3 eine wichtige Rolle in der Angiogenese spielt und diese bei {\"U}berexpression in humanen Endothelzellen stark negativ reguliert: die Migration, die Proliferation und die F{\"a}higkeit, kapillar{\"a}hnliche Strukturen in Matrix-Gelen zu bilden kommt nahezu zum Erliegen. Ein gegenl{\"a}ufiger Effekt nach siRNA induziertem Knock-down von CCM3 war weniger stark ausgepr{\"a}gt. Einzig die F{\"a}higkeit, gef{\"a}ß{\"a}hnliche Strukturen in Matrigelen zu bilden, war erh{\"o}ht. Um weiterhin Klarheit {\"u}ber die intrazellul{\"a}ren, von CCM3 beeinflussten Signalwege zu schaffen, wurden Tyrosin Kinase Arrays durchgef{\"u}hrt, bei welchen CCM3-{\"u}berexprimierende HUVEC Lysate mit Kontrolllysaten verglichen wurden. Dabei stellte sich heraus, dass 5 Substrate signifikant erh{\"o}ht phosphoryliert wurden: der Discoidin Dom{\"a}nen Rezeptor 1 (discoidin domain receptor; DDR1), die duale spezifit{\"a}tstyrosinphosphorylierungsregulierte Kinase 1A (dual specificity tyrosine-phosphorylation-regulated kinase 1A; DYR1A), die Protoonkogen Tyrosin- Protein Kinase FER (proto-oncogene tyrosine-protein kinase FER; FER), die fynbezogene Kinase (Fyn-related kinase; FRK) und die phosphoinositolabh{\"a}ngige Kinase 1 (Phosphoinositide-dependent kinase 1, PDPK-1). Im Folgenden best{\"a}tigten Western Blot, dass die {\"U}berexpression von CCM3 in Endothelzellen die phosphoinositolabh{\"a}ngige Kinase 1 und die nachgeschaltete Serin-Threonin Kinase Akt/PBK aktiviert, welche ein bedeutsames {\"U}berlebenssignal der Zelle darstellt. Schließlich konnte gezeigt werden, dass CCM3 nicht nur antiangiogen, sondern auch antiapoptotisch wirkt. Die Ergebnisse der vorliegenden Arbeit legen nahe, dass CCM3 f{\"u}r die Integrit{\"a}t des ruhenden, adulten Endothelbettes wichtig ist.}, subject = {Blutgef{\"a}ß}, language = {de} } @article{ArgentieroSolimandoKrebsetal.2020, author = {Argentiero, Antonella and Solimando, Antonio Giovanni and Krebs, Markus and Leone, Patrizia and Susca, Nicola and Brunetti, Oronzo and Racanelli, Vito and Vacca, Angelo and Silvestris, Nicola}, title = {Anti-angiogenesis and immunotherapy: novel paradigms to envision tailored approaches in renal cell-carcinoma}, series = {Journal of Clinical Medicine}, volume = {9}, journal = {Journal of Clinical Medicine}, number = {5}, issn = {2077-0383}, doi = {10.3390/jcm9051594}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-205846}, year = {2020}, abstract = {Although decision making strategy based on clinico-histopathological criteria is well established, renal cell carcinoma (RCC) represents a spectrum of biological ecosystems characterized by distinct genetic and molecular alterations, diverse clinical courses and potential specific therapeutic vulnerabilities. Given the plethora of drugs available, the subtype-tailored treatment to RCC subtype holds the potential to improve patient outcome, shrinking treatment-related morbidity and cost. The emerging knowledge of the molecular taxonomy of RCC is evolving, whilst the antiangiogenic and immunotherapy landscape maintains and reinforces their potential. Although several prognostic factors of survival in patients with RCC have been described, no reliable predictive biomarkers of treatment individual sensitivity or resistance have been identified. In this review, we summarize the available evidence able to prompt more precise and individualized patient selection in well-designed clinical trials, covering the unmet need of medical choices in the era of next-generation anti-angiogenesis and immunotherapy.}, language = {en} } @article{RathBrandlHilleretal.2014, author = {Rath, Subha N. and Brandl, Andreas and Hiller, Daniel and Hoppe, Alexander and Gbureck, Uwe and Horch, Raymund E. and Boccaccini, Aldo R. and Kneser, Ulrich}, title = {Bioactive Copper-Doped Glass Scaffolds Can Stimulate Endothelial Cells in Co-Culture in Combination with Mesenchymal Stem Cells}, series = {PLOS ONE}, volume = {9}, journal = {PLOS ONE}, number = {12}, issn = {1932-6203}, doi = {10.1371/journal.pone.0113319}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-114339}, year = {2014}, abstract = {Bioactive glass (BG) scaffolds are being investigated for bone tissue engineering applications because of their osteoconductive and angiogenic nature. However, to increase the in vivo performance of the scaffold, including enhancing the angiogenetic growth into the scaffolds, some researchers use different modifications of the scaffold including addition of inorganic ionic components to the basic BG composition. In this study, we investigated the in vitro biocompatibility and bioactivity of Cu2+-doped BG derived scaffolds in either BMSC (bone-marrow derived mesenchymal stem cells)-only culture or co-culture of BMSC and human dermal microvascular endothelial cells (HDMEC). In BMSC-only culture, cells were seeded either directly on the scaffolds (3D or direct culture) or were exposed to ionic dissolution products of the BG scaffolds, kept in permeable cell culture inserts (2D or indirect culture). Though we did not observe any direct osteoinduction of BMSCs by alkaline phosphatase (ALP) assay or by PCR, there was increased vascular endothelial growth factor (VEGF) expression, observed by PCR and ELISA assays. Additionally, the scaffolds showed no toxicity to BMSCs and there were healthy live cells found throughout the scaffold. To analyze further the reasons behind the increased VEGF expression and to exploit the benefits of the finding, we used the indirect method with HDMECs in culture plastic and Cu2+-doped BG scaffolds with or without BMSCs in cell culture inserts. There was clear observation of increased endothelial markers by both FACS analysis and acetylated LDL (acLDL) uptake assay. Only in presence of Cu2+-doped BG scaffolds with BMSCs, a high VEGF secretion was demonstrated by ELISA; and typical tubular structures were observed in culture plastics. We conclude that Cu2+-doped BG scaffolds release Cu2+, which in turn act on BMSCs to secrete VEGF. This result is of significance for the application of BG scaffolds in bone tissue engineering approaches.}, language = {en} } @article{KleefeldtUpcinBoemmeletal.2022, author = {Kleefeldt, Florian and Upcin, Berin and B{\"o}mmel, Heike and Schulz, Christian and Eckner, Georg and Allmanritter, Jan and Bauer, Jochen and Braunger, Barbara and Rueckschloss, Uwe and Erg{\"u}n, S{\"u}leyman}, title = {Bone marrow-independent adventitial macrophage progenitor cells contribute to angiogenesis}, series = {Cell Death \& Disease}, volume = {13}, journal = {Cell Death \& Disease}, number = {3}, doi = {10.1038/s41419-022-04605-2}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-299724}, year = {2022}, abstract = {Pathological angiogenesis promotes tumor growth, metastasis, and atherosclerotic plaque rupture. Macrophages are key players in these processes. However, whether these macrophages differentiate from bone marrow-derived monocytes or from local vascular wall-resident stem and progenitor cells (VW-SCs) is an unresolved issue of angiogenesis. To answer this question, we analyzed vascular sprouting and alterations in aortic cell populations in mouse aortic ring assays (ARA). ARA culture leads to the generation of large numbers of macrophages, especially within the aortic adventitia. Using immunohistochemical fate-mapping and genetic in vivo-labeling approaches we show that 60\% of these macrophages differentiate from bone marrow-independent Ly6c\(^{+}\)/Sca-1\(^{+}\) adventitial progenitor cells. Analysis of the NCX\(^{-/-}\) mouse model that genetically lacks embryonic circulation and yolk sac perfusion indicates that at least some of those progenitor cells arise yolk sac-independent. Macrophages represent the main source of VEGF in ARA that vice versa promotes the generation of additional macrophages thereby creating a pro-angiogenetic feedforward loop. Additionally, macrophage-derived VEGF activates CD34\(^{+}\) progenitor cells within the adventitial vasculogenic zone to differentiate into CD31\(^{+}\) endothelial cells. Consequently, depletion of macrophages and VEGFR2 antagonism drastically reduce vascular sprouting activity in ARA. In summary, we show that angiogenic activation induces differentiation of macrophages from bone marrow-derived as well as from bone marrow-independent VW-SCs. The latter ones are at least partially yolk sac-independent, too. Those VW-SC-derived macrophages critically contribute to angiogenesis, making them an attractive target to interfere with pathological angiogenesis in cancer and atherosclerosis as well as with regenerative angiogenesis in ischemic cardiovascular disorders.}, language = {en} } @phdthesis{Scheller2012, author = {Scheller, Katharina}, title = {Charakterisierung und Anwendung von humanen, prim{\"a}ren mikrovaskul{\"a}ren Endothelzellen mit erweiterter Proliferationsf{\"a}higkeit}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-76577}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Das Arbeitsgebiet Tissue Engineering befasst sich mit der Kl{\"a}rung der Mechanismen, die der Funktionen verschiedener Gewebearten zu Grunde liegen sowie mit der Entwicklung alternativer Strategien zur Behandlung von Organversagen bzw. Organverlusten. Einer der kritischsten Punkte im Tissue Engineering ist die ausreichende Versorgung der Zellen mit N{\"a}hrstoffen und Sauerstoff. Bioartifizielle Gewebe mit einer Dicke von bis zu 200 µm k{\"o}nnen mittels Diffusion ausreichend versorgt werden. F{\"u}r dickere Transplantate ist die Versorgung der Zellen alleine durch Diffusion jedoch nicht gegeben. Hierf{\"u}r m{\"u}ssen Mechanismen und Strategien zur Pr{\"a}vaskularisierung der artifiziellen Gewebekonstrukte entwickelt werden, damit die N{\"a}hrstoff- und Sauerstoffversorgung aller Zellen, auch im Inneren des Transplantates, von Anfang an gew{\"a}hrleistet ist. Eine wichtige Rolle bei der Pr{\"a}vaskularisierung spielt die Angiogenese. Dabei ist die Wahl einer geeigneten Zellquelle entscheidend, da die Zellen die Basis f{\"u}r die Angiogenese darstellen. Mikrovaskul{\"a}re Endothelzellen (mvEZ) sind maßgeblich an der Angiogenese beteiligt. Das Problem bei der Verwendung von humanen prim{\"a}ren mvEZ ist ihre geringe Verf{\"u}gbarkeit, ihre limitierte Proliferationskapazit{\"a}t und der schnelle Verlust ihrer typischen Endothelzellmarker in-vitro. Der Aufbau standardisierter in-vitro Testsysteme ist durch die geringe Zellausbeute auch nicht m{\"o}glich. Die upcyte® Technologie bietet hierf{\"u}r einen L{\"o}sungsansatz. In der vorliegenden Arbeit konnten upcyte® mvEZ als Alternative zu prim{\"a}ren mvEZ generiert werden. Es konnte gezeigt werden, dass die Zellen eine erweiterte Proliferationsf{\"a}higkeit aufweisen und im Vergleich zu prim{\"a}ren mvEZ durchschnittlich 15 zus{\"a}tzliche Populationsverdopplungen leisten k{\"o}nnen. Dadurch ist es m{\"o}glich 3x104-fach mehr upcyte® mvEZ eines Spenders zu generieren verglichen mit den korrespondierenden Prim{\"a}rzellen. Die gute und ausreichende Verf{\"u}gbarkeit der Zellen macht sie interessant f{\"u}r die Standardisierung von in-vitro Testsystemen, ebenso k{\"o}nnen die Zellen zur Pr{\"a}vaskularisierung von Transplantaten eingesetzt werden. Upcyte® mvEZ zeigen zahlreiche Prim{\"a}rzellmerkmale, die in der Literatur beschrieben sind. Im konfluenten Zustand zeigen sie die f{\"u}r prim{\"a}re mvEZ spezifische pflastersteinartige Morphologie. Dar{\"u}ber hinaus exprimieren upcyte® mvEZ typische Endothelzellmarker wie CD31, vWF, eNOS, CD105, CD146 und VEGFR-2 vergleichbar zu prim{\"a}ren mvEZ. Eine weitere endothelzellspezifische Eigenschaft ist die Bindung von Ulex europaeus agglutinin I Lektin an die alpha-L-Fucose enthaltene Kohlenhydratstrukturen von mvEZs. Auch hier wurden upcyte® Zellen mit prim{\"a}ren mvEZ verglichen und zeigten die hierf{\"u}r charkteristischen Strukturen. Zus{\"a}tzlich zu Morphologie, Proliferationskapazit{\"a}t und endothelzellspezifischen Markern, zeigen upcyte® mvEZ auch mehrere funktionelle Eigenschaften, welche in prim{\"a}ren mvEZ beobachtet werden k{\"o}nnen, wie beispielsweise die Aufnahme von Dil-markiertem acetyliertem Low Density Lipoprotein (Dil-Ac-LDL) oder die F{\"a}higkeit den Prozess der Angiognese zu unterst{\"u}tzen. Zus{\"a}tzlich bilden Sph{\"a}roide aus upcyte® mvEZ dreidimensionale lumin{\"a}re Zellformationen in einer Kollagenmatrix aus. Diese Charakteristika zeigen den quasi-prim{\"a}ren Ph{\"a}notyp der upcyte® mvEZs. Upcyte® mvEZ stellen dar{\"u}ber hinaus eine neuartige m{\"o}gliche Zellquelle f{\"u}r die Generierung pr{\"a}vaskularisierter Tr{\"a}germaterialien im Tissue Engineering dar. In der vorliegenden Arbeit konnte die Wiederbesiedlung der biologisch vaskularisierte Matrix (BioVaSc) mit upcyte® mvEZ vergleichbar zu prim{\"a}ren mvEZ gezeigt werden. Der Einsatz von upcyte® mvEZ in der BioVaSc stellt einen neuen, vielversprechenden Ansatz zur Herstellung eines vaskularisierten Modells f{\"u}r Gewebekonstrukte dar, wie beispielsweise einem Leberkonstrukt. Zusammenfassend konnte in der vorliegenden Arbeit gezeigt werden, dass upcyte® mvEZ vergleichbar zu prim{\"a}ren mvEZs sind und somit eine geeignete Alternative f{\"u}r die Generierung pr{\"a}vaskulierter Tr{\"a}germaterialien und Aufbau von in-vitro Testsystemen darstellen. Dar{\"u}ber hinaus wurde ein neues, innovatives System f{\"u}r die Generierung einer perfundierten, mit Endothelzellen wiederbesiedelten Matrix f{\"u}r k{\"u}nstliches Gewebe in-vitro entwickelt.}, subject = {Tissue Engineering}, language = {de} } @article{OuhaddiCharbonnierPorgeetal.2023, author = {Ouhaddi, Yassine and Charbonnier, Baptiste and Porge, Juliette and Zhang, Yu-Ling and Garcia, Isadora and Gbureck, Uwe and Grover, Liam and Gilardino, Mirko and Harvey, Edward and Makhoul, Nicholas and Barralet, Jake}, title = {Development of neovasculature in axially vascularized calcium phosphate cement scaffolds}, series = {Journal of Functional Biomaterials}, volume = {14}, journal = {Journal of Functional Biomaterials}, number = {2}, issn = {2079-4983}, doi = {10.3390/jfb14020105}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-304026}, year = {2023}, abstract = {Augmenting the vascular supply to generate new tissues, a crucial aspect in regenerative medicine, has been challenging. Recently, our group showed that calcium phosphate can induce the formation of a functional neo-angiosome without the need for microsurgical arterial anastomosis. This was a preclinical proof of concept for biomaterial-induced luminal sprouting of large-diameter vessels. In this study, we investigated if sprouting was a general response to surgical injury or placement of an inorganic construct around the vessel. Cylindrical biocement scaffolds of differing chemistries were placed around the femoral vein. A contrast agent was used to visualize vessel ingrowth into the scaffolds. Cell populations in the scaffold were mapped using immunohistochemistry. Calcium phosphate scaffolds induced 2.7-3 times greater volume of blood vessels than calcium sulphate or magnesium phosphate scaffolds. Macrophage and vSMC populations were identified that changed spatially and temporally within the scaffold during implantation. NLRP3 inflammasome activation peaked at weeks 2 and 4 and then declined; however, IL-1β expression was sustained over the course of the experiment. IL-8, a promoter of angiogenesis, was also detected, and together, these responses suggest a role of sterile inflammation. Unexpectedly, the effect was distinct from an injury response as a result of surgical placement and also was not simply a foreign body reaction as a result of placing a rigid bioceramic next to a vein, since, while the materials tested had similar microstructures, only the calcium phosphates tested elicited an angiogenic response. This finding then reveals a potential path towards a new strategy for creating better pro-regenerative biomaterials.}, language = {en} } @article{HackerEscalonaEspinosaConsalvoetal.2016, author = {Hacker, Ulrich T. and Escalona-Espinosa, Laura and Consalvo, Nicola and Goede, Valentin and Schiffmann, Lars and Scherer, Stefan J. and Hedge, Priti and Van Cutsem, Eric and Coutelle, Oliver and B{\"u}ning, Hildegard}, title = {Evaluation of Angiopoietin-2 as a biomarker in gastric cancer: results from the randomised phase III AVAGAST trial}, series = {British Journal of Cancer}, volume = {114}, journal = {British Journal of Cancer}, number = {8}, doi = {10.1038/bjc.2016.30}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-189578}, pages = {855-862}, year = {2016}, abstract = {Background: In the phase III AVAGAST trial, the addition of bevacizumab to chemotherapy improved progression-free survival (PFS) but not overall survival (OS) in patients with advanced gastric cancer. We studied the role of Angiopoietin-2 (Ang-2), a key driver of tumour angiogenesis, metastasis and resistance to antiangiogenic treatment, as a biomarker. Methods: Previously untreated, advanced gastric cancer patients were randomly assigned to receive bevacizumab (n = 387) or placebo (n = 387) in combination with chemotherapy. Plasma collected at baseline and at progression was analysed by ELISA. The role of Ang-2 as a prognostic and a predictive biomarker of bevacizumab efficacy was studied using a Cox proportional hazards model. Logistic regression analysis was applied for correlations with metastasis. Results: Median baseline plasma Ang-2 levels were lower in Asian (2143 pg ml\(^-\)\(^1\)) vs non-Asian patients (3193 pg ml\(^-\)\(^1\)), P<0.0001. Baseline plasma Ang-2 was identified as an independent prognostic marker for OS but did not predict bevacizumab efficacy alone or in combination with baseline VEGF. Baseline plasma Ang-2 correlated with the frequency of liver metastasis (LM) at any time: Odds ratio per 1000 pg ml\(^-\)\(^1\) increase: 1.19; 95\% CI 1.10-1.29; P<0.0001 (non-Asians) and 1.37; 95\% CI 1.13-1.64; P = 0.0010 (Asians). Conclusions: Baseline plasma Ang-2 is a novel prognostic biomarker for OS in advanced gastric cancer strongly associated with LM. Differences in Ang-2 mediated vascular response may, in part, account for outcome differences between Asian and non-Asian patients; however, data have to be further validated. Ang-2 is a promising drug target in gastric cancer.}, language = {en} } @article{Meierjohann2015, author = {Meierjohann, Svenja}, title = {Hypoxia independent drivers of melanoma angiogenesis}, series = {Frontiers in Oncology}, volume = {5}, journal = {Frontiers in Oncology}, number = {120}, doi = {10.3389/fonc.2015.00102}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-125586}, year = {2015}, abstract = {Tumor angiogenesis is a process which is traditionally regarded as the tumor's response to low nutrient supply occurring under hypoxic conditions. However, hypoxia is not a pre-requisite for angiogenesis. The fact that even single tumor cells or small tumor cell aggregates are capable of attracting blood vessels reveals the early metastatic capability of tumor cells. This review sheds light on the hypoxia-independent mechanisms of tumor angiogenesis in melanoma.}, language = {en} } @phdthesis{Voss2008, author = {Voß, Katrin}, title = {Identifizierung und Charakterisierung von Interaktiopnspartnern des humanen CCM3-Proteins}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-28188}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2008}, abstract = {Zerebrale kavern{\"o}se Malfomationen (CCM) sind vaskul{\"a}re Fehlbildungen im Gehirn. Sie sind gekennzeichnet durch stark dilatierte, blutgef{\"u}llte Gef{\"a}ße mit einschichtigem Endothel, denen Merkmale ausgereifter Blutgef{\"a}ße fehlen. Die klinischen Symptome reichen von Kopfschmerz bis hin zu h{\"a}morraghischem Schlaganfall. Eine genaue Vorhersage des Krankheitsverlaufs ist nicht m{\"o}glich und die neurochirurgische Dissektion ist in der Regel die Therapieform der Wahl. Die genauen molekularen Mechanismen der CCM-Pathogenese sind unbekannt. CCMs treten sporadisch oder famili{\"a}r geh{\"a}uft auf und folgen einem autosomal-dominanten Erbgang. Drei krankheitsverursachende Gene wurden in famili{\"a}ren CCMs identifiziert: CCM1/KRIT1, CCM2/MGC4607 und CCM3/PDCD10. Da Patienten mit einer Mutation in einem der drei CCM-Gene denselben klinischen Ph{\"a}notyp aufweisen, wurde angenommen, dass die CCM-Proteine (CCM1, CCM2 und CCM3) Bestandteile eines molekularen Signalwegs sind. In dieser Arbeit wurde erstmals gezeigt, dass CCM3 mit CCM2 interagiert und zusammen mit CCM1 einen tern{\"a}ren Proteinkomplex bildet. Untersuchungen mit der humanen in-frame CCM2-Deletionsmutante CCM2:p.P11_K68del belegten, dass CCM2 das zentrale Ger{\"u}stprotein des CCM1/CCM2/CCM3-Proteinkomplexes ist. Weiterhin konnte gezeigt werden, dass CCM3 an die Serin/Threonin-Kinase STK25 und an die Fas-assoziierte Phosphatase-1 (FAP-1) bindet. STK25 phosphoryliert CCM3 am Serin 39 und am Threonin 43. Die katalytische Dom{\"a}ne von FAP-1 dephosphoryliert CCM3. Untersuchungen mit der einzig bekannten humanen CCM3-Deletionsmutante, der aufgrund einer in-frame Deletion von Exon 5 im CCM3-Gen 18 Aminos{\"a}uren (CCM3:p.L33_K50del) fehlen, belegten zudem, dass in vitro dephosphoryliertes CCM3 Bestandteil des tern{\"a}ren CCM-Proteinkomplexes ist. W{\"a}hrend STK25 die Deletionsmutante nicht mehr binden und phosphorylieren konnte, war die Interaktion mit CCM2 und die Bildung des tern{\"a}ren CCMKomplexes nicht beeintr{\"a}chtigt. Somit k{\"o}nnte CCM3 {\"u}ber die Dephosphorylierung durch FAP-1 und die Phosphorylierung durch STK25 funktionell reguliert werden. Es stellte sich zudem heraus, dass CCM3 durch Induktion von oxidativem Stress mittels H2O2-Behandlung in humanen dermalen mikrovaskul{\"a}ren Endothelzellen herunterreguliert wird. Die in dieser Arbeit beschriebene Charakterisierung von CCM3-Interaktionen bringt CCM3 {\"u}ber seine Interaktionspartner erstmals in Zusammenhang mit molekularen Signalwegen, die an Prozessen der Angiogenese und vaskul{\"a}ren Entwicklung beteiligt sind. Die Ergebnisse liefern wichtige Hinweise f{\"u}r die Entschl{\"u}sselung der pathogenen Mechanismen zerebraler kavern{\"o}ser Malformationen und stellen einen ersten Schritt dar, um andere Behandlungsans{\"a}tze als den bisher angewandten chirurgischen Eingriff, der multiple Risiken birgt, entwickeln zu k{\"o}nnen.}, subject = {Angiogenese}, language = {de} } @phdthesis{Partzsch2004, author = {Partzsch, Bernhard}, title = {Identifizierung und Isolierung von Angiostatin aus dem Urin bei Patienten mit Prostatakarzinom}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-17814}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2004}, abstract = {Die Angiogenese beschreibt einen entscheidenden Schritt f{\"u}r Tumorwachstum und Metastasierung. Die Tendenz, neue Blutgef{\"a}ße zu bilden, wird durch das Gleichgewicht angiogener und nicht-angiogener Faktoren bestimmt. In einer Reihe eleganter tierexperimenteller Versuche gelang es O`Reilly erstmals einen tumorassoziierten Inhibitor der Angiogenese, den er Angiostatin nannte, nachzuweisen und zu isolieren. Uns gelang es, im Western-Blot Angiostatin und Angiostatin-Spaltprodukte sowohl aus dem Urin von PCa-Patienten als auch aus dem Urin gesunder Probanden nachzuweisen und zu isolieren. Die anti-angiogene Wirksamkeit des von uns isolierten Proteins wurde im Endothelzellkultur-Assay best{\"a}tigt. Eine Differenzierung gesunder Personen von PCa-Patienten war aufgrund der kleinen Fallzahlen nicht m{\"o}glich. Der Nachweis von Angiostatin bei Gesunden belegt aber, dass anti-angiogene Proteine unabh{\"a}ngig vom Vorhandensein maligner Tumore im Urin ausgeschieden werden. Es bleibt zu vermuten, dass Angiogenese-Inhibitoren {\"a}hnlich den Gerinnungsfaktoren bei Bedarf aktiviert und inaktiviert werden k{\"o}nnen. Der Angiogenese zugrunde liegende Mechanismen und beteiligte Faktoren sind Bestandteil intensiver Forschung. Unklar ist, ob Angiogenese-Inhibitoren in Zukunft in der Krebstherapie die Rolle spielen werden, die man ihnen bei ihrer Entdeckung zuschrieb.}, language = {de} } @article{KrebsSolimandoKalogirouetal.2020, author = {Krebs, Markus and Solimando, Antonio Giovanni and Kalogirou, Charis and Marquardt, Andr{\´e} and Frank, Torsten and Sokolakis, Ioannis and Hatzichristodoulou, Georgios and Kneitz, Susanne and Bargou, Ralf and K{\"u}bler, Hubert and Schilling, Bastian and Spahn, Martin and Kneitz, Burkhard}, title = {miR-221-3p Regulates VEGFR2 Expression in High-Risk Prostate Cancer and Represents an Escape Mechanism from Sunitinib In Vitro}, series = {Journal of Clinical Medicine}, volume = {9}, journal = {Journal of Clinical Medicine}, number = {3}, issn = {2077-0383}, doi = {10.3390/jcm9030670}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-203168}, year = {2020}, abstract = {Downregulation of miR-221-3p expression in prostate cancer (PCa) predicted overall and cancer-specific survival of high-risk PCa patients. Apart from PCa, miR-221-3p expression levels predicted a response to tyrosine kinase inhibitors (TKI) in clear cell renal cell carcinoma (ccRCC) patients. Since this role of miR-221-3p was explained with a specific targeting of VEGFR2, we examined whether miR-221-3p regulated VEGFR2 in PCa. First, we confirmed VEGFR2/KDR as a target gene of miR-221-3p in PCa cells by applying Luciferase reporter assays and Western blotting experiments. Although VEGFR2 was mainly downregulated in the PCa cohort of the TCGA (The Cancer Genome Atlas) database, VEGFR2 was upregulated in our high-risk PCa cohort (n = 142) and predicted clinical progression. In vitro miR-221-3p acted as an escape mechanism from TKI in PC3 cells, as displayed by proliferation and apoptosis assays. Moreover, we confirmed that Sunitinib induced an interferon-related gene signature in PC3 cells by analyzing external microarray data and by demonstrating a significant upregulation of miR-221-3p/miR-222-3p after Sunitinib exposure. Our findings bear a clinical perspective for high-risk PCa patients with low miR-221-3p levels since this could predict a favorable TKI response. Apart from this therapeutic niche, we identified a partially oncogenic function of miR-221-3p as an escape mechanism from VEGFR2 inhibition.}, language = {en} } @article{NeuhausSchlundtFehrholzetal.2015, author = {Neuhaus, Winfried and Schlundt, Marian and Fehrholz, Markus and Ehrke, Alexander and Kunzmann, Steffen and Liebner, Stefan and Speer, Christian P. and F{\"o}rster, Carola Y.}, title = {Multiple Antenatal Dexamethasone Treatment Alters Brain Vessel Differentiation in Newborn Mouse Pups}, series = {PLoS One}, volume = {10}, journal = {PLoS One}, number = {8}, doi = {10.1371/journal.pone.0136221}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-125471}, pages = {e0136221}, year = {2015}, abstract = {Antenatal steroid treatment decreases morbidity and mortality in premature infants through the maturation of lung tissue, which enables sufficient breathing performance. However, clinical and animal studies have shown that repeated doses of glucocorticoids such as dexamethasone and betamethasone lead to long-term adverse effects on brain development. Therefore, we established a mouse model for antenatal dexamethasone treatment to investigate the effects of dexamethasone on brain vessel differentiation towards the blood-brain barrier (BBB) phenotype, focusing on molecular marker analysis. The major findings were that in total brains on postnatal day (PN) 4 triple antenatal dexamethasone treatment significantly downregulated the tight junction protein claudin-5, the endothelial marker Pecam-1/CD31, the glucocorticoid receptor, the NR1 subunit of the N-methyl-D-aspartate receptor, and Abc transporters (Abcb1a, Abcg2 Abcc4). Less pronounced effects were found after single antenatal dexamethasone treatment and in PN10 samples. Comparisons of total brain samples with isolated brain endothelial cells together with the stainings for Pecam-1/CD31 and claudin-5 led to the assumption that the morphology of brain vessels is affected by antenatal dexamethasone treatment at PN4. On the mRNA level markers for angiogenesis, the sonic hedgehog and the Wnt pathway were downregulated in PN4 samples, suggesting fundamental changes in brain vascularization and/or differentiation. In conclusion, we provided a first comprehensive molecular basis for the adverse effects of multiple antenatal dexamethasone treatment on brain vessel differentiation.}, language = {en} } @article{AdelfingerBesslerCeciletal.2015, author = {Adelfinger, Marion and Bessler, Simon and Cecil, Alexander and Langbein-Laugwitz, Johanna and Frentzen, Alexa and Gentschev, Ivaylo and Szalay, Aladar A.}, title = {Preclinical Testing Oncolytic Vaccinia Virus Strain GLV-5b451 Expressing an Anti-VEGF Single-Chain Antibody for Canine Cancer Therapy}, series = {Viruses}, volume = {7}, journal = {Viruses}, doi = {10.3390/v7072811}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-125705}, pages = {4075-4092}, year = {2015}, abstract = {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.}, language = {en} } @article{MarquardtHartrampfKollmannsbergeretal.2023, author = {Marquardt, Andr{\´e} and Hartrampf, Philipp and Kollmannsberger, Philip and Solimando, Antonio G. and Meierjohann, Svenja and K{\"u}bler, Hubert and Bargou, Ralf and Schilling, Bastian and Serfling, Sebastian E. and Buck, Andreas and Werner, Rudolf A. and Lapa, Constantin and Krebs, Markus}, title = {Predicting microenvironment in CXCR4- and FAP-positive solid tumors — a pan-cancer machine learning workflow for theranostic target structures}, series = {Cancers}, volume = {15}, journal = {Cancers}, number = {2}, issn = {2072-6694}, doi = {10.3390/cancers15020392}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-305036}, year = {2023}, abstract = {(1) Background: C-X-C Motif Chemokine Receptor 4 (CXCR4) and Fibroblast Activation Protein Alpha (FAP) are promising theranostic targets. However, it is unclear whether CXCR4 and FAP positivity mark distinct microenvironments, especially in solid tumors. (2) Methods: Using Random Forest (RF) analysis, we searched for entity-independent mRNA and microRNA signatures related to CXCR4 and FAP overexpression in our pan-cancer cohort from The Cancer Genome Atlas (TCGA) database — representing n = 9242 specimens from 29 tumor entities. CXCR4- and FAP-positive samples were assessed via StringDB cluster analysis, EnrichR, Metascape, and Gene Set Enrichment Analysis (GSEA). Findings were validated via correlation analyses in n = 1541 tumor samples. TIMER2.0 analyzed the association of CXCR4 / FAP expression and infiltration levels of immune-related cells. (3) Results: We identified entity-independent CXCR4 and FAP gene signatures representative for the majority of solid cancers. While CXCR4 positivity marked an immune-related microenvironment, FAP overexpression highlighted an angiogenesis-associated niche. TIMER2.0 analysis confirmed characteristic infiltration levels of CD8+ cells for CXCR4-positive tumors and endothelial cells for FAP-positive tumors. (4) Conclusions: CXCR4- and FAP-directed PET imaging could provide a non-invasive decision aid for entity-agnostic treatment of microenvironment in solid malignancies. Moreover, this machine learning workflow can easily be transferred towards other theranostic targets.}, language = {en} } @phdthesis{Schneider2014, author = {Schneider, Magdalena}, title = {Synthese, Radiomarkierung und biochemische sowie pr{\"a}klinische Evaluierung neuer Aminopeptidase N- und Fibroblasten-Aktivierungs-Protein alpha- affiner Verbindungen f{\"u}r die molekulare Bildgebung mittels Positronen-Emissions-Tomographie}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-102562}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {Nach einem Myokardinfarkt setzen Wundheilungsprozesse ein, um die Durchblutung wieder herzustellen und nekrotisches Muskelgewebe durch Narbengewebe zu ersetzen. Die Einsprossung neuer Kapillaren vom bestehenden Gef{\"a}ßnetz aus wird als Angiogenese bezeichnet. Das dabei vermehrt exprimierte proteolytische Enzym Aminopeptidase N (APN) spielt eine entscheidende Rolle bei der Einsprossung von Endothelzellen. Beim kardialen Remodeling werden abgestorbene Myozyten mithilfe der Einwanderung von Fibroblasten durch Binde- oder St{\"u}tzgewebe ersetzt, dabei {\"u}bernimmt das Fibroblasten-Aktivierungs-Protein alpha (FAP) Aufgaben bei der Proliferation und Fortbewegung von Fibroblasten. Durch ihre erh{\"o}hte Expression bei den Wundheilungs- und Remodelingprozessen nach einem Herzinfarkt stellen die Metalloprotease APN und die Serinprotease FAP molekulare Targets f{\"u}r die Diagnostik und Therapie dar. Als Diagnosemethode besonders geeignet ist die Positronen-Emissions-Tomographie (PET), die es erm{\"o}glicht, biochemische Prozesse in Echtzeit im zu untersuchenden Organismus zu visualisieren und zu quantifizieren. Eine als Radiopharmakon oder Tracer bezeichnete biochemische Sonde kann im Falle eines Enzyms dessen radioaktiv markiertes Substrat oder ein Inhibitor sein. Ziel dieser Arbeit war es, spezifische APN- und FAP-affine Tracer f{\"u}r die nicht-invasive Untersuchung der APN- und FAP-Expression mittels PET zu entwickeln und dadurch die Rolle von APN und FAP bei Remodelingprozessen nach Myokardinfarkt besser verstehen bzw. kl{\"a}ren zu k{\"o}nnen. Um die Protease APN mittels PET zu untersuchen, wurden die f{\"u}r APN affine Verbindung NOTA-NGR (Komplexbildner + cyclisches Peptid inkl. Asparagin-Glycin-Arginin) mit dem Positronen-emittierenden Nuklid Gallium-68 (68Ga) markiert. Das Potential von 68Ga-NOTA-NGR als PET-Tracer wurde in vivo am Infarktmodell mittels Kleintier-PET untersucht und mit 68Ga-NOTA-RGD, einem zur Visualisierung des neo-angiogenetischen alphavbeta3-Integrins etablierten Tracer, verglichen. Untersuchungen ergaben, dass 68Ga-NOTA-NGR einen vielversprechenden neuen PET-Tracer f{\"u}r die Visualisierung und Quantifizierung der APN-Expression im Rahmen der Angiogenese nach einem Myokardinfarkt darstellt. 68Ga-NOTA-NGR zeigte eine erh{\"o}hte Aufnahme im Bereich des Myokardinfarkts im Sinne einer vermehrten Angiogenese. Die Aufnahme des Tracers in infarzierten Arealen war quantitativ h{\"o}her als in der Untersuchung mit 68Ga-NOTA-RGD. In Autoradiographie-Experimenten wurde 68Ga-NOTA-NGR ex vivo untersucht. Die Akkumulation von 68Ga-NOTA-NGR im isch{\"a}mischen Bereich war deutlich h{\"o}her als im gesunden Myokard. Der Nachweis der unterschiedlichen Bereiche des Herzens erfolgte mit HE-F{\"a}rbung. Die Expression von APN wurde immunohistochemisch mittels spezifischer Antik{\"o}rper best{\"a}tigt. Zum Vergleich wurden ebenso einige andere an der Angiogenese beteiligte Faktoren untersucht. APN stellte sich auch hier als geeignetes Target zum Nachweis der Angiogenese heraus. Um die Protease FAP mittels PET zu untersuchen, wurden eine Reihe peptidomimetischer Inhibitoren, die die Erkennungssequenz Glycin-Prolin mit einer Carbonitril-Gruppe als elektrophiler Einheit zur kovalent-reversiblen Hemmung des Enzyms enthalten, entwickelt. Ausgehend vom N-Acetylglycin-pyrrolidin-(2S)-carbonitril als Leitstruktur wurden Inhibitoren und Vorstufen zur Radiomarkierung inkl. verschieden substituierter Benzoes{\"a}uren dargestellt. Zus{\"a}tzlich wurden noch bereits bekannte Inhibitoren synthetisiert, die zum Vergleich in den Enzymassays dienten. Drei Verbindungen zeigten gute inhibitorische Wirkung an FAP und außerdem Selektivit{\"a}t gegen{\"u}ber DPP IV. Keine der entwickelten Verbindungen zeigte einen KI-Wert im nanomolaren Bereich, erforderlich f{\"u}r einen potentiellen Tracer zur in-vivo-Visualisierung einer Enzymexpression mittels PET. Um die Inhibitoren mit der besten Hemmung an FAP zum PET-Tracer weiterzuentwickeln, mussten sie mit einem Positronenemitter markiert werden. Die Markierung erfolgte {\"u}ber Isotopenaustausch, bei dem nicht-radioaktives Iod am aromatischen Ring des Precursors durch das radioaktive Iod-124 (124I) substituiert wurde. Es konnten dadurch die radioiodierten Verbindungen 1-(2-[124I]Iodhippurs{\"a}ure)-pyrrolidin-(2S)-carbonitril und 1-(4-[124I]Iod-hippurs{\"a}ure)-pyrrolidin-(2S)-carbonitril synthetisiert werden. Trotz der relativ niedrigen Affinit{\"a}t f{\"u}r FAP wurde das neue 1-(2-[124I]Iodhippurs{\"a}ure)-pyrrolidin-(2S)-carbonitril in Ratten am Infarktmodell mittels Kleintier-PET getestet. Die Lage der isch{\"a}mischen Zone wurde im Anschluss durch HE-F{\"a}rbung bestimmt. In vivo zeigte sich eine nur sehr geringe Aufnahme des Radiopharmakons in der isch{\"a}mischen Zone des Myokards. Damit ist 1-(2-[124I]Iod-hippurs{\"a}ure)-pyrrolidin-(2S)-carbonitril kein f{\"u}r den gew{\"u}nschten Zweck geeigneter PET-Tracer. Nichtsdestotrotz war der Ansatz vielversprechend und es wurde zum ersten Mal ein PET-Tracer dieser Art zur Untersuchung des FAP im Myokardinfarkt hergestellt.}, subject = {Positronen-Emissions-Tomographie}, language = {de} } @article{BugaMargaritescuScholzetal.2014, author = {Buga, Ana Maria and Margaritescu, Claudiu and Scholz, Claus J{\"u}rgen and Radu, Eugen and Zelenak, Christine and Popa-Wagner, Aurel}, title = {Transcriptomics of Post-Stroke Angiogenesis in the Aged Brain}, series = {Frontiers in Aging Neuroscience}, volume = {6}, journal = {Frontiers in Aging Neuroscience}, number = {44}, doi = {10.3389/fnagi.2014.00044}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-120700}, year = {2014}, abstract = {Despite the obvious clinical significance of post-stroke angiogenesis in aged subjects, a detailed transcriptomic analysis of post-stroke angiogenesis has not yet been undertaken in an aged experimental model. In this study, by combining stroke transcriptomics with immunohistochemistry in aged rats and post-stroke patients, we sought to identify an age-specific gene expression pattern that may characterize the angiogenic process after stroke. We found that both young and old infarcted rats initiated vigorous angiogenesis. However, the young rats had a higher vascular density by day 14 post-stroke. "New-for-stroke" genes that were linked to the increased vasculature density in young animals included Angpt2, Angptl2, Angptl4, Cib1, Ccr2, Col4a2, Cxcl1, Lef1, Hhex, Lamc1, Nid2, Pcam1, Plod2, Runx3, Scpep1, S100a4, Tgfbi, and Wnt4, which are required for sprouting angiogenesis, reconstruction of the basal lamina (BL), and the resolution phase. The vast majority of genes involved in sprouting angiogenesis (Angpt2, Angptl4, Cib1, Col8a1, Nrp1, Pcam1, Pttg1ip, Rac2, Runx1, Tnp4, Wnt4); reconstruction of a new BL (Col4a2, Lamc1, Plod2); or tube formation and maturation (Angpt1, Gpc3, Igfbp7, Sparc, Tie2, Tnfsf10), had however, a delayed upregulation in the aged rats. The angiogenic response in aged rats was further diminished by the persistent upregulation of "inflammatory" genes (Cxcl12, Mmp8, Mmp12, Mmp14, Mpeg1, Tnfrsf1a, Tnfrsf1b) and vigorous expression of genes required for the buildup of the fibrotic scar (Cthrc1, Il6ra, Il13ar1, Il18, Mmp2, Rassf4, Tgfb1, Tgfbr2, Timp1). Beyond this barrier, angiogenesis in the aged brains was similar to that in young brains. We also found that the aged human brain is capable of mounting a vigorous angiogenic response after stroke, which most likely reflects the remaining brain plasticity of the aged brain.}, language = {en} } @article{SchaeferWeibelDonatetal.2012, author = {Sch{\"a}fer, Simon and Weibel, Stephanie and Donat, Ulrike and Zhang, Quian and Aguilar, Richard J. and Chen, Nanhai G. and Szalay, Aladar A.}, title = {Vaccinia virus-mediated intra-tumoral expression of matrix metalloproteinase 9 enhances oncolysis of PC-3 xenograft tumors}, series = {BMC Cancer}, volume = {12}, journal = {BMC Cancer}, number = {366}, doi = {10.1186/1471-2407-12-366}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-140800}, year = {2012}, abstract = {Background Oncolytic viruses, including vaccinia virus (VACV), are a promising alternative to classical mono-cancer treatment methods such as surgery, chemo- or radiotherapy. However, combined therapeutic modalities may be more effective than mono-therapies. In this study, we enhanced the effectiveness of oncolytic virotherapy by matrix metalloproteinase (MMP-9)-mediated degradation of proteins of the tumoral extracellular matrix (ECM), leading to increased viral distribution within the tumors. Methods For this study, the oncolytic vaccinia virus GLV-1h255, containing the mmp-9 gene, was constructed and used to treat PC-3 tumor-bearing mice, achieving an intra-tumoral over-expression of MMP-9. The intra-tumoral MMP-9 content was quantified by immunohistochemistry in tumor sections. Therapeutic efficacy of GLV-1h255 was evaluated by monitoring tumor growth kinetics and intra-tumoral virus titers. Microenvironmental changes mediated by the intra-tumoral MMP-9 over-expression were investigated by microscopic quantification of the collagen IV content, the blood vessel density (BVD) and the analysis of lymph node metastasis formation. Results GLV-1h255-treatment of PC-3 tumors led to a significant over-expression of intra-tumoral MMP-9, accompanied by a marked decrease in collagen IV content in infected tumor areas, when compared to GLV-1h68-infected tumor areas. This led to considerably elevated virus titers in GLV-1h255 infected tumors, and to enhanced tumor regression. The analysis of the BVD, as well as the lumbar and renal lymph node volumes, revealed lower BVD and significantly smaller lymph nodes in both GLV-1h68- and GLV-1h255- injected mice compared to those injected with PBS, indicating that MMP-9 over-expression does not alter the metastasis-reducing effect of oncolytic VACV. Conclusions Taken together, these results indicate that a GLV-1h255-mediated intra-tumoral over-expression of MMP-9 leads to a degradation of collagen IV, facilitating intra-tumoral viral dissemination, and resulting in accelerated tumor regression. We propose that approaches which enhance the oncolytic effect by increasing the intra-tumoral viral load, may be an effective way to improve therapeutic outcome.}, language = {en} } @article{PatilGentschevAdelfingeretal.2012, author = {Patil, Sandeep S. and Gentschev, Ivaylo and Adelfinger, Marion and Donat, Ulrike and Hess, Michael and Weibel, Stephanie and Nolte, Ingo and Frentzen, Alexa and Szalay, Aladar A.}, title = {Virotherapy of Canine Tumors with Oncolytic Vaccinia Virus GLV-1h109 Expressing an Anti-VEGF Single-Chain Antibody}, series = {PLoS One}, volume = {7}, journal = {PLoS One}, number = {10}, doi = {10.1371/journal.pone.0047472}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130039}, pages = {e47472}, year = {2012}, abstract = {Virotherapy using oncolytic vaccinia virus (VACV) strains is one promising new strategy for cancer therapy. We have previously reported that oncolytic vaccinia virus strains expressing an anti-VEGF (Vascular Endothelial Growth Factor) single-chain antibody (scAb) GLAF-1 exhibited significant therapeutic efficacy for treatment of human tumor xenografts. Here, we describe the use of oncolytic vaccinia virus GLV-1h109 encoding GLAF-1 for canine cancer therapy. In this study we analyzed the virus-mediated delivery and production of scAb GLAF-1 and the oncolytic and immunological effects of the GLV-1h109 vaccinia virus strain against canine soft tissue sarcoma and canine prostate carcinoma in xenograft models. Cell culture data demonstrated that the GLV-1h109 virus efficiently infect, replicate in and destroy both tested canine cancer cell lines. In addition, successful expression of GLAF-1 was demonstrated in virus-infected canine cancer cells and the antibody specifically recognized canine VEGF. In two different xenograft models, the systemic administration of the GLV-1h109 virus was found to be safe and led to anti-tumor and immunological effects resulting in the significant reduction of tumor growth in comparison to untreated control mice. Furthermore, tumor-specific virus infection led to a continued production of functional scAb GLAF-1, resulting in inhibition of angiogenesis. Overall, the GLV-1h109-mediated cancer therapy and production of immunotherapeutic anti-VEGF scAb may open the way for combination therapy concept i.e. vaccinia virus mediated oncolysis and intratumoral production of therapeutic drugs in canine cancer patients.}, language = {en} }