@article{WilhelmSmetakSchaeferEckartetal.2014, author = {Wilhelm, Martin and Smetak, Manfred and Schaefer-Eckart, Kerstin and Kimmel, Brigitte and Birkmann, Josef and Einsele, Hermann and Kunzmann, Volker}, title = {Successful adoptive transfer and in vivo expansion of haploidentical γδ T cells}, series = {Journal of Translational Medicine}, volume = {12}, journal = {Journal of Translational Medicine}, number = {45}, doi = {10.1186/1479-5876-12-45}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-117290}, year = {2014}, abstract = {Background: The primary aim of this pilot study was to determine the feasibility and safety of an adoptive transfer and in vivo expansion of human haploidentical gamma delta T lymphocytes. Methods: Patients with advanced haematological malignancies who are not eligible for allogeneic transplantation received peripheral blood mononuclear cells from half-matched family donors. For that, a single unstimulated leukapheresis product was incubated with both the anti-CD4 and anti-CD8 antibodies conjugated to paramagnetic particles. The depletion procedure was performed on a fully automated CliniMACS (R) device according to the manufacturer's instructions. On average, patients received 2.17 x 10(6)/kg (range 0.9-3.48) γδ T cells with <1\% CD4-or CD8-positive cells remaining in the product. All patients received prior lymphopenia-inducing chemotherapy (fludarabine 20-25 mg/m(2) day -6 until day -2 and cyclophosphamide 30-60 mg/kg day -6 and -5) and were treated with 4 mg zoledronate on day 0 and 1.0x10(6) IU/m(2) IL-2 on day +1 until day +6 for the induction of gamma delta T cell proliferation in vivo. Results: This resulted in a marked in vivo expansion of donor γδ T cells and, to a lower extent, natural killer cells and double-negative αβ T cells (mean 68-fold, eight-fold, and eight-fold, respectively). Proliferation peaked by around day +8 and donor cells persisted up to 28 days. Although refractory to all prior therapies, three out of four patients achieved a complete remission, which lasted for 8 months in a patient with plasma cell leukaemia. One patient died from an infection 6 weeks after treatment. Conclusion: This pilot study shows that adoptive transfer and in vivo expansion of haploidentical γδ T lymphocytes is feasible and suggests a potential role of these cells in the treatment of haematological diseases.}, language = {en} } @article{StoevesandtTrautmann2022, author = {Stoevesandt, Johanna and Trautmann, Axel}, title = {Risk factors in bee and Vespula venom allergy: state of the art}, series = {Allergo Journal International}, volume = {31}, journal = {Allergo Journal International}, number = {1}, issn = {2197-0378}, doi = {10.1007/s40629-021-00187-1}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-270498}, pages = {1-10}, year = {2022}, abstract = {Background Correct recognition of risk factors enables individualized management and treatment of venom allergic patients. Methods Systematic research and review of current literature regarding the risk of (1) severe sting-induced anaphylaxis, (2) anaphylactic adverse event during venom immunotherapy (VIT), and (3) treatment failure. Results and discussion (1) Mastocytosis is the most important risk factor for severe sting-induced anaphylaxis. Hereditary α‑tryptasemia was recently identified as a genetic predictor of severe reactions. Older age is clearly associated with an increased risk; the respective impact of defined cardiovascular comorbidities has yet to be determined. Recent data do not support an aggravation of venom-induced anaphylaxis by intake of β‑blockers or angiotensin-converting enzyme (ACE) inhibitors. A higher risk in men can be attributed to more intensive exposure to stinging insects. (2) Anaphylactic side effects of VIT are most common during the buildup phase, particularly in the course of (ultra-)rush protocols involving a high number of injections and high cumulative daily doses. They are significantly more frequent during honeybee compared to Vespula VIT. Data supporting a negative effect of mastocytosis on the tolerability of VIT are scarce. Older age and cardiovascular medication are not associated with a higher incidence of VIT-induced anaphylaxis. (3) Relapsing anaphylactic reactions to both field and challenge stings are significantly more common during and after honeybee compared to Vespula VIT. Reports of severe field-sting reactions in mastocytosis patients suggest an increased risk of treatment failure which may be overcome by higher maintenance doses and longer duration of VIT.}, language = {en} } @article{SolimandoKalogirouKrebs2022, author = {Solimando, Antonio Giovanni and Kalogirou, Charis and Krebs, Markus}, title = {Angiogenesis as therapeutic target in metastatic prostate cancer - narrowing the gap between bench and bedside}, series = {Frontiers in Immunology}, volume = {13}, journal = {Frontiers in Immunology}, issn = {1664-3224}, doi = {10.3389/fimmu.2022.842038}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-263061}, year = {2022}, abstract = {Angiogenesis in metastatic castration-resistant prostate cancer (mCRPC) has been extensively investigated as a promising druggable biological process. Nonetheless, targeting angiogenesis has failed to impact overall survival (OS) in patients with mCRPC despite promising preclinical and early clinical data. This discrepancy prompted a literature review highlighting the tumor heterogeneity and biological context of Prostate Cancer (PCa). Narrowing the gap between the bench and bedside appears critical for developing novel therapeutic strategies. Searching clinicaltrials.gov for studies examining angiogenesis inhibition in patients with PCa resulted in n=20 trials with specific angiogenesis inhibitors currently recruiting (as of September 2021). Moreover, several other compounds with known anti-angiogenic properties - such as Metformin or Curcumin - are currently investigated. In general, angiogenesis-targeting strategies in PCa include biomarker-guided treatment stratification - as well as combinatorial approaches. Beyond established angiogenesis inhibitors, PCa therapies aiming at PSMA (Prostate Specific Membrane Antigen) hold the promise to have a substantial anti-angiogenic effect - due to PSMA´s abundant expression in tumor vasculature.}, language = {en} } @article{SchaeferZernecke2020, author = {Sch{\"a}fer, Sarah and Zernecke, Alma}, title = {CD8\(^+\) T cells in atherosclerosis}, series = {Cells}, volume = {10}, journal = {Cells}, number = {1}, issn = {2073-4409}, doi = {10.3390/cells10010037}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-220170}, year = {2020}, abstract = {Atherosclerotic lesions are populated by cells of the innate and adaptive immune system, including CD8\(^+\) T cells. The CD8\(^+\) T cell infiltrate has recently been characterized in mouse and human atherosclerosis and revealed activated, cytotoxic, and possibly dysfunctional and exhausted cell phenotypes. In mouse models of atherosclerosis, antibody-mediated depletion of CD8\(^+\) T cells ameliorates atherosclerosis. CD8\(^+\) T cells control monopoiesis and macrophage accumulation in early atherosclerosis. In addition, CD8\(^+\) T cells exert cytotoxic functions in atherosclerotic plaques and contribute to macrophage cell death and necrotic core formation. CD8\(^+\) T cell activation may be antigen-specific, and epitopes of atherosclerosis-relevant antigens may be targets of CD8\(^+\) T cells and their cytotoxic activity. CD8\(^+\) T cell functions are tightly controlled by costimulatory and coinhibitory immune checkpoints. Subsets of regulatory CD25\(^+\)CD8\(^+\) T cells with immunosuppressive functions can inhibit atherosclerosis. Importantly, local cytotoxic CD8\(^+\) T cell responses may trigger endothelial damage and plaque erosion in acute coronary syndromes. Understanding the complex role of CD8\(^+\) T cells in atherosclerosis may pave the way for defining novel treatment approaches in atherosclerosis. In this review article, we discuss these aspects, highlighting the emerging and critical role of CD8\(^+\) T cells in atherosclerosis.}, language = {en} } @article{SchilbachAlkhaledWelkeretal.2015, author = {Schilbach, Karin and Alkhaled, Mohammed and Welker, Christian and Eckert, Franziska and Blank, Gregor and Ziegler, Hendrik and Sterk, Marco and M{\"u}ller, Friederike and Sonntag, Katja and Wieder, Thomas and Braum{\"u}ller, Heidi and Schmitt, Julia and Eyrich, Matthias and Schleicher, Sabine and Seitz, Christian and Erbacher, Annika and Pichler, Bernd J. and M{\"u}ller, Hartmut and Tighe, Robert and Lim, Annick and Gillies, Stephen D. and Strittmatter, Wolfgang and R{\"o}cken, Martin and Handgretinger, Rupert}, title = {Cancer-targeted IL-12 controls human rhabdomyosarcoma by senescence induction and myogenic differentiation}, series = {OncoImmunology}, volume = {4}, journal = {OncoImmunology}, number = {7}, doi = {10.1080/2162402X.2015.1014760}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-154579}, pages = {e1014760}, year = {2015}, abstract = {Stimulating the immune system to attack cancer is a promising approach, even for the control of advanced cancers. Several cytokines that promote interferon-γ-dominated immune responses show antitumor activity, with interleukin 12 (IL-12) being of major importance. Here, we used an antibody-IL-12 fusion protein (NHS-IL12) that binds histones of necrotic cells to treat human sarcoma in humanized mice. Following sarcoma engraftment, NHS-IL12 therapy was combined with either engineered IL-7 (FcIL-7) or IL-2 (IL-2MAB602) for continuous cytokine bioavailability. NHS-IL12 strongly induced innate and adaptive antitumor immunity when combined with IL-7 or IL-2. NHS-IL12 therapy significantly improved survival of sarcoma-bearing mice and caused long-term remissions when combined with IL-2. NHS-IL12 induced pronounced cancer cell senescence, as documented by strong expression of senescence-associated p16\(^{INK4a}\) and nuclear translocation of p-HP1γ, and permanent arrest of cancer cell proliferation. In addition, this cancer immunotherapy initiated the induction of myogenic differentiation, further promoting the hypothesis that efficient antitumor immunity includes mechanisms different from cytotoxicity for efficient cancer control in vivo.}, language = {en} } @phdthesis{Rechtenwald2011, author = {Rechtenwald, Christian}, title = {Charakterisierung und Weiterentwicklung eines Balanced Lethal Systems in \({Salmonella}\) \({spp}\)}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-71092}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2011}, abstract = {Charakterisierung und Weiterentwicklung eines Systems zur antibiotikafreien Plasmidstabilisierung und Sekretion heterologer Antigene {\"u}ber das H{\"a}molysin a-Sekretionssystem in attenuiereten Salmonella-St{\"a}mmen. Ziel ist die Entwicklung tumorspezifischer Vakzine.}, subject = {Salmonella}, language = {de} } @article{RascheKortuemRaabetal.2019, author = {Rasche, Leo and Kort{\"u}m, K. Martin and Raab, Marc S. and Weinhold, Niels}, title = {The impact of tumor heterogeneity on diagnostics and novel therapeutic strategies in multiple myeloma}, series = {International Journal of Molecular Sciences}, volume = {20}, journal = {International Journal of Molecular Sciences}, number = {5}, issn = {1422-0067}, doi = {10.3390/ijms20051248}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-285402}, year = {2019}, abstract = {Myeloma is characterized by extensive inter-patient genomic heterogeneity due to multiple different initiating events. A recent multi-region sequencing study demonstrated spatial differences, with progression events, such as TP53 mutations, frequently being restricted to focal lesions. In this review article, we describe the clinical impact of these two types of tumor heterogeneity. Target mutations are often dominant at one site but absent at other sites, which poses a significant challenge to personalized therapy in myeloma. The same holds true for high-risk subclones, which can be locally restricted, and as such not detectable at the iliac crest, which is the usual sampling site. Imaging can improve current risk classifiers and monitoring of residual disease, but does not allow for deciphering the molecular characteristics of tumor clones. In the era of novel immunotherapies, the clinical impact of heterogeneity certainly needs to be re-defined. Yet, preliminary observations indicate an ongoing impact of spatial heterogeneity on the efficacy of monoclonal antibodies. In conclusion, we recommend combining molecular tests with imaging to improve risk prediction and monitoring of residual disease. Overcoming intra-tumor heterogeneity is the prerequisite for curing myeloma. Novel immunotherapies are promising but research addressing their impact on the spatial clonal architecture is highly warranted.}, language = {en} } @phdthesis{Piger2001, author = {Piger, Veronika}, title = {Einfluss immunologischer Mechanismen bei der Implantation von Embryonen und deren therapeutischer Nutzen im Rahmen des IVF-Programms}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-1181788}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2001}, abstract = {Trotz des hohen Standards in der IVF-Behandlung gibt es bestimmte Patientinnen bzw. Paare, bei denen es nach der Durchf{\"u}hrung von drei oder mehr optimalen Behandlungszyklen mit einem Transfer von mehr als sechs Embryonen insgesamt, nicht zum Eintritt einer Schwangerschaft kommt, vermutlich liegen hierf{\"u}r die Probleme in der Implantationsphase. Dies hieße, daß ein Embryo entweder gar nicht erst implantiert wird oder, daß er kurz nach der Implantation wieder abstirbt. Hieraus erg{\"a}be sich eine {\"A}hnlichkeit zum klinischen Abortgeschehen. Heute weiß man, daß dem klinischen Abortgeschehen in 60 bis 70 Prozent Chromosomenst{\"o}rungen zu Grunde liegen, in den {\"u}brigen F{\"a}llen wird haupts{\"a}chlich eine immunologische Ursache angenommen. Somit k{\"o}nnte auch die Ursache wiederholter frustraner IVF-Behandlungen im immunologischen Bereich zu suchen sein. In unserer Untersuchung wurden 100 sterile Paare untersucht, die von 1987 bis 1995 das IVF-Programm durchliefen. Das Kollektiv umfaßt Patientinnen zwischen 22 und 42 Jahren, mit folgenden gyn{\"a}kologischen Diagnosen: Normalbefund, idiopathische, tubare und ovarielle Sterilit{\"a}t. Bei den Partnern wurden die folgenden Befunde erhoben: Normozoospermie, Oligo-Astheno-Teratozoospermie und isolierte, aber sehr ausgepr{\"a}gte Asthenozoospermie. Bei beiden Partnern wurden die HLA-Antigene bestimmt, sowie zytotoxische Antik{\"o}rper im Serum der Frau gesucht. Anschließend wurden der Patientin intrakutan Lymphozyten ihres Partners in den Unterarm injiziert. Bei zu großer {\"U}bereinstimmung im HLA-System wurden Lymphozyten eines Fremdspenders verwendet, der in den wichtigsten Blutgruppenmerkmalen mit der Patientin {\"u}bereinstimmmte. Nach 4 Wochen wurde im Serum der Patientin nach sog. "sch{\"u}tzenden Antik{\"o}rpern" gesucht. Je nach Testergebnis wurde ein ausreichender Schutz angenommen oder eine Auffrischung empfohlen. Es wurde eine kumulative Schwangerschaftsrate von 53 Prozent erzielt. In {\"u}ber 70 Prozent trat diese bereits im ersten Zyklus nach Immunisierung ein. Folgende Schwangerschaftsverl{\"a}ufe konnten beobachtet werden: je ein Drittel Geburt, intakte Schwangerschaft (>12. SSW), Abort bzw. EUG. Die Immunisierungstherapie, wie von uns durchgef{\"u}hrt, scheint eher unspezifisch zu sein und auch zeitlich begrenzt. Der genaue Wirkmechanismus bleibt noch zu kl{\"a}ren. Die vorherrschenden Erkl{\"a}rungsmodelle in der Literatur f{\"u}r das immunologische Abortgeschehen sind die Folgenden: Ausbildung zytotoxischer Antik{\"o}rper, Fehlen Blockierender Faktoren, erh{\"o}htes HLA-Sharing der Partner.}, language = {de} } @article{PellegrinoDelBufaloDeAngelisetal.2020, author = {Pellegrino, Marsha and Del Bufalo, Francesca and De Angelis, Biagio and Quintarelli, Concetta and Caruana, Ignazio and de Billy, Emmanuel}, title = {Manipulating the metabolism to improve the efficacy of CAR T-cell immunotherapy}, series = {Cells}, volume = {10}, journal = {Cells}, number = {1}, issn = {2073-4409}, doi = {10.3390/cells10010014}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-220140}, year = {2020}, abstract = {The adoptive transfer of the chimeric antigen receptor (CAR) expressing T-cells has produced unprecedented successful results in the treatment of B-cell malignancies. However, the use of this technology in other malignancies remains less effective. In the setting of solid neoplasms, CAR T-cell metabolic fitness needs to be optimal to reach the tumor and execute their cytolytic function in an environment often hostile. It is now well established that both tumor and T cell metabolisms play critical roles in controlling the immune response by conditioning the tumor microenvironment and the fate and activity of the T cells. In this review, after a brief description of the tumoral and T cell metabolic reprogramming, we summarize the latest advances and new strategies that have been developed to improve the metabolic fitness and efficacy of CAR T-cell products.}, language = {en} } @article{PageWallstabeLotheretal.2021, author = {Page, Lukas and Wallstabe, Julia and Lother, Jasmin and Bauser, Maximilian and Kniemeyer, Olaf and Strobel, Lea and Voltersen, Vera and Teutschbein, Janka and Hortschansky, Peter and Morton, Charles Oliver and Brakhage, Axel A. and Topp, Max and Einsele, Hermann and Wurster, Sebastian and Loeffler, Juergen}, title = {CcpA- and Shm2-Pulsed Myeloid Dendritic Cells Induce T-Cell Activation and Enhance the Neutrophilic Oxidative Burst Response to Aspergillus fumigatus}, series = {Frontiers in Immunology}, volume = {12}, journal = {Frontiers in Immunology}, issn = {1664-3224}, doi = {10.3389/fimmu.2021.659752}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-239493}, year = {2021}, abstract = {Aspergillus fumigatus causes life-threatening opportunistic infections in immunocompromised patients. As therapeutic outcomes of invasive aspergillosis (IA) are often unsatisfactory, the development of targeted immunotherapy remains an important goal. Linking the innate and adaptive immune system, dendritic cells are pivotal in anti-Aspergillus defense and have generated interest as a potential immunotherapeutic approach in IA. While monocyte-derived dendritic cells (moDCs) require ex vivo differentiation, antigen-pulsed primary myeloid dendritic cells (mDCs) may present a more immediate platform for immunotherapy. To that end, we compared the response patterns and cellular interactions of human primary mDCs and moDCs pulsed with an A. fumigatus lysate and two A. fumigatus proteins (CcpA and Shm2) in a serum-free, GMP-compliant medium. CcpA and Shm2 triggered significant upregulation of maturation markers in mDCs and, to a lesser extent, moDCs. Furthermore, both A. fumigatus proteins elicited the release of an array of key pro-inflammatory cytokines including TNF-α, IL-1β, IL-6, IL-8, and CCL3 from both DC populations. Compared to moDCs, CcpA- and Shm2-pulsed mDCs exhibited greater expression of MHC class II antigens and stimulated stronger proliferation and IFN-γ secretion from autologous CD4\(^+\) and CD8\(^+\) T-cells. Moreover, supernatants of CcpA- and Shm2-pulsed mDCs significantly enhanced the oxidative burst in allogeneic neutrophils co-cultured with A. fumigatus germ tubes. Taken together, our in vitro data suggest that ex vivo CcpA- and Shm2-pulsed primary mDCs have the potential to be developed into an immunotherapeutic approach to tackle IA.}, language = {en} } @article{MunawarZhouPrommersbergeretal.2023, author = {Munawar, Umair and Zhou, Xiang and Prommersberger, Sabrina and Nerreter, Silvia and Vogt, Cornelia and Steinhardt, Maximilian J. and Truger, Marietta and Mersi, Julia and Teufel, Eva and Han, Seungbin and Haertle, Larissa and Banholzer, Nicole and Eiring, Patrick and Danhof, Sophia and Navarro-Aguadero, Miguel Angel and Fernandez-Martin, Adrian and Ortiz-Ruiz, Alejandra and Barrio, Santiago and Gallardo, Miguel and Valeri, Antonio and Castellano, Eva and Raab, Peter and Rudert, Maximilian and Haferlach, Claudia and Sauer, Markus and Hudecek, Michael and Martinez-Lopez, J. and Waldschmidt, Johannes and Einsele, Hermann and Rasche, Leo and Kort{\"u}m, K. Martin}, title = {Impaired FADD/BID signaling mediates cross-resistance to immunotherapy in Multiple Myeloma}, series = {Communications Biology}, volume = {6}, journal = {Communications Biology}, doi = {10.1038/s42003-023-05683-4}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-357609}, year = {2023}, abstract = {The treatment landscape in multiple myeloma (MM) is shifting from genotoxic drugs to immunotherapies. Monoclonal antibodies, immunoconjugates, T-cell engaging antibodies and CART cells have been incorporated into routine treatment algorithms, resulting in improved response rates. Nevertheless, patients continue to relapse and the underlying mechanisms of resistance remain poorly understood. While Impaired death receptor signaling has been reported to mediate resistance to CART in acute lymphoblastic leukemia, this mechanism yet remains to be elucidated in context of novel immunotherapies for MM. Here, we describe impaired death receptor signaling as a novel mechanism of resistance to T-cell mediated immunotherapies in MM. This resistance seems exclusive to novel immunotherapies while sensitivity to conventional anti-tumor therapies being preserved in vitro. As a proof of concept, we present a confirmatory clinical case indicating that the FADD/BID axis is required for meaningful responses to novel immunotherapies thus we report impaired death receptor signaling as a novel resistance mechanism to T-cell mediated immunotherapy in MM.}, language = {en} } @phdthesis{Lutz2017, author = {Lutz, Mathias}, title = {T-Zell-Immunit{\"a}t gegen die tumorassoziierten Antigene HER2/neu, MUC1, PRAME und WT1 bei gesunden Blutspendern und Schwangeren als ein immuntherapeutisches Modell f{\"u}r die allogene Blutstammzelltransplantation}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-156587}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {Der Erfolg der allogenen h{\"a}matopoetischen Stammzelltransplantation (HSZT) als Immuntherapie basiert neben den Minorantigendifferenzen zwischen Spender und Empf{\"a}nger entscheidend auf einer spendervermittelten Immunit{\"a}t gegen tumorassoziierte Antigene (TAA), {\"u}ber deren Herkunft und Frequenz bei gesunden Blutspendern die derzeitige Studienlage kaum Informationen bietet. Da f{\"u}r viele klinisch relevante TAA eine Expression im fetalen und plazentaren Gewebe bekannt ist, wurde in dieser Arbeit die Schwangerschaft als m{\"o}glicher Ausl{\"o}ser dieser T-lymphozyt{\"a}ren Ged{\"a}chtnisimmunantworten im Sinne eines Tumor- und Transplantationsmodells untersucht. Hierf{\"u}r wurden insgesamt 114 gesunde Blutspender in drei Subgruppen aus 38 Frauen mit negativer Schwangerschaftsanamnese, 38 Frauen mit positiver Geburtenanamnese und 38 M{\"a}nnern in einer Querschnittsstudie betrachtet, daneben wurden 44 Frauen longitudinal w{\"a}hrend und nach ihrer ersten Schwangerschaft untersucht. Dabei wurden die CD8-positiven T-Lymphozyten der Probanden isoliert, mit Peptiden der vier klinisch relevanten TAA HER2/neu (human epidermal growth factor receptor 2), MUC1 (Mucin 1), PRAME (preferentially expressed antigen of melanoma) und WT1 (Wilms tumor protein 1) stimuliert und die Produktion von IFN-γ-mRNA mittels RT-qPCR gemessen. Daneben wurden zum Vergleich durchflusszytometrische und ELISPOT-basierte Verfahren durchgef{\"u}hrt. Bei den gesunden Blutspendern konnten CD8-positive Ged{\"a}chtnisimmunantworten von niedriger und/oder hoher funktioneller Avidit{\"a}t gegen alle vier untersuchten TAA gemessen werden: Die Frequenz der dabei als „positiv" definierten Immunantworten betrug bei HER2/neu 5 \%, bei MUC1 14 \%, bei PRAME 7 \% und bei WT1 15 \%. M{\"a}nner wiesen insgesamt h{\"o}here absolute Level der Immunantworten gegen die untersuchten TAA auf, was auf eine testikul{\"a}re Expression dieser Antigene zur{\"u}ckzuf{\"u}hren sein k{\"o}nnte. In der Longitudinalanalyse bei den erstschwangeren Frauen ließen sich die st{\"a}rksten Immunantworten zu Beginn der Schwangerschaft nachweisen, so dass es hier zu einem „Boost" pr{\"a}existenter TAA-spezifischer Autoimmunit{\"a}t zu kommen scheint. Durch das immunsuppressive hormonelle Milieu im Verlauf der Schwangerschaft und den Verlust der Zielantigene der feto-plazentaren Einheit durch die Geburt und Nachgeburt scheint diese Immunit{\"a}t aber nicht zu persistieren. Dadurch erkl{\"a}rt sich auch die Beobachtung, dass Frauen mit einer positiven Geburtenanamnese keine st{\"a}rkeren Immunantworten gegen die untersuchten TAA aufwiesen als Frauen mit einer negativen Schwangerschaftsanamnese. Die Schwangerschaft hinterl{\"a}sst diesbez{\"u}glich also ohne die Anwesenheit der vermittelnden Antigene keinen regelhaft bleibenden Effekt. Diese Resultate decken sich mit Beobachtungen aus der Tumorimmuntherapie, bei denen Vakzinierungen gegen TAA zwar eine kurzfristige Immunit{\"a}t generieren konnten, die aber nicht persistierte. Im Rahmen der HSZT kann eine solche TAA-spezifische Immunit{\"a}t vom Spender auf den Empf{\"a}nger transferiert werden und vermag dann aufgrund des proinflammatorischen Immunmilieus sehr wohl zu expandieren und in einem begrenzten Ausmaß auch zu persistieren. Dementsprechend ergeben sich aus den in dieser Arbeit gewonnenen Resultaten relevante Implikationen f{\"u}r die allogene und in geringerem Ausmaß die autologe HSZT, daneben aber auch f{\"u}r innovative Tumortherapien wie die Immuncheckpoint-Blockade, da die Persistenz von tumorspezifischer Immunit{\"a}t letztendlich hochrelevant f{\"u}r eine langfristige Tumorkontrolle und damit f{\"u}r ein tumorfreies {\"U}berleben ist. Das vorliegende Modell tr{\"a}gt somit zum Verst{\"a}ndnis der komplexen immunregulatorischen Vorg{\"a}nge bei der Tumorkontrolle bei. Ob die hierbei aufgezeigten Immunantworten generell zu einer verbesserten TAA-spezifischen Immunrekonstitution und konsekutiv zu einem besseren klinischen Ergebnis beitragen, bleibt offen und wird in klinischen Studien gekl{\"a}rt werden m{\"u}ssen.}, subject = {Immuntherapie}, language = {de} } @phdthesis{Lorenz2004, author = {Lorenz, Judith}, title = {Immunhistochemische und zellbiologische Analyse humaner monoklonaler Antik{\"o}rper gegen kolorektale Karzinome}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-11532}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2004}, abstract = {Drei verschiedene mit Hilfe der humanen Hybridomatechnologie aus einem Rektumkarzinompatienten isolierte humane monoklonale Antik{\"o}rper wurden immunhistochemisch und mit Hilfe zellbiologischer Tests untersucht. Die Ergebnisse der Immunperoxidasef{\"a}rbung geben erste Hinweise auf die Art und Verteilung der von den Antik{\"o}rpern erkannten Antigene. HH 98/81-33-154 und HH 99/71-81-149 f{\"a}rben nur wenige kolorektale Karzinome sowie einige Tumoren verschiedenen histologischen Ursprungs an und zeigen keine Reaktion mit normalen adulten oder fetalen Geweben. HH 101/99-14 reagiert dagegen außer mit zahlreichen Dickdarmkarzinomen auch mit einigen fetalen Geweben, was auf die Expression des spezifischen Antigens w{\"a}hrend der Fetalperiode schließen l{\"a}ßt. Neben den Kreuzreaktionen mit verschiedenen zentralnerv{\"o}sen Strukturen adulter Normalgewebe, die im Hinblick auf paraneoplastische neurologische Krankheitsbilder von Interesse sind, zeigt der Antik{\"o}rper weiterhin eine Affinit{\"a}t zu dr{\"u}sig differenzierten Tumoren. Im MTT-Assay auf Zellen der Kolonkarzinomzellinie CACO-2 bewirkte unverd{\"u}nnter Zellkultur{\"u}berstand der Antik{\"o}rper HH 98/81-33-154 und HH 101/99-14 eine starke Proliferationshemmung. Dieser Effekt konnte sowohl nach 24- als auch nach 48st{\"u}ndiger Inkubation, nicht jedoch mit verd{\"u}nntem {\"U}berstand nachgewiesen werden. Mit {\"U}berstand von HH 99/71-81-149 war hingegen nur eine schwache Hemmung mitochondrialer Dehydrogenasen zu verzeichnen. Zellen der Linien HT-29, COLO 206F sowie COLO 320 zeigten sich hingegen resistent gegen die antik{\"o}rpervermittelte Wachstumshemmung. Mit Hilfe des Cell Death Detection ELISAPLUS{\`O} konnte weiterhin belegt werden, daß HH 98/81-33-154 und HH 101/99-14 nicht nur das Wachstum von Tumorzellen hemmen, sondern {\"u}berdies auf der Zellinie CACO-2 Apoptose induzieren. In beiden funktionellen Tests {\"u}berstieg die gemessene Aktivit{\"a}t des niedriger konzentrierten HH 98/81-33-154 die von HH 101/99-14. Die relativ geringe Anzahl von Kreuzreaktionen mit gesundem Gewebe sowie die apoptoseinduzierende Aktivit{\"a}t von HH 98/81-33-154 und HH 101/99-14 unterstreichen die Eignung der Antik{\"o}rper als potentielle Immuntherapeutika f{\"u}r die adjuvante Behandlung des kolorektalen Karzinoms, einer weltweit zu den h{\"a}ufigsten Krebsarten z{\"a}hlenden Tumorerkrankung.}, language = {de} } @article{LangZaitsevaWajant2022, author = {Lang, Isabell and Zaitseva, Olena and Wajant, Harald}, title = {FcγRs and their relevance for the activity of anti-CD40 antibodies}, series = {International Journal of Molecular Sciences}, volume = {23}, journal = {International Journal of Molecular Sciences}, number = {21}, issn = {1422-0067}, doi = {10.3390/ijms232112869}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-290458}, year = {2022}, abstract = {Simple Summary Targeting of CD40 with antibodies attracts significant translational interest. While inhibitory CD40 targeting appears particularly attractive in the field of organ transplantation and for the treatment of autoimmune diseases, stimulatory CD40 targeting is the aim in tumor immunotherapy and vaccination against infectious pathogens. It turned out that lack of FcγR-binding is the crucial factor for the development of safe and well-tolerated inhibitory anti-CD40 antibodies. In striking contrast, FcγR-binding is of great importance for the CD40 stimulatory capacity of the majority of anti-CD40 antibodies. Typically, anti-CD40 antibodies only robustly stimulate CD40 when presented by FcγRs. However, FcγR-binding of anti-CD40 antibodies also triggers unwanted activities such as destruction of CD40 expressing cells by ADCC or ADCP. Based on a brief discussion of the mechanisms of CD40 activation, we give an overview of the ongoing activities in the development of anti-CD40 antibodies under special consideration of attempts aimed at the development of anti-CD40 antibodies with FcγR-independent agonism or FcγR subtype selectivity. Abstract Inhibitory targeting of the CD40L-CD40 system is a promising therapeutic option in the field of organ transplantation and is also attractive in the treatment of autoimmune diseases. After early complex results with neutralizing CD40L antibodies, it turned out that lack of Fcγ receptor (FcγR)-binding is the crucial factor for the development of safe inhibitory antibodies targeting CD40L or CD40. Indeed, in recent years, blocking CD40 antibodies not interacting with FcγRs, has proven to be well tolerated in clinical studies and has shown initial clinical efficacy. Stimulation of CD40 is also of considerable therapeutic interest, especially in cancer immunotherapy. CD40 can be robustly activated by genetically engineered variants of soluble CD40L but also by anti-CD40 antibodies. However, the development of CD40L-based agonists is biotechnologically and pharmacokinetically challenging, and anti-CD40 antibodies typically display only strong agonism in complex with FcγRs or upon secondary crosslinking. The latter, however, typically results in poorly developable mixtures of molecule species of varying stoichiometry and FcγR-binding by anti-CD40 antibodies can elicit unwanted side effects such as antibody-dependent cellular cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP) of CD40 expressing immune cells. Here, we summarize and compare strategies to overcome the unwanted target cell-destroying activity of anti-CD40-FcγR complexes, especially the use of FcγR type-specific mutants and the FcγR-independent cell surface anchoring of bispecific anti-CD40 fusion proteins. Especially, we discuss the therapeutic potential of these strategies in view of the emerging evidence for the dose-limiting activities of systemic CD40 engagement.}, language = {en} } @article{LandwehrAltieriSchreineretal.2020, author = {Landwehr, Laura-Sophie and Altieri, Barbara and Schreiner, Jochen and Sbiera, Iuliu and Weigand, Isabel and Kroiss, Matthias and Fassnacht, Martin and Sbiera, Silviu}, title = {Interplay between glucocorticoids and tumor-infiltrating lymphocytes on the prognosis of adrenocortical carcinoma}, series = {Journal for ImmunoTherapy of Cancer}, volume = {8}, journal = {Journal for ImmunoTherapy of Cancer}, doi = {10.1136/jitc-2019-000469}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-229893}, year = {2020}, abstract = {Background Adrenocortical carcinoma (ACC) is a rare endocrine malignancy. Tumor-related glucocorticoid excess is present in similar to 60\% of patients and associated with particularly poor prognosis. Results of first clinical trials using immune checkpoint inhibitors were heterogeneous. Here we characterize tumor-infiltrating T lymphocytes (TILs) in ACC in association with glucocorticoids as potential explanation for resistance to immunotherapy. Methods We performed immunofluorescence analysis to visualize tumor-infiltrating T cells (CD3\(^+\)), T helper cells (CD3\(^+\)CD4\(^+\)), cytotoxic T cells (CD3\(^+\)CD8\(^+\)) and regulatory T cells (Tregs; CD3\(^+\)CD4\(^+\)FoxP3\(^+\)) in 146 ACC tissue specimens (107 primary tumors, 16 local recurrences, 23 metastases). Quantitative data of immune cell infiltration were correlated with clinical data (including glucocorticoid excess). Results 86.3\% of ACC specimens showed tumor infiltrating T cells (7.7 cells/high power field (HPF)), including T helper (74.0\%, 6.7 cells/HPF), cytotoxic T cells (84.3\%, 5.7 cells/HPF) and Tregs (49.3\%, 0.8 cells/HPF). The number of TILs was associated with better overall survival (HR for death: 0.47, 95\% CI 0.25 to 0.87), which was true for CD4\(^+\)- and CD8\(^+\) subpopulations as well. In localized, non-metastatic ACC, the favorable impact of TILs on overall and recurrence-free survival was manifested even independently of ENSAT (European Network for the Study of Adrenal Tumors) stage, resection status and Ki67 index. T helper cells were negatively correlated with glucocorticoid excess (Phi=-0.290, p=0.009). Patients with glucocorticoid excess and low TILs had a particularly poor overall survival (27 vs. 121 months in patients with TILs without glucocorticoid excess). Conclusion Glucocorticoid excess is associated with T cell depletion and unfavorable prognosis. To reactivate the immune system in ACC by checkpoint inhibitors, an inhibition of adrenal steroidogenesis might be pivotal and should be tested in prospective studies.}, language = {en} } @phdthesis{Kuehnemundt2024, author = {K{\"u}hnemundt, Johanna}, title = {Defined microphysiologic 3D tumour models with aspects from the tumour microenvironment for the evaluation of cellular immunotherapies}, doi = {10.25972/OPUS-27667}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-276674}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2024}, abstract = {Adoptive cellular immunotherapy with chimeric antigen receptor (CAR) T cells is highly effective in haematological malignancies. This success, however, has not been achieved in solid tumours so far. In contrast to hematologic malignancies, solid tumours include a hostile tumour microenvironment (TME), that poses additional challenges for curative effects and consistent therapeutic outcome. These challenges manifest in physical and immunological barriers that dampen efficacy of the CAR T cells. Preclinical testing of novel cellular immunotherapies is performed mainly in 2D cell culture and animal experiments. While 2D cell culture is an easy technique for efficacy analysis, animal studies reveal information about toxicity in vivo. However, 2D cell culture cannot fully reflect the complexity observed in vivo, because cells are cultured without anchorage to a matrix and only short-term periods are feasible. Animal studies provide a more complex tissue environment, but xenografts often lack human stroma and tumour inoculation occurs mostly ectopically. This emphasises the need for standardisable and scalable tumour models with incorporated TME-aspects, which enable preclinical testing with enhanced predictive value for the clinical outcome of immunotherapies. Therefore, microphysiologic 3D tumour models based on the biological SISmuc (Small Intestinal mucosa and Submucosa) matrix with preserved basement membrane were engaged and improved in this work to serve as a modular and versatile tumour model for efficacy testing of CAR T cells. In order to reflect a variety of cancer entities, TME-aspects, long-term stability and to enhance the read-out options they were further adapted to achieve scalable and standardisable defined microphysiologic 3D tumour models. In this work, novel culture modalities (semi-static, sandwich-culture) were characterised and established that led to an increased and organised tissue generation and long-term stability. Application of the SISmuc matrix was extended to sarcoma and melanoma models and serial bioluminescence intensity (BLI)-based in vivo imaging analysis was established in the microphysiologic 3D tumour models, which represents a time-efficient read-out method for quality evaluation of the models and treatment efficacy analysis, that is independent of the cell phenotype. Isolation of cancer-associated-fibroblasts (CAFs) from lung (tumour) tissue was demonstrated and CAF-implementation further led to stromal-enriched microphysiologic 3D tumour models with in vivo-comparable tissue-like architecture. Presence of CAFs was confirmed by CAF-associated markers (FAP, α-SMA, MMP-2/-9) and cytokines correlated with CAF phenotype, angiogenesis, invasion and immunomodulation. Additionally, an endothelial cell barrier was implemented for static and dynamic culture in a novel bioreactor set-up, which is of particular interest for the analysis of immune cell diapedesis. Studies in microphysiologic 3D Ewing's sarcoma models indicated that sarcoma cells could be sensitised for GD2-targeting CAR T cells. After enhancing the scale of assessment of the microphysiologic 3D tumour models and improving them for CAR T cell testing, the tumour models were used to analyse their sensitivity towards differently designed receptor tyrosine kinase-like orphan receptor 1 (ROR1) CAR T cells and to study the effects of the incorporated TME-aspects on the CAR T cell treatment respectively. ROR1 has been described as a suitable target for several malignancies including triple negative breast cancer (TNBC), as well as lung cancer. Therefore, microphysiologic 3D TNBC and lung cancer models were established. Analysis of ROR1 CAR T cells that differed in costimulation, spacer length and targeting domain, revealed, that the microphysiologic 3D tumour models are highly sensitive and can distinguish optimal from sub-optimal CAR design. Here, higher affinity of the targeting domain induced stronger anti-tumour efficacy and anti-tumour function depended on spacer length, respectively. Long-term treatment for 14 days with ROR1 CAR T cells was demonstrated in dynamic microphysiologic 3D lung tumour models, which did not result in complete tumour cell removal, whereas direct injection of CAR T cells into TNBC and lung tumour models represented an alternative route of application in addition to administration via the medium flow, as it induced strong anti-tumour response. Influence of the incorporated TME-aspects on ROR1 CAR T cell therapy represented by CAF-incorporation and/or TGF-β supplementation was analysed. Presence of TGF-β revealed that the specific TGF-β receptor inhibitor SD-208 improves ROR1 CAR T cell function, because it effectively abrogated immunosuppressive effects of TGF-β in TNBC models. Implementation of CAFs should provide a physical and immunological barrier towards ROR1 CAR T cells, which, however, was not confirmed, as ROR1 CAR T cell function was retained in the presence of CAFs in stromal-enriched microphysiologic 3D lung tumour models. The absence of an effect of CAF enrichment on CAR T cell efficacy suggests a missing component for the development of an immunosuppressive TME, even though immunomodulatory cytokines were detected in co-culture models. Finally, improved gene-edited ROR1 CAR T cells lacking exhaustion-associated genes (PD-1, TGF-β-receptor or both) were challenged by the combination of CAF-enrichment and TGF-β in microphysiologic 3D TNBC models. Results indicated that the absence of PD-1 and TGF-β receptor leads to improved CAR T cells, that induce strong tumour cell lysis, and are protected against the hostile TME. Collectively, the microphysiologic 3D tumour models presented in this work reflect aspects of the hostile TME of solid tumours, engage BLI-based analysis and provide long-term tissue homeostasis. Therefore, they present a defined, scalable, reproducible, standardisable and exportable model for translational research with enhanced predictive value for efficacy testing and candidate selection of cellular immunotherapy, as exemplified by ROR1 CAR T cells.}, subject = {Immuntherapie}, language = {en} } @article{KouhestaniGeisAlsourietal.2021, author = {Kouhestani, Dina and Geis, Maria and Alsouri, Saed and Bumm, Thomas G. P. and Einsele, Hermann and Sauer, Markus and Stuhler, Gernot}, title = {Variant signaling topology at the cancer cell-T-cell interface induced by a two-component T-cell engager}, series = {Cellular \& Molecular Immunology}, volume = {18}, journal = {Cellular \& Molecular Immunology}, doi = {10.1038/s41423-020-0507-7}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-241189}, pages = {1568-1570}, year = {2021}, abstract = {No abstract available.}, language = {en} } @phdthesis{Hofmann2011, author = {Hofmann, Bernd}, title = {Bienen-/Wespengiftallergie: Wirksamkeit und Sicherheit einer Immuntherapie mit 100 µg Gift {\"u}ber 3 bis 5 Jahre}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-57500}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2011}, abstract = {Die Bienen-/Wespengiftallergie ist auf der einen Seite eine potentiell lebensbedrohliche IgE-vermittelte Allergiekrankheit, in Deutschland neben den Nahrungsmittelallergien die h{\"a}ufigste Ursache f{\"u}r eine t{\"o}dlich verlaufende Anaphylaxie. Auf der anderen Seite steht mit der Bienen-/Wespengift-spezifischen Immuntherapie (SIT) gerade f{\"u}r diese Allergie seit Jahrzehnten eine hochwirksame kausale Therapie zur Verf{\"u}gung. Placebokontrollierte Studien mit unbehandelten Patienten sind daher aus ethischen Gr{\"u}nden nicht vertretbar. Neue Erkenntnisse zur Sicherheit dieser Therapieform k{\"o}nnen nur aus der Verlaufsbeobachtung standardisiert behandelter Patientenkollektive gewonnen werden. Der besondere Wert der hier analysierten großen Patientenserie liegt vor allem darin, dass alle Patienten in einer Allergieambulanz (der Klinik und Poliklinik f{\"u}r Dermatologie, Venerologie und Allergologie) betreut wurden. Im gesamten Behandlungszeitraum aller 679 Patienten wurde die Diagnostik und Therapie der Bienen-/Wespengiftallergie hoch standardisiert durchgef{\"u}hrt und nicht ver{\"a}ndert. Die dadurch gleichbleibende Betreuung garantierten Qualit{\"a}t, Umfang und Homogenit{\"a}t der Dokumentation und damit die Vergleichbarkeit der retrospektiv erfassten Daten {\"u}ber den gesamten Zeitraum 1988 bis 2008. F{\"u}r die besonders wichtige Verlaufsbeobachtung nach Ende der spezifischen Immuntherapie (SIT) wurden zus{\"a}tzlich 616 der 679 Patienten aus Unterfranken und Umgebung direkt telefonisch befragt. Die bekannten charakteristischen Merkmale von Patienten mit Bienen-/Wespengiftallergie zeigten sich auch in dem untersuchten Kollektiv. Imkert{\"a}tigkeit ist die wahrscheinlichste Erkl{\"a}rung warum Bienengiftallergiker j{\"u}nger und h{\"a}ufiger m{\"a}nnlich sind. Die prognostisch bedeutsamen Schweregrade der allergischen Indikatorstichreaktion (das Stichereignis mit der schwersten anaphylaktischen Reaktion vor SIT) unterscheiden sich zwischen Bienen- und Wespengiftallergikern dagegen nicht. In den diagnostischen Untersuchungen vor und auch am Ende der SIT waren die Schwellenwertkonzentrationen des Intrakutan- und Pricktests bei Bienengiftallergikern im Vergleich zu den Wespengiftallergikern signifikant niedriger, die spezifischen IgE-Serumspiegel h{\"o}her. Die Wirksamkeit der SIT mit Bienengift betrug 89,0 \%. Nur 11,0 \% der Patienten (8 von 73 Patienten mit erneutem Bienenstich) hatten nach Beginn der SIT erneut eine}, subject = {Bienengiftallergie}, language = {de} } @article{HenkeNandigamaErguen2020, author = {Henke, Erik and Nandigama, Rajender and Erg{\"u}n, S{\"u}leyman}, title = {Extracellular matrix in the tumor microenvironment and its impact on cancer therapy}, series = {Frontiers in Molecular Biosciences}, volume = {6}, journal = {Frontiers in Molecular Biosciences}, number = {160}, issn = {2296-889X}, doi = {10.3389/fmolb.2019.00160}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-199341}, year = {2020}, abstract = {Solid tumors are complex organ-like structures that consist not only of tumor cells but also of vasculature, extracellular matrix (ECM), stromal, and immune cells. Often, this tumor microenvironment (TME) comprises the larger part of the overall tumor mass. Like the other components of the TME, the ECM in solid tumors differs significantly from that in normal organs. Intratumoral signaling, transport mechanisms, metabolisms, oxygenation, and immunogenicity are strongly affected if not controlled by the ECM. Exerting this regulatory control, the ECM does not only influence malignancy and growth of the tumor but also its response toward therapy. Understanding the particularities of the ECM in solid tumor is necessary to develop approaches to interfere with its negative effect. In this review, we will also highlight the current understanding of the physical, cellular, and molecular mechanisms by which the pathological tumor ECM affects the efficiency of radio-, chemo-, and immunotherapy. Finally, we will discuss the various strategies to target and modify the tumor ECM and how they could be utilized to improve response to therapy.}, language = {en} } @article{HaakeHaackSchaeferetal.2023, author = {Haake, Markus and Haack, Beatrice and Sch{\"a}fer, Tina and Harter, Patrick N. and Mattavelli, Greta and Eiring, Patrick and Vashist, Neha and Wedekink, Florian and Genssler, Sabrina and Fischer, Birgitt and Dahlhoff, Julia and Mokhtari, Fatemeh and Kuzkina, Anastasia and Welters, Marij J. P. and Benz, Tamara M. and Sorger, Lena and Thiemann, Vincent and Almanzar, Giovanni and Selle, Martina and Thein, Klara and Sp{\"a}th, Jacob and Gonzalez, Maria Cecilia and Reitinger, Carmen and Ipsen-Escobedo, Andrea and Wistuba-Hamprecht, Kilian and Eichler, Kristin and Filipski, Katharina and Zeiner, Pia S. and Beschorner, Rudi and Goedemans, Renske and Gogolla, Falk Hagen and Hackl, Hubert and Rooswinkel, Rogier W. and Thiem, Alexander and Romer Roche, Paula and Joshi, Hemant and P{\"u}hringer, Dirk and W{\"o}ckel, Achim and Diessner, Joachim E. and R{\"u}diger, Manfred and Leo, Eugen and Cheng, Phil F. and Levesque, Mitchell P. and Goebeler, Matthias and Sauer, Markus and Nimmerjahn, Falk and Schuberth-Wagner, Christine and Felten, Stefanie von and Mittelbronn, Michel and Mehling, Matthias and Beilhack, Andreas and van der Burg, Sjoerd H. and Riedel, Angela and Weide, Benjamin and Dummer, Reinhard and Wischhusen, J{\"o}rg}, title = {Tumor-derived GDF-15 blocks LFA-1 dependent T cell recruitment and suppresses responses to anti-PD-1 treatment}, series = {Nature Communications}, volume = {14}, journal = {Nature Communications}, doi = {10.1038/s41467-023-39817-3}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-357333}, year = {2023}, abstract = {Immune checkpoint blockade therapy is beneficial and even curative for some cancer patients. However, the majority don't respond to immune therapy. Across different tumor types, pre-existing T cell infiltrates predict response to checkpoint-based immunotherapy. Based on in vitro pharmacological studies, mouse models and analyses of human melanoma patients, we show that the cytokine GDF-15 impairs LFA-1/β2-integrin-mediated adhesion of T cells to activated endothelial cells, which is a pre-requisite of T cell extravasation. In melanoma patients, GDF-15 serum levels strongly correlate with failure of PD-1-based immune checkpoint blockade therapy. Neutralization of GDF-15 improves both T cell trafficking and therapy efficiency in murine tumor models. Thus GDF-15, beside its known role in cancer-related anorexia and cachexia, emerges as a regulator of T cell extravasation into the tumor microenvironment, which provides an even stronger rationale for therapeutic anti-GDF-15 antibody development.}, language = {en} } @article{GinzkeyEickerMargetetal.2013, author = {Ginzkey, Christian and Eicker, Sven and Marget, Matthias and Krause, J{\"o}rg and Brecht, Stefan and Westphal, Manfred and Hugo, Heinz-Hermann and Mehdorn, Maximilian and Steinmann, J{\"o}rg and Hamel, Wolfgang}, title = {Incomplete tumour control following DNA vaccination against rat gliomas expressing a model antigen}, series = {Acta Neurochirurgica}, volume = {155}, journal = {Acta Neurochirurgica}, number = {1}, doi = {10.1007/s00701-012-1526-7}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-126775}, pages = {51-59}, year = {2013}, abstract = {Background Vaccination against tumour-associated antigens is one approach to elicit anti-tumour responses. We investigated the effect of polynucleotide (DNA) vaccination using a model antigen (E. coli lacZ) in a syngeneic gliosarcoma model (9L). Methods Fisher 344 rats were vaccinated thrice by intramuscular injection of a lacZ-encoding or a control plasmid in weekly intervals. One week after the last vaccination, lacZ-expressing 9L cells were implanted into the striatum. Results After 3 weeks, in lacZ-vaccinated animals the tumours were significantly smaller than in control-vaccinated animals. In cytotoxic T cell assays lysis rates of >50 \% could only be observed in a few of the lacZ-vaccinated animals. This response was directed against lacZ-expressing and parental 9L cells but not against syngeneic MADB 106 adenocarcinoma cells. In Elispot assays interferon-γ production was observed upon stimulation with 9LlacZ and 9L wild-type but not MADB 106 cells. This response was higher for lacZ-immunized animals. All animals revealed dense infiltrates with CD8+ lymphocytes and, to a lesser extent, with NK cells. CD25-staining indicated cells possibly associated with the maintenance of peripheral tolerance to self-antigens. All tumours were densely infiltrated by microglia consisting mostly of ramified cells. Only focal accumulation of macrophage-like cells expressing ED1, a marker for phagocytic activity, was observed. Conclusion Prophylactic DNA vaccination resulted in effective but incomplete suppression of brain tumour formation. Mechanisms other than cytotoxic T cell responses as measured in the generally used in vitro assays appear to play a role in tumour suppression.}, language = {en} } @article{FeiglStahringerPeindletal.2023, author = {Feigl, Frederik Fabian and Stahringer, Anika and Peindl, Matthias and Dandekar, Gudrun and Koehl, Ulrike and Fricke, Stephan and Schmiedel, Dominik}, title = {Efficient redirection of NK cells by genetic modification with chemokine receptors CCR4 and CCR2B}, series = {International Journal of Molecular Sciences}, volume = {24}, journal = {International Journal of Molecular Sciences}, number = {4}, issn = {1422-0067}, doi = {10.3390/ijms24043129}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-304049}, year = {2023}, abstract = {Natural killer (NK) cells are a subset of lymphocytes that offer great potential for cancer immunotherapy due to their natural anti-tumor activity and the possibility to safely transplant cells from healthy donors to patients in a clinical setting. However, the efficacy of cell-based immunotherapies using both T and NK cells is often limited by a poor infiltration of immune cells into solid tumors. Importantly, regulatory immune cell subsets are frequently recruited to tumor sites. In this study, we overexpressed two chemokine receptors, CCR4 and CCR2B, that are naturally found on T regulatory cells and tumor-resident monocytes, respectively, on NK cells. Using the NK cell line NK-92 as well as primary NK cells from peripheral blood, we show that genetically engineered NK cells can be efficiently redirected using chemokine receptors from different immune cell lineages and migrate towards chemokines such as CCL22 or CCL2, without impairing the natural effector functions. This approach has the potential to enhance the therapeutic effect of immunotherapies in solid tumors by directing genetically engineered donor NK cells to tumor sites. As a future therapeutic option, the natural anti-tumor activity of NK cells at the tumor sites can be increased by co-expression of chemokine receptors with chimeric antigen receptors (CAR) or T cell receptors (TCR) on NK cells can be performed in the future.}, language = {en} } @article{EyrichRachorSchreiberetal.2013, author = {Eyrich, Matthias and Rachor, Johannes and Schreiber, Susanne C. and W{\"o}lfl, Matthias and Schlegel, Paul G.}, title = {Dendritic cell vaccination in pediatric gliomas: lessons learnt and future perspectives}, series = {Frontiers in Pediatrics}, journal = {Frontiers in Pediatrics}, doi = {10.3389/fped.2013.00012}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-96937}, year = {2013}, abstract = {Immunotherapy of malignant gliomas with autologous dendritic cells (DCs) in addition to surgery and radiochemotherapy has been a focus of intense research during the past decade. Since both children and adults are affected by this highly aggressive brain tumor, 10-15\% of the several hundred vaccinated patients represent children, making pediatric glioma patients the largest uniform pediatric vaccination cohort so far. In general, DC vaccination in malignant gliomas has been shown to be safe and several studies with a non-vaccinated control group could clearly demonstrate a survival benefit for the vaccinated patients. Interestingly, children and adolescents below 21 years of age seem to benefit even more than adult patients. This review summarizes the findings of the 25 clinical trials published so far and gives a perspective how DC vaccination could be implemented as part of multimodal therapeutic strategies in the near future.}, language = {en} } @article{DraganovSantidrianMinevetal.2019, author = {Draganov, Dobrin D. and Santidrian, Antonio F. and Minev, Ivelina and Duong, Nguyen and Kilinc, Mehmet Okyay and Petrov, Ivan and Vyalkova, Anna and Lander, Elliot and Berman, Mark and Minev, Boris and Szalay, Aladar A.}, title = {Delivery of oncolytic vaccinia virus by matched allogeneic stem cells overcomes critical innate and adaptive immune barriers}, series = {Journal of Translational Medicine}, volume = {17}, journal = {Journal of Translational Medicine}, issn = {100}, doi = {10.1186/s12967-019-1829-z}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-226312}, year = {2019}, abstract = {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.}, language = {en} } @phdthesis{Diessner2009, author = {Diessner, Ernst Joachim}, title = {Konstruktion eines Balanced lethal Systems f{\"u}r Salmonella typhi Ty21a}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-51638}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2009}, abstract = {Ziel der Arbeit ist unter anderem die Entwicklung von Vakzinen gegen maligne Neoplasien auf der Basis von attenuierten Bakterien. Sie dienen hierbei als Tr{\"a}ger von tumorspezifischen Antigenen. Diese k{\"o}nnen mit Hilfe des E. coli H{\"a}molysin-a-Sekretionssystems von Salmonella-Bakterienst{\"a}mmen sezerniert werden und eine spezifische Immunreaktion induzieren. Sowohl die Gene, die f{\"u}r das Sekretionssystem kodieren als auch die Gensequenzen des tumorspezifische Antigens sind bei dem Projekt auf dem detailliert beschriebenen Antigendelivery Plasmid pMO kodiert. Die bis dato angewandte Methode der pMO-Plasmidstabilisierung mit Hilfe von Antibiotikaresistenzgenen beinhaltet jedoch zahlreiche, beschriebene Probleme und wird seitens der Beh{\"o}rden in Impfst{\"a}mmen zunehmend restriktiv gehandhabt. Im Rahmen der Entwicklung eines bakteriellen Tumorimpfstoffes war es daher das Ziel dieser Arbeit ein System zur Stabilisierung des Antigendelivery Plasmids pMO zu etablieren, das auf den Einsatz von Antibiotikaresistenzgenen verzichtet.}, subject = {Salmonella}, language = {de} } @article{ArdeltEbbingAdamsetal.2015, author = {Ardelt, Peter U. and Ebbing, Jan and Adams, Fabian and Reiss, Cora and Arap, Wadih and Pasqualini, Renata and Bachmann, Alexander and Wetterauer, Ulrich and Riedmiller, Hubertus and Kneitz, Burkard}, title = {An anti-ubiquitin antibody response in transitional cell carcinoma of the urinary bladder}, series = {PLoS ONE}, volume = {10}, journal = {PLoS ONE}, number = {3}, doi = {10.1371/journal.pone.0118646}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-143711}, pages = {e0118646}, year = {2015}, abstract = {Background To use combinatorial epitope mapping ("fingerprinting") of the antibody response to identify targets of the humoral immune response in patients with transitional cell carcinoma (TCC) of the bladder. Methods A combinatorial random peptide library was screened on the circulating pool of immunoglobulins purified from an index patient with a high risk TCC (pTa high grade plus carcinoma in situ) to identify corresponding target antigens. A patient cohort was investigated for antibody titers against ubiquitin. Results We selected, isolated, and validated an immunogenic peptide motif from ubiquitin as a dominant epitope of the humoral response. Patients with TCC had significantly higher antibody titers against ubiquitin than healthy donors (p<0.007), prostate cancer patients (p<0.0007), and all patients without TCC taken together (p<0.0001). Titers from superficial tumors were not significantly different from muscle invasive tumors (p = 0.0929). For antibody response against ubiquitin, sensitivity for detection of TCC was 0.44, specificity 0.96, positive predictive value 0.96 and negative predictive value 0.41. No significant titer changes were observed during the standard BCG induction immunotherapy. Conclusions This is the first report to demonstrate an anti-ubiquitin antibody response in patients with TCC. Although sensitivity of antibody production was low, a high specificity and positive predictive value make ubiquitin an interesting candidate for further diagnostic and possibly immune modulating studies.}, language = {en} } @article{AdamKircherSbieraetal.2021, author = {Adam, Pia and Kircher, Stefan and Sbiera, Iuliu and Koehler, Viktoria Florentine and Berg, Elke and Kn{\"o}sel, Thomas and Sandner, Benjamin and Fenske, Wiebke Kristin and Bl{\"a}ker, Hendrik and Smaxwil, Constantin and Zielke, Andreas and Sipos, Bence and Allelein, Stephanie and Schott, Matthias and Dierks, Christine and Spitzweg, Christine and Fassnacht, Martin and Kroiss, Matthias}, title = {FGF-Receptors and PD-L1 in Anaplastic and Poorly Differentiated Thyroid Cancer: Evaluation of the Preclinical Rationale}, series = {Frontiers in Endocrinology}, volume = {12}, journal = {Frontiers in Endocrinology}, issn = {1664-2392}, doi = {10.3389/fendo.2021.712107}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-244653}, year = {2021}, abstract = {Background Treatment options for poorly differentiated (PDTC) and anaplastic (ATC) thyroid carcinoma are unsatisfactory and prognosis is generally poor. Lenvatinib (LEN), a multi-tyrosine kinase inhibitor targeting fibroblast growth factor receptors (FGFR) 1-4 is approved for advanced radioiodine refractory thyroid carcinoma, but response to single agent is poor in ATC. Recent reports of combining LEN with PD-1 inhibitor pembrolizumab (PEM) are promising. Materials and Methods Primary ATC (n=93) and PDTC (n=47) tissue samples diagnosed 1997-2019 at five German tertiary care centers were assessed for PD-L1 expression by immunohistochemistry using Tumor Proportion Score (TPS). FGFR 1-4 mRNA was quantified in 31 ATC and 14 PDTC with RNAscope in-situ hybridization. Normal thyroid tissue (NT) and papillary thyroid carcinoma (PTC) served as controls. Disease specific survival (DSS) was the primary outcome variable. Results PD-L1 TPS≥50\% was observed in 42\% of ATC and 26\% of PDTC specimens. Mean PD-L1 expression was significantly higher in ATC (TPS 30\%) than in PDTC (5\%; p<0.01) and NT (0\%, p<0.001). 53\% of PDTC samples had PD-L1 expression ≤5\%. FGFR mRNA expression was generally low in all samples but combined FGFR1-4 expression was significantly higher in PDTC and ATC compared to NT (each p<0.001). No impact of PD-L1 and FGFR 1-4 expression was observed on DSS. Conclusion High tumoral expression of PD-L1 in a large proportion of ATCs and a subgroup of PDTCs provides a rationale for immune checkpoint inhibition. FGFR expression is low thyroid tumor cells. The clinically observed synergism of PEM with LEN may be caused by immune modulation.}, language = {en} }