@article{Prelog2013, author = {Prelog, Martina}, title = {Vaccination in Patients with Rheumatoid Arthritis Receiving Immunotherapies}, series = {Clinical \& Cellular Immunology}, journal = {Clinical \& Cellular Immunology}, doi = {10.4172/2155-9899.S6-007}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-96446}, year = {2013}, abstract = {Patients with rheumatoid arthritis (RA) are at higher risk to suffer from morbidity due to vaccine-preventable diseases and, thus, display an important target population to receive vaccines for protection from infectious complications. There have been only a few studies focusing on the administration of vaccines in RA patients with immunotherapy. Overall, antibody response rates against influenza or pneumococcal disease appeared to be only slightly lower than expected in healthy individuals. Crucial problems in the interpretation of data from studies in RA patients vaccinated against influenza and pneumococcal disease are the impaired comparability of studies due to different study designs and type of vaccines used, different health states among RA patients, heterogeneity in treatments including concomitant therapy with conventional DMARDs and glucocorticoids in addition to biological agents. Assessment of vaccination status should be performed in the initial work-up of patients with RA and should ideally be administered before initiation of immunotherapies or during stable disease. Due to differences in antibody responses and uncertainty regarding maintenance of protective antibodies, routine controls for antibody titers and specific strategies for earlier re-vaccination might be scheduled for patients with RA.}, language = {en} } @phdthesis{Rasche2008, author = {Rasche, Leo}, title = {Tumortherapie mit Cocktails aus humanen monoklonalen Antik{\"o}rpern}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-35809}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2008}, abstract = {Humane oder humanisierte monoklonale Antik{\"o}rper haben sich in den letzten zehn Jahren als Arzneimittel etabliert. Sie sind hochspezifisch und zeigen in ihrer Anwendung im Vergleich zu konventionellen Therapeutika viel weniger Nebenwirkungen. In den 80er Jahren gelang es am Pathologischen Institut der Universit{\"a}t W{\"u}rzburg eine Reihe von humanen Antik{\"o}rpern aus Patienten zu isolieren, die hochspezifisch mit malignen Zellen reagieren und diese sowohl in vitro als auch im experimentellen Tiermodel selektiv durch Induktion von Apoptose t{\"o}ten. Um die Wirkungsweise von monoklonalen Antik{\"o}rpern in der Krebstherapie zu erh{\"o}hen, werden die meisten in Kombination mit herk{\"o}mmlichen Methoden, wie Chemotherapie, eingesetzt. Die ideale Therapieform sind hinsichtlich der Nebenwirkungen sog. Cocktails aus verschiedenen monoklonalen Antik{\"o}rpern. Allerdings sind die Studien hierzu noch wenig fortgeschritten. Das Ziel dieser Arbeit war es, in pr{\"a}klinischen Versuchsreihen den Einsatz verschiedener tumorspezifischer humaner monoklonaler Antik{\"o}rper als Cocktail und in Kombination mit Chemotherapie zu evaluieren. Hierzu wurden neun Antik{\"o}rper in 32 verschiedenen Antik{\"o}rperkombinationen hinsichtlich ihrer Auswirkungen auf die in vitro Proliferation einer Pankreaskarzinom-Zellinie untersucht. In Immunfluoreszenz-Aufnahmen ließ sich zeigen, dass kombinierte Antik{\"o}rper an unterschiedlichen Stellen an der Zelle binden, was eindeutig auf verschiedene Zielstrukturen hinweist. Einige werden dabei endozytiert, w{\"a}hrend andere auf der Zellmembran bleiben. Interessanterweise ließen sich Kombinationen identifizieren, deren antiproliferative Wirkung sowohl additiv als auch synergistisch ist, das heißt gr{\"o}ßer als die Summe ihrer Einzelaktivit{\"a}ten. Wurden Antik{\"o}rper mit Zytostatika (5-Flurouracil) kombiniert, so ließen sich ebenfalls synergistische Effekte beobachten. In FACS-Analysen zeigt sich ein gesteigertes Bindungsverhalten der Antik{\"o}rper, wenn die Zellen mit 5-FU vorinkubiert wurden. Zusammenfassend best{\"a}tigen die Ergebnisse dieser Arbeit die Beobachtung, dass die Wirkung humaner monoklonaler Antik{\"o}rper in Kombination mit Chemotherapie erh{\"o}ht werden kann. F{\"u}r die Zukunft humaner Antik{\"o}rper als Therapiemittel gegen maligne Erkrankungen mag allerdings noch wichtiger sein, dass Antik{\"o}per in Cocktails tats{\"a}chlich synergistische Wirkung zeigen k{\"o}nnen.}, subject = {Monoklonaler Antik{\"o}rper}, language = {de} } @phdthesis{GarciaGuerrero2017, author = {Garcia Guerrero, Estefania}, title = {Strategies to Obtain Tumor-Reactive Cells for Cancer Immunotherapy by Cell Sorting and Genetic Modifications of T Lymphocytes}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-150547}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {Recent advances in the field of cancer immunotherapy have enabled this therapeutic approach to enter the mainstream of modern cancer treatment. In particular, adoptive T cell therapy (ACT) is a potentially powerful immunotherapy approach that relies on the administration of tumor-specific T cells into the patient. There are several strategies to obtain tumor-reactive cytotoxic T lymphocytes (CTLs), which have already been shown to induce remarkable responses in the clinical setting. However, there are concerns and limitations regarding the conventional approaches to obtain tumor-reactive T cells, such as accuracy of the procedure and reproducibility. Therefore, we aimed to develop two approaches to improve the precision and efficacy of tumor-reactive T cells therapy. These two techniques could constitute effective, safe and broadly applicable alternatives to the conventional methods for obtaining tumor-specific CTLs. The first approach of this study is the so called "Doublet Technology". Here, we demonstrate that peptide-human leukocyte antigen-T cell receptor (pHLA-TCR) interactions that involve immune reactive peptides are stable and strong. Therefore, the CTLs that are bound by their TCR to tumor cells can be selected and isolated through FACS-based cell sorting taking advantage of this stable interaction between the CTLs and the target cells. The CTLs from acute myeloid leukemia (AML) patients obtained with this technique show cytolytic activity against blast cells suggesting a potential clinical use of these CTLs. "Doublet Technology" offers a personalized therapy in which there is no need for a priori knowledge of the exact tumor antigen. The second approach of this study is the Chimeric Antigen Receptor (CAR) Technology. We design several CARs targeting the B-Cell Maturation Antigen (BCMA). BCMA CAR T cells show antigen-specific cytolytic activity, production of cytokines including IFN-γ and IL-2, as well as productive proliferation. Although we confirm the presence of soluble BCMA in serum of multiple myeloma (MM) patients, we demonstrate that the presence of soluble protein does not abrogate the efficacy of BCMA CAR T cells suggesting that BCMA CAR T cells can be used in the clinical setting to treat MM patients. The high antigen specificity of CAR T cells allows efficient tumor cell eradication and makes CAR Technology attractive for broadly applicable therapies.}, subject = {Immunotherapy}, language = {en} } @article{StoevesandtHofmannHainetal.2013, author = {Stoevesandt, Johanna and Hofmann, Bernd and Hain, Johannes and Kerstan, Andreas and Trautmann, Axel}, title = {Single venom-based immunotherapy effectively protects patients with double positive tests to honey bee and Vespula venom}, series = {Allergy, Asthma \& Clinical Immunology}, journal = {Allergy, Asthma \& Clinical Immunology}, doi = {10.1186/1710-1492-9-33}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-96808}, year = {2013}, abstract = {Background Referring to individuals with reactivity to honey bee and Vespula venom in diagnostic tests, the umbrella terms "double sensitization" or "double positivity" cover patients with true clinical double allergy and those allergic to a single venom with asymptomatic sensitization to the other. There is no international consensus on whether immunotherapy regimens should generally include both venoms in double sensitized patients. Objective We investigated the long-term outcome of single venom-based immunotherapy with regard to potential risk factors for treatment failure and specifically compared the risk of relapse in mono sensitized and double sensitized patients. Methods Re-sting data were obtained from 635 patients who had completed at least 3 years of immunotherapy between 1988 and 2008. The adequate venom for immunotherapy was selected using an algorithm based on clinical details and the results of diagnostic tests. Results Of 635 patients, 351 (55.3\%) were double sensitized to both venoms. The overall re-exposure rate to Hymenoptera stings during and after immunotherapy was 62.4\%; the relapse rate was 7.1\% (6.0\% in mono sensitized, 7.8\% in double sensitized patients). Recurring anaphylaxis was statistically less severe than the index sting reaction (P = 0.004). Double sensitization was not significantly related to relapsing anaphylaxis (P = 0.56), but there was a tendency towards an increased risk of relapse in a subgroup of patients with equal reactivity to both venoms in diagnostic tests (P = 0.15). Conclusions Single venom-based immunotherapy over 3 to 5 years effectively and long-lastingly protects the vast majority of both mono sensitized and double sensitized Hymenoptera venom allergic patients. Double venom immunotherapy is indicated in clinically double allergic patients reporting systemic reactions to stings of both Hymenoptera and in those with equal reactivity to both venoms in diagnostic tests who have not reliably identified the culprit stinging insect.}, language = {en} } @phdthesis{Oesterreich2017, author = {Oesterreich, Babett}, title = {Preclinical development of an immunotherapy against antibiotic-resistant Staphylococcus aureus}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-123237}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {The Gram-positive bacterium Staphylococcus aureus is the leading cause of nosocomial infections. In particular, diseases caused by methicillin-resistant S. aureus (MRSA) are associated with higher morbidity, mortality and medical costs due to showing resistance to several classes of established antibiotics and their ability to develop resistance mechanisms against new antibiotics rapidly. Therefore, strategies based on immunotherapy approaches have the potential to close the gap for an efficient treatment of MRSA. In this thesis, a humanized antibody specific for the immunodominant staphylococcal antigen A (IsaA) was generated and thoroughly characterized as potential candidate for an antibody based therapy. A murine monoclonal antibody was selected for humanization based on its binding characteristics and the ability of efficient staphylococcal killing in mouse infection models. The murine antibody was humanized by CDR grafting and mouse and humanized scFv as well as scFv-Fc fragments were constructed for comparative binding studies to analyse the successful humanization. After these studies, the full antibody with the complete Fc region was constructed as isotype IgG1, IgG2 and IgG4, respectively to assess effector functions, including antibody-dependent killing of S. aureus. The biological activity of the humanized antibody designated hUK-66 was analysed in vitro with purified human PMNs and whole blood samples taken from healthy donors and patients at high risk of S. aureus infections, such as those with diabetes, end-stage renal disease, or artery occlusive disease (AOD). Results of the in vitro studies show, that hUK-66 was effective in antibody-dependent killing of S. aureus in blood from both healthy controls and patients vulnerable to S. aureus infections. Moreover, the biological activity of hUK-66 and hUK-66 combined with a humanized anti-alpha-toxin antibody (hUK-tox) was investigated in vivo using a mouse pneumonia model. The in vivo results revealed the therapeutic efficacy of hUK-66 and the antibody combination of hUK-66 and hUK-tox to prevent staphylococcal induced pneumonia in a prophylactic set up. Based on the experimental data, hUK-66 represents a promising candidate for an antibody-based therapy against antibiotic resistant MRSA.}, language = {en} } @phdthesis{Gesser2011, author = {Gesser, Martin Benedikt Ambros}, title = {Optimierung eines Balanced Lethal Systems f{\"u}r Salmonella Typhi Ty21a}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-55740}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2011}, abstract = {Das Projekt „Balanced Lethal System in Salmonella typhi Ty21a" der AG Bakterielle Tumortherapie des MSZ zielt auf die Entwicklung eines Vakzinstammes, der in der humanen Krebstherapie zum Einsatz kommen soll. Ziel dieser Arbeit war das zuvor etablierte Balanced Lethal System in Salmonella typhi Ty21a zu optimieren.}, subject = {Immuntherapie}, language = {de} } @article{GoncharovaRuzhenkovaPetrovetal.2016, author = {Goncharova, Elena P. and Ruzhenkova, Julia S. and Petrov, Ivan S. and Shchelkunov, Sergey N. and Zenkova, Marina A.}, title = {Oncolytic virus efficiency inhibited growth of tumour cells with multiple drug resistant phenotype in vivo and in vitro}, series = {Journal of Translational Medicine}, volume = {14}, journal = {Journal of Translational Medicine}, number = {241}, doi = {10.1186/s12967-016-1002-x}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-165714}, pages = {1-14}, year = {2016}, abstract = {Background Tumour resistance to a wide range of drugs (multiple drug resistant, MDR) acquired after intensive chemotherapy is considered to be the main obstacle of the curative treatment of cancer patients. Recent work has shown that oncolytic viruses demonstrated prominent potential for effective treatment of diverse cancers. Here, we evaluated whether genetically modified vaccinia virus (LIVP-GFP) may be effective in treatment of cancers displaying MDR phenotype. Methods LIVP-GFP replication, transgene expression and cytopathic effects were analysed in human cervical carcinomas KB-3-1 (MDR-), KB-8-5 (MDR+) and in murine melanoma B-16 (MDR-), murine lymphosarcomas RLS and RLS-40 (MDR+). To investigate the efficacy of this therapy in vivo, we treated immunocompetent mice bearing murine lymphosarcoma RLS-40 (MDR+) (6- to 8-week-old female CBA mice; n = 10/group) or melanoma B-16 (MDR-) (6- to 8-week-old female C57Bl mice; n = 6/group) with LIVP-GFP (5 × 107 PFU of virus in 0.1 mL of IMDM immediately and 4 days after tumour implantation). Results We demonstrated that LIVP-GFP replication was effective in human cervical carcinomas KB-3-1 (MDR-) and KB-8-5 (MDR+) and in murine melanoma B-16 (MDR-), whereas active viral production was not detected in murine lymphosarcomas RLS and RLS-40 (MDR+). Additionally, it was found that in tumour models in immunocompetent mice under the optimized regimen intratumoural injections of LIVP-GFP significantly inhibited melanoma B16 (33 \% of mice were with complete response after 90 days) and RLS-40 tumour growth (fourfold increase in tumour doubling time) as well as metastasis. Conclusion The anti-tumour activity of LIVP-GFP is a result of direct oncolysis of tumour cells in case of melanoma B-16 because the virus effectively replicates and destroys these cells, and virus-mediated activation of the host immune system followed by immunologically mediated destruction of of tumour cells in case of lymphosarcoma RLS-40. Thus, the recombinant vaccinia virus LIVP-GFP is able to inhibit the growth of malignant cells with the MDR phenotype and tumour metastasis when administered in the early stages of tumour development.}, language = {en} } @article{WeberGlutschGeissingeretal.2020, author = {Weber, J. and Glutsch, V. and Geissinger, E. and Haug, L. and Lock, J.F. and Schneider, F. and Kneitz, H. and Goebeler, M. and Schilling, B. and Gesierich, A.}, title = {Neoadjuvant immunotherapy with combined ipilimumab and nivolumab in patients with melanoma with primary or in transit disease}, series = {British Journal of Dermatology}, volume = {183}, journal = {British Journal of Dermatology}, number = {3}, doi = {10.1111/bjd.18739}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-213520}, pages = {559-563}, year = {2020}, abstract = {The introduction of new therapeutic agents has revolutionized the treatment of metastatic melanoma. The approval of adjuvant anti-programmed death-1 monotherapy with nivolumab or pembrolizumab, and dabrafenib plus trametinib has recently set a new landmark in the treatment of stage III melanoma. Now, clinical trials have shown that immune checkpoint blockade can be performed in a neoadjuvant setting, an approach established as a standard therapeutic approach for other tumour entities such as breast cancer. Recent studies suggest that a pathological response achieved by neoadjuvant immunotherapy is associated with long-term tumour control and that short neoadjuvant application of checkpoint inhibitors may be superior to adjuvant therapy. Most recently, neoadjuvant ipilimumab plus nivolumab in stage III melanoma was reported. With two courses of dose-optimized ipilimumab (1 mg kg-1) combined with nivolumab (3 mg kg-1), pathological responses were observed in 77\% of patients, while only 20\% of patients experienced grade 3 or 4 adverse events. However, the neoadjuvant trials employing combined immune checkpoint blockade conducted so far have excluded patients with in transit metastases, a common finding in stage III melanoma. Here we report four patients with in transit metastases or an advanced primary tumour who have been treated with neoadjuvant ipilimumab plus nivolumab according to the OpACIN-neo trial scheme (arm B). All patients achieved radiological disease control and a pathological response. None of the patients has relapsed so far.}, language = {en} } @phdthesis{Steinfatt2023, author = {Steinfatt, Tim Alexander}, title = {Modulation of regulatory T cells for the immunotherapy of inflammatory diseases and cancer}, doi = {10.25972/OPUS-19260}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-192600}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Regulatory T cells (Tregs) are the masters of immune regulation controlling inflammation and tolerance, tissue repair and homeostasis. Multiple immunological diseases result from altered Treg frequencies and Treg dysfunction. We hypothesized that augmenting Treg function and numbers would prevent inflammatory disease whereas inhibiting or depleting Tregs would improve cancer immunotherapy. In the first part of this thesis, we explored whether in vivo activation and expansion of Tregs would impair acute graft-versus-host disease (aGvHD). In this inflammatory disease, Tregs are highly pathophysiological relevant and their adoptive transfer proved beneficial on disease outcome in preclinical models and clinical studies. IL-2 has been recognized as a key cytokine for Treg function. Yet, attempts in translating Treg expansion via IL-2 have remained challenging, due to IL-2s extremely broad action on other cell types including effector T cells, NK cells, eosinophils and vascular leakage syndrome, and importantly, due to poor pharmacokinetics in vivo. We addressed the latter issue using an IL-2-IgG-fusion protein (irrIgG-IL-2) with improved serum retention and demonstrated profound Treg expansion in vivo in FoxP3-luciferase reporter mice. Further, we augmented Treg numbers and function via the selective-TNF based agonists of TNFR2 (STAR2). Subsequently, we tested a next-generation TNFR2 agonist, termed NewSTAR, which proved even more effective. TNFR2 stimulation augmented Treg numbers and function and was as good as or even superior to the IL-2 strategy. Finally, in a mouse model of aGvHD we proved the clinical relevance of Treg expansion and activation with irrIgG-IL-2, STAR2 and NewSTAR. Notably, the TNFR2 stimulating constructs were outstanding as we observed not the IL-2 prototypic effects on other cell populations and no severe side effects. In the second part of this thesis, we explored Tregs in pancreatic ductal adenocarcinoma (PDAC) and developed targeting strategies. Among several tumor entities in which Tregs impact survival, preclinical and clinical data demonstrated their negative role on PDAC. In our studies we employed the orthotopic syngeneic Panc02 model in immunocompetent mice. Based on flow cytometric analysis of the tumor microenvironment we propose TIGIT and TNFRSF members as novel therapeutic targets. Surprisingly, we found that blocking TNFR2 did not interfere with intratumoral Treg accumulation. However, we decreased the highly abundant intratumoral Tregs when we disrupted the tumor extracellular matrix. In PDAC, Treg manipulation alone did not lead to tumor regression and we propose that an additional immune boost may be necessary for efficient tumor immune surveillance and cancer clearance. This contrasts with aGvHD, in which Treg manipulation alone was sufficient to improve disease outcome. Conclusively, we demonstrated the enormous medical benefit of Treg manipulation. Our promising data obtained with our newly developed powerful tools highlight the potential to translate our findings into clinical practice to therapeutically target human Tregs in patients. With novel TNFR2 agonists (STAR2, NewSTAR) we augmented Treg numbers and function as (or even more) effectively than with IL-2, without causing adverse side effects. Importantly, exogenous in vivo Treg expansion protected mice from aGvHD. For the therapy of PDAC, we identified novel targets on Tregs, notably TIGIT and members of the TNFRSF. We demonstrated that altering the extracellular tumor matrix can efficiently disrupt the Treg abundance in tumors. These novel targeting strategies appear as attractive new treatment options and they may benefit patients suffering from inflammatory disease and cancer in the future.}, language = {en} } @article{LoehrMolcanyiPoggenborgetal.2013, author = {L{\"o}hr, Mario and Molcanyi, Marek and Poggenborg, J{\"o}rg and Spuentrup, Elmar and Runge, Matthias and R{\"o}hn, Gabriele and H{\"a}rtig, Wolfgang and Hescheler, J{\"u}rgen and Hampl, J{\"u}rgen A.}, title = {Intracerebral Administration of Heat-Inactivated Staphylococcus Epidermidis Enhances Oncolysis and Prolongs Survival in a 9L Orthotopic Gliosarcoma Model}, series = {Cellular Physiology and Biochemistry}, journal = {Cellular Physiology and Biochemistry}, doi = {10.1159/000350081}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-96754}, year = {2013}, abstract = {Background/Aims: The association between postoperative infection and prolonged survival in high-grade glioma is still a matter of debate. Previously we demonstrated that the intracerebral (i.c.) injection of heat-inactivated staphylococcal epitopes (HISE) resulted in a well-defined infux of immunocompetent cells across the blood-brain barrier. The present study investigated the potential antitumoral effect of HISE-immunostimulation in an experimental glioma model. Methods: Wistar rats were intracerebrally implanted with 9L gliosarcoma cells (n=6), 9L cells mixed with HISE (n=12), or phosphate buffered saline (n=4). Tumor growth was measured by serial magnetic resonance imaging (MRI). After death due to the tumor burden, the brains were histopathologically assessed for inflammation and oncolysis. A toxicity assay was performed to quantify potential impairment of HISE on tumor cell growth in vitro. Results: Animals treated by HISE showed a significant increase in average survival and even complete regression of an already established mass in one case. Na{\"i}ve 9L gliosarcomas failed to recruit significant numbers of systemic immune cells. In contrast, concomitant intracerebral HISE inoculation lead to a oncolysis and a distinct peri- and intratumoral infiltration of macrophages, CD8 and CD4 co-expressing T-lymphocytes in two thirds of the tumor-bearing animals. The toxicity screening showed HISE-mediated oncolysis to be ineffective ex vivo. Conclusion: This study describes a novel approach for combatting malignant glioma using inactivated staphylococci as potent immunomodulators. Our results provide an outline for investigating the strategic potential of bacteria as emerging future therapeutics.}, language = {en} }