Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ulrike Gnad-Vogt is active.

Publication


Featured researches published by Ulrike Gnad-Vogt.


European Journal of Cancer | 2013

A phase I dose-escalation study of the immunocytokine EMD 521873 (Selectikine) in patients with advanced solid tumours

Silke Gillessen; Ulrike Gnad-Vogt; E. Gallerani; Joachim Beck; Cristiana Sessa; Aurelius Omlin; Maria R. Mattiacci; Bernd Liedert; Daniel Kramer; Julien Laurent; Daniel E. Speiser; Roger Stupp

BACKGROUND EMD 521873 (Selectikine), an immunocytokine comprising a DNA-targeting antibody, aimed at tumour necrosis, fused with a genetically modified interleukin-2 (IL-2) moiety, was investigated in this first-in-human phase I study. METHODS Patients had metastatic or locally advanced solid tumours failing previous standard therapy. Selectikine was administered as a 1-hour intravenous infusion on 3 consecutive days, every 3 weeks. A subgroup of patients also received 300 mg/m(2) cyclophosphamide on day 1 of each cycle. Escalating doses of Selectikine were investigated with the primary objective of determining the maximum tolerated dose (MTD). RESULTS Thirty-nine patients were treated with Selectikine alone at dose levels from 0.075 to 0.9 mg/kg, and nine were treated at doses of 0.45 and 0.6 mg/kg in combination with cyclophosphamide. A dose-dependent linear increase of peak serum concentrations and area under curve was found. The dose-limiting toxicity was grade 3 skin rash at the 0.9 mg/kg dose-level; the MTD was 0.6 mg/kg. Rash and flu-like symptoms were the most frequent side-effects. No severe cardiovascular side-effects (hypotension or vascular leak) were observed. At all dose-levels, transient increases in total lymphocyte, eosinophil and monocyte counts were recorded. No objective tumour responses, but long periods of disease stabilisation were observed. Transient and non-neutralising Selectikine antibodies were detected in 69% of patients. CONCLUSIONS The MTD of Selectikine with or without cyclophosphamide administered under this schedule was 0.6 mg/kg. The recommended phase II dose was 0.45-0.6 mg/kg. Selectikine had a favourable safety profile and induced biological effects typical for IL-2.


Journal of Translational Medicine | 2013

T-cell activation by treatment of cancer patients with EMD 521873 (Selectikine), an IL-2/anti-DNA fusion protein

Julien Laurent; Cédric Touvrey; Silke Gillessen; Magali Joffraud; Manuela Vicari; Caroline Bertrand; Stefano Ongarello; Bernd Liedert; E. Gallerani; Joachim Beck; Aurelius Omlin; Cristiana Sessa; Sonia Quaratino; Roger Stupp; Ulrike Gnad-Vogt; Daniel E. Speiser

BackgroundEMD 521873 (Selectikine or NHS-IL2LT) is a fusion protein consisting of modified human IL-2 which binds specifically to the high-affinity IL-2 receptor, and an antibody specific for both single- and double-stranded DNA, designed to facilitate the enrichment of IL-2 in tumor tissue.MethodsAn extensive analysis of pharmacodynamic (PD) markers associated with target modulation was assessed during a first-in-human phase I dose-escalation trial of Selectikine.ResultsThirty-nine patients with metastatic or locally advanced tumors refractory to standard treatments were treated with increasing doses of Selectikine, and nine further patients received additional cyclophosphamide. PD analysis, assessed during the first two treatment cycles, revealed strong activation of both CD4+ and CD8+ T-cells and only weak NK cell activation. No dose response was observed. As expected, Treg cells responded actively to Selectikine but remained at lower frequency than effector CD4+ T-cells. Interestingly, patient survival correlated positively with both high lymphocyte counts and low levels of activated CD8+ T-cells at baseline, the latter of which was associated with enhanced T-cell responses to the treatment.ConclusionsThe results confirm the selectivity of Selectikine with predominant T-cell and low NK cell activation, supporting follow-up studies assessing the clinical efficacy of Selectikine for cancer patients.


The Lancet | 2017

Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial

Martin Alberer; Ulrike Gnad-Vogt; Henoch S. Hong; Keyvan Tadjalli Mehr; Linus Backert; Greg Finak; Raphael Gottardo; Mihai Alexandru Bica; Aurelio Garofano; Sven Koch; Mariola Fotin-Mleczek; Ingmar Hoerr; Ralf Clemens; Frank von Sonnenburg

BACKGROUND Vaccines based on mRNA coding for antigens have been shown to be safe and immunogenic in preclinical models. We aimed to report results of the first-in-human proof-of-concept clinical trial in healthy adults of a prophylactic mRNA-based vaccine encoding rabies virus glycoprotein (CV7201). METHODS We did an open-label, uncontrolled, prospective, phase 1 clinical trial at one centre in Munich, Germany. Healthy male and female volunteers (aged 18-40 years) with no history of rabies vaccination were sequentially enrolled. They received three doses of CV7201 intradermally or intramuscularly by needle-syringe or one of three needle-free devices. Escalating doses were given to subsequent cohorts, and one cohort received a booster dose after 1 year. The primary endpoint was safety and tolerability. The secondary endpoint was to determine the lowest dose of CV7201 to elicit rabies virus neutralising titres equal to or greater than the WHO-specified protective antibody titre of 0·5 IU/mL. The study is continuing for long-term safety and immunogenicity follow-up. This trial is registered with ClinicalTrials.gov, number NCT02241135. FINDINGS Between Oct 21, 2013, and Jan 11, 2016, we enrolled and vaccinated 101 participants with 306 doses of mRNA (80-640 μg) by needle-syringe (18 intradermally and 24 intramuscularly) or needle-free devices (46 intradermally and 13 intramuscularly). In the 7 days post vaccination, 60 (94%) of 64 intradermally vaccinated participants and 36 (97%) of 37 intramuscularly vaccinated participants reported solicited injection site reactions, and 50 (78%) of 64 intradermally vaccinated participants and 29 (78%) of 37 intramuscularly vaccinated participants reported solicited systemic adverse events, including ten grade 3 events. One unexpected, possibly related, serious adverse reaction that occurred 7 days after a 640 μg intramuscular dose resolved without sequelae. mRNA vaccination by needle-free intradermal or intramuscular device injection induced virus neutralising antibody titres of 0·5 IU/mL or more across dose levels and schedules in 32 (71%) of 45 participants given 80 μg or 160 μg CV7201 doses intradermally and six (46%) of 13 participants given 200 μg or 400 μg CV7201 doses intramuscularly. 1 year later, eight (57%) of 14 participants boosted with an 80 μg needle-free intradermal dose of CV7201 achieved titres of 0·5 IU/mL or more. Conversely, intradermal or intramuscular needle-syringe injection was ineffective, with only one participant (who received 320 μg intradermally) showing a detectable immune response. INTERPRETATION This first-ever demonstration in human beings shows that a prophylactic mRNA-based candidate vaccine can induce boostable functional antibodies against a viral antigen when administered with a needle-free device, although not when injected by a needle-syringe. The vaccine was generally safe with a reasonable tolerability profile. FUNDING CureVac AG.


Cancer immunology research | 2016

Short Peptide Vaccine Induces CD4+ T Helper Cells in Patients with Different Solid Cancers

Stefanie Gross; Volker Lennerz; E. Gallerani; Nicolas Mach; Steffen Böhm; Dagmar Hess; Lotta von Boehmer; Alexander Knuth; Adrian F. Ochsenbein; Ulrike Gnad-Vogt; Ulf Forssmann; Thomas Woelfel; Eckhart Kaempgen

The possibility that short peptide vaccines induce antitumor CD4+ T-cell responses has been widely ignored. Peripheral blood from vaccinated patients revealed that short peptides often activate specific T helper cells, facilitating a strong combined CD4+ and CD8+ T-cell response. Previous cancer vaccination trials often aimed to activate CD8+ cytotoxic T-cell (CTL) responses with short (8–10mer) peptides and targeted CD4+ helper T cells (TH) with HLA class II–binding longer peptides (12–16 mer) that were derived from tumor antigens. Accordingly, a study of immunomonitoring focused on the detection of CTL responses to the short, and TH responses to the long, peptides. The possible induction of concurrent TH responses to short peptides was widely neglected. In a recent phase I vaccination trial, 53 patients with different solid cancers were vaccinated with EMD640744, a cocktail of five survivin-derived short (9- or 10-mer) peptides in Montanide ISA 51VG. We monitored 49 patients and found strong CD8+ T-cell responses in 63% of the patients. In addition, we unexpectedly found CD4+ TH cell responses against at least two of the five short peptides in 61% (23/38) of the patients analyzed. The two peptides were recognized by HLA-DP4– and HLA-DR–restricted TH1 cells. Some short peptide–reactive (sp)CD4 T cells showed high functional avidity. Here, we show that a short peptide vaccine is able to activate a specific CD4+ T-cell repertoire in many patients, facilitating a strong combined CD4+/CD8+ T-cell response. Cancer Immunol Res; 4(1); 18–25. ©2015 AACR.


Cancer immunology research | 2016

Abstract A043: Discovery to first-in-man studies of a multi-peptide-based hepatocellular carcinoma vaccine adjuvanted with CV8102 (RNAdjuvant®): HEPAVAC

Andrea Mayer-Mokler; Roberto S. Accolla; Yuk Ting Ma; Regina Heidenreich; Francesco Izzo; Alfred Koenigsrainer; Markus Loeffler; Christian Flohr; Phillip Mueller; Sarah Kutscher; Hans-Georg Rammensee; Bruno Sangro; Sven Francque; Danila Valmori; Toni Weinschenk; Carsten Reinhardt; Ulrike Gnad-Vogt; Harpreet Singh; Luigi Buonaguro

Hepatocellular (HCC)/normal adjacent tissue matched samples have been collected for HLA immunopeptidome analysis. 17 HCC samples from HLA-A*02+ patients and 15 samples from HLA-A*24+ patients have been analysed by mass spectrometry (LC-MS/MS). RNA-expression profiles have been established for 12 HCC samples. HLA-presentation/expression of peptides on primary HCC samples (as well as mRNA expression) were compared to normal tissue samples from relevant organs (including heart, brain, lung, kidney, liver, nerve, skin etc.) present in the Immatics9 database. A total of 16 peptides have been selected and confirmed for immunogenicity for the HepaVac vaccine and are currently synthesized according to GMP standard. Of these, 7 are restricted to HLA-A*02; 5 to HLA-A*24 and 4 to HLA class II. Formulation development studies have been undertaken leading to a suitable and stable pharmaceutical form. An analytical method was developed which allows the characterization of each individual peptide within the HepaVac vaccine (IMA970A). At present, preclinical studies assessing the combination of the immunological RNA-based adjuvant (RNAdjuvant®) with the peptide-based HepaVac vaccine IMA970 are conducted. A single-arm, first-in-man trial entitled HepaVac-101 is designed to investigate in patients with very early, early and intermediate stage of HCC the off-the-shelf multi-peptide-based HCC vaccine (IMA970) plus the CV8102 adjuvant (RNAdjuvant®) following a single pre-vaccination infusion of low-dose cyclophosphamide acting as an immunomodulator. The study drugs are applied without concomitant anti-tumor therapy with the intention to reduce risk of tumor recurrence/progression in patients who have received all indicated standard treatments. The primary endpoints are safety, tolerability, and immunogenicity. Secondary/exploratory endpoints are additional immunological parameters in blood (e.g. regulatory T-cells, myeloid-derived suppressor cells, impact of the standard therapy on the natural immune response), infiltrating T-lymphocytes in tumor tissue, biomarkers in blood and tissue, disease-free survival/progression-free survival and overall survival. Once safety of this vaccination approach has been determined in the first 10-20 patients the addition of a checkpoint inhibitor will be considered. Suitable patients enrolled in Tuebingen are invited to participate in a trial extension investigating an actively personalized vaccine (APVAC) plus CV8102. The HepaVac project started in September 2013 and is supported by the European Commission9s 7th Framework Program under the Grant Agreement Nr. 602893 (www.hepavac.eu). The clinical trial HepaVac-101 will be conduct in 6 centers located in 5 European countries, i.e. Italy (Naples and Varese), Germany (Tubingen), UK (Birmingham), Spain (Pamplona) and Belgium (Antwerpen). Citation Format: Andrea Mayer-Mokler, Roberto Accolla, Yuk T. Ma, Regina Heidenreich, Francesco Izzo, Alfred Koenigsrainer, Markus Loeffler, Christian Flohr, Phillip Mueller, Sarah Kutscher, Hans-Georg Rammensee, Bruno Sangro, Sven Francque, Danila Valmori, Toni Weinschenk, Carsten Reinhardt, Ulrike Gnad-Vogt, Harpreet Singh, Luigi Buonaguro. Discovery to first-in-man studies of a multi-peptide-based hepatocellular carcinoma vaccine adjuvanted with CV8102 (RNAdjuvant®): HEPAVAC [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr A043.


Cancer Research | 2011

Abstract 5516: Therapeutic vaccination with the survivin-derived multi-epitope vaccine EMD640744 in patients with advanced solid tumors

Volker Lennerz; Stefanie Gross; E. Gallerani; Cristiana Sessa; Nicolas Mach; Steffen Boehm; Dagmar Hess; Lotta von Boehmer; Alexander Knuth; Adrian F. Ochsenbein; Ulrike Gnad-Vogt; Juergen Zieschang; Ulf Forssmann; Thomas Wölfel; Eckhart Kaempgen

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Background: Survivin is a promising tumor-associated antigen for cancer immunotherapy that fulfills two major criteria for this purpose: (1) tumor cells depend on the actions of survivin (inhibition of apoptosis, unrestricted proliferation and angiogenesis); and (2) the protein has demonstrated immunogenicity in patients with different cancers. Here we report results of a first-in-man Phase I study with EMD640744, a cocktail of survivin-derived partially modified HLA class I-restricted peptides in Montanide® ISA 51 VG. Methods: This multicenter, open-label, parallel group, randomized study compared the immunologic efficacy, safety, tolerability and clinical activity of three dosages of EMD640744 (30, 100, or 300 µg) in patients with different types of metastatic or locally advanced solid tumors who were positive for at least one relevant HLA antigen (A1, A2, A3, A24, B7). Patients received weekly s.c. injections of EMD640744 for 8 weeks followed by injections every 4 weeks until tumor progression. For the assessment of the primary endpoint, PBMC samples were prepared at baseline and after 4, 8, 12, 16, and 17 weeks. Peptide-specific T cell responses were analyzed by IFN-γ ELISpot. As a secondary analysis, peptide/HLA-multimer staining was performed. Frequencies of CD8+ T cells specific for patient-relevant single HLA-restricted peptides and EMD640744 were determined ex vivo and after short-term in vitro presensitization with EMD640744. Native homologues of modified vaccination peptides were also included in the analysis. Results: Of 66 patients screened, 53 were treated, and a majority was eligible for response analysis: 38 patients were analyzed by ELISpot and 42 by multimers, with positive ex vivo responses observed in 7 (18%) and 14 (33%) patients, respectively. Combining ex vivo data with results after in vitro stimulation, T cell responses were detected in 14 (37%) and 31 (74%) patients by ELISpot and multimer analysis, respectively. Multimer staining revealed a de novo induction of T cells against survivin peptides in at least 16 patients (38%) whereas pre-existent responses were seen in only 4 patients (10%). T cell responses were detected in all dose groups with similar frequencies. Five out of 10 HLA-A2+ patients reacting against the modified A2-binding peptide in ELISpot assays showed responses against its native homologue. The best tumor response according to RECIST was stable disease in 28% of patients. The most frequent treatment-related adverse events (AEs) were Grade 1-2 local injection site reactions and 2 patients experienced treatment-related Grade 3 AEs (granuloma at injection site and thrombosis). Conclusion: Vaccination with EMD640744 was safe and well tolerated and elicited de novo T cell responses against survivin peptides, demonstrating immunological efficacy of EMD640744 in cancer patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5516. doi:10.1158/1538-7445.AM2011-5516


Cancer Immunology, Immunotherapy | 2014

Immunologic response to the survivin-derived multi-epitope vaccine EMD640744 in patients with advanced solid tumors.

Volker Lennerz; Stefanie Gross; E. Gallerani; Cristiana Sessa; Nicolas Mach; Steffen Boehm; Dagmar Hess; Lotta von Boehmer; Alexander Knuth; Adrian F. Ochsenbein; Ulrike Gnad-Vogt; Juergen Zieschang; Ulf Forssmann; Thomas Woelfel; Eckhart Kaempgen


Archive | 2016

COMPOSITION AND VACCINE FOR TREATING PROSTATE CANCER

Karl-Josef Kallen; Mariola Fotin-Mleczek; Ulrike Gnad-Vogt; Thomas Lander


Archive | 2016

COMPOSITION AND VACCINE FOR TREATING LUNG CANCER

Karl-Josef Kallen; Mariola Fotin-Mleczek; Ulrike Gnad-Vogt


Journal of Clinical Oncology | 2011

First-in-human trial focusing on the immunologic effects of the survivin-derived multiepitope vaccine EMD640744.

Stefanie Gross; Volker Lennerz; E. Gallerani; Cristiana Sessa; Nicolas Mach; Steffen Boehm; Dagmar Hess; L. von Boehmer; Alexander Knuth; Adrian F. Ochsenbein; Ulrike Gnad-Vogt; Juergen Zieschang; Ulf Forssmann; Thomas Woelfel; Eckhart Kaempgen

Collaboration


Dive into the Ulrike Gnad-Vogt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge