TY - JOUR AB - BACKGROUND: Obese subjects undergoing weight loss often fear the Yoyo dieting effect, which involves regaining or even surpassing their initial weight. To date, our understanding of such long-term obesity and weight cycling effects is still limited and often based on only short-term murine weight gain and loss studies. This study aimed to investigate the long-term impacts of weight cycling on glycemic control and metabolic health, focusing on adipose tissue, liver, and hypothalamus. METHODS: Chow-fed mice and mice subjected to prolonged high-fat diet (HFD) consumption for 20 weeks, followed by 24 weeks of dietary interventions to either induce weight gain, weight loss, or weight cycling were monitored for perturbations in feeding efficiency and glucose homeostasis. Post-mortem analyses included qPCR, Western Blotting, biochemical and microscopical assessments for hepatic steatosis and insulin resistance, hypothalamic and adipose tissue inflammation, and circulating lipid, leptin and IL-6 levels. RESULTS: Weight cycling led to hyperphagia and rapid weight regain, matching the weights of mice continuously on HFD. Despite weight loss, adipose tissue inflammation persisted with elevated pro-inflammatory markers, macrophage infiltration, and impaired Glut4 expression. HFD-induced dysregulation in hypothalamic expression of orexigenic peptides and synaptic plasticity markers persisted also after weight normalization suggesting long-lasting neural alterations. Weight-cycled mice exhibited higher circulating IL-6 and leptin levels, increased hepatic lipid storage, and dysregulated glucose metabolism compared to those with consistent diets, indicating worsened metabolic effects by Yoyo dieting. CONCLUSION: In sum, our study highlights significant metabolic risks associated with weight cycling, particularly following prolonged obesity. Persistent adipose tissue inflammation, perturbed neural peptide and plasticity markers and impaired glucose tolerance emphasize the need for effective and sustainable weight loss strategies to mitigate the adverse outcomes of weight regain and improve long-term metabolic health. AU - Bernecker, M. AU - Lin, A. AU - Feuchtinger, A. AU - Molenaar, A. AU - Schriever, S.C. AU - Pfluger, P.T. C1 - 72943 C2 - 56885 CY - Campus, 4 Crinan St, London N1 9xw, England TI - Weight cycling exacerbates glucose intolerance and hepatic triglyceride storage in mice with a history of chronic high fat diet exposure. JO - J. Transl. Med. VL - 23 IS - 1 PB - Bmc PY - 2025 SN - 1479-5876 ER - TY - JOUR AB - BACKGROUND: Long-chain acyl-carnitines (ACs) are potential arrhythmogenic metabolites. Their role in atrial fibrillation (AF) remains incompletely understood. Using a systems medicine approach, we assessed the contribution of C18:1AC to AF by analysing its in vitro effects on cardiac electrophysiology and metabolism, and translated our findings into the human setting. METHODS AND RESULTS: Human iPSC-derived engineered heart tissue was exposed to C18:1AC. A biphasic effect on contractile force was observed: short exposure enhanced contractile force, but elicited spontaneous contractions and impaired Ca2+ handling. Continuous exposure provoked an impairment of contractile force. In human atrial mitochondria from AF individuals, C18:1AC inhibited respiration. In a population-based cohort as well as a cohort of patients, high C18:1AC serum concentrations were associated with the incidence and prevalence of AF. CONCLUSION: Our data provide evidence for an arrhythmogenic potential of the metabolite C18:1AC. The metabolite interferes with mitochondrial metabolism, thereby contributing to contractile dysfunction and shows predictive potential as novel circulating biomarker for risk of AF. AU - Krause, J.* AU - Nickel, A.* AU - Madsen, A.* AU - Aitken-Buck, H.M.* AU - Stoter, A.M.S.* AU - Schrapers, J.* AU - Ojeda, F.* AU - Geiger, K.* AU - Kern, M.* AU - Kohlhaas, M.* AU - Bertero, E.* AU - Hofmockel, P.* AU - Hübner, F.* AU - Assum, I. AU - Heinig, M. AU - Müller, C.* AU - Hansen, A.* AU - Krause, T.* AU - Park, D.D.* AU - Just, S.* AU - Aïssi, D.* AU - Börnigen, D.* AU - Lindner, D.* AU - Friedrich, N.* AU - Alhussini, K.* AU - Bening, C.* AU - Schnabel, R.B.* AU - Karakas, M.* AU - Iacoviello, L.* AU - Salomaa, V.* AU - Linneberg, A.* AU - Tunstall-Pedoe, H.* AU - Kuulasmaa, K.* AU - Kirchhof, P.* AU - Blankenberg, S.* AU - Christ, T.* AU - Eschenhagen, T.* AU - Lamberts, R.R.* AU - Maack, C.* AU - Stenzig, J.* AU - Zeller, T.* C1 - 67959 C2 - 54437 CY - Campus, 4 Crinan St, London N1 9xw, England TI - An arrhythmogenic metabolite in atrial fibrillation. JO - J. Transl. Med. VL - 21 IS - 1 PB - Bmc PY - 2023 SN - 1479-5876 ER - TY - JOUR AB - BACKGROUND: The aim of this study was to investigate the association between inflammatory markers and 28-day mortality in patients with ST-elevation myocardial infarction (STEMI). METHODS: In 398 STEMI patients recorded between 2009 and 2013 by the population-based Myocardial Infarction Registry Augsburg, 92 protein biomarkers were measured in admission arterial blood samples using the OLINK inflammatory panel. In multivariable-adjusted logistic regression models, the association between each marker and 28-day mortality was investigated. The values of the biomarkers most significantly associated with mortality were standardized and summarized to obtain a prediction score for 28-day mortality. The predictive ability of this biomarker score was compared to the established GRACE score using ROC analysis. Finally, a combined total score was generated by adding the standardized biomarker score to the standardized GRACE score. RESULTS: The markers IL-6, IL-8, IL-10, FGF-21, FGF-23, ST1A1, MCP-1, 4E-BP1, and CST5 were most significantly associated with 28-day mortality, each with FDR-adjusted (false discovery rate adjusted) p-values of < 0.01 in the multivariable logistic regression model. In a ROC analysis, the biomarker score and the GRACE score showed comparable predictive ability for 28-day mortality (biomarker score AUC: 0.7859 [CI: 0.6735-0.89], GRACE score AUC: 0.7961 [CI: 0.6965-0.8802]). By combining the biomarker score and the Grace score, the predictive ability improved with an AUC of 0.8305 [CI: 0.7269-0.9187]. A continuous Net Reclassification Improvement (cNRI) of 0.566 (CI: 0.192-0.94, p-value: 0.003) and an Integrated Discrimination Improvement (IDI) of 0.083 ((CI: 0.016-0.149, p-value: 0.015) confirmed the superiority of the combined score over the GARCE score. CONCLUSIONS: Inflammatory biomarkers may play a significant role in the pathophysiology of acute myocardial infarction (AMI) and AMI-related mortality and might be a promising starting point for personalized medicine, which aims to provide each patient with tailored therapy. AU - Schmitz, T.* AU - Harmel, E.* AU - Heier, M. AU - Peters, A. AU - Linseisen, J.* AU - Meisinger, C.* C1 - 66338 C2 - 52793 TI - Inflammatory plasma proteins predict short-term mortality in patients with an acute myocardial infarction. JO - J. Transl. Med. VL - 20 IS - 1 PY - 2022 SN - 1479-5876 ER - TY - JOUR AB - BackgroundIn pre-clinical research, systematic reviews have the potential to mitigate translational challenges by facilitating understanding of how pre-clinical studies can inform future clinical research. Yet their conduct is encumbered by heterogeneity in the outcomes measured and reported, and those outcomes may not always relate to the most clinically important outcomes. We aimed to systematically review outcomes measured and reported in pre-clinical in vivo studies of pharmacological interventions to treat high blood glucose in mouse models of type 2 diabetes.MethodsA systematic review of pre-clinical in vivo studies of pharmacological interventions aimed at addressing elevated blood glucose in mouse models of type 2 diabetes was completed. Studies were screened for eligibility and outcomes extracted from the included studies. The outcomes were recorded verbatim and classified into outcome domains using an existing outcome taxonomy. Outcomes were also compared to those identified in a systematic review of registered phase 3/4 clinical trials for glucose lowering interventions in people with type 2 diabetes.ResultsReview of 280 included studies identified 532 unique outcomes across 19 domains. No single outcome, or domain, was measured in all studies and only 132 (21%) had also been measured in registered phase 3/4 clinical trials. A core outcome set, representing the minimum that should be measured and reported, developed for type 2 diabetes effectiveness clinical trials includes 18 core outcomes, of these 12 (71%) outcomes were measured and reported in one or more of the included pre-clinical studies.ConclusionsThere is heterogeneity of outcomes reported in pre-clinical research. Harmonisation of outcomes across the research pathway using a core outcome set may facilitate interpretation, evidence synthesis and translational success, and may contribute to the refinement of the use of animals in research.Systematic review registration: The study was prospectively registered on the PROSPERO Database, registration number CRD42018106831 AU - Harman, N.L.* AU - Sanz-Moreno, A. AU - Papoutsopoulou, S.* AU - Lloyd, K.A.* AU - Ameen-Ali, K.E.* AU - MacLeod, M.* AU - Williamson, P.R.* C1 - 60718 C2 - 49487 CY - Campus, 4 Crinan St, London N1 9xw, England TI - Can harmonisation of outcomes bridge the translation gap for pre-clinical research? A systematic review of outcomes measured in mouse models of type 2 diabetes. JO - J. Transl. Med. VL - 18 IS - 1 PB - Bmc PY - 2020 SN - 1479-5876 ER - TY - JOUR AB - BackgroundPools of overlapping synthetic peptides are routinely used for ex vivo monitoring of antigen-specific T-cell responses. However, it is rather unlikely that these peptides match those resulting from naturally processed antigens. T-activated proteins have been described as immunogenic and more natural stimulants, since they have to pass through antigen processing and comprise activation of all clinically relevant effector cell populations.MethodsWe performed comparative analysis of numbers and cytokine expression pattern of CD4 and CD8 T cells after stimulation with recombinant, urea-formulated T-activated EBV-BZLF1, -EBNA3A, and HCMV-IE1, and -pp65 proteins or corresponding overlapping peptide pools. Freshly isolated and cryopreserved PBMC of 30 EBV- and 19 HCMV-seropositive and seven EBV- and HCMV-seronegative subjects were stimulated ex vivo and analysed for IFN-gamma, TNF and IL-2 production by flow cytometry-based intracellular cytokine staining.ResultsT-activated proteins showed a high specificity of 100% (EBV-BZLF1, HCMV-IE1, and -pp65) and 86% (EBV-EBNA3A), and a high T-cell stimulatory capacity of 73-95% and 67-95% using freshly isolated and cryopreserved PBMC, respectively. The overall CD4 T-cell response rates in both cohorts were comparable after stimulation with either T-activated protein or peptide pools with the exception of lower numbers of CD8 T cells detected after stimulation with T-activated EBV-EBNA3A- (p=0.038) and HCMV-pp65- (p=0.0006). Overall, the number of detectable antigen-specific T cells varied strongly between individuals. Cytokine expression patterns in response to T-activated protein and peptide pool-based stimulation were similar for CD4, but significantly different for CD8 T-cell responses.ConclusionEBV and HCMV-derived T-activated proteins represent innovative, highly specific recall antigens suitable for use in immunological endpoint assays to evaluate success or failure in immunotherapy clinical trials (e.g. to assess the risk of EBV and/or HCMV reactivation after allogenic hematopoietic stem cell transplantation). T-activated proteins could be of particular importance, if an impaired antigen processing (e.g. in a post-transplant setting) must be taken into account. AU - Körber, N. AU - Behrends, U. AU - Protzer, U. AU - Bauer, T. C1 - 59424 C2 - 48810 CY - Campus, 4 Crinan St, London N1 9xw, England TI - Evaluation of T-activated proteins as recall antigens to monitor Epstein-Barr virus and human cytomegalovirus-specific T cells in a clinical trial setting. JO - J. Transl. Med. VL - 18 IS - 1 PB - Bmc PY - 2020 SN - 1479-5876 ER - TY - JOUR AB - Background Breast cancer is the most common malignancy in female patients worldwide. Because of its heterogeneity in terms of prognosis and therapeutic response, biomarkers with the potential to predict survival or assist in making treatment decisions in breast cancer patients are essential for an individualised therapy. Epigenetic alterations in the genome of the cancer cells, such as changes in DNA methylation pattern, could be a novel marker with an important role in the initiation and progression of breast cancer. Method DNA methylation and RNA-seq datasets from The Cancer Genome Atlas (TCGA) were analysed using the Least Absolute Shrinkage and Selection Operator (LASSO) Cox model. Applying gene ontology (GO) and single sample gene set enrichment analysis (ssGSEA) an epigenetic signature associated with the survival of breast cancer patients was constructed that yields the best discrimination between tumour and normal breast tissue. A predictive nomogram was built for the optimal strategy to distinguish between high- and low-risk cases. Results The combination of mRNA-expression and of DNA methylation datasets yielded a 13-gene epigenetic signature that identified subset of breast cancer patients with low overall survival. This high-risk group of tumor cases was marked by upregulation of known cancer-related pathways (e.g. mTOR signalling). Subgroup analysis indicated that this epigenetic signature could distinguish high and low-risk patients also in different molecular or histological tumour subtypes (by Her2-, EGFR- or ER expression or different tumour grades). Using Gene Expression Omnibus (GEO) the 13-gene signature was confirmed in four external breast cancer cohorts. Conclusion An epigenetic signature was discovered that effectively stratifies breast cancer patients into low and high-risk groups. Since its efficiency appears independent of other known classifiers (such as staging, histology, metastasis status, receptor status), it has a high potential to further improve likely individualised therapy in breast cancer. AU - Bao, X. AU - Anastasov, N. AU - Wang, Y.* AU - Rosemann, M. C1 - 57390 C2 - 47733 CY - Campus, 4 Crinan St, London N1 9xw, England TI - A novel epigenetic signature for overall survival prediction in patients with breast cancer. JO - J. Transl. Med. VL - 17 IS - 1 PB - Bmc PY - 2019 SN - 1479-5876 ER - TY - JOUR AB - BackgroundEarly diagnosis is critical to reduce the mortality caused by nasopharyngeal carcinoma (NPC). MicroRNAs (miRNAs) are dysregulated and play important roles in carcinogenesis. Therefore, this study aimed to identify diagnostically relevant circulating miRNA signatures in patients with NPC.MethodsTotal RNA was extracted from whole blood samples obtained from 120 patients with NPC, 30 patients with head-neck tumors (HNT), and 30 healthy subjects (HSs), and examined by using a custom microarray. The expression levels of four miRNAs identified by using the microarray were validated with quantitative real-time reverse transcription polymerase chain reaction. The 120 patients with NPC and 30 HSs were randomly assigned to training group-1 and validation group-1, respectively. By using significance analysis of microarray (SAM), the specific miRNA expression profiles in whole blood from patients with NPC are obtained. By using lasso regression and adaptive boosting, a diagnostic signature was identified in training group-1, and its accuracy was verified in validation group-1. By using the same methods, another signature to distinguish patients with NPC from those with HNT and HSs was identified in training group-2 and confirmed in validation group-2.ResultsThere were 117 differentially expressed miRNAs (upregulated and downregulated fold change 1.5) between the patients with NPC and HSs, among which an 8-miRNA signature was identified with 96.43% sensitivity and 100% specificity [area under the curve (AUC)=0.995] to diagnose NPC in training group-1 and 86.11% sensitivity and 88.89% specificity (AUC=0.941) in validation group-1. Compared with traditional Epstein-Barr virus (EBV) seromarkers, this signature was more specific for NPC. Furthermore, a 16-miRNA signature to differentiate NPC from HNT and HS (HNT-HS) was established from 164 differentially expressed miRNAs, which diagnosed NPC and HNT-HS with 100% accuracy (AUC=1.000) in training group-2 and 87.04% (AUC=0.924) in validation group-2.ConclusionsThe present study identified two miRNA signatures for the highly accurate diagnosis and differential diagnosis of patients with NPC from HSs and patients with HNT. The identified miRNAs might represent novel serological biomarkers and potential therapeutic targets for NPC. AU - Wen, W.* AU - Mai, S.J.* AU - Lin, H.X.* AU - Zhang, M.Y.* AU - Huang, J. AU - Hua, X.* AU - Lin, C.* AU - Long, Z.Q.* AU - Lu, Z.J.* AU - Sun, X.Q.* AU - Liu, S.-L.* AU - Yang, Q.* AU - Zhu, Q.* AU - Wang, H.Y.* AU - Guo, L.* C1 - 56212 C2 - 46904 CY - Campus, 4 Crinan St, London N1 9xw, England TI - Identification of two microRNA signatures in whole blood as novel biomarkers for diagnosis of nasopharyngeal carcinoma. JO - J. Transl. Med. VL - 17 IS - 1 PB - Bmc PY - 2019 SN - 1479-5876 ER - TY - JOUR AB - Background: A major complication after allogeneic hematopoietic stem cell transplantation (aSCT) is the reactivation of herpesviruses such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV). Both viruses cause significant mortality and compromise quality of life after aSCT. Preventive transfer of virus-specific T cells can suppress reactivation by re-establishing functional antiviral immune responses in immunocompromised hosts. Methods: We have developed a good manufacturing practice protocol to generate CMV/EBV-peptide-stimulated T cells from leukapheresis products of G-CSF mobilized and non-mobilized donors. Our procedure selectively expands virus-specific CD8+ und CD4+ T cells over 9 days using a generic pool of 34 CMV and EBV peptides that represent well-defined dominant T-cell epitopes with various HLA restrictions. For HLA class I, this set of peptides covers at least 80% of the European population. Results: CMV/EBV-specific T cells were successfully expanded from leukapheresis material of both G-CSF mobilized and non-mobilized donors. The protocol allows administration shortly after stem cell transplantation (d30+), storage over liquid nitrogen for iterated applications, and protection of the stem cell donor by avoiding a second leukapheresis. Conclusion: Our protocol allows for rapid and cost-efficient production of T cells for early transfusion after aSCT as a preventive approach. It is currently evaluated in a phase I/IIa clinical trial. AU - Gary, R.* AU - Aigner, M.* AU - Moi, S.* AU - Schaffer, S.* AU - Gottmann, A.* AU - Maas, S.* AU - Zimmermann, R.* AU - Zingsem, J.* AU - Strobel, J.* AU - Mackensen, A.* AU - Mautner, J. AU - Moosmann, A. AU - Gerbitz, A.* C1 - 53503 C2 - 44701 TI - Clinical-grade generation of peptide-stimulated CMV/EBV-specific T cells from G-CSF mobilized stem cell grafts. JO - J. Transl. Med. VL - 16 IS - 1 PY - 2018 SN - 1479-5876 ER - TY - JOUR AB - Following publication of the original article [1], the authors reported that for one of the authors, Stephanie E. Combs, the middle name was accidentally omitted. They also reported that for two of the authors, Daniel Habermehl and Stephanie E. Combs, two affiliations were accidentally omitted. In this Correction the incorrect and correct author name are shown and the two omitted affiliations are listed. AU - Slotta-Huspenina, J.* AU - Drecoll, E.* AU - Feith, M.* AU - Habermehl, D. AU - Combs, S.E. AU - Weichert, W.* AU - Bettstetter, M.* AU - Becker, K.* AU - Langer, R.* C1 - 53805 C2 - 45032 CY - 233 Spring St, New York, Ny 10013 Usa TI - Correction: MicroRNA expression profiling for the prediction of resistance to neoadjuvant radiochemotherapy in squamous cell carcinoma of the esophagus [J Transl Med., 16, (2018) (109)] DOI: 10.1186/s12967-018-1492-9. JO - J. Transl. Med. VL - 16 IS - 1 PB - Springer PY - 2018 SN - 1479-5876 ER - TY - JOUR AB - Background: Diabetes testing using saliva, rather than blood and urine, could facilitate diabetes screening in public spaces. We previously identified 1,5-anhydro-d-glucitol (1,5-AG) in saliva as a diabetes biomarker. The Glycomark (TM) assay kit is FDA approved for 1,5-AG measurement in blood. Here we evaluated its applicability for 1,5-AG quantification in saliva. Methods: Using pooled saliva samples, we validated Glycomark (TM) assay use with a RX Daytona(+) clinical chemistry analyser. We then used this set-up to analyse 82 paired blood and saliva samples from a diabetes case-control study, for which broad mass spectrometry-based characterization of the blood and saliva metabolome was also available. Osmolality was measured to account for potential variability in saliva samples. Results: The technical variability of the read-outs for the pooled saliva samples (CV = 2.05 %) was comparable to that obtained with manufacturer-provided blood surrogate quality controls (CV = 1.38-1.8 %). We found a high correlation between Glycomark assay and mass spectrometry measurements of serum 1,5-AG (r(2) = 0.902), showing reproducibility of the non-targeted metabolomics results. The significant correlation between the osmolality measurements performed at two independent platforms with the time interval of 2 years (r(2) = 0.887), also indicates the sample integrity. The assay read-out for saliva was not correlated with the mass spectrometry-based 1,5-AG saliva measurements. Comparison with the full saliva metabolome revealed a high correlation of the saliva assay read-outs with galactose. Conclusions: Glycomark (TM) assay read-outs for saliva were stable and replicable. However, the signal was dominated by galactose, which is biochemically similar to 1,5-AG and absent in blood. Adapting the 1,5-AG kit for saliva analysis will require enzymatic depletion of galactose. This should be feasible, since the assay already includes a similar step for glucose depletion from blood samples. AU - Halama, A.* AU - Kulinski, M.* AU - Kader, S.A.* AU - Satheesh, N.J.* AU - Abou-Samra, A.B.* AU - Suhre, K. AU - Mohammad, R.M.* C1 - 48766 C2 - 41407 CY - London TI - Measurement of 1,5-anhydroglucitol in blood and saliva: From non-targeted metabolomics to biochemical assay. JO - J. Transl. Med. VL - 14 IS - 1 PB - Biomed Central Ltd PY - 2016 SN - 1479-5876 ER - TY - JOUR AB - BACKGROUND: The FluoroSpot assay, an advancement of the ELISpot assay, enables simultaneous measurement of different analytes secreted at a single-cell level. This allows parallel detection of several cytokines secreted by immune cells upon antigen recognition. Easier standardization, higher sensitivity and reduced labour intensity render FluoroSpot assays an interesting alternative to flow-cytometry based assays for analysis of clinical samples. While the use of immunoassays to study immunological primary and secondary endpoints becomes increasingly attractive, assays used require pre-trial validation. Here we describe the assay validation (precision, specificity and linearity) of a FluoroSpot immunological endpoint assay detecting Interferon γ (IFNγ) and Interleukin 2 (IL2) for use in clinical trial immune monitoring. METHODS: We validated an IFNγ/IL2 FluoroSpot assay to determine Epstein-Barr virus (EBV)-specific cellular immune responses (IFNγ, IL2 and double positive IFNγ + IL2 responses), using overlapping peptide pools corresponding to EBV-proteins BZLF1 and EBNA3A. Assay validation was performed using cryopreserved PBMC of 16 EBV-seropositive and 6 EBV-seronegative donors. Precision was assessed by (i) testing 16 donors using three replicates per assay (intra-assay precision/repeatability) (ii) using two plates in parallel (intermediate precision/plate-to-plate variability) and (iii) by performing the assays on three different days (inter-assay precision/reproducibility). In addition, we determined specificity, linearity and quantification limits of the assay. Further we tested precision across the two assay systems, IFNγ/IL2 FluoroSpot and the corresponding enzymatic single cytokine ELISpot. RESULTS: The validation revealed: (1) a high intra-assay precision (coefficient of variation (CV) 9.96, 8.85 and 13.05 %), intermediate precision (CV 6.48, 10.20 and 12.97 %) and reproducibility (CV 20.81 %, 12,75 % and 12.07 %) depending on the analyte and antigen used; (2) a specificity of 100 %; (3) a linearity with R (2) values from 0.93 to 0.99 depending on the analyte. The testing of the precision across the two assay systems, adduced a concordance correlation coefficient p c  = 0.99 for IFNγ responses and p c  = 0.93 for IL2 responses, indicating a large agreement between both assay methods. CONCLUSIONS: The validated primary endpoint assay, an EBV peptide pool specific IFNγ/IL2 FluoroSpot assay was found to be suitable for the detection of EBV-specific immune responses subject to the requirement of standardized assay procedure and data analysis. AU - Körber, N. AU - Behrends, U. AU - Hapfelmeier, A.* AU - Protzer, U. AU - Bauer, T. C1 - 48816 C2 - 41432 CY - London TI - Validation of an IFNγ/IL2 FluoroSpot assay for clinical trial monitoring. JO - J. Transl. Med. VL - 14 IS - 1 PB - Biomed Central Ltd PY - 2016 SN - 1479-5876 ER - TY - JOUR AB - Background: In this era of precision medicine, the deep and comprehensive characterization of tumor phenotypes will lead to therapeutic strategies beyond classical factors such as primary sites or anatomical staging. Recently, "-omics" approached have enlightened our knowledge of tumor biology. Such approaches have been extensively implemented in order to provide biomarkers for monitoring of the disease as well as to improve readouts of therapeutic impact. The application of metabolomics to the study of cancer is especially beneficial, since it reflects the biochemical consequences of many cancer type-specific pathophysiological processes. Here, we characterize metabolic profiles of colon and ovarian cancer cell lines to provide broader insight into differentiating metabolic processes for prospective drug development and clinical screening. Methods: We applied non-targeted metabolomics-based mass spectroscopy combined with ultrahigh-performance liquid chromatography and gas chromatography for the metabolic phenotyping of four cancer cell lines: two from colon cancer (HCT15, HCT116) and two from ovarian cancer (OVCAR3, SKOV3). We used the MetaP server for statistical data analysis. Results: A total of 225 metabolites were detected in all four cell lines; 67 of these molecules significantly discriminated colon cancer from ovarian cancer cells. Metabolic signatures revealed in our study suggest elevated tricarboxylic acid cycle and lipid metabolism in ovarian cancer cell lines, as well as increased beta-oxidation and urea cycle metabolism in colon cancer cell lines. Conclusions: Our study provides a panel of distinct metabolic fingerprints between colon and ovarian cancer cell lines. These may serve as potential drug targets, and now can be evaluated further in primary cells, biofluids, and tissue samples for biomarker purposes. AU - Halama, A.* AU - Guerrouahen, B.S.* AU - Pasquier, J.* AU - Diboun, I.* AU - Karoly, E.D.* AU - Suhre, K. AU - Rafii, A.* C1 - 46434 C2 - 37583 CY - London TI - Metabolic signatures differentiate ovarian from colon cancer cell lines. JO - J. Transl. Med. VL - 13 IS - 1 PB - Biomed Central Ltd PY - 2015 SN - 1479-5876 ER - TY - JOUR AB - Background: Individualized Medicine aims at providing optimal treatment for an individual patient at a given time based on his specific genetic and molecular characteristics. This requires excellent clinical stratification of patients as well as the availability of genomic data and biomarkers as prerequisites for the development of novel diagnostic tools and therapeutic strategies. The University Medicine Greifswald, Germany, has launched the " Greifswald Approach to Individualized Medicine" (GANI_MED) project to address major challenges of Individualized Medicine. Herein, we describe the implementation of the scientific and clinical infrastructure that allows future translation of findings relevant to Individualized Medicine into clinical practice.Methods/design: Clinical patient cohorts (N > 5,000) with an emphasis on metabolic and cardiovascular diseases are being established following a standardized protocol for the assessment of medical history, laboratory biomarkers, and the collection of various biosamples for bio-banking purposes. A multi-omics based biomarker assessment including genome-wide genotyping, transcriptome, metabolome, and proteome analyses complements the multi-level approach of GANI_MED. Comparisons with the general background population as characterized by our Study of Health in Pomerania (SHIP) are performed. A central data management structure has been implemented to capture and integrate all relevant clinical data for research purposes. Ethical research projects on informed consent procedures, reporting of incidental findings, and economic evaluations were launched in parallel. AU - Grabe, H.J.* AU - Assel, H.* AU - Bahls, T.A.* AU - Dörr, M.* AU - Endlich, K.H.* AU - Endlich, N.* AU - Erdmann, P.* AU - Ewert, R.* AU - Felix, S.B.* AU - Fiene, B.* AU - Fischer, T.* AU - Fleßa, S.* AU - Friedrich, N.* AU - Gadebusch-Bondio, M.* AU - Salazar, M.G.* AU - Hammer, E.* AU - Haring, R.* AU - Havemann, C.* AU - Hecker, M.* AU - Hoffmann, W.J.* AU - Holtfreter, B.* AU - Kacprowski, T.* AU - Klein, K.* AU - Kocher, T.M.* AU - Kock, H.* AU - Krafczyk, J.* AU - Kuhn, J.* AU - Langanke, M.* AU - Lendeckel, U.* AU - Lerch, M.M.* AU - Lieb, W.* AU - Lorbeer, R.* AU - Mayerle, J.* AU - Meissner, K.* AU - Meyer zu Schwabedissen, H.E.* AU - Nauck, M.A.* AU - Ott, K.* AU - Rathmann, W.G.* AU - Rettig, R.* AU - Richardt, C.* AU - Saljé, K.* AU - Schminke, U.* AU - Schulz, A.* AU - Schwab, M.* AU - Siegmund, W.* AU - Stracke, S.* AU - Suhre, K. AU - Ueffing, M. AU - Ungerer, S.* AU - Völker, U.* AU - Völzke, H.* AU - Wallaschofski, H.* AU - Werner, V.* AU - Zygmunt, M.T.* AU - Kroemer, H.K.* C1 - 31624 C2 - 34667 TI - Cohort profile: Greifswald approach to individualized medicine (GANI_MED). JO - J. Transl. Med. VL - 12 IS - 1 PY - 2014 SN - 1479-5876 ER - TY - JOUR AB - High-throughput screening techniques that analyze the metabolic endpoints of biological processes can identify the contributions of genetic predisposition and environmental factors to the development of common diseases. Studies applying controlled physiological challenges can reveal dysregulation in metabolic responses that may be predictive for or associated with these diseases. However, large-scale epidemiological studies with well controlled physiological challenge conditions, such as extended fasting periods and defined food intake, pose logistic challenges. Culturally and religiously motivated behavioral patterns of life style changes provide a natural setting that can be used to enroll a large number of study volunteers. Here we report a proof of principle study conducted within a Muslim community, showing that a metabolomics study during the Holy Month of Ramadan can provide a unique opportunity to explore the pre-prandial and postprandial response of human metabolism to nutritional challenges. Up to five blood samples were obtained from eleven healthy male volunteers, taken directly before and two hours after consumption of a controlled meal in the evening on days 7 and 26 of Ramadan, and after an over-night fast several weeks after Ramadan. The observed increases in glucose, insulin and lactate levels at the postprandial time point confirm the expected physiological response to food intake. Targeted metabolomics further revealed significant and physiologically plausible responses to food intake by an increase in bile acid and amino acid levels and a decrease in long-chain acyl-carnitine and polyamine levels. A decrease in the concentrations of a number of phospholipids between samples taken on days 7 and 26 of Ramadan shows that the long-term response to extended fasting may differ from the response to short-term fasting. The present study design is scalable to larger populations and may be extended to the study of the metabolic response in defined patient groups such as individuals with type 2 diabetes. AU - Mathew, S.* AU - Krug, S.* AU - Skurk, T.* AU - Halama, A. AU - Stank, A.* AU - Artati, A. AU - Prehn, C. AU - Malek, J.A.* AU - Kastenmüller, G. AU - Römisch-Margl, W. AU - Adamski, J. AU - Hauner, H.* AU - Suhre, K. C1 - 31566 C2 - 34602 CY - London TI - Metabolomics of Ramadan fasting: An opportunity for the controlled study of physiological responses to food intake. JO - J. Transl. Med. VL - 12 IS - 1 PB - Biomed Central Ltd PY - 2014 SN - 1479-5876 ER - TY - JOUR AB - Background: Multiple myeloma is characterized by clonal expansion of B cells producing monoclonal immunoglobulins or fragments thereof, which can be detected in the serum and/or urine and are ideal target antigens for patient-specific immunotherapies. Methods: Using phage particles as immunological carriers, we employed a novel chemically linked idiotype vaccine in a clinical phase I/II trial including 15 patients with advanced multiple myeloma. Vaccines composed of purified paraproteins linked to phage were manufactured successfully for each patient. Patients received six intradermal immunizations with phage idiotype vaccines in three different dose groups. Results: Phage idiotype was well tolerated by all study participants. A subset of patients (80% in the middle dose group) displayed a clinical response indicated by decrease or stabilization of paraprotein levels. Patients exhibiting a clinical response to phage vaccines also raised idiotype-specific immunoglobulins. Induction of a cellular immune response was demonstrated by a cytotoxicity assay and delayed type hypersensitivity tests. Conclusion: We present a simple, time-and cost-efficient phage idiotype vaccination strategy, which represents a safe and feasible patient-specific therapy for patients with advanced multiple myeloma and produced promising anti-tumor activity in a subset of patients. AU - Roehnisch, T.* AU - Then, C.* AU - Nagel, W. AU - Blumenthal, C.* AU - Braciak, T.* AU - Donzeau, M.* AU - Boehm, T.* AU - Flaig, M.* AU - Bourquin, C.* AU - Oduncu, F.* C1 - 31699 C2 - 34671 CY - London TI - Phage idiotype vaccination: First phase I/II clinical trial in patients with multiple myeloma. JO - J. Transl. Med. VL - 12 IS - 1 PB - Biomed Central Ltd PY - 2014 SN - 1479-5876 ER - TY - JOUR AB - BACKGROUND: Patients with B cell malignancies refractory to allogeneic stem cell transplantation (SCT) can be treated by subsequent immunotherapy with donor lymphocyte infusions (DLI). But unlike myeloid leukemia, B cell leukemia and lymphoma are less sensitive to allogeneic adoptive immunotherapy. Moreover, the beneficial graft-versus-lymphoma (GVL) effect may be associated with moderate to severe graft-versus-host disease (GVHD). Thus, novel therapeutic approaches augmenting the anti-tumor efficacy of DLI and dissociating the GVL effect from GVHD are needed. The anti-CD20 x anti-CD3 trifunctional bispecific antibody (trAb) FBTA05 may improve the targeting of tumor cells by redirecting immune allogeneic effector cells while reducing the risk of undesirable reactivity against normal host cells. Hence, FBTA05 may maximize GVL effects by simultaneously decreasing the incidence and severity of GVHD. METHODS/DESIGN: Based on this underlying treatment concept and on promising data taken from preclinical results and a small pilot study, an open-label, non-randomized, uncontrolled, dose-escalating phase I/II-study is conducted to evaluate safety and preliminary efficacy of the investigational antibody FBTA05 in combination with DLI for patients suffering from rituximab- and/or alemtuzumab-refractory, CD20-positive low- or high-grade lymphoma after allogeneic SCT. During the first trial phase with emphasis on dose escalation a maximum of 24 patients distributed into 4 cohorts will be enrolled. For the evaluation of preliminary efficacy data a maximum of 12 patients (6 patients with low-grade lymphoma and/or Chronic Lymphocytic Leukemia (CLL) / 6 patients with high-grade or aggressive lymphoma) will attend the second phase of this clinical trial. DISCUSSION: Promising data (e.g. induction of cellular immunity; GVL predominance over GVHD; achievement of partial or complete responses; prolongation of time-to-progression) obtained from this phase I/II trial would represent the first milestone in the clinical evaluation of a novel immunotherapeutic concept for treatment-resistant low- and high-grade lymphoma and NHL patients in relapse. TRIAL REGISTRATION: NCT01138579. AU - Buhmann, R. AU - Stanglmaier, M.* AU - Hess J.* AU - Lindhofer, H.* AU - Peschel, C.* AU - Kolb, H.-J. C1 - 26110 C2 - 32074 TI - Immunotherapy with FBTA05 (Bi20), a trifunctional bispecific anti-CD3 x anti-CD20 antibody and donor lymphocyte infusion (DLI) in relapsed or refractory B-cell lymphoma after allogeneic stem cell transplantation: Study protocol of an investigator-driven, open-label, non-randomized, uncontrolled, dose-escalating phase I/II-trial. JO - J. Transl. Med. VL - 11 IS - 1 PB - BioMed Central PY - 2013 SN - 1479-5876 ER - TY - JOUR AB - Background: B cell malignancies are characterized by clonal expansion of B cells expressing tumor-specific idiotypes on their surface. These idiotypes are ideal target antigens for an individualized immunotherapy. However, previous idiotype vaccines mostly lacked efficiency due to a low immunogenicity of the idiotype. The objective of the present study was the determination of the feasibility, safety and immunogenicity of a novel chemically linked phage idiotype vaccine. Methods: In the murine B cell lymphoma 1 model, tumor idiotypes were chemically linked to phage particles used as immunological carriers. For comparison, the idiotype was genetically expressed on the major phage coat protein g8 or linked to keyhole limpet hemocynanin. After intradermal immunizations with idiotype vaccines, tolerability and humoral immune responses were assessed. Results: Feasibility and tolerability of the chemically linked phage idiotype vaccine was demonstrated. Vaccination with B cell lymphoma 1 idiotype expressing phage resulted in a significant survival benefit in the murine B cell lymphoma 1 protection model (60.2 +/- 23.8 days vs. 41.8 +/- 1.6 days and 39.8 +/- 3.8 days after vaccination with wild type phage or phosphate buffered saline, respectively). Superior immunogenicity of the chemically linked phage idiotype vaccine compared to the genetically engineered phage idiotype and keyhole limpet hemocynanin-coupled idiotype vaccine was demonstrated by significantly higher B cell lymphoma 1 idiotype-specific IgG levels after vaccination with chemically linked phage idiotype. Conclusion: We present a novel, simple, time-and cost-efficient phage idiotype vaccination strategy, which represents a safe and feasible therapy and may produce a superior immune response compared to previously employed idiotype vaccination strategies. AU - Roehnisch, T.* AU - Then, C.* AU - Nagel, W. AU - Blumenthal, C.* AU - Braciak, T.* AU - Donzeau, M.* AU - Bohm, T.* AU - Bourquin, C.* AU - Oduncu, F.* C1 - 29265 C2 - 33252 TI - Chemically linked phage idiotype vaccination in the murine B cell lymphoma 1 model. JO - J. Transl. Med. VL - 11 IS - 1 PB - Biomed Central Ltd PY - 2013 SN - 1479-5876 ER - TY - JOUR AB - BACKGROUND: Trifunctional bispecific antibodies (trAb) are a special class of bispecific molecules recruiting and activating T cells and accessory immune cells simultaneously at the targeted tumor. The new trAb Ektomab that targets the melanoma-associated ganglioside antigen GD2 and the signaling molecule human CD3 (hCD3) on T cells demonstrated potent T-cell activation and tumor cell destruction in vitro. However, the relatively low affinity for the GD2 antigen raised the question of its therapeutic capability. To further evaluate its efficacy in vivo it was necessary to establish a mouse model. METHODS: We generated the surrogate trAb Surek, which possesses the identical anti-GD2 binding arm as Ektomab, but targets mouse CD3 (mCD3) instead of hCD3, and evaluated its chemical and functional quality as a therapeutic antibody homologue. The therapeutic and immunizing potential of Surek was investigated using B78-D14, a B16 melanoma transfected with GD2 and GD3 synthases and showing strong GD2 surface expression. The induction of tumor-associated and autoreactive antibodies was evaluated. RESULTS: Despite its low affinity of approximately 107 M-1 for GD2, Surek exerted efficient tumor cell destruction in vitro at an EC50 of 70ng/ml [0.47nM]. Furthermore, Surek showed strong therapeutic efficacy in a dose-dependent manner and is superior to the parental GD2 mono-specific antibody, while the use of a control trAb with irrelevant target specificity had no effect. The therapeutic activity of Surek was strictly dependent on CD4+ and CD8+ T cells, and cured mice developed a long-term memory response against a second challenge even with GD2-negative B16 melanoma cells. Moreover, tumor protection was associated with humoral immune responses dominated by IgG2a and IgG3 tumor-reactive antibodies indicating a Th1-biased immune response. Autoreactive antibodies against the GD2 target antigen were not induced. CONCLUSION: Our data suggest that Surek revealed strong tumor elimination and anti-tumor immunization capabilities. The results warrant further clinical development of the human therapeutic equivalent antibody Ektomab. AU - Ruf, P.* AU - Schäfer, B.* AU - Eissler, N. AU - Mocikat, R. AU - Hess J.* AU - Plöscher, M.* AU - Wosch, S.* AU - Suckstorff, I.* AU - Zehetmeier, C.* AU - Lindhofer, H.* C1 - 11402 C2 - 30650 TI - Ganglioside GD2-specific trifunctional surrogate antibody Surek demonstrates therapeutic activity in a mouse melanoma model. JO - J. Transl. Med. VL - 10 IS - 1 PB - Biomed Central PY - 2012 SN - 1479-5876 ER - TY - JOUR AB - BACKGROUND: To date very few systems have been described for preclinical investigations of human cellular therapeutics in vivo. However, the ability to carry out comparisons of new cellular vaccines in vivo would be of substantial interest for design of clinical studies. Here we describe a humanized mouse model to assess the efficacy of various human dendritic cell (DC) preparations. Two reconstitution regimes of NOD/scid IL2Rg(null) (NSG) mice with adult human peripheral blood mononuclear cells (PBMC) were evaluated for engraftment using 4-week and 9-week schedules. This led to selection of a simple and rapid protocol for engraftment and vaccine evaluation that encompassed 4 weeks. METHODS: NSG recipients of human PBMC were engrafted over 14 days and then vaccinated twice with autologous DC via intravenous injection. Three DC vaccine formulations were compared that varied generation time in vitro (3 days versus 7 days) and signals for maturation (with or without Toll-like receptor (TLR)3 and TLR7/8 agonists) using MART-1 as a surrogate antigen, by electroporating mature DC with in vitro transcribed RNA encoding full length protein. After two weekly vaccinations, the splenocyte populations containing human lymphocytes were recovered 7 days later and assessed for MART-1-specific immune responses using MHC-multimer-binding assays and functional assessment of specific killing of melanoma tumor cell lines. RESULTS: Human monocyte-derived DC generated in vitro in 3 days induced better MART-1-specific immune responses in the autologous donor T cells present in the humanized NSG mice. Moreover, consistent with our in vitro observations, vaccination using mature DC activated with TLR3 and TLR7/8 agonists resulted in enhanced immune responses in vivo. These findings led to a ranking of the DC vaccine effects in vivo that reflected the hierarchy previously found for these mature DC variations in vitro. CONCLUSIONS: This humanized mouse model system enables comparisons among different DC vaccine types to be rapidly assessed in vivo. In addition, ex vivo analyses of human CD3+ T cells recovered from the spleens of these mice are also possible, including studies on lymphocyte subsets, Th1/Th2 polarization, presence of regulatory T cells and the impact of DC vaccination on their functions. AU - Spranger, S. AU - Frankenberger, B. AU - Schendel, D.J. C1 - 7506 C2 - 29765 TI - NOD/scid IL-2Rgnull mice: A preclinical model system to evaluate human dendritic cell-based vaccine strategies in vivo. JO - J. Transl. Med. VL - 10 IS - 1 PB - BioMed Central PY - 2012 SN - 1479-5876 ER - TY - JOUR AB - Active dendritic cell (DC) immunization protocols are rapidly gaining interest as therapeutic options in patients with acute myeloid leukemia (AML). Here we present for the first time a GMP-compliant 3-day protocol for generation of monocyte-derived DCs using different synthetic Toll-like receptor (TLR) agonists in intensively pretreated patients with AML. Four different maturation cocktails were compared for their impact on cell recovery, phenotype, cytokine secretion, migration, and lymphocyte activation in 20 AML patients and 25 healthy controls. Maturation cocktails containing the TLR7/8 agonists R848 or CL075, with and without the addition of the TLR3 agonist poly(I:C), induced DCs that had a positive costimulatory profile, secreted high levels of IL-12(p70), showed chemotaxis to CCR7 ligands, had the ability to activate NK cells, and efficiently stimulated antigen-specific CD8+ T cells. Our results demonstrate that this approach translates into biologically improved DCs, not only in healthy controls but also in AML patients. This data supports the clinical application of TLR-matured DCs in patients with AML for activation of innate and adaptive immune responses. AU - Beck, B.* AU - Dörfel, D.* AU - Lichtenegger, F.S.* AU - Geiger, C. AU - Lindner, L.* AU - Merk, M.* AU - Schendel, D.J. AU - Subklewe, M.* C1 - 6612 C2 - 28975 TI - Effects of TLR agonists on maturation and function of 3-day dendritic cells from AML patients in complete remission. JO - J. Transl. Med. VL - 9 IS - 1 PB - BioMed Central PY - 2011 SN - 1479-5876 ER - TY - JOUR AB - Scientific discoveries that provide strong evidence of antitumor effects in preclinical models often encounter significant delays before being tested in patients with cancer. While some of these delays have a scientific basis, others do not. We need to do better. Innovative strategies need to move into early stage clinical trials as quickly as it is safe, and if successful, these therapies should efficiently obtain regulatory approval and widespread clinical application. In late 2009 and 2010 the Society for Immunotherapy of Cancer (SITC), convened an "Immunotherapy Summit" with representatives from immunotherapy organizations representing Europe, Japan, China and North America to discuss collaborations to improve development and delivery of cancer immunotherapy. One of the concepts raised by SITC and defined as critical by all parties was the need to identify hurdles that impede effective translation of cancer immunotherapy. With consensus on these hurdles, international working groups could be developed to make recommendations vetted by the participating organizations. These recommendations could then be considered by regulatory bodies, governmental and private funding agencies, pharmaceutical companies and academic institutions to facilitate changes necessary to accelerate clinical translation of novel immune-based cancer therapies. The critical hurdles identified by representatives of the collaborating organizations, now organized as the World Immunotherapy Council, are presented and discussed in this report. Some of the identified hurdles impede all investigators, others hinder investigators only in certain regions or institutions or are more relevant to specific types of immunotherapy or first-in-humans studies. Each of these hurdles can significantly delay clinical translation of promising advances in immunotherapy yet be overcome to improve outcomes of patients with cancer. AU - Fox, B.A.* AU - Schendel, D.J. AU - Butterfield, L.H.* AU - Aamdal, S. AU - Allison, J.P.* AU - Ascierto, P.A.* AU - Atkins, M.B.* AU - Bartunkova, J.* AU - Bergmann, L.* AU - Berinstein, N.* AU - Bonorino, C.C.* AU - Borden, E.* AU - Bramson, J.L.* AU - Britten, C.M.* AU - Cao, X.* AU - Carson, W.E.* AU - Chang, A.E.* AU - Characiejus, D.* AU - Choudhury, A.R.* AU - Coukos, G.* AU - de Gruijl, T.* AU - Dillman, R.O.* AU - Dolstra, H.* AU - Dranoff, G.* AU - Durrant, L.G.* AU - Finke, J.H.* AU - Galon, J.* AU - Gollob, J.A.* AU - Gouttefangeas, C.* AU - Grizzi, F.* AU - Guida, M.* AU - Håkansson, L.* AU - Hege, K.* AU - Herberman, R.B.* AU - Hodi, F.S.* AU - Hoos, A.* AU - Huber, C.* AU - Hwu, P.* AU - Imai, K.* AU - Jaffee, E.M.* AU - Janetzki, S.* AU - June, C.H.* AU - Kalinski, P.* AU - Kaufman, H.L.* AU - Kawakami, K.* AU - Kawakami, Y.* AU - Keilholtz, U.* AU - Khleif, S.N.* AU - Kiessling, R.* AU - Kotlan, B.* AU - Kroemer, G.* AU - Lapointe, R.* AU - Levitsky, H.I.* AU - Lotze, M.T.* AU - Maccalli, C.* AU - Maio, M.* AU - Marschner, J.P.* AU - Mastrangelo, M.J.* AU - Masucci, G.* AU - Melero, I.* AU - Nelief, C.* AU - Murphy, W.J.* AU - Nelson, B.* AU - Nicolini, A.* AU - Nishimura, MI.* AU - Odunsi, K.* AU - Ohashi, P.S.* AU - O'Donnell-Tormey, J.* AU - Old, L.J.* AU - Ottensmeier, C.* AU - Papamichail, M.* AU - Parmiani, G.* AU - Pawelec, G. AU - Proietti, E.* AU - Qin, S.* AU - Rees, R.* AU - Ribas, A.* AU - Ridolfi, R.* AU - Ritter, G.* AU - Rivoltini, L.* AU - Romero, P.J.* AU - Salem, M.L.* AU - Scheper, R.J.* AU - Seliger, B.* AU - Sharma, P.* AU - Shiku, H.* AU - Singh-Jasuja, H.* AU - Song, W.* AU - Straten, P.T.* AU - Tahara, H.* AU - Tian, Z.* AU - van Der Burg, S.H.* AU - von Hoegen, P.* AU - Wang, E.* AU - Welters, M.J.* AU - Winter, H.* AU - Withington, T.* AU - Wolchok, J.D.* AU - Xiao, W.* AU - Zitvogel, L.* AU - Zwierzina, H.* AU - Marincola, F.M.* AU - Gajewski, T.F.* AU - Wigginton, J.M.* AU - Disis, ML.* C1 - 7028 C2 - 29455 TI - Defining the critical hurdles in cancer immunotherapy. JO - J. Transl. Med. VL - 9 IS - 1 PB - BioMed Central PY - 2011 SN - 1479-5876 ER - TY - JOUR AB - BACKGROUND: Antigen-loaded dendritic cells (DC) are capable of priming naïve T cells and therefore represent an attractive adjuvant for vaccine development in anti-tumor immunotherapy. Numerous protocols have been described to date using different maturation cocktails and time periods for the induction of mature DC (mDC) in vitro. For clinical application, the use of mDC that can be generated in only three days saves on the costs of cytokines needed for large scale vaccine cell production and provides a method to produce cells within a standard work-week schedule in a GMP facility. METHODS: In this study, we addressed the properties of antigen uptake, processing and presentation by monocyte-derived DC prepared in three days (3d mDC) compared with conventional DC prepared in seven days (7d mDC), which represent the most common form of DC used for vaccines to date. RESULTS: Although they showed a reduced capacity for spontaneous antigen uptake, 3d mDC displayed higher capacity for stimulation of T cells after loading with an extended synthetic peptide that requires processing for MHC binding, indicating they were more efficient at antigen processing than 7d DC. We found, however, that 3d DC were less efficient at expressing protein after introduction of in vitro transcribed (ivt)RNA by electroporation, based on published procedures. This deficit was overcome by altering electroporation parameters, which led to improved protein expression and capacity for T cell stimulation using low amounts of ivtRNA. CONCLUSIONS: This new procedure allows 3d mDC to replace 7d mDC for use in DC-based vaccines that utilize long peptides, proteins or ivtRNA as sources of specific antigen. AU - Bürdek, M. AU - Spranger, S. AU - Wilde, S. AU - Frankenberger, B. AU - Schendel, D.J. AU - Geiger, C. C1 - 5653 C2 - 27732 TI - Three-day dendritic cells for vaccine development: Antigen uptake, processing and presentation. JO - J. Transl. Med. VL - 8 PB - BioMed Central Ltd. PY - 2010 SN - 1479-5876 ER - TY - JOUR AB - Background: Membrane-bound heat shock protein 70 (Hsp70) serves as a tumor-specific recognition structure for Hsp70-peptide (TKD) plus IL-2 activated NK cells. A phase I clinical trial has shown that repeated re-infusions of ex vivo TKD/IL-2-activated, autologous leukapheresis product is safe. This study investigated the maintenance of the cytolytic activity of NK cells against K562 cells and autologous tumor after 6 plus 3 infusions of TKD/IL-2-activated effector cells. Methods: A stable tumor cell line was generated from the resected anastomotic relapse of a patient with colon carcinoma (pT3, N2, M0, G2). Two months after surgery, the patient received the first monthly i.v. infusion of his ex vivo TKD/IL-2-activated peripheral blood mononuclear cells (PBMNC). After 6 infusions and a pause of 3 months, the patient received another 3 cell infusions. The phenotypic characteristics and activation status of tumor and effector cells were determined immediately before and at times after each infusion. Results: The NK cell ligands Hsp70, MICA/B, and ULBP-1,2,3 were expressed on the patient's anastomotic relapse. An increased density of activatory NK cell receptors following ex vivo stimulation correlated with an enhanced anti-tumoricidal activity. After 4 re-infusion cycles, the intrinsic cytolytic activity of non-stimulated PBMNC was significantly elevated and this heightened responsiveness persisted for up to 3 months after the last infusion. Another 2 re-stimulations with TKD/IL-2 restored the cytolytic activity after the therapeutic pause. Conclusion: In a patient with colon carcinoma, repeated infusions of ex vivo TKD/IL-2-activated PBMNC initiate an intrinsic NK cell-mediated cytolytic activity against autologous tumor cells. AU - Milani, V. AU - Stangl, S.* AU - Issels, R.D. AU - Gehrmann, M.* AU - Wagner, B.* AU - Hube, K.* AU - Mayr, D. AU - Hiddemann, W. AU - Molls, M.* AU - Multhoff, G. C1 - 1626 C2 - 26622 TI - Anti-tumor activity of patient-derived NK cells after cell-based immunotherapy - a case report. JO - J. Transl. Med. VL - 7 PB - Biomed Central Ltd PY - 2009 SN - 1479-5876 ER - TY - JOUR AB - The International Society for the Biological Therapy of Cancer (iSBTc) has initiated in collaboration with the United States Food and Drug Administration (FDA) a programmatic look at innovative avenues for the identification of relevant parameters to assist clinical and basic scientists who study the natural course of host/tumor interactions or their response to immune manipulation. The task force has two primary goals: 1) identify best practices of standardized and validated immune monitoring procedures and assays to promote inter-trial comparisons and 2) develop strategies for the identification of novel biomarkers that may enhance our understating of principles governing human cancer immune biology and, consequently, implement their clinical application. Two working groups were created that will report the developed best practices at an NCI/FDA/iSBTc sponsored workshop tied to the annual meeting of the iSBTc to be held in Washington DC in the Fall of 2009. This foreword provides an overview of the task force and invites feedback from readers that might be incorporated in the discussions and in the final document. AU - Butterfield, L.H.* AU - Disis, M.L.* AU - Fox, BA.* AU - Lee, PP.* AU - Khleif, SN.* AU - Thurin, M.* AU - Trinchieri, G.* AU - Wang, E.* AU - Wigginton, J.* AU - Chaussabel, D.* AU - Coukos, G.* AU - Dhodapkar, M.* AU - Håkansson, L.* AU - Janetzki, S.* AU - Kleen, T.O.* AU - Kirkwood, J.M.* AU - Maccalli, C.* AU - Maecker, H.* AU - Maio, M.* AU - Malyguine, A.* AU - Masucci, G.* AU - Palucka, A.K.* AU - Potter, D.M.* AU - Ribas, A.* AU - Rivoltini, L.* AU - Schendel, D.J.* AU - Seliger, B.* AU - Selvan, S.* AU - Slingluff Jr., C.L.* AU - Stroncek, D.F.* AU - Streicher, H.* AU - Wu, X.* AU - Zeskind, B.* AU - Zhao, Y.* AU - Zocca, M.B.* AU - Zwierzina, H.* AU - Marincola, F.M.* C1 - 1060 C2 - 25925 TI - A systematic approach to biomarker discovery; Preamble to "the iSBTc-FDA taskforce on immunotherapy biomarkers". JO - J. Transl. Med. VL - 6 PB - Biomed Central PY - 2008 SN - 1479-5876 ER - TY - JOUR AB - Dendritic cells (DC) pulsed with tumor-derived antigenic material have widely been used in antitumor vaccination protocols. However, the optimal strategy of DC loading has not yet been established. Our aim was to define requirements of optimal DC vaccines in terms of in vivo protection in a murine B-cell lymphoma model. METHODS: We compare various loading reagents including whole parental and modified tumor cells and a single tumor-specific antigen, namely the lymphoma idiotype (Id). Bone marrow-derived DC were pulsed in vitro and used for therapy of established A20 lymphomas. RESULTS: We show that a vaccine with superior antitumor efficacy can be generated when DC are loaded with whole modified tumor cells which provide both (i) antigenic polyvalency and (ii) receptor-mediated antigen internalization. Uptake of cellular material was greatly enhanced when the tumor cells used for DC pulsing were engineered to express an anti-Fc receptor immunoglobulin specificity. Upon transfer of these DC, established tumor burdens were eradicated in 50% of mice. By contrast, pulsing DC with unmodified lymphoma cells or with the lymphoma Id, even when it was endowed with the anti-Fc receptor binding arm, was far less effective. A specific humoral anti-Id response could be detected, particularly following delivery of Id protein-pulsed DC, but it was not predictive of tumor protection. Instead a T-cell response was pivotal for successful tumor protection. Interaction of the transferred DC with CD8+ T lymphocytes seemed to play a role for induction of the immune response but was dispensable when DC had received an additional maturation stimulus. CONCLUSION: Our analyses show that the advantages of specific antigen redirection and antigenic polyvalency can be combined to generate DC-based vaccines with superior antitumor efficacy. This mouse model may provide information for the standardization of DC-based vaccination protocols. AU - Adam, C. AU - Mysliwietz, J. AU - Mocikat, R. C1 - 3818 C2 - 24827 TI - Specific targeting of whole lymphoma cells to dendritic cells ex vivo provides a potent antitumor vaccine. JO - J. Transl. Med. VL - 5 PB - Biomed Central PY - 2007 SN - 1479-5876 ER - TY - JOUR AB - Given the considerable toxicity and modest benefit of adjuvant chemotherapy for non-small cell lung cancer (NSCLC), there is clearly a need for new treatment modalities in the adjuvant setting. Active specific immunotherapy may represent such an option. However, clinical responses have been rare so far. Manipulating the host by inducing lymphopenia before vaccination resulted in a magnification of the immune response in the preclinical setting. To evaluate feasibility and safety of an irradiated, autologous tumor cell vaccine given following induction of lymphopenia by chemotherapy and reinfusion of autologous peripheral blood mononuclear cells (PBMC), we are currently conducting a pilot-phase I clinical trial in patients with NSCLC following surgical resection. This paper reports on the first clinical experience and evidence of an immune response in patients suffering from NSCLC. METHODS: NSCLC patients stages I-IIIA are recruited. Vaccines are generated from their resected lung specimens. Patients undergo leukapheresis to harvest their PBMC prior to or following the surgical procedure. Furthermore, patients receive preparative chemotherapy (cyclophosphamide 350 mg/m2 and fludarabine 20 mg/m2 on 3 consecutive days) for induction of lymphopenia followed by reconstitution with their autologous PBMC. Vaccines are administered intradermally on day 1 following reconstitution and every two weeks for a total of up to five vaccinations. Granulocyte-macrophage-colony-stimulating-factor (GM-CSF) is given continuously (at a rate of 50 mug/24 h) at the site of vaccination via minipump for six consecutive days after each vaccination. RESULTS: To date, vaccines were successfully manufactured for 4 of 4 patients. The most common toxicities were local injection-site reactions and mild constitutional symptoms. Immune responses to chemotherapy, reconstitution and vaccination are measured by vaccine site and delayed type hypersensitivity (DTH) skin reactions. One patient developed positive DTH skin tests so far. Immunohistochemical assessment of punch biopsies taken at the local vaccine site reaction revealed a dense lymphocyte infiltrate. Further immunohistochemical differentiation showed that CD1a+ cells had been attracted to the vaccine site as well as predominantly CD4+ lymphocytes. The 3-day combination chemotherapy consisting of cyclophosphamide and fludarabine induced a profound lymphopenia in all patients. Sequential FACS analysis revealed that different T cell subsets (CD4, CD8, CD4CD25) as well as granulocytes, B cells and NK cells were significantly reduced. Here, we report on clinical safety and feasibility of this vaccination approach during lymphoid recovery and demonstrate a patient example. CONCLUSION: Thus far, all vaccines were well tolerated. The overall trial design seems safe and feasible. Vaccine site reactions associated with infusion of GM-CSF via mini-pump are consistent with the postulated mechanism of action. More detailed immune-monitoring is required to evaluate a potential systemic immune response. Further studies to exploit homeostasis-driven T cell proliferation for the induction of a specific anti-tumor immune response in this clinical setting are warranted. AU - Rüttinger, D.* AU - van den Engel, N.K.* AU - Winter, H.* AU - Schlemmer, M.* AU - Pohla, H. AU - Grützner, S.* AU - Wagner, B.* AU - Schendel, D.J. AU - Fox, BA.* AU - Jauch, K.W.* AU - Hatz, R.A.* C1 - 2203 C2 - 24767 TI - Adjuvant therapeutic vaccination in patients with non-small cell lung cancer made lymphopenic and reconstituted with autologous PBMC: First clinical experience and evidence of an immune response. JO - J. Transl. Med. VL - 5 PB - Biomed Central PY - 2007 SN - 1479-5876 ER - TY - JOUR AB - For optimal T cell activation it is desirable that dendritic cells (DCs) display peptides within MHC molecules as signal 1, costimulatory molecules as signal 2 and, in addition, produce IL-12p70 as signal 3. IL-12p70 polarizes T cell responses towards CD4+ T helper 1 cells, which then support the development of CD8+ cytotoxic T lymphocytes. We therefore developed new maturation cocktails allowing DCs to produce biologically active IL-12p70 for large-scale cancer vaccine development. METHODS: After elutriation of leukapheresis products in a closed bag system, enriched monocytes were cultured with GM-CSF and IL-4 for six days to generate immature DCs that were then matured with cocktails, containing cytokines, interferon-gamma, prostaglandin E2, and a ligand for Toll-like receptor 8, with or without poly (I:C). RESULTS: Mature DCs expressed appropriate maturation markers and the lymph node homing chemokine receptor, CCR7. They retained full maturity after culture for two days without maturation cocktails and following cryopreservation. TLR ligand stimulation induced DCs capable of secreting IL-12p70 in primary cultures and after one day of coculture with CD40L-expressing fibroblasts, mimicking an encounter with T cells. DCs matured with our new cocktails containing TLR8 ligand, with or without poly (I:C), induced alloresponses and stimulated virus-specific T cells after peptide-pulsing. DCs matured in cocktails containing TLR8 ligand without poly (I:C) could also be loaded with RNA as a source of antigen, whereas DCs matured in cocktails containing poly (I:C) were unable to express proteins following RNA transfer by electroporation. CONCLUSION: Our new maturation cocktails allowed easy DC harvesting, stable maturation and substantial recoveries of mature DCs after cryopreservation. Our procedure for generating DCs is easily adaptable for GMP-compliance and yields IL-12p70-secreting DCs suitable for development of cancer vaccines using peptides or RNA as sources of immunizing antigens. AU - Zobywalski, A. AU - Javorovic, M. AU - Frankenberger, B. AU - Pohla, H.* AU - Kremmer, E. AU - Bigalke, I. AU - Schendel, D.J. C1 - 5148 C2 - 24766 TI - Generation of clinical grade dendritic cells with capacity to produce biologically active IL-12p70. JO - J. Transl. Med. VL - 5 PB - Biomed Central PY - 2007 SN - 1479-5876 ER - TY - JOUR AU - Geiger, C. AU - Regn, S. AU - Weinzierl, A.* AU - Nößner, E. AU - Schendel, D.J. C1 - 5485 C2 - 24116 SP - 1-15 TI - A generic RNA-pulsed dendritic cell vaccine strategy for renal cell carcinoma. JO - J. Transl. Med. VL - 3 PY - 2005 SN - 1479-5876 ER -