Network


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

Hotspot


Dive into the research topics where Hermann Pelzer is active.

Publication


Featured researches published by Hermann Pelzer.


Blood | 2013

A novel B-domain O-glycoPEGylated FVIII (N8-GP) demonstrates full efficacy and prolonged effect in hemophilic mice models

Henning R. Stennicke; Marianne Kjalke; Ditte M. Karpf; Kristoffer W. Balling; Peter B. Johansen; Torben Elm; Kristine Øvlisen; Flemming Möller; Heidi L. Holmberg; Charlotte N. Gudme; Egon Persson; Ida Hilden; Hermann Pelzer; Henrik Rahbek-Nielsen; Christina Jespersgaard; Are Bogsnes; Anette A. Pedersen; Anne K. Kristensen; Bernd Peschke; Wendy Kappers; Frederik Rode; Lars Thim; Mikael Tranholm; Mirella Ezban; Eva H. N. Olsen; Søren E. Bjørn

Frequent infusions of intravenous factor VIII (FVIII) are required to prevent bleeding associated with hemophilia A. To reduce the treatment burden, recombinant FVIII with a longer half-life was developed without changing the protein structure. FVIII-polyethylene glycol (PEG) conjugates were prepared using an enzymatic process coupling PEG (ranging from 10 to 80 kDa) selectively to a unique O-linked glycan in the FVIII B-domain. Binding to von Willebrand factor (VWF) was maintained for all conjugates. Upon cleavage by thrombin, the B-domain and the associated PEG were released, generating activated FVIII (FVIIIa) with the same primary structure and specific activity as native FVIIIa. In both FVIII- and VWF-deficient mice, the half-life was found to increase with the size of PEG. In vivo potency and efficacy of FVIII conjugated with a 40-kDa PEG (N8-GP) and unmodified FVIII were not different. N8-GP had a longer duration of effect in FVIII-deficient mouse models, approximately a twofold prolonged half-life in mice, rabbits, and cynomolgus monkeys; however, the prolongation was less pronounced in rats. Binding capacity of N8-GP on human monocyte-derived dendritic cells was reduced compared with unmodified FVIII, resulting in several-fold reduced cellular uptake. In conclusion, N8-GP has the potential to offer efficacious prevention and treatment of bleeds in hemophilia A at reduced dosing frequency.


Journal of Thrombosis and Haemostasis | 2011

Recombinant human factor VIIa (rFVIIa) cleared principally by antithrombin following IV administration in haemophilia patients.

Henrik Agersø; Donald F. Brophy; Hermann Pelzer; Erika J. Martin; Marcus E. Carr; Ulla Hedner; Mirella Ezban

Summary.  Objective: The objective of the present study was to evaluate the pharmacokinetics and the clearance pathways of rFVIIa after intravenous administration to hemophilia patients. Methods: Ten severe hemophilia patients were included in the study; all patients were intravenously administered a clinically relevant dose of 90 μg kg−1 (1.8 nmol kg−1) rFVIIa. Blood samples were collected consecutively to describe the pharmacokinetics of rFVIIa. All samples were analyzed using three different assays: a clot assay to measure the activity (FVIIa:C), an enzyme immunoassay (EIA) to measure the antigen levels (FVII:Ag), and an EIA (FVIIa‐AT) to measure the FVIIa antithrombin III (AT) complex. Pharmacokinetic parameters were evaluated both by use of standard non‐compartmental methods and by use of mixed effects methods. A population pharmacokinetic model was used to simultaneously model all three datasets. The total body clearance of rFVIIa:C was estimated to be 38 mL h−1 kg−1. The rFVII‐AT complex formation was responsible for 65% of the total rFVIIa:C clearance. The initial and the terminal half‐life of rFVIIa:C was estimated to be 0.6 and 2.6 h, respectively. The formation of rFVII‐AT complex was able to explain the difference observed between the rFVIIa:C and the rFVII:Ag concentration. The non‐compartmental analysis resulted in almost identical parameters.


Haemophilia | 2012

Pharmacokinetics and pharmacodynamics of turoctocog alfa and N8-GP in haemophilia A dogs.

Henrik Agersø; Henning R. Stennicke; Hermann Pelzer; E. H. N. Olsen; Elizabeth P. Merricks; N. Defriess; Timothy C. Nichols; Mirella Ezban

The objective of the present study was to evaluate the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of the new recombinant FVIII compound turoctocog alfa and a Glyco‐PEGylated FVIII derivative thereof (N8‐GP) in Haemophilia A dogs. Six haemophilic dogs divided into two groups were included in the study. Each dog was administered a dose of 125 U kg−1, blood samples were collected at predetermined time points for both pharmacokinetic (FVIII measured by one‐stage aPTT assay) and pharmacodynamic [whole blood clotting time (WBCT)] evaluations. After intravenous administration to haemophilic dogs, the plasma concentration at the first sampling point was comparable for turoctocog alfa and N8‐GP, and the clearance was estimated to be 6.5 and 3.9 mL h−1kg−1 for turoctocog alfa and N8‐GP respectively. Both turoctocog alfa and N8‐GP were able to reduce the WBCT time to normal levels (<20 min), however, the reduced clearance was reflected in the WBCT, which returned to baseline at a later time point for N8‐GP as compared with dogs dosed with turoctocog alfa. The clearance was 40% reduced for N8‐GP as compared with turoctocog alfa. Simulations of a multiple dosing regimen in dogs, suggest that to maintain WBCT <20 min N8‐GP can be dosed at reduced intervals, e.g. with 4 days between doses, whereas turoctocog alfa will have to be dosed with 2½ day between doses. Data thereby supports N8‐GP as an alternative to standard rFVIII replacement therapy, with a more convenient dosing regimen.


Haemophilia | 2012

Pharmacokinetics and pharmacodynamics of a new recombinant FVIII (N8) in haemophilia A mice

T. Elm; Ditte M. Karpf; K. Øvlisen; Hermann Pelzer; Mirella Ezban; Marianne Kjalke; Mikael Tranholm

Summary.  N8 is a new recombinant factor VIII (rFVIII) compound produced and formulated without human‐ or animal‐derived protein. The aims of the present studies were to evaluate the pharmacokinetics and pharmacodynamics properties of N8 and to compare with a commercially available rFVIII product (Advate®) in haemophilia A mice. The pharmacokinetics were evaluated after single i.v. administration of 80, 120 and 280 IU kg−1 of N8 and Advate® and measurements of FVIII blood concentrations as a function of time. The efficacy and dose response curves of N8 and Advate® (1–200 IU kg−1) were evaluated in a tail bleeding model. Furthermore, the effects in a newly developed haemophilia knee joint haemarthrosis model were investigated. No significant differences were found in the pharmacokinetic parameters between N8 and Advate®. The clearances were 11 ± 1 vs. 10 ± 2 mL h−1 kg−1 (P = 0.14) and the half‐lives 7.2 ± 0.9 vs. 7.7 ± 1.4 h (P = 0.31) after administration of N8 and Advate® respectively. Dose‐independent pharmacokinetics was shown, and comparable efficacy and potency were shown between N8 and Advate® in the tail bleeding model. Both compounds normalized the bleeding at the dose of 200 IU kg−1, and for blood loss ED50 values of 27 IU kg−1 (N8) and 28 IU/kg (Advate®) were found (P = 0.97). In the haemarthrosis model, treatment with N8 and Advate® at 200 IU kg−1 reduced the mean increase in the joint diameter significantly from 1.23 ± 0.19 to 0.32 ± 0.08 mm (P < 0.01) and 0.25 ± 0.08 mm (P < 0.001) respectively. Pharmacokinetics and pharmacodynamics of N8 and Advate® were comparable after i.v. administration to haemophilia A mice.


British Journal of Haematology | 2011

Intravascular inhibition of factor VIIa and the analogue NN1731 by antithrombin

Lars C. Petersen; Ditte M. Karpf; Henrik Agersø; Mette B. Hermit; Hermann Pelzer; Egon Persson; Timothy C. Nichols; Mirella Ezban

NN1731 is a recombinant activated factor VII (rFVIIa) analogue with increased intrinsic activity. This also applies to its reactivity towards antithrombin (AT), the role of which was investigated in a pharmacokinetic (PK) study. NN1731 or rFVIIa was administered to normal and haemophilia A dogs and elimination was measured by FVIIa clot activity, FVIIa‐ and FVIIa‐AT antigen. In vitro AT complex formation was studied in canine plasma spiked with NN1731 or rFVIIa. Based on FVIIa antigen concentrations, PK profiles in normal and haemophilia A dogs were similar for NN1731 and rFVIIa with antigen half lives, t½ ≈ 1·8 h. In contrast, PK profiles based on activity measurements were distinctly different. NN1731 induced a strong, short lasting (t½ ≈ 0·5 h) pro‐coagulant response, whereas rFVIIa induced a lower, longer lasting (t½ ≈ 1·1 h) response. Western Blot and FVIIa‐AT antigen analysis demonstrated in vivo AT complex formation that accounted for these divergences. AT complex formation with FVIIa or NN1731 in vitro in canine plasma was considerably slower than the in vivo reaction. The results suggest that in vivo inhibition by AT contributes significantly to define drug duration in haemophilia treatment with rFVIIa and in particular with the NN1731 analogue.


European Journal of Pharmaceutical Sciences | 2011

Clearance of rFVIIa and NN1731 after intravenous administration to Beagle dogs

Henrik Agersø; Niels Rode Kristensen; Henrik Østergaard; Ditte M. Karpf; Mette B. Hermit; Hermann Pelzer; Lars C. Petersen; Mirella Ezban

AIM NN1731 is a recombinant activated factor VII (rFVIIa) analogue with enhanced activity. The objective of the present study was to evaluate the clearance mechanisms of rFVIIa and NN1731 after intravenous administration to Beagle dogs. METHODS The study was performed in Beagle dogs administered with a single dose of 5.4 nmol/kg rFVIIa or NN1731 intravenously. Plasma samples collected up to 12-h post-administration were analysed using three different assays to determine FVIIa clot activity (FVIIa:C), total FVIIa antigen, and levels of FVIIa-antithrombin (AT) complexes. Pharmacokinetic parameters were determined by use of standard non-compartmental and non-linear mixed effects methods. RESULTS For both compounds, complex formation with AT accounted for the observed difference between the activity and the antigen curves and constituted 60-70% of the total clearance. The clearance of rFVIIa and NN1731 was estimated to be 73 and 214 mL/h/kg, respectively, accordingly, AT complex formation occurred around three times faster for NN1731. The difference in activity observed in the initial phase, resulting in distribution half-lives of 0.71 and 0.22 h for rFVIIa and NN1731, was mainly caused by the 3-fold difference in clearance. The terminal half-life of rFVIIa and NN1731 was estimated to be 2.1 and 2.5 h, respectively. The non-compartmental analysis resulted in almost identical parameters. CONCLUSION The present study demonstrates that the difference between the activity and the antigen profiles of rFVIIa and NN1731 in Beagle dogs is the result of complex formation with AT which constitutes a major pathway for the clearance of rFVIIa activity.


Blood | 2011

Activation peptides prolong the murine plasma half-life of human factor VII.

Linda Johansson; Ditte M. Karpf; Lene Hansen; Hermann Pelzer; Egon Persson

Coagulation factors VII (FVII), IX (FIX), X (FX), and protein C share the same domain organization but display very different plasma half-lives. It is plausible that the half-life is influenced by the activation peptide, differing in length and glycosylation and missing in FVII. To test this hypothesis, the influence of activation peptides on the plasma half-life of human FVII was studied by administering human FVII variants containing activation peptide motifs to mice. Insertion of the activation peptide from FX gave 4-fold longer terminal half-life (5.5 hours vs 1.4 hours for FVII), whereas the activation peptide from FIX and protein C resulted in half-lives of 4.3 and 1.7 hours, respectively. Using FXs activation peptide we identified the N-linked glycans as structural features important for the half-life. The peptide location within the FVII molecule appeared not to be critical because similar prolongation was obtained with the activation peptide inserted immediately before the normal site of activation and at the C-terminus. However, only the latter variant was activatable, yielding full amidolytic activity and reduced proteolytic activity with preserved long half-life. Our data support that activation peptides function as plasma retention signals and constitute a new manner to extend the half-life of FVII(a).


PLOS ONE | 2015

Factor XIII Transglutaminase Supports the Resolution of Mucosal Damage in Experimental Colitis.

Christina Andersson; Peter Helding Kvist; Kathryn E. McElhinney; Richard Baylis; Luise K. Gram; Hermann Pelzer; Brian Lauritzen; Thomas Lindebo Holm; Simon P. Hogan; David Wu; Brian Turpin; Whitney Miller; Joseph S. Palumbo

The thrombin-activated transglutaminase factor XIII (FXIII) that covalently crosslinks and stablizes provisional fibrin matrices is also thought to support endothelial and epithelial barrier function and to control inflammatory processes. Here, gene-targeted mice lacking the FXIII catalytic A subunit were employed to directly test the hypothesis that FXIII limits colonic pathologies associated with experimental colitis. Wildtype (WT) and FXIII-/- mice were found to be comparable in their initial development of mucosal damage following exposure to dextran sulfate sodium (DSS) challenge. However, unlike FXIII-sufficient mice, FXIII-deficient cohorts failed to efficiently resolve colonic inflammatory pathologies and mucosal damage following withdrawal of DSS. Consistent with prior evidence of ongoing coagulation factor activation and consumption in individuals with active colitis, plasma FXIII levels were markedly decreased in colitis-challenged WT mice. Treatment of colitis-challenged mice with recombinant human FXIII-A zymogen significantly mitigated weight loss, intestinal bleeding, and diarrhea, regardless of whether cohorts were FXIII-sufficient or were genetically devoid of FXIII. Similarly, both qualitative and quantitative microscopic analyses of colonic tissues revealed that exogenous FXIII improved the resolution of multiple colitis disease parameters in both FXIII-/- and WT mice. The most striking differences were seen in the resolution of mucosal ulceration, the most severe histopathological manifestation of DSS-induced colitis. These findings directly demonstrate that FXIII is a significant determinant of mucosal healing and clinical outcome following inflammatory colitis induced mucosal injury and provide a proof-of-principle that clinical interventions supporting FXIII activity may be a means to limit colitis pathology and improve resolution of mucosal damage.


Haemophilia | 2011

Monitoring rFVIIa 90 μg kg−1 dosing in haemophiliacs: comparing laboratory response using various whole blood assays over 6 h

Donald F. Brophy; Erika J. Martin; J Christian Barrett; Mindy Nolte; Janice Kuhn; P M Gerk; Marcus E. Carr; Hermann Pelzer; Henrik Agersø; Mirella Ezban; Ulla Hedner

Summary.  Recombinant FVIIa is a haemostatic agent administered to patients with severe FVIII or FIX deficiency with inhibitors. Although rFVIIa is effective at stopping bleeding, a reliable assay to monitor its effect is lacking. To characterize the pharmacokinetics and global coagulation effects of rFVIIa for 6 h following a IV dose of 90 μg kg−1. Ten non‐bleeding subjects with severe FVIII or FIX deficiency were infused with a single‐dose of rFVIIa 90 μg kg−1 body weight and blood was collected before and at 0.5, 1, 2, 4 and 6 h postdose. Global haemostasis was characterized throughout the study utilizing whole blood analyses (Hemodyne HAS, TEG, ROTEM). The clearance and half‐life of factor FVII:C was estimated as 39.0 ± 8.8 mL h−1 kg−1 and 2.1 ± 0.2 h respectively. There was good inter‐assay agreement with respect to clot initiation parameters (R, CT and FOT) and these parameters all fell to a mean of approximately 9 min following rFVIIa dosing. The platelet contractile force (PCF) and clot elastic modulus (CEM) were positively correlated to FVII:C (P < 0.0001), and these parameters were dynamic throughout the 6‐h period. The MA and MCF did not correlate to FVII:C nor did they significantly change during the study. Prothrombin F1 + 2 significantly increased following rFVIIa dosing (P < 0.001), but remained steady throughout the study. There was no change in D‐dimer concentrations over time. The FOT, R and CT characterized clot initiation following rFVIIa dosing. The PCF and CEM were correlated to FVII:C and characterized the dynamics of platelet function and clot strength over the rFVIIa dosing interval. The clinical significance of these findings needs additional study.


Journal of Gastroenterology | 2016

Systemic and intestinal levels of factor XIII-A: the impact of inflammation on expression in macrophage subtypes

Christoffer Soendergaard; Peter Helding Kvist; Jakob Benedict Seidelin; Hermann Pelzer; Ole Haagen Nielsen

BackgroundSubunit A of coagulation factor XIII (FXIII-A) is important for clot stability and acts in the subsequent wound healing process. Loss of plasma FXIII-A has been reported after surgery, sepsis, and inflammatory conditions. In the intestinal mucosa, FXIII-A is expressed by macrophages and cellular FXIII-A has been associated with phagocytosis and migration of macrophages. The objective was to evaluate the consequences of intestinal inflammation on resident mucosal macrophages, focusing on the level and distribution of FXIII-A.MethodsPlasma and colonic biopsies were collected from 67 patients with ulcerative colitis and controls. Intestinal samples were stained using immunohistochemistry for FXIII-A and macrophages (CD68, CD163 and iNOS). In situ hybridization were used to assess the intestinal expression of FXIII-A. FXIII-A antigen and activity levels were measured in plasma.ResultsIncreased infiltration of CD68 positive macrophages in the inflamed mucosa coincided with increased extracellular deposited FXIII-A and decreased expression and intracellular protein levels of FXIII-A. A decreased proportion of FXIII-A/CD68/CD163 triple-positive macrophages was observed in inflamed mucosa, indicating a reduction of the M2 phenotype with consequent loss of FXIII-A. No induction of iNOS positive macrophages was observed. Stimulation of naïve monocytes with physiological concentrations of pro-inflammatory mediators negatively affected the expression of FXIII-A. Measurements in plasma confirmed the loss of both FXIII antigen and activity during active disease.ConclusionsIntestinal inflammation in UC induces loss of M2 macrophages with subsequent loss of FXIII-A synthesis. The loss of cellular FXIII-A may impact migration and phagocytosis, and hence limit pathogen eradication in UC.

Collaboration


Dive into the Hermann Pelzer'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
Researchain Logo
Decentralizing Knowledge