Network


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

Hotspot


Dive into the research topics where Ulrich Jaehde is active.

Publication


Featured researches published by Ulrich Jaehde.


American Journal of Pathology | 2005

Cisplatin Nephrotoxicity Is Critically Mediated via the Human Organic Cation Transporter 2

Giuliano Ciarimboli; Thomas Ludwig; Detlef Lang; Hermann Pavenstädt; Hermann Koepsell; Hansjürgen Piechota; Jörg Haier; Ulrich Jaehde; Jochen Zisowsky; Eberhard Schlatter

Cis-platin is an effective anti-neoplastic agent, but it is also highly nephrotoxic. Here, we clearly identify the human organic cation transporter 2 (hOCT2) as the critical transporter for cis-platin nephrotoxicity in isolated human proximal tubules and offer a potential mechanism for reducing nephrotoxicity in clinical practice. Interaction of cis-platin with hOCT2 in kidney or hOCT1 in liver was investigated with the fluorescent cation 4-[4-(dimethyl-amino)styril]-methylpyridinium in stably transfected HEK293 cells and for the first time in tissues physiologically expressing these transporters, human proximal tubules, and human hepatocyte couplets. Cis-platin (100 micromol/L) inhibited transport via hOCT2-HEK293 but not hOCT1-HEK293. In human proximal tubules cis-platin competed with basolateral organic cation transport, whereas it had no effect in tubules from a diabetic kidney or in hepatocytes. In hOCT2-HEK293 cells treated for 15 hours, incubation with cis-platin induced apoptosis, which was completely suppressed by contemporaneous incubation with the hOCT2 substrate cimetidine (100 micromol/L). These findings demonstrate that uptake of cis-platin is mediated by hOCT2 in renal proximal tubules, explaining its organ-specific toxicity. A combination of cis-platin with other substrates that compete for hOCT2 offers an effective option to decrease nephrotoxicity in the clinical setting.


Chemotherapy | 2003

Biochemical and Clinical Aspects of Methotrexate Neurotoxicity

Sandra Vezmar; Achim Becker; Udo Bode; Ulrich Jaehde

Acute, subacute and chronic neurotoxicity have been observed after the administration of high-dose and/or intrathecal methotrexate (MTX). Acute toxicity is usually transient without permanent damage. Subacute and chronic toxicity are associated with changes in the brain and/or the spinal cord which may be progressive and even lead to coma and death in severe cases. It is believed that MTX can induce direct toxic effects to the CNS by damaging the neuronal tissue. Moreover, MTX interferes with the metabolic pathways of folates, excitatory amino acids, homocysteine, S-adenosylmethionine/S-adenosylhomocysteine, adenosine and biopterins, inducing biochemical alterations which have been associated with neurotoxic symptoms. It has been suggested that acute toxicity is partly mediated by adenosine, whereas homocysteine, S-adenosylmethionine/S-adenosylhomocysteine, excitatory amino acids and biopterins may play an important role in the development of subacute and chronic toxicity. A better understanding of the pathogenesis of MTX neurotoxicity would offer the possibility of developing new therapeutic strategies for its treatment or prevention.


Anti-Cancer Drugs | 2009

Pharmacokinetics of a novel anticancer ruthenium complex (KP1019, FFC14A) in a phase I dose-escalation study.

Frederike Lentz; Anne Drescher; Andreas Lindauer; Magdalena Henke; Ralf A. Hilger; Christian G. Hartinger; Max E. Scheulen; Christian Dittrich; Bernhard K. Keppler; Ulrich Jaehde

A phase I and pharmacokinetic study was carried out with the new ruthenium complex indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019, FFC14A). Seven patients with various types of solid tumours refractory to standard therapy were treated with escalating doses of KP1019 (25–600 mg) twice weekly for 3 weeks. No dose-limiting toxicity occurred. Ruthenium plasma concentration–time profiles after the first dose and under multiple-dose conditions were analysed using a compartmental approach. The pharmacokinetic disposition was characterised by a small volume of distribution, low clearance and long half-life. Only a small fraction of ruthenium was excreted renally. The area under the curve values increased proportionally with dose indicating linear pharmacokinetics.


Clinical Pharmacokinectics | 2011

Clinical Pharmacokinetics of Tyrosine Kinase Inhibitors

Paola Di Gion; Friederike Kanefendt; Andreas Lindauer; Matthias Scheffler; Oxana Doroshyenko; Uwe Fuhr; Jürgen Wolf; Ulrich Jaehde

Pyrimidine (imatinib, dasatinib, nilotinib and pazopanib), pyridine (sorafenib) and pyrrole (sunitinib) tyrosine kinase inhibitors (TKIs) are multi-targeted TKIs with high activity towards several families of receptor and non-receptor tyrosine kinases involved in angiogenesis, tumour growth and metastatic progression of cancer. These orally administered TKIs have quite diverse characteristics with regard to absorption from the gastrointestinal tract. Absolute bioavailability in humans has been investigated only for imatinib (almost 100%) and pazopanib (14–39%; n = 3). On the basis of human radioactivity data, dasatinib is considered to be well absorbed after oral administration (19% and 0.1% of the total radioactivity were excreted as unchanged dasatinib in the faeces and urine, respectively). Quite low absolute bioavailability under fasted conditions is assumed for nilotinib (31%), sorafenib (50%) and sunitinib (50%). Imatinib, dasatinib and sunitinib exhibit dose-proportional increases in their area under the plasma concentration-time curve values over their therapeutic dose ranges. Less than dose-proportional increases were observed for nilotinib at doses ≥400 mg/day and for sorafenib and pazopanib at doses ≥800 mg/day. At steady state, the accumulation ratios are 1.5–2.5 (unchanged imatinib), 2.0 (nilotinib once-daily dosing), 3.4 (nilotinib twice-daily dosing), 1.2–4.5 (pazopanib), 5.7–6.4 (sorafenib) and 3.0–4.5 (sunitinib). Concomitant intake of a high-fat meal does not alter exposure to imatinib, dasatinib and sunitinib but leads to considerably increased bioavailability of nilotinib and pazopanib and decreased bioavailability of sorafenib. With the exception of pazopanib, the TKIs described here have large apparent volumes of distribution, exceeding the volume of body water by at least 4-fold.Very low penetration into the central nervous system in humans has been reported for imatinib and dasatinib, but there are currently no published human data for nilotinib, pazopanib, sorafenib or sunitinib. All TKIs that have been described are more than 90% bound to the plasma proteins: α1-acid glycoprotein and/or albumin. They are metabolized primarily via cytochrome P450 (CYP) 3A4, the only exception being sorafenib, for which uridine diphosphate glucuronosyltransferase 1A9 is the other main enzyme involved. Active metabolites of imatinib and sunitinib contribute to their antitumour activity. Although some patient demographics have been identified as significant co-factors that partly explain interindividual variability in exposure to TKIs, these findings have not been regarded as sufficient to recommend age-, sex-, bodyweight-or ethnicity-specific dose adjustment. Systemic exposure to imatinib, sorafenib and pazopanib increases in patients with hepatic impairment, and reduction of the initial therapeutic dose is recommended in this subpopulation. The starting dose of imatinib should also be reduced in renally impaired subjects. Because the solubility of dasatinib is pH dependent, co-administration of histamine H2-receptor antagonists and proton pump inhibitors with dasatinib should be avoided. With the exception of sorafenib, systemic exposure to TKIs is significantly decreased/increased by co-administration of potent CYP3A4 inducers/inhibitors, and so it is strongly recommended that the TKI dose is adjusted or that such co-administration is avoided. Caution is also recommended for co-administration of CYP3A4 substrates with TKIs, especially for those with a narrow therapeutic index. However, current recommendations with regard to dose adjustment of TKIs need to be validated in clinical studies. Further investigations are needed to explain the large interindividual variability in the pharmacokinetics of these drugs and to assess the clinical relevance of their interaction potential and inhibitory effects on metabolizing enzymes and transporters.


European Journal of Clinical Pharmacology | 2007

Evaluation of probe drugs and pharmacokinetic metrics for CYP2D6 phenotyping

Dorothee Frank; Ulrich Jaehde; Uwe Fuhr

IntroductionCytochrome P450 2D6 (CYP2D6) is one of the most important enzymes catalyzing biotransformation of xenobiotics in the human liver. This enzyme’s activity shows a high degree of interindividual variability caused in part by its genetic polymorphism, the so-called debrisoquine/sparteine polymorphism. The genetic component influencing CYP2D6 activity can be determined by genotyping. However, genotyping alone is not sufficient to accurately predict an individual’s actual CYP2D6 activity, as this is also influenced by other factors. For the determination of the exact actual enzymatic activity (“phenotyping”), adequate probe drugs have to be administered prior to measurements of these compounds and/or their metabolites in body fluids.Probe drugsDebrisoquine, sparteine, metoprolol or dextromethorphan represent well-established probe drugs while tramadol has been recently investigated for this purpose. The enzymatic activity is reflected by various pharmacokinetic metrics such as the partial clearance of a parent compound to the respective CYP2D6-mediated metabolite or metabolic ratios. Appropriate metrics need to fulfill pre-defined validation criteria.MethodsIn this review, we have compiled a list of such criteria useful to select the best metrics to reflect CYP2D6 activity. A comprehensive Medline search for reports on CYP2D6 phenotyping trials with the above mentioned probe drugs was carried out.ConclusionApplication of the validation criteria suggests that dextromethorphan and debrisoquine are the best CYP2D6 phenotyping drugs, with debrisoquine having the problem of very limited availability as a therapeutic drug. However, the assessment of the best dextromethorphan CYP2D6 phenotyping metric/procedure is still ongoing.


International Journal of Cancer | 2004

Long-term activation of SAPK/JNK, p38 kinase and fas-L expression by cisplatin is attenuated in human carcinoma cells that acquired drug resistance.

Anamaria Brozovic; Gerhard Fritz; Markus Christmann; Jochen Zisowsky; Ulrich Jaehde; Maja Osmak; Bernd Kaina

Tumor cells chronically exposed to cisplatin (cDDP) acquire cDDP resistance that impacts tumor therapy. To elucidate the mechanism of acquired cDDP resistance (ACR), we compared HeLa cells that gained ACR upon chronic cDDP treatment with the parental strain. We show that ACR is due to a lower level of induced apoptosis. Further, upon cDDP treatment, the levels of Fas, Bax and Bid remained unchanged, whereas Bcl‐2 and p‐Bad were reduced at late times (120 hr) after treatment. At early times, Fas ligand (fas‐L) expression was significantly enhanced in sensitive compared to resistant cells and remained upregulated up to the onset of apoptosis. Thus, activation of the Fas system is critical, which is in line with the finding that in sensitive cells, caspase‐8 along with caspase‐9 and ‐3 were activated by cDDP. cDDP provoked the activation of stress‐activated protein kinase/c‐Jun N‐terminal kinase (SAPK/JNK) and p38 kinase dose‐dependently, with significantly lower levels in ACR cells than in the sensitive parental line. cDDP induces c‐Jun and AP‐1 activity, as measured by a reporter gene assay, which was again attenuated in ACR cells. Time course analysis revealed that SAPK/JNK and p38 kinase activity was sustained upregulated (> 72 hr postexposure), which occurred at much higher level in sensitive than in ACR cells. Inhibition of either JNK or p38 kinase (by JNK inhibitor II and SB 203580, respectively) attenuated cDDP‐induced apoptosis, supporting the role of JNK and p38 kinase in the cDDP response. Since several independently derived cDDP‐resistant cell lines displayed attenuated MAPK signaling, sustained SAPK/JNK and p38 kinase activation may be a general mechanism of cDDP‐induced cell death. ACR cells displayed a reduced level of DNA damage, indicating long‐term stimulation of SAPK/JNK and p38 kinase is triggered by nonrepaired cDDP‐induced DNA lesions.


Clinical Pharmacokinectics | 1989

Pharmacokinetic Disposition of Quinolones in Human Body Fluids and Tissues

Fritz Sörgel; Ulrich Jaehde; Kurt G. Naber; U. Stephan

SummaryThe unique pharmacokinetic properties as well as the body fluid and tissue penetration of quinolones are discussed.Quinolones are well absorbed from the gastrointestinal tract and are eliminated with considerable differences in their terminal half-lives. The major elimination pathways of quinolones are renal excretion and hepatic metabolism. Renally, these drugs undergo the potential excretion mechanisms (glomerular filtration, tubular secretion, reabsorption). In the liver, they are metabolised primarily by oxidation as well as by conjugative pathways. However, the metabolic pattern and extent of metabolism differ significantly between individual agents.Alterations in the pharmacokinetic disposition of these agents in liver and renal failure as well as in elderly patients are observed as predicted from their excretion pattern. In addition, quinolones can interact with a number of other compounds at hepatic (e.g. with xanthine derivatives), renal (with probenecid) and gastrointestinal (with antacids) sites.The volume of distribution of quinolones is considerably higher than body volume, which suggests intracellular penetration. Studies on tissue penetration show that concentrations exceeding plasma levels are obtained in most tissues. The highest tissue/plasma concentration ratios are achieved in lung and kidney, whereas concentrations in fat are considerably lower than in plasma.Body fluid penetration is introduced as a new approach to evaluate distribution kinetics of quinolones. With the exception of those in nasal secretions and ejaculate, body fluid levels of these drugs rarely reach plasma levels. The body fluid penetration model allows for differentiation among individual agents. There is no apparent relationship between differences in body fluid penetration of quinolones and differences in volume of distribution. For the clinical use of these drugs it is important that the concentrations achieved in body fluids and tissues are sufficient to kill most pathogens. A discussion on the relationship between plasma and tissue levels and the MICs of quinolones is, however, beyond the scope of this article.


Clinical Pharmacokinectics | 1995

Clinical pharmacokinetics in patients with burns.

Ulrich Jaehde; Fritz Sörgel

SummaryBurn injury induces many different pathological changes in the human body, which potentially alter pharmacokinetic parameters such as bioavailability, protein binding, volume of distribution (Vd) and clearance. The extent of these alterations depends on the drug, the type and extent of injury and the time that elapsed between injury and drug administration. Bioavailability of large and hydrophilic molecules may be increased because of enhanced intestinal permeability. The free fraction of a drug in plasma can be increased (when primarily bound to albumin) or decreased (when primarily bound to α1-acid glycoprotein). Vd may change as a consequence of altered protein binding or an enlarged extracellular fluid volume. Alterations in clearance may be due to changes in glomerular filtration, tubular secretion, hepatic blood flow, drug-metabolising activity, protein binding and to the presence of additional elimination pathways. Elimination half-life changes when Vd and/or clearance is affected following burn injury.The therapeutic consequences of pharmacokinetic alterations are discussed in principle, and for specific treatment with antibacterials, anti-ulcer drugs, analgesics, muscle relaxants, anxiolytics, phenytoin and cyclosporin. If significant changes in pharmacokinetic disposition occur following thermal injury, therapeutic drug monitoring and dosage adjustment may be required to ensure rational and well tolerated drug therapy in patients with burns. Future studies should focus on the impact of specific patient variables (e.g. type of injury and size of burn) on the extent of pharmacokinetic alterations.


Journal of Biological Chemistry | 2008

Epidermal Growth Factor Receptor Pathway Analysis Identifies Amphiregulin as a Key Factor for Cisplatin Resistance of Human Breast Cancer Cells

Niels Eckstein; Kati Servan; Luc Girard; Di Cai; Georg von Jonquieres; Ulrich Jaehde; Matthias U. Kassack; Adi F. Gazdar; John D. Minna; Hans Dieter Royer

The use of platinum complexes for the therapy of breast cancer is an emerging new treatment modality. To gain insight into the mechanisms underlying cisplatin resistance in breast cancer, we used estrogen receptor-positive MCF-7 cells as a model system. We generated cisplatin-resistant MCF-7 cells and determined the functional status of epidermal growth factor receptor (EGFR), MAPK, and AKT signaling pathways by phosphoreceptor tyrosine kinase and phospho-MAPK arrays. The cisplatin-resistant MCF-7 cells are characterized by increased EGFR phosphorylation, high levels of AKT1 kinase activity, and ERK1 phosphorylation. In contrast, the JNK and p38 MAPK modules of the MAPK signaling pathway were inactive. These conditions were associated with inactivation of the p53 pathway and increased BCL-2 expression. We investigated the expression of genes encoding the ligands for the ERBB signaling cascade and found a selective up-regulation of amphiregulin expression, which occurred at later stages of cisplatin resistance development. Amphiregulin is a specific ligand of the EGFR (ERBB1) and a potent mitogen for epithelial cells. After exposure to cisplatin, the resistant MCF-7 cells secreted amphiregulin protein over extended periods of time, and knockdown of amphiregulin expression by specific short interfering RNA resulted in a nearly complete reversion of the resistant phenotype. To demonstrate the generality and importance of our findings, we examined amphiregulin expression and cisplatin resistance in a variety of human breast cancer cell lines and found a highly significant correlation. In contrast, amphiregulin levels did not significantly correlate with cisplatin resistance in a panel of lung cancer cell lines. We have thus identified a novel function of amphiregulin for cisplatin resistance in human breast cancer cells.


BMC Cancer | 2008

Altered localisation of the copper efflux transporters ATP7A and ATP7B associated with cisplatin resistance in human ovarian carcinoma cells

Ganna V. Kalayda; Christina H. Wagner; Irina Buß; Jan Reedijk; Ulrich Jaehde

BackgroundCopper homeostasis proteins ATP7A and ATP7B are assumed to be involved in the intracellular transport of cisplatin. The aim of the present study was to assess the relevance of sub cellular localisation of these transporters for acquired cisplatin resistance in vitro. For this purpose, localisation of ATP7A and ATP7B in A2780 human ovarian carcinoma cells and their cisplatin-resistant variant, A2780cis, was investigated.MethodsSub cellular localisation of ATP7A and ATP7B in sensitive and resistant cells was investigated using confocal fluorescence microscopy after immunohistochemical staining. Co-localisation experiments with a cisplatin analogue modified with a carboxyfluorescein-diacetate residue were performed. Cytotoxicity of the fluorescent cisplatin analogue in A2780 and A2780cis cells was determined using an MTT-based assay. The significance of differences was analysed using Students t test or Mann-Whitney test as appropriate, p values of < 0.05 were considered significant.ResultsIn the sensitive cells, both transporters are mainly localised in the trans-Golgi network, whereas they are sequestrated in more peripherally located vesicles in the resistant cells. Altered localisation of ATP7A and ATP7B in A2780cis cells is likely to be a consequence of major abnormalities in intracellular protein trafficking related to a reduced lysosomal compartment in this cell line. Changes in sub cellular localisation of ATP7A and ATP7B may facilitate sequestration of cisplatin in the vesicular structures of A2780cis cells, which may prevent drug binding to genomic DNA and thereby contribute to cisplatin resistance.ConclusionOur results indicate that alterations in sub cellular localisation of transport proteins may contribute to cisplatin resistance in vitro. Investigation of intracellular protein localisation in primary tumour cell cultures and tumour tissues may help to develop markers of clinically relevant cisplatin resistance. Detection of resistant tumours in patients may in turn enable individualization of the chemotherapy in the early stage of treatment.

Collaboration


Dive into the Ulrich Jaehde's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charlotte Kloft

Free University of Berlin

View shared research outputs
Top Co-Authors

Avatar

Walter Schunack

Free University of Berlin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Uwe Fuhr

University of Cologne

View shared research outputs
Top Co-Authors

Avatar

Markus Joerger

University of St. Gallen

View shared research outputs
Top Co-Authors

Avatar

Fritz Sörgel

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Ralf A. Hilger

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge