Network


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

Hotspot


Dive into the research topics where Stefanie Kraff is active.

Publication


Featured researches published by Stefanie Kraff.


Clinical Pharmacokinectics | 2012

Evaluation of a Pharmacology-Driven Dosing Algorithm of 3-Weekly Paclitaxel Using Therapeutic Drug Monitoring

Markus Joerger; Stefanie Kraff; Alwin D. R. Huitema; Gary Feiss; Berta Moritz; Jan H. M. Schellens; Jos H. Beijnen; Ulrich Jaehde

Background and ObjectiveSevere neutropenia is the most frequent and important toxicity of 3-weekly paclitaxel and puts patients at substantial risk of infectious complications. It is well known that the time during which paclitaxel plasma concentrations exceed 0.05 μmol/L (TC>0.05) correlates with the extent of neutropenia. This study was initiated to develop a dosing algorithm that would be able to reduce severe neutropenia by targeting an individual paclitaxel TC>0.05 between 26 and 31 hours, and could be validated in a prospective randomized trial by comparing it to conventional dosing of paclitaxel.MethodsPaclitaxel plasma concentration-time (n = 273) and absolute neutrophil count (ANC) data (152 of the 273 patients) were pooled from two previous studies and submitted to population pharmacokinetic and pharmacodynamic modelling using nonlinear mixed-effects modelling software NONMEM® version VII. To fit the data, we used a previously described 3-compartment model with saturable elimination and distribution, coupled to a semiphysiological model with a linear function to describe the myelotoxic effect of paclitaxel (Epaclitaxel) on circulating neutrophils (neutropenia). Patient age, sex, body surface area (BSA), bilirubin and renal function were tested as potential covariates on the maximum elimination capacity of paclitaxel (VMEL). Limited sampling strategies were tested on the pharmacokinetic model for their accuracy to predict paclitaxel TC>0.05. Subsequently, we proposed a first-cycle dosing algorithm that accounted for BSA, patient age and sex, while later cycles accounted for the previous-cycle paclitaxel TC>0.05 (target: 26 to 31 hours) and ANC nadir to adapt the paclitaxel dose for the next treatment cycle. To test the adequacy of the proposed dosing algorithm, we used extensive data simulations on the final pharmacokinetic/pharma-codynamic model, generating datasets of 1000 patients for six subsequent treatment cycles. Grade 4 neutropenia was tested as a potential endpoint for a prospective clinical trial and simulated for two scenarios, i.e. conventional dosing of paclitaxel 200 mg/m2 every 3 weeks, and personalized, pharmacology-driven dosing as outlined above.ResultsConcentration-time data for paclitaxel were adequately described by the 3-compartment model. Also, individual ANC counts were adequately described by the semiphysiological model using a linear function to describe Epaclitaxel on neutropenia. Patient age, sex, bilirubin and BSA were significant and independent covariates on the elimination of paclitaxel. Paclitaxel VMEL was 16% higher in males than in female patients, and a 10-year increase in age led to a 13% decrease in VMEL. A single paclitaxel plasma concentration 24 hours after the start of infusion was adequate to predict paclitaxel TC>0.05 (root squared mean error [RSME] = +0.5%), and the addition of an end-of-infusion sample did not further improve precision (RSME = −0.6%). Data simulations on the final pharmacokinetic/pharmacodynamic model and using the proposed dosing algorithm resulted in a first-cycle paclitaxel dose ranging from 150 to 185 mg/m2 for women and from 165 to 200 mg/m2 for men. Dose adaptations for cycles two to six ranged from −40% to +30%, with a final median paclitaxel dose of 167 mg/m2 (range 76 to 311 mg/m2). When compared with conventional dosing (paclitaxel 200 mg/m2 every 3 weeks), personalized dosing reduced grade 4 neutropenia in cycle one from 15% to 7%, and further to 4% in cycle 2.ConclusionThis study proposes a pharmacology-driven dosing algorithm of 3-weekly paclitaxel to reduce the incidence of grade 4 neutropenia. A randomized clinical trial comparing this dosing algorithm with conventional BSA-based dosing of paclitaxel in patients with advanced non-small cell lung cancer is currently ongoing.


Clinical Pharmacokinectics | 2012

Evaluation of a pharmacology-driven dosing algorithm of 3-weekly paclitaxel using therapeutic drug monitoring: a pharmacokinetic-pharmacodynamic simulation study.

Markus Joerger; Stefanie Kraff; Alwin D. R. Huitema; Gary Feiss; Berta Moritz; Jan H. M. Schellens; Jos H. Beijnen; Ulrich Jaehde

BACKGROUND AND OBJECTIVE Severe neutropenia is the most frequent and important toxicity of 3-weekly paclitaxel and puts patients at substantial risk of infectious complications. It is well known that the time during which paclitaxel plasma concentrations exceed 0.05 μmol/L (T(C>0.05)) correlates with the extent of neutropenia. This study was initiated to develop a dosing algorithm that would be able to reduce severe neutropenia by targeting an individual paclitaxel T(C>0.05) between 26 and 31 hours, and could be validated in a prospective randomized trial by comparing it to conventional dosing of paclitaxel. METHODS Paclitaxel plasma concentration-time (n = 273) and absolute neutrophil count (ANC) data (152 of the 273 patients) were pooled from two previous studies and submitted to population pharmacokinetic and pharmacodynamic modelling using nonlinear mixed-effects modelling software NONMEM® version VII. To fit the data, we used a previously described 3-compartment model with saturable elimination and distribution, coupled to a semiphysiological model with a linear function to describe the myelotoxic effect of paclitaxel (E(paclitaxel)) on circulating neutrophils (neutropenia). Patient age, sex, body surface area (BSA), bilirubin and renal function were tested as potential covariates on the maximum elimination capacity of paclitaxel (VM(EL)). Limited sampling strategies were tested on the pharmacokinetic model for their accuracy to predict paclitaxel T(C>0.05). Subsequently, we proposed a first-cycle dosing algorithm that accounted for BSA, patient age and sex, while later cycles accounted for the previous-cycle paclitaxel T(C>0.05) (target: 26 to 31 hours) and ANC nadir to adapt the paclitaxel dose for the next treatment cycle. To test the adequacy of the proposed dosing algorithm, we used extensive data simulations on the final pharmacokinetic/pharmacodynamic model, generating datasets of 1000 patients for six subsequent treatment cycles. Grade 4 neutropenia was tested as a potential endpoint for a prospective clinical trial and simulated for two scenarios, i.e. conventional dosing of paclitaxel 200 mg/m(2) every 3 weeks, and personalized, pharmacology-driven dosing as outlined above. RESULTS Concentration-time data for paclitaxel were adequately described by the 3-compartment model. Also, individual ANC counts were adequately described by the semiphysiological model using a linear function to describe E(paclitaxel) on neutropenia. Patient age, sex, bilirubin and BSA were significant and independent covariates on the elimination of paclitaxel. Paclitaxel VM(EL) was 16% higher in males than in female patients, and a 10-year increase in age led to a 13% decrease in VM(EL). A single paclitaxel plasma concentration 24 hours after the start of infusion was adequate to predict paclitaxel T(C>0.05) (root squared mean error [RSME] = +0.5%), and the addition of an end-of-infusion sample did not further improve precision (RSME = -0.6%). Data simulations on the final pharmacokinetic/pharmacodynamic model and using the proposed dosing algorithm resulted in a first-cycle paclitaxel dose ranging from 150 to 185 mg/m(2) for women and from 165 to 200 mg/m(2) for men. Dose adaptations for cycles two to six ranged from -40% to +30%, with a final median paclitaxel dose of 167 mg/m(2) (range 76 to 311 mg/m(2)). When compared with conventional dosing (paclitaxel 200 mg/m(2) every 3 weeks), personalized dosing reduced grade 4 neutropenia in cycle one from 15% to 7%, and further to 4% in cycle 2. CONCLUSION This study proposes a pharmacology-driven dosing algorithm of 3-weekly paclitaxel to reduce the incidence of grade 4 neutropenia. A randomized clinical trial comparing this dosing algorithm with conventional BSA-based dosing of paclitaxel in patients with advanced non-small cell lung cancer is currently ongoing.


Annals of Oncology | 2016

Open-label, randomized study of individualized, pharmacokinetically (PK)-guided dosing of paclitaxel combined with carboplatin or cisplatin in patients with advanced non-small-cell lung cancer (NSCLC).

Markus Joerger; J. von Pawel; Stefanie Kraff; Juergen R. Fischer; Wilfried Eberhardt; Thomas Gauler; Lothar Mueller; Niels Reinmuth; M. Reck; Martin Kimmich; Frank Mayer; Hans-Georg Kopp; Dirk Behringer; Yon-Dschun Ko; Ralf A. Hilger; Max Roessler; C. Kloft; A. Henrich; Berta Moritz; M. C. Miller; S. J. Salamone; Ulrich Jaehde

BACKGROUND Variable chemotherapy exposure may cause toxicity or lack of efficacy. This study was initiated to validate pharmacokinetically (PK)-guided paclitaxel dosing in patients with advanced non-small-cell lung cancer (NSCLC) to avoid supra- or subtherapeutic exposure. PATIENTS AND METHODS Patients with newly diagnosed, advanced NSCLC were randomly assigned to receive up to 6 cycles of 3-weekly carboplatin AUC 6 or cisplatin 80 mg/m(2) either with standard paclitaxel at 200 mg/m(2) (arm A) or PK-guided dosing of paclitaxel (arm B). In arm B, initial paclitaxel dose was adjusted to body surface area, age, sex, and subsequent doses were guided by neutropenia and previous-cycle paclitaxel exposure [time above a plasma concentration of 0.05 µM (Tc>0.05)] determined from a single blood sample on day 2. The primary end point was grade 4 neutropenia; secondary end points included neuropathy, radiological response, progression-free survival (PFS) and overall survival (OS). RESULTS Among 365 patients randomly assigned, grade 4 neutropenia was similar in both arms (19% versus 16%; P = 0.10). Neuropathy grade ≥2 (38% versus 23%, P < 0.001) and grade ≥3 (9% versus 2%, P < 0.001) was significantly lower in arm B, independent of the platinum drug used. The median final paclitaxel dose was significantly lower in arm B (199 versus 150 mg/m(2), P < 0.001). Response rate was similar in arms A and B (31% versus 27%, P = 0.405), as was adjusted median PFS [5.5 versus 4.9 months, hazard ratio (HR) 1.16, 95% confidence interval (CI) 0.91-1.49, P = 0.228] and OS (10.1 versus 9.5 months, HR 1.05, 95% CI 0.81-1.37, P = 0.682). CONCLUSION PK-guided dosing of paclitaxel does not improve severe neutropenia, but reduces paclitaxel-associated neuropathy and thereby improves the benefit-risk profile in patients with advanced NSCLC. CLINICAL TRIAL INFORMATION NCT01326767 (https://clinicaltrials.gov/ct2/show/NCT01326767).


Therapeutic Drug Monitoring | 2015

Excel-Based Tool for Pharmacokinetically Guided Dose Adjustment of Paclitaxel.

Stefanie Kraff; Andreas Lindauer; Markus Joerger; Salvatore J. Salamone; Ulrich Jaehde

Background: Neutropenia is a frequent and severe adverse event in patients receiving paclitaxel chemotherapy. The time above a paclitaxel threshold concentration of 0.05 &mgr;mol/L (Tc > 0.05 &mgr;mol/L) is a strong predictor for paclitaxel-associated neutropenia and has been proposed as a target pharmacokinetic (PK) parameter for paclitaxel therapeutic drug monitoring and dose adaptation. Up to now, individual Tc > 0.05 &mgr;mol/L values are estimated based on a published PK model of paclitaxel by using the software NONMEM. Because many clinicians are not familiar with the use of NONMEM, an Excel-based dosing tool was developed to allow calculation of paclitaxel Tc > 0.05 &mgr;mol/L and give clinicians an easy-to-use tool. Methods: Population PK parameters of paclitaxel were taken from a published PK model. An Alglib VBA code was implemented in Excel 2007 to compute differential equations for the paclitaxel PK model. Maximum a posteriori Bayesian estimates of the PK parameters were determined with the Excel Solver using individual drug concentrations. Concentrations from 250 patients were simulated receiving 1 cycle of paclitaxel chemotherapy. Predictions of paclitaxel Tc > 0.05 &mgr;mol/L as calculated by the Excel tool were compared with NONMEM, whereby maximum a posteriori Bayesian estimates were obtained using the POSTHOC function. Results: There was a good concordance and comparable predictive performance between Excel and NONMEM regarding predicted paclitaxel plasma concentrations and Tc > 0.05 &mgr;mol/L values. Tc > 0.05 &mgr;mol/L had a maximum bias of 3% and an error on precision of <12%. The median relative deviation of the estimated Tc > 0.05 &mgr;mol/L values between both programs was 1%. Conclusions: The Excel-based tool can estimate the time above a paclitaxel threshold concentration of 0.05 &mgr;mol/L with acceptable accuracy and precision. The presented Excel tool allows reliable calculation of paclitaxel Tc > 0.05 &mgr;mol/L and thus allows target concentration intervention to improve the benefit–risk ratio of the drug. The easy use facilitates therapeutic drug monitoring in clinical routine.


Antimicrobial Agents and Chemotherapy | 2014

Pharmacokinetics of Ganciclovir during Continuous Venovenous Hemodiafiltration in Critically Ill Patients

Thomas Horvatits; Reinhard Kitzberger; Andreas Drolz; Christian Zauner; Walter Jäger; Michaela Böhmdorfer; Stefanie Kraff; Achim Fritsch; Florian Thalhammer; Valentin Fuhrmann; Peter Schenk

ABSTRACT Ganciclovir is an antiviral agent that is frequently used in critically ill patients with cytomegalovirus (CMV) infections. Continuous venovenous hemodiafiltration (CVVHDF) is a common extracorporeal renal replacement therapy in intensive care unit patients. The aim of this study was to investigate the pharmacokinetics of ganciclovir in anuric patients undergoing CVVHDF. Population pharmacokinetic analysis was performed for nine critically ill patients with proven or suspected CMV infection who were undergoing CVVHDF. All patients received a single dose of ganciclovir at 5 mg/kg of body weight intravenously. Serum and ultradiafiltrate concentrations were assessed by high-performance liquid chromatography, and these data were used for pharmacokinetic analysis. Mean peak and trough prefilter ganciclovir concentrations were 11.8 ± 3.5 mg/liter and 2.4 ± 0.7 mg/liter, respectively. The pharmacokinetic parameters elimination half-life (24.2 ± 7.6 h), volume of distribution (81.2 ± 38.3 liters), sieving coefficient (0.76 ± 0.1), total clearance (2.7 ± 1.2 liters/h), and clearance of CVVHDF (1.5 ± 0.2 liters/h) were determined. Based on population pharmacokinetic simulations with respect to a target area under the curve (AUC) of 50 mg · h/liter and a trough level of 2 mg/liter, a ganciclovir dose of 2.5 mg/kg once daily seems to be adequate for anuric critically ill patients during CVVHDF.


Journal of Pharmacology and Experimental Therapeutics | 2017

Semi-mechanistic bone marrow exhaustion pharmacokinetic/pharmacodynamic model for chemotherapy-induced cumulative neutropenia

Andrea Henrich; Markus Joerger; Stefanie Kraff; Ulrich Jaehde; Wilhelm Huisinga; Charlotte Kloft; Zinnia P. Parra-Guillen

Paclitaxel is a commonly used cytotoxic anticancer drug with potentially life-threatening toxicity at therapeutic doses and high interindividual pharmacokinetic variability. Thus, drug and effect monitoring is indicated to control dose-limiting neutropenia. Joerger et al. (2016) developed a dose individualization algorithm based on a pharmacokinetic (PK)/pharmacodynamic (PD) model describing paclitaxel and neutrophil concentrations. Furthermore, the algorithm was prospectively compared in a clinical trial against standard dosing (Central European Society for Anticancer Drug Research Study of Paclitaxel Therapeutic Drug Monitoring; 365 patients, 720 cycles) but did not substantially improve neutropenia. This might be caused by misspecifications in the PK/PD model underlying the algorithm, especially without consideration of the observed cumulative pattern of neutropenia or the platinum-based combination therapy, both impacting neutropenia. This work aimed to externally evaluate the original PK/PD model for potential misspecifications and to refine the PK/PD model while considering the cumulative neutropenia pattern and the combination therapy. An underprediction was observed for the PK (658 samples), the PK parameters, and these parameters were re-estimated using the original estimates as prior information. Neutrophil concentrations (3274 samples) were overpredicted by the PK/PD model, especially for later treatment cycles when the cumulative pattern aggravated neutropenia. Three different modeling approaches (two from the literature and one newly developed) were investigated. The newly developed model, which implemented the bone marrow hypothesis semiphysiologically, was superior. This model further included an additive effect for toxicity of carboplatin combination therapy. Overall, a physiologically plausible PK/PD model was developed that can be used for dose adaptation simulations and prospective studies to further improve paclitaxel/carboplatin combination therapy.


Antimicrobial Agents and Chemotherapy | 2014

Single dose pharmacokinetics of cidofovir in continuous venovenous hemofiltration

Matthias G. Vossen; Klaus-Bernhard Gattringer; Walter Jäger; Stefanie Kraff; Florian Thalhammer

ABSTRACT Dosage recommendations for cidofovir are available for renally competent as well as impaired patients; however, there are no data for patients undergoing continuous renal replacement therapy. We determined the single-dose concentration-versus-time profile of cidofovir in a critically ill patient undergoing continuous venovenous hemofiltration (CVVH). One dose of 450 mg cidofovir (5 mg/kg) was administered intravenously due to a proven cytomegalovirus (CMV) infection and failure of first-line antiviral therapy. Additionally, 2 g of probenecid was administered orally 3 h prior to and 1 g was administered 2 h as well as 8 h after completion of the infusion. The concentrations of cidofovir in serum and ultrafiltrate were assessed by high-performance liquid chromatography. The peak serum concentration measured at 60 min postinfusion was 28.01 mg/liter at the arterial port. The trough serum level was 19.33 mg/liter at the arterial port after 24 h. The value of the area under the concentration-versus-time curve from 0 to 24 h was 543.8 mg · h/liter. The total body clearance was 2.46 ml/h/kg, and the elimination half-life time was 53.32 h. The sieving coefficient was 0.138 ± 0.022. Total removal of the drug was 30.99% after 24 h. Because of these data, which give us a rough idea of the concentration profile of cidofovir in patients undergoing CVVH, a toxic accumulation of the drug following repeated doses may be expected. Further trials have to be done to determine the right dosage of cidofovir in patients undergoing CVVH to avoid toxic accumulation of the drug.


Cancer Chemotherapy and Pharmacology | 2015

Pharmacokinetically based dosing of weekly paclitaxel to reduce drug‑related neurotoxicity based on a single sample strategy

Stefanie Kraff; Annemieke J.M. Nieuweboer; Ron H.J. Mathijssen; Florent Baty; Anne-Joy M. de Graan; Ron H.N. van Schaik; Ulrich Jaehde; Markus Joerger


Therapeutic Drug Monitoring | 2017

Validation of a commercial assay and decision support tool for routine paclitaxel therapeutic drug monitoring (TDM).

Markus Joerger; Stefanie Kraff; Ulrich Jaehde; Ralf A. Hilger; Jodi Blake Courtney; Daniel J. Cline; Sonali Jog; Irina Baburina; Michael Craig Miller; Salvatore J. Salamone


Journal of Clinical Oncology | 2017

A prospective, multi-center study of individualized, pharmacokinetically (PK)-guided dosing of 5-fluorouracil (5-FU) in metastatic colorectal cancer (mCRC) patients treated with weekly or biweekly 5-FU/oxaliplatin containing regimens.

Volker Kunzmann; Karin Link; Michael Craig Miller; Stefan Holdenrieder; Thomas Bertsch; Lothar Mueller; Yon-Dschun Ko; Oliver J. Stoetzer; Ingo Suttmann; Jan Braess; Ulrich Jaehde; Max Roessler; Berta Moritz; Stefanie Kraff; Achim Fritsch; Salvatore J. Salamone; Martin Wilhelm

Collaboration


Dive into the Stefanie Kraff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Markus Joerger

University of St. Gallen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ralf A. Hilger

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Thomas Gauler

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Niels Reinmuth

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge