Neeltje Steeghs
Netherlands Cancer Institute
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Neeltje Steeghs.
Clinical Cancer Research | 2008
Neeltje Steeghs; Hans Gelderblom; Jos op 't Roodt; Olaf Christensen; Prabhu Rajagopalan; Marcel Hovens; Hein Putter; Ton J. Rabelink; Eelco J.P. de Koning
Purpose: Hypertension is a commonly reported side effect in antiangiogenic therapy. We investigated the hypothesis that telatinib, a small molecule angiogenesis inhibitor, impairs vascular function, induces rarefaction, and causes hypertension. Experimental Design: A side-study was done in a phase I trial of telatinib, a small molecule tyrosine kinase inhibitor of vascular endothelial growth factor receptors 2 and 3, platelet-derived growth factor receptor, and c-KIT in patients with advanced solid tumors. Measurements of blood pressure, flow-mediated dilation, nitroglycerin-mediated dilation, aortic pulse wave velocity, skin blood flux with laser Doppler flow, and capillary density with sidestream dark field imaging were done at baseline and after 5 weeks of treatment. Blood pressure and proteinuria were measured weekly. Results: Mean systolic and diastolic blood pressure values increased significantly at +6.6 mm Hg (P = 0.009) and +4.7 mm Hg (P = 0.016), respectively. Mean flow-mediated dilation and mean nitroglycerin-mediated dilation values significantly decreased by −2.1% (P = 0.003) and −5.1% (P = 0.001), respectively. After 5 weeks of treatment, mean pulse wave velocity significantly increased by 1.2 m/s (P = 0.001). A statistically significant reduction of mean skin blood flux of 532.8% arbitrary units was seen (P = 0.015). Capillary density statistically significantly decreased from 20.8 to 16.7 capillary loops (P = 0.015). Proteinuria developed or increased in six patients during telatinib treatment. Conclusion: The increase in blood pressure observed in the treatment with telatinib, an angiogenesis inhibitor, may be caused by functional or structural rarefaction.
Clinical Pharmacology & Therapeutics | 2014
M H M Diekstra; Heinz-Josef Klümpen; M P J K Lolkema; Huixin Yu; Jacqueline S. L. Kloth; Hans Gelderblom; R.H.N. van Schaik; Howard Gurney; Jesse J. Swen; Alwin D. R. Huitema; Neeltje Steeghs; Ron H.J. Mathijssen
Interpatient variability in the pharmacokinetics (PK) of sunitinib is high. Single nucleotide polymorphisms (SNPs) in PK candidate genes have been associated with the efficacy and toxicity of sunitinib, but whether these SNPs truly affect the PK of sunitinib remains to be elucidated. This multicenter study involving 114 patients investigated whether these SNPs and haplotypes in genes encoding metabolizing enzymes or efflux transporters are associated with the clearance of sunitinib and its active metabolite SU12662. SNPs were tested as covariates in a population PK model. From univariate analysis, we found that the SNPs in CYP3A4, CYP3A5, and ABCB1 were associated with the clearance of both sunitinib and SU12662. In multivariate analysis, CYP3A4*22 was found to be eliminated last with an effect size of −22.5% on clearance. Observed effect sizes are below the interindividual variability in clearance and are therefore too limited to directly guide individual dosing of sunitinib.
Therapeutic Drug Monitoring | 2014
Nienke A.G. Lankheet; Lotte M Knapen; Jan H. M. Schellens; Jos H. Beijnen; Neeltje Steeghs; Alwin D. R. Huitema
Background: The objectives of this study were to evaluate the plasma concentrations of the tyrosine kinase inhibitors (TKIs), imatinib, erlotinib, and sunitinib, in a cohort of patients with cancer in routine clinical practice and to find the possible factors related to plasma concentrations below the target level. Methods: An observational study was performed in an unselected cohort of patients using TKIs for cancer treatment. Randomly timed plasma samples were drawn together with regular laboratory investigations during routine outpatient clinic visits. The plasma concentrations of TKIs were determined using a validated high-performance liquid chromatography coupled with tandem mass spectrometry detection method. Trough concentrations were estimated using the interval between the last dose intake and blood sampling and the mean elimination half-life of the TKIs and were compared with target trough concentrations. Outpatient medical records were reviewed to collect data on patient- and medication-related factors that could have contributed to the variation in TKI plasma concentrations. Results: Only 26.8%, 88.9%, and 51.4% of the calculated trough plasma concentrations of imatinib, erlotinib, and sunitinib samples, respectively, reached the predefined target concentration (imatinib: 1100 ng/mL, erlotinib: 500 ng/mL, and sunitinib: 50 ng/mL). Interpatient variability was high with coefficients of variation of 39.1%, 40.1%, and 29.2% for imatinib, erlotinib, and sunitinib, respectively. High variation in plasma concentrations could only partly be explained by patient- or medication related factors. Conclusions: Almost half of the plasma concentrations in the outpatient population seemed to be below the target level with a risk of treatment failure. It is not possible to predict which patients are at a risk of plasma concentrations below the target level based on patient- or medication-related factors. Thus, therapeutic drug monitoring could play a crucial role in routine cancer care to identify patients that are in need of individual adjusted dosages. Further research is required to investigate the safety and efficacy of therapeutic drug monitoring.
Journal of Clinical Oncology | 2009
Ferry A.L.M. Eskens; Neeltje Steeghs; Jaap Verweij; Johan L. Bloem; Olaf Christensen; Leni van Doorn; Jan Ouwerkerk; Maja J.A. de Jonge; Johan W.R. Nortier; Joern Kraetzschmar; Prabhu Rajagopalan; Hans Gelderblom
PURPOSE Telatinib (BAY 57-9352) is an orally available tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR) -2, VEGFR-3, platelet-derived growth factor receptor-beta, and c-Kit. This phase I dose escalation study was conducted to evaluate the safety and tolerability of telatinib, with additional pharmacokinetic, pharmacodynamic, and efficacy assessments. PATIENTS AND METHODS Patients with solid tumors refractory to standard therapies or with no standard therapy available were enrolled. Doses of continuously administered telatinib were escalated from 20 mg once daily to 1,500 mg twice daily. RESULTS Fifty-three patients were enrolled. Most frequently observed drug-related adverse events were nausea (26.4%; grade >or= 3, 0%) and hypertension (20.8%; grade 3, 11.3%; grade 4, 0%). Two dose-limiting toxicities were observed: one poorly controlled hypertension (600 mg twice daily), and one grade 2 weight loss, anorexia, and fatigue (1,500 mg twice daily). A formal maximum-tolerated dose was not reached. Telatinib was rapidly absorbed, with median time to peak concentration (t(max)) lower than 3 hours after dose. A nearly dose-proportional increase in exposure was observed with substantial variability. Telatinib half-life averaged 5.5 hours. Biomarker analyses showed dose-dependent increase in VEGF levels and decrease in plasma soluble VEGFR-2 levels, with a plateau at 900 mg twice daily. A decrease in tumor blood flow (K(trans) and IAUC(60)) was observed with dynamic contrast-enhanced magnetic resonance imaging. Best tumor response was stable disease, observed in 50.9% of patients. CONCLUSION Telatinib was safe and well tolerated up to 1,500 mg twice daily. Based on pharmacodynamic and pharmacokinetic end points, telatinib 900 mg twice daily is the recommended dose for subsequent phase II studies.
British Journal of Cancer | 2015
Ruud van der Noll; Serena Marchetti; Neeltje Steeghs; Jos H. Beijnen; Marja Mergui-Roelvink; Emmy Harms; Harriet Rehorst; Gabe S. Sonke; Jan H. M. Schellens
Background:Olaparib (AZD2281), a PARP-1/2 inhibitor, has been extensively investigated in clinical trials. However, limited clinical data are available about its long-term safety and anti-tumour activity.Methods:Patients had first participated in a phase I study of olaparib combined with carboplatin and/or paclitaxel. They continued with olaparib monotherapy in their best interest if they failed to tolerate the combination due to the treatment-related adverse events (TRAEs). Safety data were collected by physical examination and regular laboratory evaluations. Disease evaluations were performed by CT scan.Results:At data cutoff, 21 patients were included; 10 with breast, 9 with ovarian and 2 with fallopian tube cancer of whom 16 patients had a BRCA mutation (13 BRCA1; 3 BRCA2). TRAEs were mostly haematological and most prominent shortly after switching from combination to monotherapy, probably due to carry-over effects of chemotherapy. Over time, both severity and frequency of TRAEs decreased. Responses to olaparib were durable with a median treatment duration of 52 (range 7–183) weeks. In total, nine (43%) patients were still on study at data cutoff.Conclusion:Continued long-term daily olaparib was found to be safe and tolerable. Encouragingly, patients who showed a favourable response on earlier combination therapy maintained this response on olaparib monotherapy.
British Journal of Cancer | 2015
Jacqueline S. L. Kloth; Ambrogio Pagani; Michiel C. Verboom; Alberto Malovini; Carlo Napolitano; Wim H. J. Kruit; Stefan Sleijfer; Neeltje Steeghs; Alberto Zambelli; Ron H.J. Mathijssen
Background:Tyrosine kinase inhibitors (TKIs) are associated with prolongation of the QTc interval on the electrocardiogram (ECG). The QTc-interval prolongation increases the risk of life-threatening arrhythmias. However, studies evaluating the effects of TKIs on QTc intervals are limited and only consist of small patient numbers.Methods:In this multicentre trial in four centres in the Netherlands and Italy we screened all patients who were treated with any TKI. To evaluate the effects of TKIs on the QTc interval, we investigated ECGs before and during treatment with erlotinib, gefitinib, imatinib, lapatinib, pazopanib, sorafenib, sunitinib, or vemurafenib.Results:A total of 363 patients were eligible for the analyses. At baseline measurement, QTc intervals were significantly longer in females than in males (QTcfemales=404 ms vs QTcmales=399 ms, P=0.027). A statistically significant increase was observed for the individual TKIs sunitinib, vemurafenib, sorafenib, imatinib, and erlotinib, after the start of treatment (median ΔQTc ranging from +7 to +24 ms, P<0.004). The CTCAE grade for QTc intervals significantly increased after start of treatment (P=0.0003). Especially patients who are treated with vemurafenib are at increased risk of developing a QTc of ⩾470 ms, a threshold associated with an increased risk for arrhythmias.Conclusions:These observations show that most TKIs significantly increase the QTc interval. Particularly in vemurafenib-treated patients, the incidence of patients at risk for arrhythmias is increased. Therefore, especially in case of combined risk factors, ECG monitoring in patients treated with TKIs is strongly recommended.
Acta Oncologica | 2015
Rick L. Haas; Hans Gelderblom; Stefan Sleijfer; Hester van Boven; Astrid Scholten; Luc Dewit; Gerben R. Borst; Jos A. van der Hage; Martijn Kerst; Remi A. Nout; Henk H. Hartgrink; Ilse de Pree; Kees Verhoef; Neeltje Steeghs; Frits van Coevorden
Abstract Accumulating evidence suggests significant synergism combining radiotherapy (RT) with angiogenesis targeted therapies. This multicenter prospective phase I clinical trial established the safety profile and recommended dose for further studies of pazopanib concurrent with preoperative RT in patients with extremity soft tissue sarcomas (ESTS) in curative setting. Methods. Patients with deep seated intermediate and high grade sarcomas, ≥ 5 cm, received once daily pazopanib (dose-escalation cohorts 400 mg, 600 mg and 800 mg) for 6 weeks and 50 Gy preoperative RT starting Day 8. Surgery was performed 5–7 weeks later. Toxicity was scored according to CTC criteria 4.0. Dose limiting toxicities (DLT) were divided into two separate sets; DLT-I being toxicities occurring during the 6-week chemoradiotherapy period within the radiation portals until day of surgery (designated as DLT-I) and those occurring perioperatively until Day 21 after surgery (DLT-II). Results. A total of 12 patients were enrolled, 11 were evaluable (3 females and 8 males, median age 58 years, range 24–78 years, median tumor size 9 cm, range 5–15 cm). Ten underwent surgery. No increased toxicity inside the radiation fields was seen, but two of 10 patients (one each in the 400 mg and 600 mg cohorts) showed delayed wound healing after surgery. None of the patients showed significant volume reductions after RT. Evaluation of the resection specimen showed pathological (near) complete responses (≥ 95% necrosis rate) in four of 10 cases. Unexpectedly, grade 3 + hepatotoxicity led to premature pazopanib interruption in three of 11 (27%) of cases. Conclusion. Apart from hepatotoxicity, neoadjuvant pazopanib 800 mg daily in combination with 50 Gy seems tolerable; the regimen appears to demonstrate promising activity in ESTS and is the recommended dose for further studies.
PLOS ONE | 2016
Wendy de Leng; Christa G. Gadellaa-van Hooijdonk; Françoise A. S. Barendregt-Smouter; Marco J. Koudijs; Ies Nijman; John W. J. Hinrichs; Edwin Cuppen; Stef van Lieshout; Robert Loberg; Maja J.A. de Jonge; Emile E. Voest; Roel A. de Weger; Neeltje Steeghs; Marlies H.G. Langenberg; Stefan Sleijfer; Stefan M. Willems; Martijn P. Lolkema
Background Targeted Next Generation Sequencing (NGS) offers a way to implement testing of multiple genetic aberrations in diagnostic pathology practice, which is necessary for personalized cancer treatment. However, no standards regarding input material have been defined. This study therefore aimed to determine the effect of the type of input material (e.g. formalin fixed paraffin embedded (FFPE) versus fresh frozen (FF) tissue) on NGS derived results. Moreover, this study aimed to explore a standardized analysis pipeline to support consistent clinical decision-making. Method We used the Ion Torrent PGM sequencing platform in combination with the Ion AmpliSeq Cancer Hotspot Panel v2 to sequence frequently mutated regions in 50 cancer related genes, and validated the NGS detected variants in 250 FFPE samples using standard diagnostic assays. Next, 386 tumour samples were sequenced to explore the effect of input material on variant detection variables. For variant calling, Ion Torrent analysis software was supplemented with additional variant annotation and filtering. Results Both FFPE and FF tissue could be sequenced reliably with a sensitivity of 99.1%. Validation showed a 98.5% concordance between NGS and conventional sequencing techniques, where NGS provided both the advantage of low input DNA concentration and the detection of low-frequency variants. The reliability of mutation analysis could be further improved with manual inspection of sequence data. Conclusion Targeted NGS can be reliably implemented in cancer diagnostics using both FFPE and FF tissue when using appropriate analysis settings, even with low input DNA.
Acta Oncologica | 2017
A. Kollar; Robin L. Jones; Silvia Stacchiotti; Hans Gelderblom; M. Guida; Giovanni Grignani; Neeltje Steeghs; Akmal Safwat; Daniela Katz; Florence Duffaud; Stefan Sleijfer; W.T.A. van der Graaf; Nathan Touati; Saskia Litière; Sandrine Marreaud; Alessandro Gronchi; Bernd Kasper
Abstract Background: Pazopanib is a multitargeted tyrosine kinase inhibitor approved for the treatment of patients with selective subtypes of advanced soft tissue sarcoma (STS) who have previously received standard chemotherapy including anthracyclines. Data on the efficacy in vascular sarcomas are limited. The main objective of this study was to investigate the activity of pazopanib in vascular sarcomas. Patients and methods: A retrospective study of patients with advanced vascular sarcomas, including angiosarcoma (AS), epithelioid hemangioendothelioma (HE) and intimal sarcoma (IS) treated with pazopanib in real life practice at EORTC centers as well as patients treated within the EORTC phase II and III clinical trials (62043/62072) was performed. Patient and tumor characteristics were collected. Response was assessed according to RECIST 1.1. and survival analysis was performed. Results: Fifty-two patients were identified, 40 (76.9%), 10 (19.2%) and two (3.8%) with AS, HE and IS, respectively. The response rate was eight (20%), two (20%) and two (100%) in the AS, HE and IS subtypes, respectively. There was no significant difference in response rate between cutaneous and non-cutaneous AS and similarly between radiation-associated and non-radiation-associated AS. Median progression-free survival (PFS) and median overall survival (OS; from commencing pazopanib) were three months (95% CI 2.1–4.4) and 9.9 months (95% CI 6.5–11.3) in AS, respectively. Conclusion: The activity of pazopanib in AS is comparable to its reported activity in other STS subtypes. In this study, the activity of pazopanib was similar in cutaneous/non-cutaneous and in radiation/non-radiation-associated AS. In addition, pazopanib showed promising activity in HE and IS, worthy of further evaluation.
Clinical Cancer Research | 2016
Remy B. Verheijen; Sander Bins; Ron H.J. Mathijssen; Martijn P. Lolkema; Leni van Doorn; Jan H. M. Schellens; Jos H. Beijnen; Marlies H.G. Langenberg; Alwin D. R. Huitema; Neeltje Steeghs
Purpose: Pazopanib is a tyrosine kinase inhibitor approved for the treatment of renal cell carcinoma and soft tissue sarcoma. Retrospective analyses have shown that an increased median progression-free survival and tumor shrinkage appear in patients with higher plasma trough levels (Cmin). Therefore, patients with low Cmin might benefit from pharmacokinetically guided individualized dosing. Experimental Design: We conducted a prospective multicenter trial in 30 patients with advanced solid tumors. Pazopanib Cmin was measured weekly by LC-MS/MS. At weeks 3, 5, and 7, the pazopanib dose was increased if the measured Cmin was <20 mg/L and toxicity was <grade 3. Results: In total, 17 patients had at least one Cmin <20 mg/L at weeks 3, 5, and 7. Of these, 10 were successfully treated with a pharmacokinetically guided dose escalation, leading to daily dosages ranging from 1,000 to 1,800 mg. Cmin in these patients increased significantly from 13.2 (38.0%) mg/L [mean (CV%)] to 22.9 mg/L (44.9%). Thirteen patients had all Cmin levels ≥20.0 mg/L. Of these, 9 patients with a high Cmin of 51.3 mg/L (45.1%) experienced ≥grade 3 toxicity and subsequently required a dose reduction to 600 or 400 mg daily, yet in these patients, Cmin remained above the threshold at 28.2 mg/L (25.3%). Conclusions: A pharmacokinetically guided individualized dosing algorithm was successfully applied and evaluated. The dosing algorithm led to patients being treated at dosages ranging from 400 to 1,800 mg daily. Further studies are needed to show a benefit of individualized dosing on clinical outcomes, such as progression-free survival. Clin Cancer Res; 22(23); 5738–46. ©2016 AACR. See related commentary by Ornstein and Rini, p. 5626