Network


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

Hotspot


Dive into the research topics where Hans Gelderblom is active.

Publication


Featured researches published by Hans Gelderblom.


Journal of Clinical Oncology | 2012

Intrapatient Cetuximab Dose Escalation in Metastatic Colorectal Cancer According to the Grade of Early Skin Reactions: The Randomized EVEREST Study

Eric Van Cutsem; Sabine Tejpar; Dirk Vanbeckevoort; Marc Peeters; Yves Humblet; Hans Gelderblom; Jan B. Vermorken; F. Viret; Bengt Glimelius; Elisa Gallerani; Alain Hendlisz; Annemieke Cats; Markus Moehler; Xavier Sagaert; Soetkin Vlassak; Michael Schlichting; Fortunato Ciardiello

PURPOSEnSkin toxicity in patients receiving cetuximab has been associated positively with clinical outcome in several tumor types. This study investigated the effect of cetuximab dose escalation in patients with irinotecan-refractory metastatic colorectal cancer who had developed no or mild skin reactions after 21 days of treatment at the standard dose. This article reports clinical and pharmacokinetic (PK) data.nnnPATIENTS AND METHODSnAfter 21 days of standard-dose cetuximab (400 mg/m(2) initial dose, then 250 mg/m(2) per week) plus irinotecan, patients with ≤ grade 1 skin reactions were randomly assigned to standard-dose (group A) or dose-escalated (to 500 mg/m(2) per week; group B) cetuximab. Patients with ≥ grade 2 skin reactions continued on standard-dose cetuximab plus irinotecan (group C).nnnRESULTSnThe intent-to-treat population comprised 157 patients. PK profiles reflected the dose increase and were predictable across the dose range investigated. Weekly cetuximab doses of up to 500 mg/m(2) were well tolerated, and grade 3 and 4 adverse events were generally comparable between treatment groups. Dose escalation (n = 44) was associated with an increase in skin reactions ≥ grade 2 compared with standard (n = 45) dosing (59% v 38%, respectively). Dose escalation, compared with standard dosing, showed some evidence for improved response rate (30% v 16%, respectively) and disease control rate (70% v 58%, respectively) but no indication of benefit in relation to overall survival. In an exploratory analysis, dose escalation seemed to increase response rate compared with standard dosing in patients with KRAS wild-type but not KRAS mutant tumors.nnnCONCLUSIONnCetuximab serum concentrations increased predictably with dose. Higher dose levels were well tolerated. The possible indication for improved efficacy in the dose-escalation group warrants further investigation.


Journal of Virology | 2005

Molecular and Biological Characterization of Human Monoclonal Antibodies Binding to the Spike and Nucleocapsid Proteins of Severe Acute Respiratory Syndrome Coronavirus

Edward Norbert van den Brink; Jan ter Meulen; Freek Cox; Mandy Jongeneelen; Alexandra Thijsse; Mark Throsby; Wilfred E. Marissen; Pauline M.L. Rood; Alexander Berthold Hendrik Bakker; Hans Gelderblom; Byron E. E. Martina; Albert D. M. E. Osterhaus; Wolfgang Preiser; Hans Wilhelm Doerr; John de Kruif; Jaap Goudsmit

ABSTRACT Human monoclonal antibodies (MAbs) were selected from semisynthetic antibody phage display libraries by using whole irradiated severe acute respiratory syndrome (SARS) coronavirus (CoV) virions as target. We identified eight human MAbs binding to virus and infected cells, six of which could be mapped to two SARS-CoV structural proteins: the nucleocapsid (N) and spike (S) proteins. Two MAbs reacted with N protein. One of the N protein MAbs recognized a linear epitope conserved between all published human and animal SARS-CoV isolates, and the other bound to a nonlinear N epitope. These two N MAbs did not compete for binding to SARS-CoV. Four MAbs reacted with the S glycoprotein, and three of these MAbs neutralized SARS-CoV in vitro. All three neutralizing anti-S MAbs bound a recombinant S1 fragment comprising residues 318 to 510, a region previously identified as the SARS-CoV S receptor binding domain; the nonneutralizing MAb did not. Two strongly neutralizing anti-S1 MAbs blocked the binding of a recombinant S fragment (residues 1 to 565) to SARS-CoV-susceptible Vero cells completely, whereas a poorly neutralizing S1 MAb blocked binding only partially. The MAb ability to block S1-receptor binding and the level of neutralization of the two strongly neutralizing S1 MAbs correlated with the binding affinity to the S1 domain. Finally, epitope mapping, using recombinant S fragments (residues 318 to 510) containing naturally occurring mutations, revealed the importance of residue N479 for the binding of the most potent neutralizing MAb, CR3014. The complete set of SARS-CoV MAbs described here may be useful for diagnosis, chemoprophylaxis, and therapy of SARS-CoV infection and disease.


Journal of Clinical Microbiology | 2004

First International Quality Assurance Study on the Rapid Detection of Viral Agents of Bioterrorism

Matthias Niedrig; Herbert Schmitz; Stephan Becker; Stephan Günther; Jan ter Meulen; Herman Meyer; Heinz Ellerbrok; Andreas Nitsche; Hans Gelderblom; Christian Drosten

ABSTRACT We have conducted an international quality assurance study of filovirus, Lassa virus, and orthopox virus PCR with 24 participants. Of the participating laboratories, 45.8 and 66.7% detected virus in all plasma samples, which contained ≥5,000 and ≥100,000 copies per ml, respectively. Sensitivity levels were not significantly different between viruses. False-negative results were attributable to a lack of sensitivity.


European Journal of Cancer | 2013

An open-label, phase 2 study evaluating the efficacy and safety of the anti-IGF-1R antibody cixutumumab in patients with previously treated advanced or metastatic soft-tissue sarcoma or Ewing family of tumours

Patrick Schöffski; Douglas D. Adkins; Jean Yves Blay; Thierry Gil; Anthony Elias; Piotr Rutkowski; Gregory K. Pennock; Hagop Youssoufian; Hans Gelderblom; R. R. Willey; Dmitri Grebennik

BACKGROUNDnCixutumumab (IMC-A12), a fully human immunoglobulin G1 (IgG1) monoclonal antibody, exerts preclinical activity in several sarcoma models and may be effective for the treatment of these tumours.nnnMETHODSnIn this open-label, multicentre, phase 2 study, patients with previously treated advanced or metastatic rhabdomyosarcoma, leiomyosarcoma, adipocytic sarcoma, synovial sarcoma or Ewing family of tumours received intravenous cixutumumab (10mg/kg) for 1h every other week until disease progression or discontinuation. The primary end-point was the progression-free survival rate (PFR), defined as stable disease or better at 12 weeks. In each tier of disease histology, Simons optimum 2-stage design was applied (PFR at 12 weeks P0=20%, P1=40%, α=0.10, β=0.10). Stage 1 enrolled 17 patients in each disease group/tier, with at least four patients with stable disease or better required at 12 weeks to proceed to stage 2.nnnRESULTSnA total of 113 patients were enrolled; all tiers except adipocytic sarcoma were closed after stage 1 due to futility. The 12-week PFR was 12% for rhabdomyosarcoma (n=17), 14% for leiomyosarcoma (n=22), 32% for adipocytic sarcoma (n=37), 18% for synovial sarcoma (n=17) and 11% for Ewing family of tumours (n=18). Median progression-free survival (weeks) was 6.1 for rhabdomyosarcoma, 6.0 for leiomyosarcoma, 12.1 for adipocytic sarcoma, 6.4 for synovial sarcoma and 6.4 for Ewing family of tumours. Among all patients, the most frequent treatment-emergent adverse events (AEs) were nausea (26%), fatigue (23%), diarrhoea (23%) and hyperglycaemia (20%).nnnCONCLUSIONSnPatients with adipocytic sarcoma may benefit from treatment with cixutumumab. Cixutumumab treatment was well tolerated, with limited gastrointestinal AEs, fatigue and hyperglycaemia.


Cancer Chemotherapy and Pharmacology | 2009

Mechanistic population pharmacokinetics of total and unbound paclitaxel for a new nanodroplet formulation versus Taxol in cancer patients

Jürgen B. Bulitta; Ping Zhao; Robert D. Arnold; Dean R. Kessler; Richard Daifuku; James Pratt; Gabriel Luciano; A.-R. Hanauske; Hans Gelderblom; Ahmad Awada; William J. Jusko

PurposeOur objectives were (1) to compare the disposition and in vivo release of paclitaxel between a tocopherol-based Cremophor-free formulation (Tocosol Paclitaxel®) and Cremophor® EL-formulated paclitaxel (Taxol®) in human subjects, and (2) to develop a mechanistic model for unbound and total paclitaxel pharmacokinetics.MethodsA total of 35 patients (averagexa0±xa0SD age: 59xa0±13xa0years) with advanced non-hematological malignancies were studied in a randomized two-way crossover trial. Patients received 175xa0mg/m2 paclitaxel as 15xa0min (Tocosol Paclitaxel) or 3xa0h (Taxol) intravenous infusion in each study period. Paclitaxel concentrations were determined by LC–MS/MS in plasma ultrafiltrate and whole blood. NONMEM VI was used for population pharmacokinetics.ResultsA linear disposition model with three compartments for unbound paclitaxel and a one-compartment model for Cremophor were applied. Total clearance of unbound paclitaxel was 845xa0L/h (variability: 25% CV). The prolonged release with Tocosol Paclitaxel was explained by the limited solubility of unbound paclitaxel of 405xa0ng/mL (estimated) in plasma. The 15xa0min Tocosol Paclitaxel infusion yielded a mean time to 90% cumulative input of 1.14xa0±xa00.16xa0h. Tocosol Paclitaxel was estimated to release 9.8% of the dose directly into the deep peripheral compartment. The model accounted for the presence of drug-containing nanodroplets in blood.ConclusionsPopulation pharmacokinetic analysis indicated linear disposition and a potentially higher bioavailability of unbound paclitaxel following Tocosol Paclitaxel administration due to direct release at the target site. The prolonged release of Tocosol Paclitaxel supports 15xa0min paclitaxel infusions. This mechanistic model may be important for development of prolonged release formulations that distribute in and from the systemic circulation.


Cancer Chemotherapy and Pharmacology | 2009

Multiple-pool cell lifespan models for neutropenia to assess the population pharmacodynamics of unbound paclitaxel from two formulations in cancer patients

Jiirgen B. Bulitta; Ping Zhao; Robert D. Arnold; Dean R. Kessler; Richard Daifuku; James Pratt; Gabriel Luciano; A.-R. Hanauske; Hans Gelderblom; Ahmad Awada; William J. Jusko

PurposeOur objective was to build a mechanism-based pharmacodynamic model for the time course of neutropenia in cancer patients following paclitaxel treatment with a tocopherol-based Cremophor-free formulation (Tocosol Paclitaxel®) and Cremophor® EL-formulated paclitaxel (Taxol®).MethodsA randomized two-way crossover trial was performed with 35 adult patients who received 175xa0mg/m2 paclitaxel as either 15xa0min (Tocosol Paclitaxel) or 3xa0h (Taxol) intravenous infusions. Paclitaxel concentrations were measured by LC–MS/MS. NONMEM VI was used for population pharmacodynamics.ResultsThe cytotoxic effect on neutrophils was described by four mechanism-based models predicated on known properties of paclitaxel that used unbound concentrations in the central, deep peripheral or an intracellular compartment as forcing functions. Tocosol Paclitaxel was estimated to release 9.8% of the dose directly into the deep peripheral compartment (DPC). All models provided reasonable fitting of neutropenic effects. The model with the best predictive performance assumed that this dose fraction was released into 22.5% of the DPC which included the site of toxicity. The second-order cytotoxic rate constant was 0.00211xa0mL/ng per hour (variability: 52% CV). The relative exposure at the site of toxicity was 2.21xa0±xa00.41 times (averagexa0±xa0SD) larger for Tocosol Paclitaxel compared to Taxol. Lifespan was 11.0xa0days for progenitor cells, 1.95xa0days for maturating cells, and 4.38xa0days for neutrophils. Total drug exposure in blood explained half of the variance in nadir to baseline neutrophil count ratio.ConclusionsThe relative exposure of unbound paclitaxel at the site of toxicity was twice as large for Tocosol Paclitaxel compared to Taxol. The proposed mechanism-based models explained the extent and time course of neutropenia jointly for both formulations.


Journal of Virology | 2003

Replication-Competent Hybrids between Murine Leukemia Virus and Foamy Virus

Evelina Shikova-Lekova; Dirk Lindemann; Thomas Pietschmann; Thomas Juretzek; Wolfram W. Rudolph; Ottmar Herchenröder; Hans Gelderblom; Axel Rethwilm

ABSTRACT Replication-competent chimeric retroviruses constructed of members of the two subfamilies of Retroviridae, orthoretroviruses and spumaretroviruses, specifically murine leukemia viruses (MuLV) bearing hybrid MuLV-foamy virus (FV) envelope (env) genes, were characterized. All viruses had the cytoplasmic tail of the MuLV transmembrane protein. In ESL-1, a truncated MuLV leader peptide (LP) was fused to the complete extracellular portion of FV Env, and ESL-2 to -4 contained the complete MuLV-LP followed by N-terminally truncated FV Env decreasing in size. ESL-1 to -4 had an extended host cell range compared to MuLV, induced a cytopathology reminiscent of FVs, and exhibited an ultrastructure that combined the features of the condensed core of MuLV with the prominent surface knobs of FVs. Replication of ESL-2 to -4 resulted in the acquisition of a stop codon at the N terminus of the chimeric Env proteins. This mutation rendered the MuLV-LP nonfunctional and indicated that the truncated FV-LP was sufficient to direct Env synthesis into the secretory pathway. Compared to the parental viruses, the chimeras replicated with only moderate cell-free titers.


Virology | 1993

Assembly of the Matrix Protein of Simian Immunodeficiency Virus into Virus-like Particles

Silvia A. González; José L. Affranchino; Hans Gelderblom; Arsène Burny


AIDS Research and Human Retroviruses | 1992

Spatial visualization of the maturing HIV-1 core and its linkage to the envelope

Stefan Höglund; Lars-Göran Öfverstedt; Åsa Nilsson; Pär Lundquist; Hans Gelderblom; Muhsin Özel; Ulf Skoglund


Clinical Chemistry | 2004

Detection of human polyomaviruses in urine from bone marrow transplant patients: comparison of electron microscopy with PCR.

Stefan S. Biel; Andreas Nitsche; Andreas Kurth; Wolfgang Siegert; Muhsin Özel; Hans Gelderblom

Collaboration


Dive into the Hans Gelderblom's collaboration.

Top Co-Authors

Avatar

Arsène Burny

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmad Awada

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony Elias

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas D. Adkins

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge