Network


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

Hotspot


Dive into the research topics where Simone Kruithof is active.

Publication


Featured researches published by Simone Kruithof.


Annals of the Rheumatic Diseases | 2013

Adalimumab elicits a restricted anti-idiotypic antibody response in autoimmune patients resulting in functional neutralisation

Pauline A. van Schouwenburg; Lotte A van de Stadt; Rob N. de Jong; Esther E. L. van Buren; Simone Kruithof; Els R. de Groot; Margreet Hart; S. Marieke van Ham; Theo Rispens; Lucien A. Aarden; Gerrit Jan Wolbink; Diana Wouters

Objectives Millions of patients worldwide are treated with therapeutic monoclonal antibodies. These biological therapeutics can be immunogenic, resulting in anti-drug antibody formation which leads to loss of response. Fully human biological agents, such as the anti-tumour necrosis factor α (anti-TNFα) antibody adalimumab, are considered to be weakly immunogenic, but anti-adalimumab antibodies (AAA) were recently detected in more than half of treated patients with rheumatoid arthritis (RA) within 28 weeks of treatment. A study was undertaken to determine the mechanism by which AAA lead to loss of response. Methods The specificity of the repertoire of AAA was investigated in a cohort of 50 AAA-positive RA patients. Inhibition experiments using TNFα and patient-derived anti-adalimumab monoclonal antibodies were performed. Results The antibody response against adalimumab is highly restricted: Fab fragments of a single monoclonal antibody specific for the idiotype of adalimumab inhibited 98.65% (25th–75th percentiles: 98.25–99.90) of the total anti-adalimumab reactivity in serum from 50 AAA-positive patients. The anti-adalimumab response was confined to the TNFα binding region of adalimumab, thereby neutralising its therapeutic efficacy. In line with this restricted specificity, small immune complexes were found in the circulation of AAA-forming patients. Conclusions The humoral immune response against adalimumab is highly restricted and limited to the idiotype of the therapeutic antibody. All antibodies result in functional neutralisation of the drug, thereby providing a mechanism by which AAA formation leads to clinical non-response.


Annals of the Rheumatic Diseases | 2015

The antibody response against human and chimeric anti-TNF therapeutic antibodies primarily targets the TNF binding region

K A van Schie; Margreet Hart; E R de Groot; Simone Kruithof; Lucien A. Aarden; Gertjan Wolbink; Theo Rispens

Background In a subset of patients, anti tumour necrosis factor (TNF) therapeutic antibodies are immunogenic, resulting in the formation of antidrug antibodies (ADAs). Neutralising ADAs compete with TNF for its binding site and reduces the effective serum concentration, causing clinical non-response. It is however unknown to which extent ADAs are neutralising. Objectives To study which proportion of antibodies to human(ised) anti-TNF (adalimumab, golimumab, certolizumab) as well as chimeric anti-TNF (infliximab) is neutralising. Methods Neutralising capacity of ADAs was assessed using a TNF competition assay in ADA-positive sera of patients treated with adalimumab (n=21), golimumab (n=4), certolizumab (n=9) or infliximab (n=34) sent in to our diagnostic department. Results In 34 sera with ADAs to adalimumab, golimumab or certolizumab, >97% of the antibodies were neutralising. In 34 sera with ADAs to infliximab >90% of the antibodies were neutralising. Further characterisation of the broader antibody response to infliximab revealed that non-neutralising antibodies to infliximab do not target murine domains, but may bind infliximab-unique domains not involved in TNF binding (located outside the paratope). Conclusions Our study shows that ADAs to human(ised) as well as chimeric anti-TNF therapeutic antibodies are largely neutralising. This highly restricted ADA response suggests an immunodominant role for the paratope of anti-TNF therapeutics.


Journal of Biological Chemistry | 2014

Functional Analysis of the Anti-Adalimumab Response using Patient-Derived Monoclonal Antibodies

Pauline A. van Schouwenburg; Simone Kruithof; Christian Votsmeier; Karin A van Schie; Margreet Hart; Rob N. de Jong; Esther E. L. van Buren; Marieke van Ham; Lucien A. Aarden; Gertjan Wolbink; Diana Wouters; Theo Rispens

Background: Therapeutic antibodies such as adalimumab can elicit anti-drug antibodies. Results: Monoclonal patient-derived antibodies were generated and found to compete for binding to adalimumab and are neutralizing, but they show markedly different fine-specificity. Conclusion: Anti-adalimumab antibodies bind to overlapping but distinct epitopes on adalimumab. Significance: Even for a fully human therapeutic antibody, there may be multiple determinants that contribute to immunogenicity. The production of antibodies to adalimumab in autoimmune patients treated with adalimumab is shown to diminish treatment efficacy. We previously showed that these antibodies are almost exclusively neutralizing, indicating a restricted response. Here, we investigated the characteristics of a panel of patient-derived monoclonal antibodies for binding to adalimumab. Single B-cells were isolated from two patients, cultured, and screened for adalimumab specificity. Analysis of variable region sequences of 16 clones suggests that the immune response against adalimumab is broad, involving multiple B-cell clones each using different combinations of V(D)J segments. A strong bias for replacement mutations in the complementarity determining regions was found, indicating an antigen-driven response. We recombinantly expressed 11 different monoclonal antibodies and investigated their affinity and specificity. All clones except one are of high affinity (Kd between 0.6 and 233 pm) and compete with TNF as well as each other for binding to adalimumab. However, binding to a panel of single-point mutants of adalimumab indicates markedly different fine specificities that also result in a differential tendency of each clone to form dimeric and multimeric immune complexes. We conclude that although all anti-adalimumab antibodies compete for binding to TNF, the response is clonally diverse and involves multiple epitopes on adalimumab. These results are important for understanding the relationship between self and non-self or idiotypic determinants on therapeutic antibodies and their potential immunogenicity.


Analytical Biochemistry | 2013

Nanomolar to sub-picomolar affinity measurements of antibody–antigen interactions and protein multimerizations: Fluorescence-assisted high-performance liquid chromatography

Theo Rispens; Pleuni Ooijevaar-de Heer; Ninotska I. L. Derksen; Gertjan Wolbink; Pauline A. van Schouwenburg; Simone Kruithof; Rob C. Aalberse

Although several techniques exist for the measurement of high-affinity interactions, it is still challenging to determine dissociation constants around or even below 1pM. During the analysis of several human-derived monoclonal antibodies to adalimumab, we found a clone with a very high affinity that could not be measured using conventional surface plasmon resonance assays. We developed a straightforward and robust method to measure affinities in the nanomolar to sub-picomolar range. The assay is based on separation of bound and free fluorescently labeled antigen using size exclusion chromatography and quantification by in-line fluorescence detection. We describe optimal conditions and procedures that result in a very sensitive assay that can be used to reliably determine ultra-high affinities. Using the method described in this article, a dissociation constant of 0.78pM could be determined for the anti-adalimumab antibody.


Journal of Pharmaceutical and Biomedical Analysis | 2016

Using monoclonal antibodies as an international standard for the measurement of anti-adalimumab antibodies

Pauline A. van Schouwenburg; Simone Kruithof; Gertjan Wolbink; Diana Wouters; Theo Rispens

Comparing studies investigating anti-drug antibody (ADA) formation is hampered by the lack of comparability between study protocols, assay formats, and standardized reference materials. In this respect, the use of an international standard would mean a major step forward. Here we compared 11 fully human monoclonal antibodies against adalimumab in two assays commonly used for ADA measurement; the bridging ELISA and the antigen binding test (ABT). Our results show non-parallel titration of the monoclonal antibodies in both assays, which we also find for polyclonal ADA sources. Moreover, we observed that the output of the bridging ELISA depends to a large degree on the affinity of the monoclonal antibody. For the ABT, results reflect a combination of affinity and avidity. This suggests that rather than reporting ADA values in nanogram per milliliter, arbitrary units may be more appropriate. Together our data highlight the difficulty of ADA standardization by identifying several pitfalls that should be taken into account when selecting a standard for ADA testing.


Scientific Reports | 2016

Therapeutic TNF Inhibitors can Differentially Stabilize Trimeric TNF by Inhibiting Monomer Exchange.

Karin A van Schie; Pleuni Ooijevaar-de Heer; Lisanne Dijk; Simone Kruithof; Gertjan Wolbink; Theo Rispens

Tumor necrosis factor (TNF) is a homotrimeric cytokine that is a key mediator of inflammation. It is unstable at physiological concentrations and slowly converts into an inactive form. Here, we investigated the mechanism of this process by using a Förster resonance energy transfer (FRET) assay that allowed monitoring of monomeric subunit exchange in time. We observed continuous exchange of monomeric subunits even at concentrations of TNF high enough to maintain its bioactivity. The kinetics of this process closely corresponds with the appearance of monomeric subunits and disappearance of trimeric TNF in time at ng/ml concentrations as monitored by high-performance size-exclusion chromatography (HP-SEC). Furthermore, of the five therapeutic TNF inhibitors that are currently used in the clinic, three (adalimumab, infliximab, etanercept) were found to completely inhibit the monomer exchange reaction and stabilize TNF trimers, whereas golimumab and certolizumab could not prevent monomer exchange, but did slow down the exchange process. These differences were not correlated with the affinities of the TNF inhibitors, measured with both surface plasmon resonance (SPR) and in fluid phase using fluorescence-assisted HP-SEC. The stabilizing effect of these TNF inhibitors might result in prolonged residual TNF bioactivity under conditions of incomplete blocking, as observed in vitro for adalimumab.


Annals of the Rheumatic Diseases | 2017

Infusion reactions during infliximab treatment are not associated with IgE anti-infliximab antibodies

Karin A van Schie; Pleuni Ooijevaar-de Heer; Simone Kruithof; C. Plasencia; T. Jurado; Dora Pascual Salcedo; Johannan F. Brandse; Geert R. D'Haens; Gerrit Jan Wolbink; Theo Rispens

Objectives Controversy exists on the role of IgE antidrug antibodies (IgE-ADA) in infusion reactions (IR) on infliximab treatment, partly due to the lack of a positive control used for assay validation. We sought to (1) develop a robust assay to measure IgE-ADA, including a positive control, (2) determine the association between IgE-ADA and IR and (3) determine the incidence of IgE-ADA in infliximab treated patients. Methods A recombinant human IgE anti-infliximab monoclonal antibody was developed as standard and positive control. With this antibody, we set up a novel robust assay to measure IgE-ADA. IgE-ADA was determined in three retrospective cohorts (n=159) containing IR+ (n=37) and IR− (n=39), and longitudinal sera of 83 spondyloarthritis. Results IgE-ADA was found in 0/39 IR−, whereas 4/37 (11%) IR+ showed low levels (0.1–0.3 IU/mL, below the 0.35 IU/mL threshold associated with elevated risk of allergic symptoms). All patients who were IgE-ADA positive also had (very) high IgG-ADA levels. The incidence of IgE-ADA in patients with infliximab-treated spondyloarthritis was estimated at less than approximately 1%. Conclusions IgE-ADA is rarely detected in infliximab-treated patients. Moreover, the absence of IgE-ADA in the majority of IR+ patients suggests that IgE-ADA is not associated with infusion reactions.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Adaptive antibody diversification through N-linked glycosylation of the immunoglobulin variable region

Fleur S. van de Bovenkamp; Ninotska I. L. Derksen; Pleuni Ooijevaar-de Heer; Karin A van Schie; Simone Kruithof; Magdalena Berkowska; C. Ellen van der Schoot; Hanna IJspeert; Mirjam van der Burg; Ann Gils; Lise Hafkenscheid; René E. M. Toes; Yoann Rombouts; Rosina Plomp; Manfred Wuhrer; S. Marieke van Ham; Gestur Vidarsson; Theo Rispens

Significance Structural variation of antibodies is generally defined in terms of amino acid composition, neglecting posttranslational modifications such as N-linked glycosylation. Little is known about the role of the glycans that are present in about 15% of variable domains. However, recent studies suggest that variable domain glycans exhibit distinct patterns according to (patho)physiological conditions, and can have immunomodulatory effects. Here we highlight a physiological role for variable domain glycans that is predetermined in the germline antibody repertoire: We show that variable domain N-linked glycans are acquired during somatic hypermutation at positions predisposed in the germline and may be positively selected during affinity maturation, representing an additional mechanism of secondary antibody diversification that contributes to the extent of the B-cell antibody repertoire. A hallmark of B-cell immunity is the generation of a diverse repertoire of antibodies from a limited set of germline V(D)J genes. This repertoire is usually defined in terms of amino acid composition. However, variable domains may also acquire N-linked glycans, a process conditional on the introduction of consensus amino acid motifs (N-glycosylation sites) during somatic hypermutation. High levels of variable domain glycans have been associated with autoantibodies in rheumatoid arthritis, as well as certain follicular lymphomas. However, the role of these glycans in the humoral immune response remains poorly understood. Interestingly, studies have reported both positive and negative effects on antibody affinity. Our aim was to elucidate the role of variable domain glycans during antigen-specific antibody responses. By analyzing B-cell repertoires by next-generation sequencing, we demonstrate that N-glycosylation sites are introduced at positions in which glycans can affect antigen binding as a result of a specific clustering of progenitor glycosylation sites in the germline sequences of variable domain genes. By analyzing multiple human monoclonal and polyclonal (auto)antibody responses, we subsequently show that this process is subject to selection during antigen-specific antibody responses, skewed toward IgG4, and positively contributes to antigen binding. Together, these results highlight a physiological role for variable domain glycosylation as an additional layer of antibody diversification that modulates antigen binding.


The Journal of Allergy and Clinical Immunology | 2017

Neutralizing capacity of monoclonal and polyclonal anti-natalizumab antibodies: The immune response to antibody therapeutics preferentially targets the antigen-binding site

Karin A van Schie; Simone Kruithof; Pauline A. van Schouwenburg; Anke Vennegoor; Joep Killestein; Gertjan Wolbink; Theo Rispens

This study shows that the immune response towards natalizumab, an IgG4 antibody targeting alpha4-integrins, is directed towards the antigen binding site of the drug. This extends previous observations for TNF inhibitors, suggesting that antibodies towards therapeutic antibodies may be inherently neutralizing


PLOS ONE | 2018

Dimeric IgG complexes from IVIg are incapable of inducing in vitro neutrophil degranulation or complement activation

Iwan Kustiawan; Ninotska I. L. Derksen; Theresa Guhr; Simone Kruithof; Wim Jiskoot; Gestur Vidarsson; Theo Rispens

Purpose Intravenous immunoglobulin (IVIg) products contain various amounts of dimeric IgG complexes. Current insights into the possible biological activities of these dimers remain controversial, and both immunemodulating and immune-activating effects have been reported. Here, we analyzed the putative immune-activating effects of dimers isolated from IVIg. Methods Dimers isolated from IVIg were purified by high-performance size-exclusion chromatography (HP-SEC) and tested for the ability to induce neutrophil degranulation in vitro. Results Dimers isolated from IVIg were found to be incapable of inducing in vitro neutrophil degranulation or complement activation, even at concentrations exceeding those expected to be reached upon administration in patients. These results depend on the removal of artefactual activation by using 0.1 micron filtration and the use of poloxamer to prevent adsorption of IgG onto the solid phase. Conclusions The data suggest dimeric IgG found in IVIg may bind to Fc-receptors without causing activation.

Collaboration


Dive into the Simone Kruithof's collaboration.

Top Co-Authors

Avatar

Theo Rispens

University of Amsterdam

View shared research outputs
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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge