Network


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

Hotspot


Dive into the research topics where Michel Vierboom is active.

Publication


Featured researches published by Michel Vierboom.


Arthritis Research & Therapy | 2005

Modeling human arthritic diseases in nonhuman primates

Michel Vierboom; Margreet Jonker; Ronald E. Bontrop; Bert A. 't Hart

Models of rheumatoid arthritis (RA) in laboratory animals are important tools for research into pathogenic mechanisms and the development of effective, safe therapies. Rodent models (rats and mice) have provided important information about the pathogenic mechanisms. However, the evolutionary distance between rodents and humans hampers the translation of scientific principles into effective therapies. The impact of the genetic distance between the species is especially seen with treatments based on biological molecules, which are usually species-specific. The outbred nature and the closer anatomical, genetic, microbiological, physiological, and immunological similarity of nonhuman primates to humans may help to bridge the wide gap between inbred rodent strain models and the heterogeneous RA patient population. Here we review clinical, immunological and pathological aspects of the rhesus monkey model of collagen-induced arthritis, which has emerged as a reproducible model of human RA in nonhuman primates.


Arthritis Research & Therapy | 2015

The preclinical pharmacology of the high affinity anti-IL-6R Nanobody® ALX-0061 supports its clinical development in rheumatoid arthritis

Maarten Van Roy; Cedric Jozef Neotere Ververken; Els Beirnaert; Sven Hoefman; Joost Kolkman; Michel Vierboom; Elia Breedveld; Bert A. 't Hart; Sofie Poelmans; Lieselot Bontinck; Alex Hemeryck; Sandy Jacobs; Judith Baumeister; Hans Ulrichts

IntroductionThe pleiotropic cytokine interleukin-6 (IL-6) plays an important role in the pathogenesis of different diseases, including rheumatoid arthritis (RA). ALX-0061 is a bispecific Nanobody® with a high affinity and potency for IL-6 receptor (IL-6R), combined with an extended half-life by targeting human serum albumin. We describe here the relevant aspects of its in vitro and in vivo pharmacology.MethodsALX-0061 is composed of an affinity-matured IL-6R-targeting domain fused to an albumin-binding domain representing a minimized two-domain structure. A panel of different in vitro assays was used to characterize the biological activities of ALX-0061. The pharmacological properties of ALX-0061 were examined in cynomolgus monkeys, using plasma levels of total soluble (s)IL-6R as pharmacodynamic marker. Therapeutic effect was evaluated in a human IL-6-induced acute phase response model in the same species, and in a collagen-induced arthritis (CIA) model in rhesus monkeys, using tocilizumab as positive control.ResultsALX-0061 was designed to confer the desired pharmacological properties. A 200-fold increase of target affinity was obtained through affinity maturation of the parental domain. The high affinity for sIL-6R (0.19 pM) translated to a concentration-dependent and complete neutralization of sIL-6R in vitro. In cynomolgus monkeys, ALX-0061 showed a dose-dependent and complete inhibition of hIL-6-induced inflammatory parameters, including plasma levels of C-reactive protein (CRP), fibrinogen and platelets. An apparent plasma half-life of 6.6 days was observed after a single intravenous administration of 10 mg/kg ALX-0061 in cynomolgus monkeys, similar to the estimated expected half-life of serum albumin. ALX-0061 and tocilizumab demonstrated a marked decrease in serum CRP levels in a non-human primate CIA model. Clinical effect was confirmed in animals with active drug exposure throughout the study duration.ConclusionsALX-0061 represents a minimized bispecific biotherapeutic of 26 kDa, nearly six times smaller than monoclonal antibodies. High in vitro affinity and potency was demonstrated. Albumin binding as a half-life extension technology resulted in describable and expected pharmacokinetics. Strong IL-6R engagement was shown to translate to in vivo effect in non-human primates, demonstrated via biomarker deregulation as well as clinical effect. Presented results on preclinical pharmacological properties of ALX-0061 are supportive of clinical development in RA.


Arthritis Research & Therapy | 2014

Type II collagen antibody response is enriched in the synovial fluid of rheumatoid joints and directed to the same major epitopes as in collagen induced arthritis in primates and mice

Ingrid Lindh; Omri Snir; Erik Lönnblom; Hüseyin Uysal; Ida Andersson; Kutty Selva Nandakumar; Michel Vierboom; Bert A. 't Hart; Vivianne Malmström; Rikard Holmdahl

IntroductionAntibodies towards type II collagen (CII) are detected in patients with rheumatoid arthritis (RA) and in non-human primates and rodents with collagen induced arthritis (CIA). We have previously shown that antibodies specific for several CII-epitopes are pathogenic using monoclonal antibodies from arthritic mice, although the role of different anti-CII epitopes has not been investigated in detail in other species. We therefore performed an inter-species comparative study of the autoantibody response to CII in patients with RA versus monkeys and mice with CIA.MethodsAnalysis of the full epitope repertoire along the disease course of CIA was performed using a library of CII triple-helical peptides. The antibody responses to the major CII epitopes were analyzed in sera and synovial fluid from RA patients, and in sera from rhesus monkeys (Macaca mulatta), common marmosets (Callithrix jacchus) and mice.ResultsMany CII epitopes including the major C1, U1, and J1 were associated with established CIA and arginine residues played an important role in the anti-CII antibody interactions. The major epitopes were also recognized in RA patients, both in sera and even more pronounced in synovial fluid: 77% of the patients had antibodies to the U1 epitope. The anti-CII immune response was not restricted to the anti-citrulline protein antibodies (ACPA) positive RA group.ConclusionCII conformational dependent antibody responses are common in RA and are likely to originate from rheumatoid joints but did not show a correlation with ACPA response. Importantly, the fine specificity of the anti-CII response is similar with CIA in monkeys and rodents where the recognized epitopes are conserved and have a major pathogenic role. Thus, anti-CII antibodies may both contribute to, as well as be the consequence of, local joint inflammation.


Transplantation | 2002

BLOCKING THE CD80 AND CD86 COSTIMULATION MOLECULES: LESSONS TO BE LEARNED FROM ANIMAL MODELS

Margreet Jonker; Miriam Ossevoort; Michel Vierboom

The induction of tolerance for allografts, obviating the need for immunosuppression, is the ultimate goal in transplantation. Immunoregulatory antibodies preventing graft rejection are promising candidates for the induction of tolerance. Costimulation blockade could be a useful approach to inducing donor-specific nonresponsiveness in organ transplantation. Rodent studies and in vitro studies using human or nonhuman primate peripheral blood mononuclear cells indicated that this approach might indeed lead to specific T-cell anergy. Nonhuman primate studies, in which the B7 costimulation pathway was blocked, have so far not led to permanent drug-free graft acceptance. The results are promising, however, because during the treatment period with B7 costimulation blockade alone or combined with anti-CD40 or cyclosporine, no graft loss was observed and donor-specific antibody formation was prevented. Based on these findings, new approaches to inducing drug-free graft acceptance should be investigated.


Arthritis Research & Therapy | 2010

Collagen-induced arthritis in common marmosets: A new nonhuman primate model for chronic arthritis

Michel Vierboom; Elia Breedveld; Ivanela Kondova; Bert A. ‘t Hart

IntroductionThere is an ever-increasing need for animal models to evaluate efficacy and safety of new therapeutics in the field of rheumatoid arthritis (RA). Particularly for the early preclinical evaluation of human-specific biologicals targeting the progressive phase of the disease, there is a need for relevant animal models. In response to this requirement we set out to develop a model of collagen-induced arthritis (CIA) in a small-sized nonhuman primate species (300 to 400 g at adult age); that is, the common marmoset (Callithrix jacchus).MethodsTwenty-two animals divided into three experiments were immunized with collagen type II (CII) of either bovine or chicken origin with different immunization strategies. The animals were analyzed for clinical manifestation of arthritis, hematology and clinical chemistry, immunological responses against CII and histopathological features of the arthritis.ResultsClinically manifest arthritis was observed in almost 100% (21 out of 22) of the animals. Fifty percent of the animals developed semi-acute CIA while the other 50% displayed a more chronic disease. Both cellular (CD3/CD4 and CD3/CD8) and humoral responses (IgM and IgG) against CII were involved in the development of the disease. Besides mild histopathological changes in bone and cartilage, severe inflammation in extraarticular tissues like periosteum and subcutaneous tissues was observed.ConclusionsThis new model in marmosets more closely resembles chronic RA with respect to the chronic disease course and pathomorphological presentation than the more acute monophasic and destructive CIA model in macaques. This model can therefore fill a niche in preclinical testing of new human specific therapeutics.


Experimental Animals | 2013

An overview of models, methods, and reagents developed for translational autoimmunity research in the common marmoset (Callithrix jacchus)

S. Anwar Jagessar; Michel Vierboom; Erwin L. A. Blezer; Jan Niklas Bauer; Bert A. 't Hart; Yolanda S. Kap

The common marmoset (Callithrix jacchus) is a small-bodied Neotropical primate and a useful preclinical animal model for translational research into autoimmune-mediated inflammatory diseases (AIMID), such as rheumatoid arthritis (RA) and multiple sclerosis (MS). The animal model for MS established in marmosets has proven their value for exploratory research into (etio) pathogenic mechanisms and for the evaluation of new therapies that cannot be tested in lower species because of their specificity for humans. Effective usage of the marmoset in preclinical immunological research has been hampered by the limited availability of blood for immunological studies and of reagents for profiling of cellular and humoral immune reactions. In this paper, we give a concise overview of the procedures and reagents that were developed over the years in our laboratory in marmoset models of the above-mentioned diseases.


Transplant Immunology | 2003

Induction of allograft tolerance through costimulatory blockade: first selection of drugs in vitro

Michel Vierboom; Miriam Ossevoort; Ella A. Sick; Krista G. Haanstra; Margreet Jonker

The development of an in vitro assay predicting the chances of graft survival after treatment with immunoregulatory agents is a major topic in transplantation. Antibodies (Abs) interfering in the costimulatory pathway are promising candidates for the induction of tolerance. To evaluate these antibodies for clinical use studies non-human primates are the only feasible option due to species specificity of the antibodies. Peripheral blood mononuclear cells, isolated from a large panel of rhesus monkeys, were used in a unidirectional mixed lymphocyte reaction to evaluate the ability of antibodies blocking the costimulatory pathway, to affect both primary and secondary proliferative and cytolytic allospecific immune responses in vitro. These blocking antibodies were also used in protocols prolonging allograft survival in a life-supporting kidney allotransplant model in rhesus macaques. The ultimate aim is to establish a correlation between parameters obtained in vitro and the success of transplantation in vivo. The combination of anti-CD80 and anti-CD86 resulted in a complete abrogation of the primary alloresponse as measured in a proliferation assay. Adding anti-CD40 significantly reduced this inhibitory effect although the in vivo effects of this antibody have been shown to be beneficial. The secondary response was most prominently inhibited by the combination of anti-CD80/86. Paradoxically, anti-CD40 alone markedly inhibited the secondary proliferative response, but did not add to the inhibitory effect of the combination of anti-CD80/86. The cytolytic response was inhibited maximally only when CsA was added to the combination of anti-CD80/86. Treatment with monoclonal antibodies alone without immunosuppressive drugs was sufficient to maintain graft survival during the time of treatment in most animals. However, rejection was initiated as soon as the treatment ceased and no tolerance, resulting in long-term graft and patient survival, was established. The complete inhibition of primary alloresponses and the partial inhibition of secondary proliferative alloresponses correlate with prolonged graft survival during treatment, but have no predictive value for the success of tolerance induction for kidney allografts in rhesus monkeys.


Clinical and Experimental Immunology | 2016

Clinical efficacy of a new CD28-targeting antagonist of T cell co-stimulation in a non-human primate model of collagen-induced arthritis.

Michel Vierboom; Elia Breedveld; Yolanda S. Kap; Caroline Mary; Nicolas Poirier; Bert A. 't Hart; Bernard Vanhove

T cells have a central pathogenic role in the aetiopathogenesis of rheumatoid arthritis (RA), and are therefore a favoured target of immunotherapy aiming at physical or functional elimination. Here we report an efficacy test of FR104, a new co‐stimulation inhibitor directly targeting CD28 on T cells, in a translationally relevant model, the rhesus monkey model of collagen‐induced arthritis (CIA). As a relevant comparator we used abatacept [cytotoxic T lymphocyte antigen immunoglobulin (CTLA Ig)], an antagonist of CTLA‐4 binding to CD80/86 clinically approved for treatment of RA. Treatment with either compound was started at the day of CIA induction. Although FR104 previously demonstrated a higher control of T cell responses in vitro than abatacept, both compounds were equally potent in the suppression of CIA symptoms and biomarkers, such as the production of C‐reactive protein (CRP) and interleukin (IL)‐6 and anti‐collagen type II (CII) serum antibody (IgM/IgG). However, in contrast to abatacept, FR104 showed effective suppression of CII‐induced peripheral blood mononuclear cell (PBMC) proliferation. The current study demonstrates a strong potential of the new selective CD28 antagonist FR104 for treatment of RA.


Drug Discovery Today | 2014

Improvement of preclinical animal models for autoimmune-mediated disorders via reverse translation of failed therapies.

Bert A. 't Hart; S. Anwar Jagessar; Yolanda S. Kap; Krista G. Haanstra; Ingrid H.C.H.M. Philippens; Che Serguera; J. A. M. Langermans; Michel Vierboom

The poor translational validity of autoimmune-mediated inflammatory disease (AIMID) models in inbred and specific pathogen-free (SPF) rodents underlies the high attrition of new treatments for the corresponding human disease. Experimental autoimmune encephalomyelitis (EAE) is a frequently used preclinical AIMID model. We discuss here how crucial information needed for the innovation of current preclinical models can be obtained from postclinical analysis of the nonhuman primate EAE model, highlighting the mechanistic reasons why some therapies fail and others succeed. These new insights can also help identify new targets for treatment.


Arthritis Research & Therapy | 2013

PDL241, a novel humanized monoclonal antibody, reveals CD319 as a therapeutic target for rheumatoid arthritis

Jacky Woo; Michel Vierboom; Hakju Kwon; Debra T. Chao; Shiming Ye; Jianmin Li; Karen Lin; Irene Tang; Nicole A. Belmar; Taymar Hartman; Elia Breedveld; Vladimir Vexler; Bert A. 't Hart; Debbie Law; Gary C. Starling

IntroductionTargeting the CD20 antigen has been a successful therapeutic intervention in the treatment of rheumatoid arthritis (RA). However, in some patients with an inadequate response to anti-CD20 therapy, a persistence of CD20- plasmablasts is noted. The strong expression of CD319 on CD20- plasmablast and plasma cell populations in RA synovium led to the investigation of the potential of CD319 as a therapeutic target.MethodsPDL241, a novel humanized IgG1 monoclonal antibody (mAb) to CD319, was generated and examined for its ability to inhibit immunoglobulin production from plasmablasts and plasma cells generated from peripheral blood mononuclear cells (PBMC) in the presence and absence of RA synovial fibroblasts (RA-SF). The in vivo activity of PDL241 was determined in a human PBMC transfer into NOD scid IL-2 gamma chain knockout (NSG) mouse model. Finally, the ability of PDL241 to ameliorate experimental arthritis was evaluated in a collagen-induced arthritis (CIA) model in rhesus monkeys.ResultsPDL241 bound to plasmablasts and plasma cells but not naïve B cells. Consistent with the binding profile, PDL241 inhibited the production of IgM from in vitro PBMC cultures by the depletion of CD319+ plasmablasts and plasma cells but not B cells. The activity of PDL241 was dependent on an intact Fc portion of the IgG1 and mediated predominantly by natural killer cells. Inhibition of IgM production was also observed in the human PBMC transfer to NSG mouse model. Treatment of rhesus monkeys in a CIA model with PDL241 led to a significant inhibition of anti-collagen IgG and IgM antibodies. A beneficial effect on joint related parameters, including bone remodeling, histopathology, and joint swelling was also observed.ConclusionsThe activity of PDL241 in both in vitro and in vivo models highlights the potential of CD319 as a therapeutic target in RA.

Collaboration


Dive into the Michel Vierboom's collaboration.

Top Co-Authors

Avatar

Bert A. 't Hart

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Margreet Jonker

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Elia Breedveld

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Krista G. Haanstra

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Ivanela Kondova

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Yolanda S. Kap

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Bert A. ‘t Hart

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Wubben

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Paul P. Tak

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar

S. Anwar Jagessar

Biomedical Primate Research Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge