Network


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

Hotspot


Dive into the research topics where Wijnand Helfrich is active.

Publication


Featured researches published by Wijnand Helfrich.


Molecular and Cellular Biology | 2001

Epidermal Growth Factor-Like Repeats Mediate Lateral and Reciprocal Interactions of Ep-CAM Molecules in Homophilic Adhesions

M Balzar; I H Briaire-de Bruijn; Ham Rees-Bakker; Frans A. Prins; Wijnand Helfrich; de Louis Leij; G Riethmuller; S Alberti; So Warnaar; Gert Jan Fleuren; Sergey V. Litvinov

ABSTRACT Ep-CAM is a new type of cell adhesion molecule (CAM) which does not structurally resemble the members of the four major families (cadherins, integrins, selectins, and CAMs of the immunoglobulin superfamily) and mediates Ca2+-independent, homophilic adhesions. The extracellular domain of Ep-CAM consists of a cysteine-rich region, containing two type II epidermal growth factor (EGF)-like repeats, followed by a cysteine-poor region. We generated mutated Ep-CAM forms with various deletions in the extracellular domain. These deletion mutants, together with monoclonal antibodies recognizing different epitopes in the extracellular domain, were used to investigate the role of the EGF-like repeats in the formation of intercellular contacts mediated by Ep-CAM molecules. We established that both EGF-like repeats are required for the formation of Ep-CAM-mediated homophilic adhesions, including the accumulation of Ep-CAM molecules at the cell-cell boundaries, and the anchorage of the Ep-CAM adhesion complex to F-actin via α-actinin. Deletion of either EGF-like repeat was sufficient to inhibit the adhesion properties of the molecule. The first EGF-like repeat of Ep-CAM is required for reciprocal interactions between Ep-CAM molecules on adjacent cells, as was demonstrated with blocking antibodies. The second EGF-like repeat was mainly required for lateral interactions between Ep-CAM molecules. Lateral interactions between Ep-CAM molecules result in the formation of tetramers, which might be the first and necessary step in the formation of Ep-CAM-mediated intercellular contacts.


Nature Biotechnology | 1999

A recombinant, fully human monoclonal antibody with antitumor activity constructed from phage-displayed antibody fragments

Gerwin Huls; Iafm Heijnen; Me Cuomo; J. C. Koningsberger; Edwin Boel; Arv de Vries; Saj Loyson; Wijnand Helfrich; Gpv Henegouwen; M van Meijer; J de Kruif; Ton Logtenberg

A single-chain Fv antibody fragment specific for the tumor-associated Ep-CAM molecule was isolated from a semisynthetic phage display library and converted into an intact, fully human IgG1 monoclonal antibody (huMab). The purified huMab had an affinity of 5 nM and effectively mediated tumor cell killing in in vitro and in vivo assays. These experiments show that nonimmunized phage antibody display libraries can be used to obtain high-affinity, functional, and clinically applicable huMabs directed against a tumor-associated antigen.


Journal of Biological Chemistry | 2005

Simultaneous Inhibition of Epidermal Growth Factor Receptor (EGFR) Signaling and Enhanced Activation of Tumor Necrosis Factor-related Apoptosis-inducing Ligand (TRAIL) Receptor-mediated Apoptosis Induction by an scFv:sTRAIL Fusion Protein with Specificity for Human EGFR

Edwin Bremer; Douwe F. Samplonius; Linda van Genne; Marike H. Dijkstra; Bart-Jan Kroesen; Lou de Leij; Wijnand Helfrich

Epidermal growth factor receptor (EGFR) signaling inhibition by monoclonal antibodies and EGFR-specific tyrosine kinase inhibitors has shown clinical efficacy in cancer by restoring susceptibility of tumor cells to therapeutic apoptosis induction. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anti-cancer agent with tumor-selective apoptotic activity. Here we present a novel approach that combines EGFR-signaling inhibition with target cell-restricted apoptosis induction using a TRAIL fusion protein with engineered specificity for EGFR. This fusion protein, scFv425:sTRAIL, comprises the EGFR-blocking antibody fragment scFv425 genetically fused to soluble TRAIL (sTRAIL). Treatment with scFv425:sTRAIL resulted in the specific accretion to the cell surface of EGFR-positive cells only. EGFR-specific binding rapidly induced a dephosphorylation of EGFR and down-stream mitogenic signaling, which was accompanied by cFLIPL down-regulation and Bad dephosphorylation. EGFR-specific binding converted soluble scFv425:sTRAIL into a membrane-bound form of TRAIL that cross-linked agonistic TRAIL receptors in a paracrine manner, resulting in potent apoptosis induction in a series of EGFR-positive tumor cell lines. Co-treatment of EGFR-positive tumor cells with the EGFR-tyrosine kinase inhibitor Iressa resulted in a potent synergistic pro-apoptotic effect, caused by the specific down-regulation of c-FLIP. Furthermore, in mixed culture experiments binding Lof scFv425:sTRAIL to EGFR-positive target cells conveyed a potent apoptotic effect toward EGFR-negative bystander tumor cells. The favorable characteristics of scFv425:sTRAIL, alone and in combination with Iressa, as well as its potent anti-tumor bystander activity indicate its potential value for treatment of EGFR-expressing cancers.


International Journal of Cancer | 2004

Target cell‐restricted and ‐enhanced apoptosis induction by a scFv:sTRAIL fusion protein with specificity for the pancarcinoma‐associated antigen EGP2

Edwin Bremer; Jos M. A. Kuijlen; Douwe F. Samplonius; Henning Walczak; Lou de Leij; Wijnand Helfrich

The apparent tumor selective apoptosis‐inducing activity of recombinant soluble TNF‐related apoptosis‐inducing ligand (TRAIL) has aroused much interest for use in clinical application. However, to exploit fully its therapeutic potential, the characteristics of both the TRAIL receptor system and soluble TRAIL (sTRAIL) should be taken into account: first, the widespread expression of the various TRAIL receptors throughout the human body; second, the differential binding affinities and crosslinking requirements of the agonistic receptors TRAIL‐R1 and TRAIL‐R2; and third, the solution behavior of particular sTRAIL preparations. Therefore, we constructed a novel TRAIL fusion protein, designated scFvC54:sTRAIL, comprising the human scFv antibody fragment C54 genetically linked to the N‐terminus of human sTRAIL. The scFvC54:sTRAIL fusion protein was designed to induce apoptosis by crosslinking of agonistic TRAIL receptors only after specific binding of scFvC54:sTRAIL to the abundantly expressed carcinoma‐associated cell surface antigen EGP2 (alias EpCAM). Target antigen‐restricted apoptosis induction was demonstrated for various EGP2‐positive tumor cells and could be inhibited by an EGP2 competing antibody. Target antigen binding converted soluble scFvC54:sTRAIL into a membrane‐bound form of TRAIL that was capable of signaling apoptosis not only through TRAIL‐R1 but also through TRAIL‐R2. Size‐exclusion fast protein liquid chromatography (FPLC) indicated that scFvC54:sTRAIL was produced as stable and homogeneous trimers in the absence of detectable TRAIL aggregates. The favorable characteristics of the scFvC54:sTRAIL fusion protein potentially reduce the amount of sTRAIL required for antitumor activity and may be of value for the treatment of various human carcinomas.


Medicinal Research Reviews | 2013

Therapeutic potential of Galectin-9 in human disease

Valerie R. Wiersma; Marco de Bruyn; Wijnand Helfrich; Edwin Bremer

In recent years, an important role has emerged for the glycan‐binding protein Galectin‐9 (Gal‐9) in health and disease. In normal physiology, Gal‐9 seems to be a pivotal modulator of T‐cell immunity by inducing apoptosis in specific T‐cell subpopulations. Because these T‐cell populations are associated with autoimmunity, inflammatory disease, and graft rejection, it was postulated that application of exogenous Gal‐9 may limit pathogenic T‐cell activity. Indeed, treatment with recombinant Gal‐9 ameliorates disease activity in various preclinical models of autoimmunity and allograft graft rejection. In many solid cancers, the loss of Gal‐9 expression is closely associated with metastatic progression. In line with this observation, treatment with recombinant Gal‐9 prevents metastatic spread in various preclinical cancer models. In addition, various hematological malignancies are sensitive to apoptotic elimination by recombinant Gal‐9. Here, we review the biology and physiological role of this versatile lectin and discuss the therapeutic potential of Gal‐9 in various diseases, including autoimmunity, asthma, infection, and cancer.


The Prostate | 2009

Expression of the Gastrin-Releasing Peptide Receptor, the Prostate Stem Cell Antigen and the Prostate-Specific Membrane Antigen in Lymph Node and Bone Metastases of Prostate Cancer

H. J. K. Ananias; Marius C. van den Heuvel; Wijnand Helfrich; Igle J. de Jong

Cell membrane antigens like the gastrin‐releasing peptide receptor (GRPR), the prostate stem cell antigen (PSCA), and the prostate‐specific membrane antigen (PSMA), expressed in prostate cancer, are attractive targets for new therapeutic and diagnostic applications. Therefore, we investigated in this study whether these antigens are expressed in metastasized prostate cancer.


Cancer Research | 2005

Target cell-restricted apoptosis induction of acute leukemic T cells by a recombinant tumor necrosis factor-related apoptosis-inducing ligand fusion protein with specificity for human CD7

Edwin Bremer; Douwe F. Samplonius; Matthias Peipp; Linda van Genne; Bart-Jan Kroesen; Georg H. Fey; Martin Gramatzki; Lou de Leij; Wijnand Helfrich

Current treatment of human T-cell leukemia and lymphoma is predominantly limited to conventional cytotoxic therapy and is associated with limited therapeutic response and significant morbidity. Therefore, more potent and leukemia-specific therapies with favorable toxicity profiles are urgently needed. Here, we report on the construction of a novel therapeutic fusion protein, scFvCD7:sTRAIL, designed to induce target antigen-restricted apoptosis in human T-cell tumors. ScFvCD7:sTRAIL consists of the death-inducing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) genetically linked to an scFv antibody fragment specific for the T-cell surface antigen CD7. Treatment with scFvCD7:sTRAIL induced potent CD7-restricted apoptosis in a series of malignant T-cell lines, whereas normal resting leukocytes, activated T cells, and vascular endothelial cells (human umbilical vein endothelial cells) showed no detectable apoptosis. The apoptosis-inducing activity of scFvCD7:sTRAIL was stronger than that of the immunotoxin scFvCD7:ETA. In mixed culture experiments with CD7-positive and CD7-negative tumor cells, scFvCD7:sTRAIL induced very potent bystander apoptosis of CD7-negative tumor cells. In vitro treatment of blood cells freshly derived from T-acute lymphoblastic leukemia patients resulted in marked apoptosis of the malignant T cells that was strongly augmented by vincristin. In conclusion, scFvCD7:sTRAIL is a novel recombinant protein causing restricted apoptosis in human leukemic T cells with low toxicity for normal human blood and endothelial cells.


Hepatology | 2008

Wnt2 acts as a cell type–specific, autocrine growth factor in rat hepatic sinusoidal endothelial cells cross‐stimulating the VEGF pathway

Daniel Klein; Alexandra Demory; Francis Peyre; Jens Kroll; Hellmut G. Augustin; Wijnand Helfrich; Julia Kzhyshkowska; Kai Schledzewski; Bernd Arnold; Sergij Goerdt

The mechanisms regulating the growth and differentiation of hepatic sinusoidal endothelial cells (HSECs) are not well defined. Because Wnt signaling has become increasingly important in developmental processes such as vascular and hepatic differentiation, we analyzed HSEC‐specific Wnt signaling in detail. Using highly pure HSECs isolated by a newly developed protocol selecting against nonsinusoidal hepatic endothelial cells, we comparatively screened the multiple components of the Wnt pathway for differential expression in HSECs and lung microvascular endothelial cells (LMECs) via reverse‐transcription polymerase chain reaction (RT‐PCR). As confirmed via quantitative RT‐PCR and northern and western blotting experiments, Wnt2 (and less so Wnt transporter wls/evi) and Wnt coreceptor Ryk were overexpressed by HSECs, whereas Wnt inhibitory factor (WIF) was strongly overexpressed by LMECs. Exogenous Wnt2 superinduced proliferation of HSECs (P < 0.05). The Wnt inhibitor secreted frizzled‐related protein 1 (sFRP1) (P < 0.005) and transfection of HSECs with Wnt2 small interfering RNA (siRNA) reduced proliferation of HSECs. These effects were rescued by exogenous Wnt2. Tube formation of HSECs on matrigel was strongly inhibited by Wnt inhibitors sFRP1 and WIF (P < 0.0005). Wnt signaling in HSECs activated the canonical pathway inducing nuclear translocation of β‐catenin. GST (glutathione transferase) pull‐down and co‐immunoprecipitation assays showed Fzd4 to be a novel Wnt2 receptor in HSECs. Gene profiling identified vascular endothelial growth factor receptor‐2 (VEGFR‐2) as a target of Wnt2 signaling in HSECs. Inhibition of Wnt signaling down‐regulated VEGFR‐2 messenger RNA and protein. Wnt2 siRNA knock‐down confirmed Wnt2 specificity of VEGFR‐2 regulation in HSECs. Conclusion: Wnt2 is an autocrine growth and differentiation factor specific for HSECs that synergizes with the VEGF signaling pathway to exert its effects. (HEPATOLOGY 2008;47:1018–1031.)


Journal of Immunology | 2007

Fas receptor clustering and involvement of the death receptor pathway in rituximab-mediated apoptosis with concomitant sensitization of lymphoma B cells to Fas-induced apoptosis

Alja J. Stel; Bram ten Cate; Susan Jacobs; Jan Willem Kok; Diana C. J. Spierings; Monica Dondorff; Wijnand Helfrich; Hanneke C. Kluin-Nelemans; Lou de Leij; Sebo Withoff; Bart-Jan Kroesen

Ab binding to CD20 has been shown to induce apoptosis in B cells. In this study, we demonstrate that rituximab sensitizes lymphoma B cells to Fas-induced apoptosis in a caspase-8-dependent manner. To elucidate the mechanism by which Rituximab affects Fas-mediated cell death, we investigated rituximab-induced signaling and apoptosis pathways. Rituximab-induced apoptosis involved the death receptor pathway and proceeded in a caspase-8-dependent manner. Ectopic overexpression of FLIP (the physiological inhibitor of the death receptor pathway) or application of zIETD-fmk (specific inhibitor of caspase-8, the initiator-caspase of the death receptor pathway) both specifically reduced rituximab-induced apoptosis in Ramos B cells. Blocking the death receptor ligands Fas ligand or TRAIL, using neutralizing Abs, did not inhibit apoptosis, implying that a direct death receptor/ligand interaction is not involved in CD20-mediated cell death. Instead, we hypothesized that rituximab-induced apoptosis involves membrane clustering of Fas molecules that leads to formation of the death-inducing signaling complex (DISC) and downstream activation of the death receptor pathway. Indeed, Fas coimmune precipitation experiments showed that, upon CD20-cross-linking, Fas-associated death domain protein (FADD) and caspase-8 were recruited into the DISC. Additionally, rituximab induced CD20 and Fas translocation to raft-like domains on the cell surface. Further analysis revealed that, upon stimulation with rituximab, Fas, caspase-8, and FADD were found in sucrose-gradient raft fractions together with CD20. In conclusion, in this study, we present evidence for the involvement of the death receptor pathway in rituximab-induced apoptosis of Ramos B cells with concomitant sensitization of these cells to Fas-mediated apoptosis via Fas multimerization and recruitment of caspase-8 and FADD to the DISC.


Journal of Biological Chemistry | 2005

Simultaneous inhibition of EGFR signaling and enhanced activation of TRAIL-R-mediated apoptosis induction by an scFv: sTRAIL fusion protein with specificity for human EGFR

Edwin Bremer; Douwe F. Samplonius; Linda van Genne; Marike H. Dijkstra; Bart-Jan Kroesen; Lou de Leij; Wijnand Helfrich

Epidermal growth factor receptor (EGFR) signaling inhibition by monoclonal antibodies and EGFR-specific tyrosine kinase inhibitors has shown clinical efficacy in cancer by restoring susceptibility of tumor cells to therapeutic apoptosis induction. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anti-cancer agent with tumor-selective apoptotic activity. Here we present a novel approach that combines EGFR-signaling inhibition with target cell-restricted apoptosis induction using a TRAIL fusion protein with engineered specificity for EGFR. This fusion protein, scFv425:sTRAIL, comprises the EGFR-blocking antibody fragment scFv425 genetically fused to soluble TRAIL (sTRAIL). Treatment with scFv425:sTRAIL resulted in the specific accretion to the cell surface of EGFR-positive cells only. EGFR-specific binding rapidly induced a dephosphorylation of EGFR and down-stream mitogenic signaling, which was accompanied by cFLIPL down-regulation and Bad dephosphorylation. EGFR-specific binding converted soluble scFv425:sTRAIL into a membrane-bound form of TRAIL that cross-linked agonistic TRAIL receptors in a paracrine manner, resulting in potent apoptosis induction in a series of EGFR-positive tumor cell lines. Co-treatment of EGFR-positive tumor cells with the EGFR-tyrosine kinase inhibitor Iressa resulted in a potent synergistic pro-apoptotic effect, caused by the specific down-regulation of c-FLIP. Furthermore, in mixed culture experiments binding Lof scFv425:sTRAIL to EGFR-positive target cells conveyed a potent apoptotic effect toward EGFR-negative bystander tumor cells. The favorable characteristics of scFv425:sTRAIL, alone and in combination with Iressa, as well as its potent anti-tumor bystander activity indicate its potential value for treatment of EGFR-expressing cancers.

Collaboration


Dive into the Wijnand Helfrich's collaboration.

Top Co-Authors

Avatar

Edwin Bremer

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Douwe F. Samplonius

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marco de Bruyn

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Lou de Leij

University of Groningen

View shared research outputs
Top Co-Authors

Avatar

Rudi Dierckx

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Bart-Jan Kroesen

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Valerie R. Wiersma

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Z. Yu

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

H. J. K. Ananias

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge