Network


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

Hotspot


Dive into the research topics where Hanneke Stegeman is active.

Publication


Featured researches published by Hanneke Stegeman.


BMC Cancer | 2012

Activation of AKT by hypoxia: a potential target for hypoxic tumors of the head and neck

Hanneke Stegeman; Johannes H.A.M. Kaanders; Deric L. Wheeler; Albert J. van der Kogel; Marieke Mg Verheijen; Stijn J Waaijer; Mari Iida; Reidar Grénman; Paul N. Span; Johan Bussink

BackgroundOnly a minority of cancer patients benefits from the combination of EGFR-inhibition and radiotherapy in head and neck squamous cell carcinoma (HNSCC). A potential resistance mechanism is activation of EGFR and/or downstream pathways by stimuli in the microenvironment. The aim of this study was to find molecular targets induced by the microenvironment by determining the in vitro and in vivo expression of proteins of the EGFR-signaling network in 6 HNSCC lines. As hypoxia is an important microenvironmental parameter associated with poor outcome in solid tumors after radiotherapy, we investigated the relationship with hypoxia in vitro and in vivo.MethodsSix human HNSCC cell lines were both cultured as cell lines (in vitro) and grown as xenograft tumors (in vivo). Expression levels were determined via western blot analysis and localization of markers was assessed via immunofluorescent staining. To determine the effect of hypoxia and pAKT-inhibition on cell survival, cells were incubated at 0.5% O2 and treated with MK-2206.ResultsWe observed strong in vitro-in vivo correlations for EGFR, pEGFR and HER2 (rs=0.77, p=0.10, rs=0.89, p=0.03) and rs=0.93, p=0.02, respectively), but not for pAKT, pERK1/2 or pSTAT3 (all rs<0.55 and p>0.30). In vivo, pAKT expression was present in hypoxic cells and pAKT and hypoxia were significantly correlated (rs=0.51, p=0.04). We confirmed in vitro that hypoxia induces activation of AKT. Further, pAKT-inhibition via MK-2206 caused a significant decrease in survival in hypoxic cells (p<0.01), but not in normoxic cells.ConclusionsThese data suggest that (p)EGFR and HER2 expression is mostly determined by intrinsic features of the tumor cell, while the activation of downstream kinases is highly influenced by the tumor microenvironment. We show that hypoxia induces activation of AKT both in vitro and in vivo, and that hypoxic cells can be specifically targeted by pAKT-inhibition. Targeting pAKT is thus a potential way to overcome therapy resistance induced by hypoxia and improve patient outcome.


Radiotherapy and Oncology | 2013

Predictive value of hypoxia, proliferation and tyrosine kinase receptors for EGFR-inhibition and radiotherapy sensitivity in head and neck cancer models.

Hanneke Stegeman; Johannes H.A.M. Kaanders; Albert J. van der Kogel; Mari Iida; Deric L. Wheeler; Paul N. Span; Johan Bussink

BACKGROUND AND PURPOSE EGFR-inhibitor Cetuximab (C225) improves the efficacy of radiotherapy in only a subgroup of HNSCC patients. Identification of predictive tumor characteristics is essential to improve patient selection. MATERIAL AND METHODS Response to C225 and/or radiotherapy was assessed with tumor growth delay assays in 4 HNSCC xenograft models with varying EGFR-expression levels. Hypoxia and proliferation were quantified with immunohistochemistry and the expression of proteins involved in C225-resistance with Western blot. RESULTS EGFR-expression did not predict response to C225 and/or radiotherapy. Reduction of hypoxia by C225 was only observed in SCCNij202, which was highly sensitive to C225. Proliferation changes correlated with response to C225 and C225 combined with radiotherapy, as proliferation decreased after C225 treatment in C225-sensitive SCCNij202 and after combined treatment in SCCNij185, which showed a synergistic effect to combined C225-radiotherapy. Furthermore, C225-resistant SCCNij153 tumors expressed high levels of (activated) HER3 and MET. CONCLUSIONS EGFR-expression is needed for C225-response, but is not sufficient to predict response to C225 with or without radiotherapy. However, basal expression of additional growth factor receptors and effects on proliferation, but not hypoxia, correlated with response to combined C225-radiotherapy treatment and are potential clinically relevant predictive biomarkers.


BMC Cancer | 2013

αB-crystallin stimulates VEGF secretion and tumor cell migration and correlates with enhanced distant metastasis in head and neck squamous cell carcinoma

Chantal van de Schootbrugge; Johan Bussink; Paul N. Span; Fred C.G.J. Sweep; Reidar Grénman; Hanneke Stegeman; Ger J. M. Pruijn; Johannes H.A.M. Kaanders; Wilbert C. Boelens

BackgroundαB-crystallin is able to modulate vascular endothelial growth factor (VEGF) secretion. In many solid tumors VEGF is associated with angiogenesis, metastasis formation and poor prognosis. We set out to assess whether αB-crystallin expression is correlated with worse prognosis and whether this is related to VEGF secretion and cell motility in head and neck squamous cell carcinoma (HNSCC).MethodsαB-crystallin expression was determined immunohistochemically in tumor biopsies of 38 HNSCC patients. Locoregional control (LRC) and metastasis-free survival (MFS) of the patients were analyzed in relation to αB-crystallin expression. Additionally, the effects of αB-crystallin knockdown on VEGF secretion and cell motility were studied in vitro.ResultsPatients with higher staining fractions of αB-crystallin exhibited a significantly shorter MFS (Log-Rank test, p < 0.005). Under normoxic conditions αB-crystallin knockdown with two different siRNAs in a HNSCC cell line reduced VEGF secretion 1.9-fold and 2.1-fold, respectively. Under hypoxic conditions, a similar reduction of VEGF secretion was observed, 1.9-fold and 2.2-fold, respectively. The effect on cell motility was assessed by a gap closure assay, which showed that αB-crystallin knockdown decreased the rate by which HNSCC cells were able to close a gap by 1.5- to 2.0-fold.ConclusionsOur data suggest that αB-crystallin expression is associated with distant metastases formation in HNSCC patients. This association might relate to the chaperone function of αB-crystallin in mediating folding and secretion of VEGF and stimulating cell migration.


Molecular Cancer | 2013

Combining radiotherapy with MEK1/2, STAT5 or STAT6 inhibition reduces survival of head and neck cancer lines

Hanneke Stegeman; Johannes H.A.M. Kaanders; Marieke Mg Verheijen; Wenny J.M. Peeters; Deric L. Wheeler; Mari Iida; Reidar Grénman; Albert J. van der Kogel; Paul N. Span; Johan Bussink

BackgroundKinases downstream of growth factor receptors have been implicated in radioresistance and are, therefore, attractive targets to improve radiotherapy outcome in head and neck squamous cell carcinoma (HNSCC) patients.MethodsAn antibody-based array was used to quantify the expression levels of multiple phospho-kinases involved in growth factor signaling in nine untreated or irradiated HNSCC lines. Radiosensitivity was assessed with clonogenic cell survival assays and correlated with the expression levels of the phospho-kinases. Inhibitors of the kinases that were associated with radiosensitivity were tested for their ability to increase radiosensitivity in the 3 most radioresistant HNSCC lines.ResultsThe basal expression of phosphorylated Yes, Src and STAT5A, and the expression after radiotherapy of phosphorylated AKT, MSK1/2, Src, Lyn, Fyn, Hck, and STAT6, were correlated with radiosensitivity in the panel of HNSCC lines. In combination with radiotherapy, inhibitors of AKT, p38 and Src Family Kinases (SFK) were variably able to reduce survival, whereas MEK1/2, STAT5 and STAT6 inhibition reduced survival in all cell lines. The combined effect of radiotherapy and the kinase inhibitors on cell survival was mostly additive, although also supra-additive effects were observed for AKT, MEK1/2, p38 and STAT5 inhibition.ConclusionsKinases of the AKT, MAPK, STAT and SFK pathways correlated with radiosensitivity in a panel of HNSCC lines. Particularly inhibitors against MEK1/2, STAT5 and STAT6 were able to decrease survival in combination with radiotherapy. Hence, inhibitors against these kinases have the potential to improve radiotherapy outcome in HNSCC patients and further research is warranted to confirm this in vivo.


Cancer Treatment Reviews | 2014

Improving chemoradiation efficacy by PI3-K/AKT inhibition

Hanneke Stegeman; Paul N. Span; Johannes H.A.M. Kaanders; Johan Bussink

For many tumor types concurrent chemoradiation is the standard of care for locally advanced disease. Despite this intense treatment overall survival is still poor in various solid tumors. To improve outcome in these patients it is essential to develop new therapeutic strategies that enhance the efficacy of chemoradiation. The PI3-K/AKT pathway is often activated in solid tumors and is known to be an important tumor cell survival pathway. It is also well established that hypoxic tumor cells are resistant to both radiotherapy and chemotherapy. Evidence is emerging that activation of the PI3-K/AKT pathway affects the hypoxia tolerance of tumor cells and is involved in hypoxia-related treatment resistance. Already, the combination of concurrent chemoradiation and PI3-K/AKT inhibition has been explored in phase I studies in non-small cell lung, pancreatic and rectal cancer. This review summarizes the currently available literature concerning PI3-K/AKT signaling in relation to hypoxia and discusses the potential of PI3-K/AKT inhibition to overcome hypoxia-related treatment resistance to chemoradiation. Clinical studies testing the combination of chemoradiation and PI3-K/AKT inhibition and potential methods to predict treatment response are discussed.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2015

Hypoxic regulation of the PERK/ATF4/LAMP3-arm of the unfolded protein response in head and neck squamous cell carcinoma

Anika Nagelkerke; Fred C.G.J. Sweep; Hanneke Stegeman; Reidar Grénman; Johannes H.A.M. Kaanders; Johan Bussink; Paul N. Span

The purpose of this study was to examine the hypoxic regulation of the PKR‐like endoplasmic reticulum kinase (PERK)/activating transcription factor‐4 (ATF4)/lysosome‐associated membrane protein 3 (LAMP3)‐arm of the unfolded protein response (UPR) in head and neck squamous cell carcinoma (HNSCC).


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2013

Hypoxia, metabolism, and growth factor signaling in head and neck squamous cell carcinoma: Correlation between primary and xenograft tumors

Hanneke Stegeman; Saskia E. Rademakers; Paul N. Span; Robert P. Takes; Albert J. van der Kogel; Johannes H.A.M. Kaanders; Johan Bussink

Hypoxia, metabolism, and growth factor signaling are important prognostic features in most solid tumors. The purpose of this study was to determine whether head and neck squamous cell carcinoma (HNSCC) xenografts show similar biological and molecular characteristics as the primary tumor they originate from.


Radiation Research | 2013

EGFR-Inhibition Enhances Apoptosis in Irradiated Human Head and Neck Xenograft Tumors Independent of Effects on DNA Repair

Hanneke Stegeman; Paul N. Span; S. C. Cockx; J.P.W. Peters; P.F.J.W. Rijken; A.J. van der Kogel; Johannes H.A.M. Kaanders; J. Bussink

Epidermal growth factor receptor (EGFR) inhibition using cetuximab improves the efficacy of radiotherapy in only a subgroup of head and neck squamous cell carcinoma (HNSCC) patients. Therefore, to improve patient selection a better understanding of tumor characteristics that affect treatment is necessary. Here, we investigated the effect of cetuximab on repair of radiation-induced DNA damage in a HNSCC xenograft model, which shows a synergistic effect to cetuximab and radiotherapy (SCCNij185) and a HNSCC model, which shows no additive effect of cetuximab to radiotherapy (SCCNij153). In both tumor models, clear increases were seen in the number of 53BP1 and Rad51 foci after irradiation. 53BP1 foci were present at comparable levels in hypoxic and normoxic tumor areas of the tumor xenografts, while the number of Rad51 foci was significantly higher in normoxic areas compared to hypoxic areas (P < 0.05). In both SCCNij185 and SCCNij153 xenografts an increased number of 53BP1 foci was observed in tumors treated with cetuximab and radiotherapy compared to radiotherapy alone. In SCCNij185 this increase was statistically significant in normoxic tumor areas (P = 0.04) and in SCCNij153 in both hypoxic and normoxic areas (P = 0.007 and P = 0.02, respectively). The number of Rad51 foci was not significantly different when cetuximab was added to radiotherapy compared to radiotherapy alone. Levels of pEGFR and pERK1/2 were decreased when cetuximab was added to radiotherapy in SCCNij185, but not in SCCNij153. Apoptosis was also only increased in SCCNij185 tumors at 4 days after cetuximab and radiotherapy treatment (P < 0.01). In conclusion, cetuximab inhibited DNA repair in both HNSCC models, but this effect was not predictive for the radiosensitizing effect of cetuximab in vivo. This lack of correlation may be related to differential effects of cetuximab and radiotherapy on ERK1/2 signaling and a decreased induction of apoptosis by cetuximab and radiotherapy in the resistant model.


International Journal of Radiation Oncology Biology Physics | 2013

Low Phosphorylated AKT Expression in Laryngeal Cancer: Indications for a Higher Metastatic Risk

Monique M. Nijkamp; Paul N. Span; Hanneke Stegeman; Reidar Grénman; Johannes H.A.M. Kaanders; Johan Bussink

PURPOSE To validate the association of phosphorylated (p)AKT with lymph node metastasis in an independent, homogeneous cohort of patients with larynx cancer. METHODS AND MATERIALS Seventy-eight patients with laryngeal cancer were included. Epidermal growth factor receptor, pAKT, vimentin, E-cadherin, hypoxia, and blood vessels were visualized in biopsy material using immunohistochemistry. Positive tumor areas and spatial relationships between markers were assessed by automated image analysis. In 6 laryngeal cancer cell lines, E-cadherin and vimentin messenger RNA was quantified by real-time polymerase chain reaction and by immunohistochemistry before and after treatment with the pAKT inhibitor MK-2206. RESULTS A significant correlation was found between low pAKT in the primary tumor and positive lymph node status (P=.0005). Tumors with lymph node metastases had an approximately 10-fold lower median pAKT value compared with tumors without lymph node metastases, albeit with large intertumor variations, validating our previous results. After inhibition of pAKT in laryngeal cancer cells with MK-2206, up-regulation of vimentin and a downregulation of E-cadherin occurred, consistent with epithelial-mesenchymal transition. CONCLUSION Low pAKT expression in larynx tumors is associated with lymph node metastases. Further, inhibition of pAKT in laryngeal cancer induces epithelial-mesenchymal transition, predisposing for an increased metastatic risk.


Future Science OA | 2016

Interaction between hypoxia, AKT and HIF-1 signaling in HNSCC and NSCLC: implications for future treatment strategies

Hanneke Stegeman; Paul N. Span; Wenny J.M. Peeters; Marieke Mg Verheijen; Reidar Grénman; Tineke W.H. Meijer; Johannes H.A.M. Kaanders; Johan Bussink

Background: Hypoxia is a negative prognostic factor and this study investigated the relationship between hypoxia, hypoxia inducible factor 1 (HIF-1) and AKT signaling in head and neck squamous cell carcinoma (HNSCC) and non-small-cell lung cancer (NSCLC). Results/methodology: pAKT was induced by hypoxia (0.5% O2) in a part of HNSCC (3/4) and squamous (2/3) and adenocarcinoma (1/3) NSCLS lines. AKT-inhibitor MK-2206 reduced hypoxic HIF-1 signaling in most HNSCC cell lines. This reduction did not correlate with hypoxic induction of pAKT or with sensitivity to MK-2206 under hypoxia. Patient biopsies revealed a hypoxia-induced expression pattern of pAKT in HNSCC (n = 16), which was not observed in squamous cell (n = 34) or adenocarcinoma (n = 41) NSCLC. Conclusion: The interaction between hypoxia, HIF-1 and AKT signaling varies between tumor types and histologies, which could significantly affect response to targeted therapies.

Collaboration


Dive into the Hanneke Stegeman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul N. Span

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Johan Bussink

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

J. Bussink

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Deric L. Wheeler

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Mari Iida

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Reidar Grénman

Turku University Hospital

View shared research outputs
Top Co-Authors

Avatar

A.J. van der Kogel

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Albert J. van der Kogel

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Gerben M. Franssen

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge