Network


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

Hotspot


Dive into the research topics where Johan Bussink is active.

Publication


Featured researches published by Johan Bussink.


Lancet Oncology | 2008

Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer

Johan Bussink; Albert J. van der Kogel; Johannes H.A.M. Kaanders

Activation of the phosphatidylinositol-3-kinase (PI3-K)/protein kinase B (AKT) pathway is associated with three major radioresistance mechanisms: intrinsic radioresistance; tumour-cell proliferation; and hypoxia. Monitoring and manipulation of this signal-transduction pathway can have important implications for the management of head and neck cancer, because activation of the PI3-K/AKT pathway is a frequent event in these tumours. PI3-K/AKT signalling regulates cellular processes, including proliferation, invasion, apoptosis, and the upregulation of hypoxia-related proteins. Activation of this pathway can be caused by stimulation of receptor tyrosine kinases, such as epidermal growth factor receptor (EGFR). In clinical trials, a strong and independent association has been noted between expression of activated AKT and treatment outcome. Therefore, the search for molecular predictors of sensitivity to EGFR-directed treatment should be extended to markers of PI3-K/AKT activation. Another strategy might be the direct targeting and inhibition of this pathway. Such inhibition will enhance the efficacy of radiotherapy, by antagonising radiation-induced cellular defense mechanisms, especially in tumours that have activated the PI3-K/AKT cascade. Thus, the activation status of this pathway might be a key element for the prediction of treatment response and for therapeutic targeting in head and neck cancer.


Clinical Cancer Research | 2012

Targeting Hypoxia, HIF-1, and Tumor Glucose Metabolism to Improve Radiotherapy Efficacy

T.W.H. Meijer; Johannes H.A.M. Kaanders; Paul N. Span; Johan Bussink

Radiotherapy, an important treatment modality in oncology, kills cells through induction of oxidative stress. However, malignant tumors vary in their response to irradiation as a consequence of resistance mechanisms taking place at the molecular level. It is important to understand these mechanisms of radioresistance, as counteracting them may improve the efficacy of radiotherapy. In this review, we describe how the hypoxia-inducible factor 1 (HIF-1) pathway has a profound effect on the response to radiotherapy. The main focus will be on HIF-1–controlled protection of the vasculature postirradiation and on HIF-1 regulation of glycolysis and the pentose phosphate pathway. This aberrant cellular metabolism increases the antioxidant capacity of tumors, thereby countering the oxidative stress caused by irradiation. From the results of translational studies and the first clinical phase I/II trials, it can be concluded that targeting HIF-1 and tumor glucose metabolism at several levels reduces the antioxidant capacity of tumors, affects the tumor microenvironment, and sensitizes various solid tumors to irradiation. Clin Cancer Res; 18(20); 5585–94. ©2012 AACR.


BMC Cancer | 2011

Metabolic markers in relation to hypoxia; staining patterns and colocalization of pimonidazole, HIF-1α, CAIX, LDH-5, GLUT-1, MCT1 and MCT4

Saskia E. Rademakers; Jasper Lok; Albert J. van der Kogel; Johan Bussink; Johannes H.A.M. Kaanders

BackgroundThe cellular response of malignant tumors to hypoxia is diverse. Several important endogenous metabolic markers are upregulated under hypoxic conditions. We examined the staining patterns and co-expression of HIF-1α, CAIX, LDH-5, GLUT-1, MCT1 and MCT4 with the exogenous hypoxic cell marker pimonidazole and the association of marker expression with clinicopathological characteristics.Methods20 biopsies of advanced head and neck carcinomas were immunohistochemically stained and analyzed. All patients were given the hypoxia marker pimonidazole intravenously 2 h prior to biopsy taking. The tumor area positive for each marker, the colocalization of the different markers and the distribution of the markers in relation to the blood vessels were assessed by semiautomatic quantitative analysis.ResultsMCT1 staining was present in hypoxic (pimonidazole stained) as well as non-hypoxic areas in almost equal amounts. MCT1 expression showed a significant overall correlation (r = 0.75, p < 0.001) and strong spatial relationship with CAIX. LDH-5 showed the strongest correlation with pimonidazole (r = 0.66, p = 0.002). MCT4 and GLUT-1 demonstrated a typical diffusion-limited hypoxic pattern and showed a high degree of colocalization. Both MCT4 and CAIX showed a higher expression in the primary tumor in node positive patients (p = 0.09 both).ConclusionsColocalization and staining patterns of metabolic and hypoxia-related proteins provides valuable additional information over single protein analyses and can improve the understanding of their functions and environmental influences.


Molecular Oncology | 2008

Molecular aspects of tumour hypoxia

Saskia E. Rademakers; Paul N. Span; Johannes H.A.M. Kaanders; Fred C.G.J. Sweep; Albert J. van der Kogel; Johan Bussink

Hypoxia is an important feature of the microenvironment of a wide range of solid tumours. Its critical role in radio‐ and chemoresistance and its significance as an adverse prognostic factor have been well established over the last decades. On a cellular level, hypoxia evokes a complex molecular response with a central role for the HIF‐1 pathway. The cellular processes under control of HIF‐1 contain important prognostic information and comprise potential candidates for directing hypoxia‐modifying therapies. This review will provide an overview of the current knowledge on the molecular aspects of tumour hypoxia and the link to clinical practice.


Breast Cancer Research | 2013

Hypoxia stimulates migration of breast cancer cells via the PERK/ATF4/LAMP3-arm of the unfolded protein response

Anika Nagelkerke; Johan Bussink; Hilda Mujcic; Bradly G. Wouters; Steffi Lehmann; Fred C.G.J. Sweep; Paul N. Span

IntroductionThe hypoxia-inducible factor (HIF)-1 pathway can stimulate tumor cell migration and metastasis. Furthermore, hypoxic tumors are associated with a poor prognosis. Besides the HIF-1 pathway, the unfolded protein response (UPR) is also induced by hypoxic conditions. The PKR-like ER kinase (PERK)/activating transcription factor 4 (ATF4)-arm of the UPR induces expression of lysosomal-associated membrane protein 3 (LAMP3), a factor that has been linked to metastasis and poor prognosis in solid tumors. In this study the role of UPR-induced LAMP3 in hypoxia-mediated migration of breast cancer cells was examined.MethodsA number of in vitro metastasis models were used to study the migration and invasion of MDA-MB-231 breast cancer cells under hypoxic conditions. PERK, ATF4 and their downstream factor LAMP3 were knocked down to examine their role in cell migration. In addition, multicellular tumor spheroids were used to study the involvement of the tumor microenvironment in invasion.ResultsUsing transwell assays, migration of different breast cancer cell lines was assessed. A direct correlation was found between cell migration and baseline LAMP3 expression. Furthermore, moderate hypoxia (1% O2) was found to be optimal in stimulating migration of MDA-MB-231 cells. siRNA mediated knockdown of PERK, ATF4 and LAMP3 reduced migration of cells under these conditions. Using gap closure assays, similar results were found. In a three-dimensional invasion assay into collagen, LAMP3 knockdown cells showed a diminished capacity to invade compared to control cells when collectively grown in multicellular spheroids.ConclusionsThus, the PERK/ATF4/LAMP3-arm of the UPR is an additional pathway mediating hypoxia-induced breast cancer cell migration.


International Journal of Cancer | 2008

Aerobic glycolysis in cancers: Implications for the usability of oxygen‐responsive genes and fluorodeoxyglucose‐PET as markers of tissue hypoxia

Morten Busk; Michael R. Horsman; Paul E.G. Kristjansen; Albert J. van der Kogel; Johan Bussink; Jens Overgaard

The hypoxia‐responsiveness of the glycolytic machinery may allow pretreatment identification of hypoxic tumors from HIF‐1 targets (e.g., Glut‐1) or [18F]‐fluorodeoxyglucose positron emission tomography but results have been mixed. We hypothesized that this discrepancy is an inevitable consequence of elevated aerobic glycolysis in tumors (Warburg effect) as energetics in predominantly glycolytic cells is little affected by hypoxia. Accordingly, we characterized glycolytic and mitochondrial ATP generation in normoxic and anoxic cell lines. Measurements demonstrated that most cancer cells rely largely on aerobic glycolysis as it accounts for 56–63% of their ATP budget, but in the cervical carcinoma SiHa, ATP synthesis was mainly mitochondrial. Moreover, the stimulatory effect of anoxia on glycolytic flux was inversely correlated to the relative reliance on aerobic glycolysis. Next, tumor cells representing a Warburg or a nonglycolytic phenotype were grown in mice and spatial patterns of hypoxia (pimonidazole‐stained), Glut‐1 expression and 18F‐FDG uptake were analysed on sectioned tumors. Only in SiHa tumors did foci of elevated glucose metabolism consistently colocalize with regions of hypoxia and elevated Glut‐1 expression. In contrast, spatial patterns of Glut‐1 and pimonidazole staining correlated reasonably well in all tumors. In conclusion, Glut‐1s value as a hypoxia marker is not severely restricted by aerobic glycolysis. In contrast, the specificity of 18F‐FDG uptake and Glut‐1 expression as markers of regional hypoxia and glucose metabolism, respectively, scales inversely with the intensity of the Warburg effect. This linkage suggests that multi‐tracer imaging combining FDG and hypoxia‐specific markers may provide therapeutically relevant information on tumor energetic phenotypes.


European Journal of Nuclear Medicine and Molecular Imaging | 2008

Cellular uptake of PET tracers of glucose metabolism and hypoxia and their linkage

Morten Busk; Michael R. Horsman; Steen Jakobsen; Johan Bussink; Albert J. van der Kogel; Jens Overgaard

PurposeTumour hypoxia and elevated glycolysis (Warburg effect) predict poor prognosis. Each parameter is assessable separately with positron emission tomography, but they are linked through anaerobic glycolysis (Pasteur effect). Here, we compare the oxygenation-dependent retention of fluoroazomycin arabinoside ([18F]FAZA), a promising but not well-characterised hypoxia-specific tracer, and fluorodeoxyglucose ([18F]FDG) in four carcinoma cell lines.MethodsCells seeded on coverslips were positioned in modified Petri dishes that allow physically separated cells to share the same tracer-containing medium pool. Following oxic, hypoxic or anoxic tracer incubation, coverslips were analysed for radioactivity ([18F]FDG + [18F]FAZA) or re-incubated in tracer-free oxygenated medium and then measured ([18F]FAZA). Next, we tested the reliability of [18F]FDG as a relative measure of glucose metabolic rate. Finally, from two cell lines, xenografts were established in mice, and the tracer distribution between hypoxic and well-oxygenated areas were deduced from tissue sections.ResultsThree hours of anoxia strongly stimulated [18F]FAZA retention with anoxic-to-oxic uptake ratios typically above 30. Three out of four cell lines displayed similar selectivity of [18F]FDG versus glucose, but oxic uptake and anoxic-to-oxic uptake ratio of [18F]FDG varied considerably. Although less pronounced, [18F]FAZA also showed superior in vivo hypoxia specificity compared with [18F]FDG.Conclusions[18F]FAZA displays excellent in vitro characteristics for hypoxia imaging including modest cell-to-cell line variability and no binding in oxic cells. In contrast, the usability of [18F]FDG as a surrogate marker for hypoxia is questionable due to large variations in baseline (oxic) glucose metabolism and magnitudes of the Pasteur effects.


Proceedings of the National Academy of Sciences of the United States of America | 2013

PERK/eIF2α signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS

Ludwig Dubois; Tom G. Keulers; Twan van den Beucken; Philippe Lambin; Johan Bussink; Albert J. van der Kogel; Marianne Koritzinsky; Bradly G. Wouters

Hypoxia is a common feature of tumors and an important contributor to malignancy and treatment resistance. The ability of tumor cells to survive hypoxic stress is mediated in part by hypoxia-inducible factor (HIF)-dependent transcriptional responses. More severe hypoxia activates endoplasmatic reticulum stress responses, including the double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2α (eIF2α)-dependent arm of the unfolded protein response (UPR). Although several studies implicate important roles for HIF and UPR in adaption to hypoxia, their importance for hypoxic cells responsible for therapy resistance in tumors is unknown. By using isogenic models, we find that HIF and eIF2α signaling contribute to the survival of hypoxic cells in vitro and in vivo. However, the eIF2α-dependent arm of the UPR is uniquely required for the survival of a subset of hypoxic cells that determine tumor radioresistance. We demonstrate that eIF2α signaling induces uptake of cysteine, glutathione synthesis, and protection against reactive oxygen species produced during periods of cycling hypoxia. Together these data imply that eIF2α signaling is a critical contributor to the tolerance of therapy-resistant cells that arise as a consequence of transient changes in oxygenation in solid tumors and thus a therapeutic target in curative treatments for solid cancers.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2007

Tumor microenvironment in head and neck squamous cell carcinomas: Predictive value and clinical relevance of hypoxic markers. A review

Ilse J. Hoogsteen; H.A.M. Marres; Johan Bussink; Albert J. van der Kogel; Johannes H.A.M. Kaanders

Hypoxia and tumor cell proliferation are important factors determining the treatment response of squamous cell carcinomas of the head and neck. Successful approaches have been developed to counteract these resistance mechanisms although usually at the cost of increased short‐ and long‐term side effects. To provide the best attainable quality of life for individual patients and the head and neck cancer patient population as a whole, it is of increasing importance that tools be developed that allow a better selection of patients for these intensified treatments.


Radiotherapy and Oncology | 2011

Expression of E-cadherin and vimentin correlates with metastasis formation in head and neck squamous cell carcinoma patients

Monique M. Nijkamp; Paul N. Span; Ilse J. Hoogsteen; Albert J. van der Kogel; Johannes H.A.M. Kaanders; Johan Bussink

PURPOSE E-cadherin is a transmembrane glycoprotein, involved in cell-cell adhesion and epithelial-mesenchymal transition (EMT). Vimentin is highly expressed in mesenchymal cells and is positively correlated with increased metastasis. Here we set out to determine the expression of E-cadherin and vimentin in head and neck squamous cell carcinomas (HNSCC). PATIENTS AND METHODS Twenty-six patients with primary stage II-IV HNSCC were included. E-cadherin and vimentin were visualised using immunohistochemistry, semi-automatically analysed for expression patterns and correlated with the clinical behaviour of these tumours. RESULTS A large variation in E-cadherin and vimentin expression was observed between tumours (median 17% range 0-51% respectively median 0% range 0-20%). Tumours with low E-cadherin expression showed a significantly higher incidence of metastasis formation compared to tumours with high expression (81% versus 19%, p=0.004). Enhanced expression of vimentin was associated with a trend towards a higher metastatic risk (33% versus 77%) compared to tumours without expression of vimentin. All patients with low E-cadherin and high vimentin expression (an EMT-phenotype) developed distant metastases versus only 44% of the other patients (p=0.008). CONCLUSION Loss of E-cadherin and gain of vimentin may be associated with enhanced migration of tumour cells, leading to higher metastatic risk of HNSCC patients.

Collaboration


Dive into the Johan Bussink'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

Albert J. van der Kogel

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Fred C.G.J. Sweep

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Hanneke Stegeman

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Anika Nagelkerke

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Wim J.G. Oyen

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jasper Lok

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Saskia E. Rademakers

Radboud University Nijmegen Medical Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge