Network


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

Hotspot


Dive into the research topics where Martha Chekenya is active.

Publication


Featured researches published by Martha Chekenya.


Molecular Cancer | 2010

Survival signalling and apoptosis resistance in glioblastomas: opportunities for targeted therapeutics.

Camilla Krakstad; Martha Chekenya

Glioblastoma multiforme (GBM) is the most common primary brain tumour in adults and one of the most aggressive cancers in man. Despite technological advances in surgical management, combined regimens of radiotherapy with new generation chemotherapy, the median survival for these patients is 14.6 months. This is largely due to a highly deregulated tumour genome with opportunistic deletion of tumour suppressor genes, amplification and/or mutational hyper-activation of receptor tyrosine kinase receptors. The net result of these genetic changes is augmented survival pathways and systematic defects in the apoptosis signalling machinery. The only randomised, controlled phase II trial conducted targeting the epidermal growth factor receptor (EGFR) signalling with the small molecule inhibitor, erlotinib, has showed no therapeutic benefit. Survival signalling and apoptosis resistance in GBMs can be viewed as two sides of the same coin. Targeting increased survival is unlikely to be efficacious without at the same time targeting apoptosis resistance. We have critically reviewed the literature regarding survival and apoptosis signalling in GBM, and highlighted experimental, preclinical and recent clinical trials attempting to target these pathways. Combined therapies simultaneously targeting apoptosis and survival signalling defects might shift the balance from tumour growth stasis to cytotoxic therapeutic responses that might be associated with greater therapeutic benefits.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Angiogenesis-independent tumor growth mediated by stem-like cancer cells

Per Øystein Sakariassen; Lars Prestegarden; Jian Wang; Kai-Ove Skaftnesmo; Rupavathana Mahesparan; Carla F. M. Molthoff; Peter Sminia; Eirik Sundlisæter; Anjan Misra; Berit B. Tysnes; Martha Chekenya; Hans Peters; Gabriel Lende; Karl-Henning Kalland; Anne Margrete Øyan; Kjell Petersen; Inge Jonassen; Albert J. van der Kogel; Burt G. Feuerstein; A. Jorge A. Terzis; Rolf Bjerkvig; Per Øyvind Enger

In this work, highly infiltrative brain tumors with a stem-like phenotype were established by xenotransplantation of human brain tumors in immunodeficient nude rats. These tumors coopted the host vasculature and presented as an aggressive disease without signs of angiogenesis. The malignant cells expressed neural stem cell markers, showed a migratory behavior similar to normal human neural stem cells, and gave rise to tumors in vivo after regrafting. Serial passages in animals gradually transformed the tumors into an angiogenesis-dependent phenotype. This process was characterized by a reduction in stem cells markers. Gene expression profiling combined with high throughput immunoblotting analyses of the angiogenic and nonangiogenic tumors identified distinct signaling networks in the two phenotypes. Furthermore, proinvasive genes were up-regulated and angiogenesis signaling genes were down-regulated in the stem-like tumors. In contrast, proinvasive genes were down-regulated in the angiogenesis-dependent tumors derived from the stem-like tumors. The described angiogenesis-independent tumor growth and the uncoupling of invasion and angiogenesis, represented by the stem-like cancer cells and the cells derived from them, respectively, point at two completely independent mechanisms that drive tumor progression. This article underlines the need for developing therapies that specifically target the stem-like cell pools in tumors.


Oncogene | 2008

The progenitor cell marker NG2/MPG promotes chemoresistance by activation of integrin-dependent PI3K/Akt signaling

Martha Chekenya; Camilla Krakstad; Agnete Svendsen; Inger Anne Netland; Vidar Staalesen; Berit B. Tysnes; Frode Selheim; Jian Wang; Per Øystein Sakariassen; T Sandal; Per Eystein Lønning; T Flatmark; Per Øyvind Enger; Rolf Bjerkvig; M Sioud; W B Stallcup

Chemoresistance represents a major problem in the treatment of many malignancies. Overcoming this obstacle will require improved understanding of the mechanisms responsible for this phenomenon. The progenitor cell marker NG2/melanoma proteoglycan (MPG) is aberrantly expressed by various tumors, but its role in cell death signaling and its potential as a therapeutic target are largely unexplored. We have assessed cytotoxic drug-induced cell death in glioblastoma spheroids from 15 patients, as well as in five cancer cell lines that differ with respect to NG2/MPG expression. The tumors were treated with doxorubicin, etoposide, carboplatin, temodal, cisplatin and tumor necrosis factor (TNF)α. High NG2/MPG expression correlated with multidrug resistance mediated by increased activation of α3β1 integrin/PI3K signaling and their downstream targets, promoting cell survival. NG2/MPG knockdown with shRNAs incorporated into lentiviral vectors attenuated β1 integrin signaling revealing potent antitumor effects and further sensitized neoplastic cells to cytotoxic treatment in vitro and in vivo. Thus, as a novel regulator of the antiapoptotic response, NG2/MPG may represent an effective therapeutic target in several cancer subtypes.


The FASEB Journal | 2002

NG2 proteoglycan promotes angiogenesis-dependent tumor growth in CNS by sequestering angiostatin

Martha Chekenya; Mari Hjelstuen; Per Øyvind Enger; Frits Thorsen; Anne L. Jacob; Beatrice Probst; Olav Haraldseth; Geoff Pilkington; Arthur Butt; Joel M. Levine; Rolf Bjerkvig

During embryogenesis, the NG2 proteoglycan is expressed on immature capillary vessels, but as the vessels mature they lose this expression. NG2 is up‐regulated in high‐grade gliomas, but it is not clear to what extent it contributes to malignant progression. Using a combination of high spatial and temporal resolution functional magnetic resonance imaging and histopathological analyses, we show here that overexpression of NG2 increases tumor initiation and growth rates, neovascularization, and cellular proliferation, which predisposes to a poorer survival outcome. By confocal microscopy and cDNA gene array expression profiles, we also show that NG2 tumors express lower levels of hypoxia inducible factor‐1α, vascular endothelial growth factor, and endogenous angiostatin in vivo compared with wild‐type tumors. Moreover, we demonstrate that NG2‐positive cells bind, internalize, and coimmunoprecipitate with angiostatin. These results indicate a unique role for NG2 in regulating the transition from small, poorly vascularized tumors to large, highly vascular gliomas in situ by sequestering angiostatin.


Acta Neuropathologica | 2011

Expression of the progenitor marker NG2/CSPG4 predicts poor survival and resistance to ionising radiation in glioblastoma

Agnete Svendsen; Joost J.C. Verhoeff; Heike Immervoll; Jan Brogger; Justyna Kmiecik; Aurélie Poli; Inger Anne Netland; Lars Prestegarden; Jesús Planagumà; Anja Torsvik; Anneli Bohne Kjersem; Per Øystein Sakariassen; Jan Ingemann Heggdal; Wouter R. van Furth; Rolf Bjerkvig; Morten Lund-Johansen; Per Øyvind Enger; Joerg Felsberg; Nicolaas H. C. Brons; Karl Johan Tronstad; Andreas Waha; Martha Chekenya

Glioblastoma (GBM) is a highly aggressive brain tumour, where patients respond poorly to radiotherapy and exhibit dismal survival outcomes. The mechanisms of radioresistance are not completely understood. However, cancer cells with an immature stem-like phenotype are hypothesised to play a role in radioresistance. Since the progenitor marker neuron-glial-2 (NG2) has been shown to regulate several aspects of GBM progression in experimental systems, we hypothesised that its expression would influence the survival of GBM patients. Quantification of NG2 expression in 74 GBM biopsies from newly diagnosed and untreated patients revealed that 50% express high NG2 levels on tumour cells and associated vessels, being associated with significantly shorter survival. This effect was independent of age at diagnosis, treatment received and hypermethylation of the O6-methylguanine methyltransferase (MGMT) DNA repair gene promoter. NG2 was frequently co-expressed with nestin and vimentin but rarely with CD133 and the NG2 positive tumour cells harboured genetic aberrations typical for GBM. 2D proteomics of 11 randomly selected biopsies revealed upregulation of an antioxidant, peroxiredoxin-1 (PRDX-1), in the shortest surviving patients. Expression of PRDX-1 was associated with significantly reduced products of oxidative stress. Furthermore, NG2 expressing GBM cells showed resistance to ionising radiation (IR), rapidly recognised DNA damage and effectuated cell cycle checkpoint signalling. PRDX-1 knockdown transiently slowed tumour growth rates and sensitised them to IR in vivo. Our data establish NG2 as an important prognostic factor for GBM patient survival, by mediating resistance to radiotherapy through induction of ROS scavenging enzymes and preferential DNA damage signalling.


PLOS ONE | 2011

Targeting the NG2/CSPG4 Proteoglycan Retards Tumour Growth and Angiogenesis in Preclinical Models of GBM and Melanoma

Jian Wang; Agnete Svendsen; Justyna Kmiecik; Heike Immervoll; Kai Ove Skaftnesmo; Jesús Planagumà; Rolf K. Reed; Rolf Bjerkvig; Hrvoje Miletic; Per Øyvind Enger; Cecilie Brekke Rygh; Martha Chekenya

Aberrant expression of the progenitor marker Neuron-glia 2 (NG2/CSPG4) or melanoma proteoglycan on cancer cells and angiogenic vasculature is associated with an aggressive disease course in several malignancies including glioblastoma multiforme (GBM) and melanoma. Thus, we investigated the mechanism of NG2 mediated malignant progression and its potential as a therapeutic target in clinically relevant GBM and melanoma animal models. Xenografting NG2 overexpressing GBM cell lines resulted in increased growth rate, angiogenesis and vascular permeability compared to control, NG2 negative tumours. The effect of abrogating NG2 function was investigated after intracerebral delivery of lentivirally encoded shRNAs targeting NG2 in patient GBM xenografts as well as in established subcutaneous A375 melanoma tumours. NG2 knockdown reduced melanoma proliferation and increased apoptosis and necrosis. Targeting NG2 in two heterogeneous GBM xenografts significantly reduced tumour growth and oedema levels, angiogenesis and normalised vascular function. Vascular normalisation resulted in increased tumour invasion and decreased apoptosis and necrosis. We conclude that NG2 promotes tumour progression by multiple mechanisms and represents an amenable target for cancer molecular therapy.


BMC Cancer | 2009

A reproducible brain tumour model established from human glioblastoma biopsies

Jian Wang; Hrvoje Miletic; Per Øystein Sakariassen; Peter C. Huszthy; Hege Karine Jacobsen; Narve Brekkå; Xingang Li; Peng Zhao; Sverre Mørk; Martha Chekenya; Rolf Bjerkvig; Per Øyvind Enger

BackgroundEstablishing clinically relevant animal models of glioblastoma multiforme (GBM) remains a challenge, and many commonly used cell line-based models do not recapitulate the invasive growth patterns of patient GBMs. Previously, we have reported the formation of highly invasive tumour xenografts in nude rats from human GBMs. However, implementing tumour models based on primary tissue requires that these models can be sufficiently standardised with consistently high take rates.MethodsIn this work, we collected data on growth kinetics from a material of 29 biopsies xenografted in nude rats, and characterised this model with an emphasis on neuropathological and radiological features.ResultsThe tumour take rate for xenografted GBM biopsies were 96% and remained close to 100% at subsequent passages in vivo, whereas only one of four lower grade tumours engrafted. Average time from transplantation to the onset of symptoms was 125 days ± 11.5 SEM. Histologically, the primary xenografts recapitulated the invasive features of the parent tumours while endothelial cell proliferations and necrosis were mostly absent. After 4-5 in vivo passages, the tumours became more vascular with necrotic areas, but also appeared more circumscribed. MRI typically revealed changes related to tumour growth, several months prior to the onset of symptoms.ConclusionsIn vivo passaging of patient GBM biopsies produced tumours representative of the patient tumours, with high take rates and a reproducible disease course. The model provides combinations of angiogenic and invasive phenotypes and represents a good alternative to in vitro propagated cell lines for dissecting mechanisms of brain tumour progression.


Neuropathology and Applied Neurobiology | 2002

The glial precursor proteoglycan, NG2, is expressed on tumour neovasculature by vascular pericytes in human malignant brain tumours

Martha Chekenya; Per Øyvind Enger; Frits Thorsen; Berit B. Tysnes; Safa Al-Sarraj; T A Read; T Furmanek; Rupavathana Mahesparan; Joel M. Levine; Arthur Butt; Geoffrey J. Pilkington; Rolf Bjerkvig

Glial precursor cells express NG2 and GD3 in the developing brain. These antigens are both over‐expressed during neoplasia, which suggests they may have specific functions in the malignant progression of human brain tumours. This study describes the expression of NG2 and GD3 in 28 paediatric and adult brain tumours. Glioblastoma biopsy spheroids were also implanted into nude rats to assess the regional distribution of the molecules within the tumour. These xenografts showed extensive infiltration and growth that mimicked the growth patterns of human gliomas in situ. NG2 was identified in 20 out of 28 brain tumours, where the expression was confined to the main mass of the tumour, and was reduced towards the tumour periphery. NG2 was mainly associated with blood vessels on both the pericyte and basement membrane components of the tumour vasculature. Ki67 (MIB‐1) labelling indicated that NG2 expression was associated with areas of high cellular proliferation. Conversely, all the tumours expressed GD3, which was present both in the tumour main mass and throughout the periphery. Thus, the expression of NG2 may be indicative of tumour progression and might be an amenable target for future therapeutic interventions.


Journal of Neurocytology | 2002

NG2 precursor cells in neoplasia: Functional, histogenesis and therapeutic implications for malignant brain tumours

Martha Chekenya; Geoff Pilkington

Diffusely infiltrating astrocytic tumours of the central nervous system (CNS) are the most frequent intracranial neoplasms and account for more than 60% of all primary brain tumours in man. Until recently, it was generally accepted that the glial component of the mature CNS, consisted of differentiated astrocytes, ependymal cells, oligodendrocytes and the non-neuro-ectodermal microglial cells. There exists a recently recognised population of glial cells that express the NG2 proteoglycan (NG2 cells). NG2 cells are dynamic and undergo rapid morphological changes in response to a variety of CNS pathologies. They are highly motile cells, which interact with various extracellular matrix (ECM) in association with the integrin receptors. During angiogenesis and response to tissue injury, NG2 precursor cells are recruited to sites where vessel growth and repair are occurring. NG2 is over-expressed by both tumour cells and pericytes on the blood vessels of malignant brain tumours. The function of NG2 cells in the CNS, and the notion of them as a source of and/or lineage marker for some gliomas are discussed. In addition, their possible role in glioma angiogenesis, proliferation and invasion will be considered as will their value in provision of targets for clinical and pre-clinical therapeutic strategies in brain tumours.


PLOS ONE | 2009

Hyperoxic Treatment Induces Mesenchymal-to-Epithelial Transition in a Rat Adenocarcinoma Model

Ingrid Moen; Anne Margrete Øyan; Karl-Henning Kalland; Karl Johan Tronstad; Lars A. Akslen; Martha Chekenya; Per Øystein Sakariassen; Rolf K. Reed; Linda Elin Birkhaug Stuhr

Tumor hypoxia is relevant for tumor growth, metabolism and epithelial-to-mesenchymal transition (EMT). We report that hyperbaric oxygen (HBO) treatment induced mesenchymal-to-epithelial transition (MET) in a dimetyl-α-benzantracene induced mammary rat adenocarcinoma model, and the MET was associated with extensive coordinated gene expression changes and less aggressive tumors. One group of tumor bearing rats was exposed to HBO (2 bar, pO2 = 2 bar, 4 exposures à 90 minutes), whereas the control group was housed under normal atmosphere (1 bar, pO2 = 0.2 bar). Treatment effects were determined by assessment of tumor growth, tumor vascularisation, tumor cell proliferation, cell death, collagen fibrils and gene expression profile. Tumor growth was significantly reduced (∼16%) after HBO treatment compared to day 1 levels, whereas control tumors increased almost 100% in volume. Significant decreases in tumor cell proliferation, tumor blood vessels and collagen fibrils, together with an increase in cell death, are consistent with tumor growth reduction and tumor stroma influence after hyperoxic treatment. Gene expression profiling showed that HBO induced MET. In conclusion, hyperoxia induced MET with coordinated expression of gene modules involved in cell junctions and attachments together with a shift towards non-tumorigenic metabolism. This leads to more differentiated and less aggressive tumors, and indicates that oxygen per se might be an important factor in the “switches” of EMT and MET in vivo. HBO treatment also attenuated tumor growth and changed tumor stroma, by targeting the vascular system, having anti-proliferative and pro-apoptotic effects.

Collaboration


Dive into the Martha Chekenya's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacques Zimmer

Ludwig Institute for Cancer Research

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
Researchain Logo
Decentralizing Knowledge