Network


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

Hotspot


Dive into the research topics where Hester van Cruijsen is active.

Publication


Featured researches published by Hester van Cruijsen.


Clinical Cancer Research | 2008

Sunitinib-Induced Myeloid Lineage Redistribution in Renal Cell Cancer Patients: CD1c+ Dendritic Cell Frequency Predicts Progression-Free Survival

Hester van Cruijsen; Astrid A.M. van der Veldt; Laura Vroling; Dinja Oosterhoff; Henk J. Broxterman; Rik J. Scheper; Giuseppe Giaccone; John B. A. G. Haanen; Alfons J.M. van den Eertwegh; Epie Boven; Klaas Hoekman; Tanja D. de Gruijl

Purpose: A disturbed myeloid lineage development with abnormally abundant neutrophils and impaired dendritic cell (DC) differentiation may contribute to tumor immune escape. We investigated the effect of sunitinib, a tyrosine kinase inhibitor of fms-like tyrosine kinase-3, KIT, and vascular endothelial growth factor receptors, on myeloid differentiation in renal cell cancer (RCC) patients. Experimental Design: Twenty-six advanced RCC patients were treated with sunitinib in a 4-week on/2-week off schedule. Enumeration and extensive phenotyping of myeloid subsets in the blood was done at baseline and at weeks 4 and 6 of the first treatment cycle. Baseline patient data were compared with sex- and age-matched healthy donor data. Results: Baseline frequencies of DC subsets were lower in RCC patients than in healthy donors. After 4 weeks of sunitinib treatment, a generalized decrease in myeloid frequencies was observed. Whereas neutrophils and monocytes, which were both abnormally high at baseline, remained low during the 2-week off period, DC rates recovered, resulting in a normalized myeloid lineage distribution. Subsequent to sunitinib treatment, an increase to high levels of myeloid DC (MDC) subset frequencies relative to other myeloid subsets, was specifically observed in patients experiencing tumor regression. Moreover, high CD1c/BDCA-1+ MDC frequencies were predictive for tumor regression and improved progression-free survival. Conclusion: The sunitinib-induced myeloid lineage redistribution observed in advanced RCC patients is consistent with an improved immune status. Immunologic recovery may contribute to clinical efficacy as suggested by the finding of highly increased MDC frequencies relative to other myeloid subsets in patients with tumor regression.


International Journal of Cancer | 2005

Epidermal growth factor receptor and angiogenesis: Opportunities for combined anticancer strategies

Hester van Cruijsen; Giuseppe Giaccone; Klaas Hoekman

Tumor‐induced angiogenesis is essential for malignant growth. This mini review focuses on the role of vascular endothelial growth factor (VEGF) and epidermal growth factor (EGF) and their receptors in this process, and the rationale to combine inhibitors of these growth factors as anticancer therapy. Concomitantly, targeting the VEGF(R) and the EGF(R) signaling pathway may circumvent the problem of acquired resistance to EGFR inhibitors. By targeting both pathways, the antiangiogenic effect may be more pronounced, which may lead to greater antitumor activity. Preliminary efficacy data from clinical trials encourage further exploration of this combined anticancer strategy.


BMC Cancer | 2009

Tissue micro array analysis of ganglioside N -glycolyl GM3 expression and signal transducer and activator of transcription (STAT)-3 activation in relation to dendritic cell infiltration and microvessel density in non-small cell lung cancer

Hester van Cruijsen; Marielle Gallegos Ruiz; Paul van der Valk; Tanja D. de Gruijl; Giuseppe Giaccone

BackgroundTumor immune escape and angiogenesis contribute to tumor progression, and gangliosides and activation of signal transducer and activator of transcription (STAT)-3 are implicated in these processes. As both are considered as novel therapeutic targets, we assessed the possible association of ganglioside GM3 expression and STAT3 activation with suppression of dendritic cell (DC) activation and angiogenesis in non-small cell lung cancer (NSCLC).MethodsImmunohistochemistry was performed on a tissue array to determine N-glycolyl GM3 (GM3) and phosphorylated STAT3 (pSTAT3) expression in 176 primary NSCLC resections. Median values of GM3 and pSTAT3 expression were used as cut off. Microvessel density (MVD) was determined by CD34 staining and morphology. CD1a and CD83 were used to determine infiltrating immature and mature dendritic cells, respectively.Results94% and 71% of the NSCLC samples expressed GM3 and nuclear pSTAT3, respectively. Median overall survival was 40.0 months. Both low GM3 expression and high pSTAT3 expression were associated with a worse survival, which reached near significance for GM3 (P = 0.08). Microvessel density (MVD), determined by CD34 staining and morphology, was lower in NSCLC samples with high GM3 expression. CD1a+ cells (immature DCs) were more frequent in NSCLC tissues as compared to peritumoral lung tissue, while CD83+ cells (mature DCs) were more frequent in peritumoral lung tissue. CD83+ DCs were less frequent in NSCLC tissues with high GM3 expression.ConclusionGM3 and pSTAT3 are widely expressed in NSCLC. Based on CD83 expression, GM3, but not pSTAT3, appeared to be involved in tumor-induced DC suppression. pSTAT3 expression was not associated with MVD, while GM3 might play an anti-angiogenic role.


European Journal of Cancer | 2010

Phase I evaluation of cediranib, a selective VEGFR signalling inhibitor, in combination with gefitinib in patients with advanced tumours

Hester van Cruijsen; Emile E. Voest; Cornelis J. A. Punt; Klaas Hoekman; Petronella O. Witteveen; Martijn R. Meijerink; Thomas Puchalski; Jane Robertson; Owain Saunders; Juliane M. Jürgensmeier; Carla M.L. van Herpen; Giuseppe Giaccone

AIM Cediranib is a highly potent inhibitor of vascular endothelial growth factor receptor (VEGFR) signalling. Preclinical and clinical data suggest that inhibition of the VEGFR and epidermal growth factor receptor (EGFR) pathways may be synergistic. Combination treatment with cediranib and gefitinib, an EGFR signalling inhibitor, was evaluated in patients with advanced solid tumours. PATIENTS AND METHODS Ninety patients received treatment in this four-part, open-label study (NCT00502060). The patients received once-daily oral doses of cediranib (20-45mg) and gefitinib 250mg (part A1; n=16) or 500mg (part B1; n=44). A cohort expansion phase investigated the potential pharmacokinetic interaction of cediranib 30mg with gefitinib 250mg (part A2; n=15) or 500mg (part B2; n=15). The primary objective was to assess the safety and tolerability of cediranib with gefitinib. Secondary assessments included pharmacokinetics, efficacy and pharmacodynamics. RESULTS Combination treatment was generally well tolerated; the protocol-defined maximum-tolerated dose of cediranib was 30mg/day with gefitinib 250mg/day (part A1) and cediranib 45mg/day was the maximum dose investigated with gefitinib 500mg/day (part B1). The most common adverse events were diarrhoea (84 [93%]), anorexia (63 [70%]) and fatigue (60 [67%]). Cediranib pharmacokinetic parameters were not substantially different when given alone or in combination with gefitinib. Gefitinib pharmacokinetic parameters were similar to those seen previously with gefitinib monotherapy. Efficacy results included eight (9%) confirmed partial responses (6 renal; 1 lung; 1 osteosarcoma) and 38 (42%) patients with stable disease. Pharmacodynamic assessments demonstrated changes in levels of VEGF and soluble VEGFR-2 following treatment. CONCLUSIONS Combination treatment was generally well tolerated and showed encouraging antitumour activity in patients with advanced solid tumours. These results merit further exploration.


Journal of Thoracic Oncology | 2007

Genetic heterogeneity in patients with multiple neoplastic lung lesions: a report of three cases.

Marielle Gallegos Ruiz; Hester van Cruijsen; Egbert F. Smit; Katrien Grünberg; Gerrit A. Meijer; Jose A. Rodriguez; Bauke Ylstra; Giuseppe Giaccone

Introduction: It is important to determine the relation among the various lesions in patients presenting with multiple malignant lung tumors to define the best treatment approach. A better understanding of the molecular alterations present in the different lesions may help in defining this relation. Methods: We performed a detailed molecular analysis of several tumor specimens obtained from three patients presenting with multiple lung lesions. Tumor specimens were analyzed for epidermal growth factor receptor (EGFR) and k-ras mutations by direct DNA sequencing. In addition, a genome-wide chromosomal copy number analysis was performed on DNA extracted from the various lesions using array-based comparative genomic hybridization. Results: In one case, a deletion of 15 base pairs in exon 19 of EGFR was present in all tumor sites analyzed. Furthermore, a similar pattern of chromosomal aberrations was observed among the various lesions, suggesting that they share the same clonal origin. In the other two cases, in contrast, we identified distinct k-ras genotypes among the various lesions from the same patient. These lesions, moreover, showed different chromosomal aberration patterns, indicating that they may have different underlying pathways of tumorigenesis. Conclusion: Our results show that EGFR and k-ras mutation analysis, combined with chromosomal copy number profiling, can help in defining the relationship among different tumors in one patient.


OncoImmunology | 2012

Tumor-mediated inhibition of human dendritic cell differentiation and function is consistently counteracted by combined p38 MAPK and STAT3 inhibition.

Dinja Oosterhoff; Sinéad M. Lougheed; Rieneke van de Ven; Jelle Lindenberg; Hester van Cruijsen; Lotte Hiddingh; Jan Kroon; Alfons J.M. van den Eertwegh; Basav N. Hangalapura; Rik J. Scheper; Tanja D. de Gruijl

Targeting dendritic cells (DC) through the release of suppressive factors is an effective means for tumors to escape immune control. We assessed the involvement of downstream signaling through the JAK2/STAT3 and p38 MAPK pathways in tumor-induced suppression of human DC development. Whereas the JAK2/STAT3 pathway has been pinpointed in mouse studies as a key regulator of myeloid suppression, in human DC this is less well established. We studied the effects of STAT3 inhibition on the suppression of monocyte-derived DC differentiation mediated by a short-list of four predominant suppressive factors and found that pharmacological STAT3 inhibition could only counteract the effects of IL-6. Accordingly, in testing a panel of supernatants derived from 11 cell lines representing various types of solid tumors, STAT3 inhibition only modestly affected the suppressive effects of a minority of supernatants. Importantly, combined interference in the STAT3 and p38 pathways completely prevented inhibition of DC differentiation by all tested supernatants and effected superior DC function, evidenced by increased allogeneic T cell reactivity with elevated IL-12p70/IL-10 ratios and Th1 skewing. Combined STAT3 and p38 inhibition also afforded superior protection against the suppressive effects of primary glioma and melanoma supernatants and induced a shift from CD14+ cells to CD1a+ cells in metastatic melanoma single-cell suspensions, indicating a potential for improved DC differentiation in the tumor microenvironment. We conclude that combined interference in the STAT3 and p38 MAPK signaling pathways is a promising approach to overcome tumor-induced inhibitory signaling in DC precursors and will likely support clinical immunotherapeutic strategies.


Immunotherapy | 2011

Cross-talk between tumor and myeloid cells: how to tip the balance in favor of antitumor immunity

Jelle J. Lindenberg; Cynthia M. Fehres; Hester van Cruijsen; Dinja Oosterhoff; Tanja D. de Gruijl

Myeloid differentiation is often disturbed in cancer, leading to reduced frequencies of immunostimulatory dendritic cells and an over-representation of immunosuppressive immature myeloid cells, granulocytes and macrophages. As a result of this skewed myeloid differentiation, a highly immunosuppressive myeloid subset becomes prevalent during cancer development; these myeloid-derived suppressor cells are also recruited as a collateral to certain protumorigenic inflammatory processes, resulting in an effective downregulation of T-cell-mediated immune surveillance and antitumor immunity. In this article, some of the important myeloid cell subsets and mediators involved in cancer-related immune suppression are reviewed. Furthermore, cross-talk between tumors and the myeloid compartment, and ways in which it can suppress effective cell-mediated immunity, are discussed, as well as possible therapeutic approaches to tip the balance in favor of antitumor immunity.


Thrombosis and Haemostasis | 2007

VEGFR2 expressing circulating (progenitor) cell populations in volunteers and cancer patients

Laura Vroling; Yuana Yuana; Gerrit Jan Schuurhuis; Victor W.M. van Hinsbergh; Chad Gundy; Richard R. de Haas; Hester van Cruijsen; Epie Boven; Klaas Hoekman; Henk J. Broxterman

Circulating cells of several lineages are thought to participate in angiogenesis and tumor growth. Experimental studies in tumor-bearing mice have pointed to the potential importance of VEGF-responding circulating (endothelial) progenitor cells in tumor growth. We have studied circulating CD31- and/or CD34-positive cell populations with a low to moderate VEGFR2 expression in human volunteers and cancer patients. We recognized four cell populations, which were further characterized by their content of major hematopoietic progenitor, monocytic, endothelial and platelet markers. After establishing the test-retest stability of the measurements in nine patients, we determined the frequencies of the various cell populations in a group of 20 volunteers and 14 cancer patients. Two populations were markedly increased in cancer patients. Small CD45(neg)/CD34(bright)/VEGFR2+ cells amounted to 12 and 64 cells/ml (P < 0.0001), respectively, and 246/ml and 578/ml VEGFR2+/CD45(bright) (/CD14+) monocytic cells were present in controls and cancer patients, respectively (P = 0.017). A third population of CD45(dim)/CD34(bright)/VEGFR2(low) cells amounted to 25 and 30 cells/ml (P = 0.38). Unexpectedly, a population of mainly anucleated CD45(low)/CD31(bright)/CD41(bright) cells was present in numbers of 9,076 and 16,697/ml (P = 0.04) in volunteers and cancer patients, which contained a VEGFR2(low) (compared to IgG isotype control) expressing population amounting to 1,142 and 1,642 cells/ml (P = 0.12). This fourth population probably reflects large platelets. The role of the herein identified VEGFR2+ circulating cell populations deserve further investigation in cancer patients treated with VEGF(R)-targeted therapies. Quantification of such cell populations in the blood of tumor patients may be valuable to monitor the efficacy of anti-angiogenic treatment.


Immunotherapy | 2011

Glioblastoma-induced inhibition of Langerhans cell differentiation from CD34+ precursors is mediated by IL-6 but unaffected by JAK2/STAT3 inhibition

Hester van Cruijsen; Dinja Oosterhoff; Jelle J. Lindenberg; Sinéad M. Lougheed; Cynthia M. Fehres; Karin Weijers; Robert van Boerdonk; Giuseppe Giaccone; Rik J. Scheper; Klaas Hoekman; Tanja D. de Gruijl

AIMS Langerhans cell (LC) infiltration has been observed in glioblastoma, but the glioblastoma microenvironment may be conditioned to resist antitumor immune responses. As little is known about how glioblastoma may affect dendritic cell differentiation, here we set out to delineate the effects of glioblastoma-derived soluble factors on LC differentiation. METHODS CD34(+) precursor cells of the human myeloid cell line MUTZ-3 were differentiated into LC in the presence of conditioned media of the human glioblastoma cell lines U251 or U373 and phenotypically and functionally characterized. RESULTS Glioblastoma-conditioned media inhibited LC differentiation, resulting in functional impairment, as determined by allogeneic mixed leukocyte reactivity, and induction of STAT3 activation. IL-6 blockade completely abrogated these glioblastoma-induced immunosuppressive effects and reduced STAT3 phosphorylation. However, neither addition of JSI-124 (cucurbitacin-I; a JAK2/STAT3 inhibitor), nor of GW5074 (a Raf-1 inhibitor), both of which interfere with signaling pathways reported to act downstream of the IL-6 receptor, prevented the observed inhibitory effects on LC differentiation. CONCLUSION Glioblastoma-derived IL-6 is responsible for the observed suppression of LC differentiation from CD34(+) precursors but appears to exert this effect in a STAT3 and Raf-1 independent fashion.


Archive | 2008

The EGF(R) and VEGF(R) Pathways as Combined Targets for Anti-Angiogenesis Trials in Cancer Therapy

Klaas Hoekman; Hester van Cruijsen; Giuseppe Giaccone

In tumor biology both the EGF(R) and the VEGF(R) pathway are constitutively activated due to genetic abnormalities and ongoing tumor-associated hypoxia. In addition, both pathways can be activated by anticancer therapies such as chemotherapy and radiotherapy, which contributes to the resistance to these treatments. Moreover, VEGF modulates EGFR signaling and EGF induces VEGF activity. Therefore, both pathways are logical targets for therapy, and because of their parallel and reciprocal activation, dual inhibition of the EGF(R) and VEGF(R) signaling makes sense. In this chapter we discuss the possibilities for integrated anti-angiogenesis therapy directed at these two biological systems.

Collaboration


Dive into the Hester van Cruijsen's collaboration.

Top Co-Authors

Avatar

Giuseppe Giaccone

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Klaas Hoekman

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tanja D. de Gruijl

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dinja Oosterhoff

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rik J. Scheper

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Epie Boven

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Henk J. Broxterman

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Laura Vroling

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge