Network


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

Hotspot


Dive into the research topics where Sinéad M. Lougheed is active.

Publication


Featured researches published by Sinéad M. Lougheed.


Cancer Immunology, Immunotherapy | 2006

In vitro priming of tumor-specific cytotoxic T lymphocytes using allogeneic dendritic cells derived from the human MUTZ-3 cell line

Saskia J. A. M. Santegoets; Marco W. J. Schreurs; Allan J. Masterson; Ying Poi Liu; Steffen Goletz; Hans Baumeister; Esther W.M. Kueter; Sinéad M. Lougheed; Alfons J.M. van den Eertwegh; Rik J. Scheper; Erik Hooijberg; Tanja D. de Gruijl

The adoptive transfer of in vitro-induced and expanded tumor-specific cytotoxic T lymphocytes (CTL) presents a promising immunotherapeutic approach for the treatment of cancer. The in vitro induction of tumor-reactive CTL requires repeated stimulation of CTL precursors with dendritic cells (DC). To circumvent problems like scarcity of blood DC precursors and donor variability, it would be attractive to use DC from a non-autologous, unlimited source. DCs derived from the human acute myeloid leukemia (AML) cell line MUTZ-3 are attractive candidates since these DCs closely resemble monocyte-derived DC (MoDC) in terms of phenotype and T cell stimulatory capacity. Here we demonstrate that functional CTL clones could be generated against multiple tumor-associated antigens, i.e., human telomerase reverse transcriptase (hTERT), ErbB3-binding protein-1 (Ebp1), carcinoembryonic antigen (CEA) and Her-2/neu, by stimulating CD8β+ CTL precursors with peptide-loaded allogeneic, HLA-A2-matched MUTZ-3-derived DC. A consistent induction capacity, as determined by MHC tetramer-binding, was found in multiple donors and comparable to autologous peptide-loaded MoDC. Functional characterization at the clonal level revealed the priming of CTL that recognized endogenously processed epitopes on tumor cell lines in an HLA-A2-restricted fashion. Our data indicate that MUTZ-3-derived DC can be used as stimulator cells for in vitro priming and expansion of functional TAA-specific effector CTL. MUTZ-3-derived DCs thus represent a ready and standardized source of allogeneic DC to generate CTL for therapeutic adoptive transfer strategies.


Cancer Immunology, Immunotherapy | 2013

T cell profiling reveals high CD4+CTLA-4+ T cell frequency as dominant predictor for survival after Prostate GVAX/ipilimumab treatment

Saskia J. A. M. Santegoets; Anita G. M. Stam; Sinéad M. Lougheed; Helen Gall; Petra E. T. Scholten; Martine Reijm; Karin Jooss; Natalie Sacks; Kristen Hege; Israel Lowy; Jean Marie Cuillerot; B. Mary E. von Blomberg; Rik J. Scheper; Alfons J.M. van den Eertwegh; Winald R. Gerritsen; Tanja D. de Gruijl

Immune checkpoint blockade enhances antitumor responses, but can also lead to severe immune-related adverse events (IRAE). To avoid unnecessary exposure to these potentially hazardous agents, it is important to identify biomarkers that correlate with clinical activity and can be used to select patients that will benefit from immune checkpoint blockade. To understand the consequences of CTLA-4 blockade and identify biomarkers for clinical efficacy and/or survival, an exploratory T cell monitoring study was performed in a phase I/II dose escalation/expansion trial (nxa0=xa028) of combined Prostate GVAX/ipilimumab immunotherapy. Phenotypic T cell monitoring in peripheral blood before and after Prostate GVAX/ipilimumab treatment revealed striking differences between patients who benefited from therapy and patients that did not. Treatment-induced rises in absolute lymphocyte counts, CD4+ T cell differentiation, and CD4+ and CD8+ T cell activation were all associated with clinical benefit. Moreover, significantly prolonged overall survival (OS) was observed for patients with high pre-treatment frequencies of CD4+CTLA-4+, CD4+PD-1+, or differentiated (i.e., non-naive) CD8+ T cells or low pre-treatment frequencies of differentiated CD4+ or regulatory T cells. Unsupervised clustering of these immune biomarkers revealed cancer-related expression of CTLA-4+ in CD4+ T cells to be a dominant predictor for survival after Prostate GVAX/ipilimumab therapy and to thus provide a putative and much-needed biomarker for patient selection prior to therapeutic CTLA4 blockade.


Journal of Leukocyte Biology | 2006

A CD34+ human cell line model of myeloid dendritic cell differentiation: evidence for a CD14+CD11b+ Langerhans cell precursor

Saskia J. A. M. Santegoets; Allan J. Masterson; Pieter C. van der Sluis; Sinéad M. Lougheed; Donna Fluitsma; Alfons J.M. van den Eertwegh; Herbert M. Pinedo; Rik J. Scheper; Tanja D. de Gruijl

The study of early events in dendritic cell (DC) differentiation is hampered by the lack of homogeneous primary cell systems that allow the study of cytokine‐driven, transitional DC differentiation steps. The CD34+ acute myeloid leukemia cell line MUTZ‐3 displays a unique ability to differentiate into interstitial DC (IDC) and Langerhans cells (LC) in a cytokine‐dependent manner. Phenotypic characterization revealed MUTZ‐3 to consist of three distinct subpopulations. Small CD34+CD14−CD11b− progenitors constitute the proliferative compartment of the cell line with the ability to differentiate through a CD34−CD14−CD11b+ stage to ultimately give rise to a morphologically large, nonproliferating CD14+CD11bhi progeny. These CD14+CD11bhi cells were identified as common, immediate myeloid DC precursors with the ability to differentiate into LC and IDC, exhibiting characteristic and mutually exclusive expression of Langerin and DC‐specific ICAM‐grabbing nonintegrin, respectively. The identity of the MUTZ‐3‐derived LC subset was confirmed further by the presence of Birbeck granules. We conclude that the MUTZ‐3 cell line provides a ready and continuous supply of common myeloid precursors, which should facilitate further study of the ontogeny of myeloid DC lineages.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Sensing of latent EBV infection through exosomal transfer of 5′pppRNA

S. Rubina Baglio; Monique van Eijndhoven; Danijela Koppers-Lalic; Jordi Berenguer; Sinéad M. Lougheed; Susan Gibbs; Nicolas Léveillé; Rico N. P. M. Rinkel; Erik S. Hopmans; Sankar Swaminathan; Sandra A. W. M. Verkuijlen; George L. Scheffer; Frank J. M. van Kuppeveld; Tanja D. de Gruijl; Irene E. M. Bultink; Ekaterina S. Jordanova; Michael Hackenberg; Sander R. Piersma; Jaco C. Knol; Alexandre E. Voskuyl; Thomas Wurdinger; Connie R. Jimenez; Jaap M. Middeldorp; D. Michiel Pegtel

Significance Increasing evidence suggests that the exosomal messenger pathway warns neighboring cells against cellular stress and infection. Recent studies have shown that viruses and cancer cells exploit exosomes to transmit functional RNAs. Our studies reveal that a viral small RNA signal for innate immunity Epstein–Barr virus (EBV)-EBER1 is produced by latent EBV-infected B cells and recognized by noninfected dendritic cells activating an inflammatory response. We detected high amounts of EBV-EBER1 transcripts and EBV-microRNAs in inflamed skin lesions of autoimmune patients that are infiltrated with dendritic cells. Importantly, we found virtually no EBV-DNA present in these tissues, suggesting that continuous cell–cell EBER1 transmission via exosomes occurs in humans. We propose that innate sensing of latent EBV in predisposed individuals may be more harmful than previously thought. Complex interactions between DNA herpesviruses and host factors determine the establishment of a life-long asymptomatic latent infection. The lymphotropic Epstein–Barr virus (EBV) seems to avoid recognition by innate sensors despite massive transcription of immunostimulatory small RNAs (EBV-EBERs). Here we demonstrate that in latently infected B cells, EBER1 transcripts interact with the lupus antigen (La) ribonucleoprotein, avoiding cytoplasmic RNA sensors. However, in coculture experiments we observed that latent-infected cells trigger antiviral immunity in dendritic cells (DCs) through selective release and transfer of RNA via exosomes. In ex vivo tonsillar cultures, we observed that EBER1-loaded exosomes are preferentially captured and internalized by human plasmacytoid DCs (pDCs) that express the TIM1 phosphatidylserine receptor, a known viral- and exosomal target. Using an EBER-deficient EBV strain, enzymatic removal of 5′ppp, in vitro transcripts, and coculture experiments, we established that 5′pppEBER1 transfer via exosomes drives antiviral immunity in nonpermissive DCs. Lupus erythematosus patients suffer from elevated EBV load and activated antiviral immunity, in particular in skin lesions that are infiltrated with pDCs. We detected high levels of EBER1 RNA in such skin lesions, as well as EBV-microRNAs, but no intact EBV-DNA, linking non–cell-autonomous EBER1 presence with skin inflammation in predisposed individuals. Collectively, our studies indicate that virus-modified exosomes have a physiological role in the host–pathogen stand-off and may promote inflammatory disease.


Journal of Immunology | 2013

Intradermal Delivery of TLR Agonists in a Human Explant Skin Model: Preferential Activation of Migratory Dendritic Cells by Polyribosinic-Polyribocytidylic Acid and Peptidoglycans

Dinja Oosterhoff; Moniek Heusinkveld; Sinéad M. Lougheed; Ilona Kosten; Malin Lindstedt; Sven C. M. Bruijns; Thomas van Es; Yvette van Kooyk; Sjoerd H. van der Burg; Tanja D. de Gruijl

TLR agonists are attractive candidate adjuvants for therapeutic cancer vaccines as they can induce a balanced humoral and T cell–mediated immune response. With a dense network of dendritic cells (DCs) and draining lymphatics, the skin provides an ideal portal for vaccine delivery. Beside direct DC activation, TLR agonists may also induce DC activation through triggering the release of inflammatory mediators by accessory cells in the skin microenvironment. Therefore, a human skin explant model was used to explore the in vivo potential of intradermally delivered TLR agonists to stimulate Langerhans cells and dermal DCs in their natural complex tissue environment. The skin-emigrated DCs were phenotyped and analyzed for T cell stimulatory capacity. We report that, of six tested TLR-agonists, the TLR2 and -3 agonists peptidoglycan (PGN) and polyribosinic-polyribocytidylic acid (Poly I:C) were uniquely able to enhance the T cell–priming ability of skin-emigrated DCs, which, in the case of PGN, was accompanied by Th1 polarization. The enhanced priming capacity of Poly I:C–stimulated DCs was associated with a strong upregulation of appropriate costimulatory molecules, including CD70, whereas that of PGN-stimulated DCs was associated with the release of a broad array of proinflammatory cytokines. Transcriptional profiling further supported the notion that the PGN- and Poly I:C–induced effects were mediated through binding to TLR2/nucleotide-binding oligomerization domain 2 and TLR3/MDA5, respectively. These data warrant further exploration of PGN and Poly I:C, alone or in combination, as DC-targeted adjuvants for intradermal cancer vaccines.


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.


Journal for ImmunoTherapy of Cancer | 2014

Myeloid derived suppressor and dendritic cell subsets are related to clinical outcome in prostate cancer patients treated with prostate GVAX and ipilimumab

Saskia J. A. M. Santegoets; Anita G. M. Stam; Sinéad M. Lougheed; Helen Gall; Karin Jooss; Natalie Sacks; Kristen Hege; Israel Lowy; Rik J. Scheper; Winald R. Gerritsen; Alfons J.M. van den Eertwegh; Tanja D. de Gruijl

BackgroundCancer-related disturbances in myeloid lineage development, marked by high levels of myeloid-derived suppressor cells (MDSC) and impaired dendritic cell (DC) development, are associated with poor clinical outcome due to immune escape and therapy resistance. Redressing this balance may therefore be of clinical benefit. Here we investigated the effects of combined Prostate GVAX/ipilimumab immunotherapy on myeloid subsets in peripheral blood of castration-resistant prostate cancer (CRPC) patients as well as the putative predictive value of baseline and on-treatment myeloid parameters on clinical outcome.MethodsPatients with CRPC (n?=?28) received thirteen intradermal administrations of Prostate GVAX, consisting of two allogeneic GM-CSF-transduced and irradiated prostate cancer cell lines (LN-CaP and PC3) and six infusions of escalating doses of anti-CTLA4/ipilimumab. Frequencies and activation status of peripheral blood DC (PBDC) and MDSC were determined before, during and after treatment by flowcytometric analysis and related to clinical benefit.ResultsSignificant treatment-induced activation of conventional and plasmacytoid DC subsets (cDC and pDC) was observed, which in the case of BDCA1/CD1c+ cDC1 and MDC8+/6-sulfoLacNAc+ inflammatory cDC3 was associated with significantly prolonged overall survival (OS), but also with the development of autoimmune-related adverse events. High pre-treatment levels of CD14+HLA-DR?monocytic MDSC (mMDSC) were associated with reduced OS. Unsupervised clustering of these myeloid biomarkers revealed particular survival advantage in a group of patients with high treatment-induced PBDC activation and low pretreatment frequencies of suppressive mMDSC in conjunction with our previously identified lymphoid biomarker of high pretreatment CD4+CTLA4+ T cell frequencies.ConclusionsOur data demonstrate that DC and MDSC subsets are affected by prostate GVAX/ipilimumab therapy and that myeloid profiling may contribute to the identification of patients with possible clinical benefit of Prostate GVAX/ipilimumab treatment.


PLOS ONE | 2013

IL-10 conditioning of human skin affects the distribution of migratory dendritic cell subsets and functional T cell differentiation

Jelle Lindenberg; Dinja Oosterhoff; Claudia C. Sombroek; Sinéad M. Lougheed; Erik Hooijberg; Anita G. M. Stam; Saskia J. A. M. Santegoets; Henk J. Tijssen; Jan Buter; Herbert M. Pinedo; Alfons J.M. van den Eertwegh; Rik J. Scheper; Hans J. P. M. Koenen; Rieneke van de Ven; Tanja D. de Gruijl

In cancer patients pervasive systemic suppression of Dendritic Cell (DC) differentiation and maturation can hinder vaccination efficacy. In this study we have extensively characterized migratory DC subsets from human skin and studied how their migration and T cell-stimulatory abilities were affected by conditioning of the dermal microenvironment through cancer-related suppressive cytokines. To assess effects in the context of a complex tissue structure, we made use of a near-physiological skin explant model. By 4-color flow cytometry, we identified migrated Langerhans Cells (LC) and five dermis-derived DC populations in differential states of maturation. From a panel of known tumor-associated suppressive cytokines, IL-10 showed a unique ability to induce predominant migration of an immature CD14+CD141+DC-SIGN+ DC subset with low levels of co-stimulatory molecules, up-regulated expression of the co-inhibitory molecule PD-L1 and the M2-associated macrophage marker CD163. A similarly immature subset composition was observed for DC migrating from explants taken from skin overlying breast tumors. Whereas predominant migration of mature CD1a+ subsets was associated with release of IL-12p70, efficient Th cell expansion with a Th1 profile, and expansion of functional MART-1-specific CD8+ T cells, migration of immature CD14+ DDC was accompanied by increased release of IL-10, poor expansion of CD4+ and CD8+ T cells, and skewing of Th responses to favor coordinated FoxP3 and IL-10 expression and regulatory T cell differentiation and outgrowth. Thus, high levels of IL-10 impact the composition of skin-emigrated DC subsets and appear to favor migration of M2-like immature DC with functional qualities conducive to T cell tolerance.


Clinical Immunology | 2010

Attenuation of invariant Natural Killer T-cell anergy induction through intradermal delivery of alpha-galactosylceramide

Hetty J. Bontkes; María Moreno; Basav N. Hangalapura; Jelle Lindenberg; Jan de Groot; Sinéad M. Lougheed; Hans J. van der Vliet; Alfons J.M. van den Eertwegh; Tanja D. de Gruijl; B. Mary E. von Blomberg; Rik J. Scheper

CD1d restricted, alpha-galactosylceramide (alphaGC) responsive invariant (i)NKT cells positively regulate immune responses. Both intravenous and intradermal administered alphaGC are known to activate iNKT cells. iNKT cells become unresponsive to a second intravenous alphaGC injection, whereas no data are available regarding potential anergy upon intradermal administration. Here, comparative analysis of two intradermal versus two intravenous injections in mice demonstrated that iNKT cell anergy was prevented by intradermal injection and when combined with a vaccine, superior tumor protection afforded by intradermally administered alphaGC. Moreover, human skin dendritic cells (DC) took up intradermally injected alphaGC and activated iNKT cells upon migration, while iNKT cells in human skin-draining lymph nodes expanded in response to alphaGC presented either by exogenously added DC or by CD1d positive antigen presenting cells in the lymph nodes. In conclusion, glycolipids such as alphaGC may greatly improve the efficacy of skin immunization strategies, targeting cutaneous and lymph node DC.


Clinical Cancer Research | 2017

Blocking tumor-educated MSC paracrine activity halts osteosarcoma progression

S. Rubina Baglio; Tonny Lagerweij; Maria Pérez-Lanzón; Xuan Dung Ho; Nicolas Léveillé; Sonia A. Melo; Anne-Marie Cleton-Jansen; Ekaterina S. Jordanova; Laura Roncuzzi; Michelina Greco; Monique van Eijndhoven; Giulia Grisendi; Massimo Dominici; Roberta Bonafede; Sinéad M. Lougheed; Tanja D. de Gruijl; Nicoletta Zini; Silvia Cervo; Agostino Steffan; Vincenzo Canzonieri; Aare Märtson; Katre Maasalu; Sulev Kõks; Tom Wurdinger; Nicola Baldini; D. Michiel Pegtel

Purpose: Human osteosarcoma is a genetically heterogeneous bone malignancy with poor prognosis despite the employment of aggressive chemotherapy regimens. Because druggable driver mutations have not been established, dissecting the interactions between osteosarcoma cells and supporting stroma may provide insights into novel therapeutic targets. Experimental Design: By using a bioluminescent orthotopic xenograft mouse model of osteosarcoma, we evaluated the effect of tumor extracellular vesicle (EV)–educated mesenchymal stem cells (TEMSC) on osteosarcoma progression. Characterization and functional studies were designed to assess the mechanisms underlying MSC education. Independent series of tissue specimens were analyzed to corroborate the preclinical findings, and the composition of patient serum EVs was analyzed after isolation with size-exclusion chromatography. Results: We show that EVs secreted by highly malignant osteosarcoma cells selectively incorporate a membrane-associated form of TGFβ, which induces proinflammatory IL6 production by MSCs. TEMSCs promote tumor growth, accompanied with intratumor STAT3 activation and lung metastasis formation, which was not observed with control MSCs. Importantly, intravenous administration of the anti-IL6 receptor antibody tocilizumab abrogated the tumor-promoting effects of TEMSCs. RNA-seq analysis of human osteosarcoma tissues revealed a distinct TGFβ-induced prometastatic gene signature. Tissue microarray immunostaining indicated active STAT3 signaling in human osteosarcoma, consistent with the observations in TEMSC-treated mice. Finally, we isolated pure populations of EVs from serum and demonstrated that circulating levels of EV-associated TGFβ are increased in osteosarcoma patients. Conclusions: Collectively, our findings suggest that TEMSCs promote osteosarcoma progression and provide the basis for testing IL6- and TGFβ-blocking agents as new therapeutic options for osteosarcoma patients. Clin Cancer Res; 23(14); 3721–33. ©2017 AACR.

Collaboration


Dive into the Sinéad M. Lougheed's collaboration.

Top Co-Authors

Avatar

Tanja D. de Gruijl

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

Anita G. M. Stam

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dinja Oosterhoff

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erik Hooijberg

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jelle Lindenberg

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge