Network


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

Hotspot


Dive into the research topics where Pieter Vader is active.

Publication


Featured researches published by Pieter Vader.


Journal of extracellular vesicles | 2015

Applying extracellular vesicles based therapeutics in clinical trials - an ISEV position paper.

Thomas Lener; Mario Gimona; Ludwig Aigner; Verena Börger; Edit I. Buzás; Giovanni Camussi; Nathalie Chaput; Devasis Chatterjee; Felipe A. Court; Hernando A. del Portillo; Lorraine O'Driscoll; Stefano Fais; Juan M. Falcon-Perez; Ursula Felderhoff-Mueser; Lorenzo Fraile; Yong Song Gho; André Görgens; Ramesh C. Gupta; An Hendrix; Dirk M. Hermann; Andrew F. Hill; Fred H. Hochberg; Peter A. Horn; Dominique P.V. de Kleijn; Lambros Kordelas; Boris W. Kramer; Eva Maria Krämer-Albers; Sandra Laner-Plamberger; Saara Laitinen; Tommaso Leonardi

Extracellular vesicles (EVs), such as exosomes and microvesicles, are released by different cell types and participate in physiological and pathophysiological processes. EVs mediate intercellular communication as cell-derived extracellular signalling organelles that transmit specific information from their cell of origin to their target cells. As a result of these properties, EVs of defined cell types may serve as novel tools for various therapeutic approaches, including (a) anti-tumour therapy, (b) pathogen vaccination, (c) immune-modulatory and regenerative therapies and (d) drug delivery. The translation of EVs into clinical therapies requires the categorization of EV-based therapeutics in compliance with existing regulatory frameworks. As the classification defines subsequent requirements for manufacturing, quality control and clinical investigation, it is of major importance to define whether EVs are considered the active drug components or primarily serve as drug delivery vehicles. For an effective and particularly safe translation of EV-based therapies into clinical practice, a high level of cooperation between researchers, clinicians and competent authorities is essential. In this position statement, basic and clinical scientists, as members of the International Society for Extracellular Vesicles (ISEV) and of the European Cooperation in Science and Technology (COST) program of the European Union, namely European Network on Microvesicles and Exosomes in Health and Disease (ME-HaD), summarize recent developments and the current knowledge of EV-based therapies. Aspects of safety and regulatory requirements that must be considered for pharmaceutical manufacturing and clinical application are highlighted. Production and quality control processes are discussed. Strategies to promote the therapeutic application of EVs in future clinical studies are addressed.


Journal of extracellular vesicles | 2015

Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting

Oscar P. B. Wiklander; Joel Z. Nordin; Aisling O'Loughlin; Ylva Gustafsson; Giulia Corso; Imre Mäger; Pieter Vader; Yi Lee; Helena Sork; Yiqi Seow; Nina Heldring; Lydia Alvarez-Erviti; C. I. Edvard Smith; Katarina Le Blanc; Paolo Macchiarini; Philipp Jungebluth; Matthew J.A. Wood; Samir El Andaloussi

Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication in a diverse range of biological processes. For future therapeutic applications and for EV biology research in general, understanding the in vivo fate of EVs is of utmost importance. Here we studied biodistribution of EVs in mice after systemic delivery. EVs were isolated from 3 different mouse cell sources, including dendritic cells (DCs) derived from bone marrow, and labelled with a near-infrared lipophilic dye. Xenotransplantation of EVs was further carried out for cross-species comparison. The reliability of the labelling technique was confirmed by sucrose gradient fractionation, organ perfusion and further supported by immunohistochemical staining using CD63-EGFP probed vesicles. While vesicles accumulated mainly in liver, spleen, gastrointestinal tract and lungs, differences related to EV cell origin were detected. EVs accumulated in the tumour tissue of tumour-bearing mice and, after introduction of the rabies virus glycoprotein-targeting moiety, they were found more readily in acetylcholine-receptor-rich organs. In addition, the route of administration and the dose of injected EVs influenced the biodistribution pattern. This is the first extensive biodistribution investigation of EVs comparing the impact of several different variables, the results of which have implications for the design and feasibility of therapeutic studies using EVs.


Trends in Molecular Medicine | 2014

Extracellular vesicles: emerging targets for cancer therapy

Pieter Vader; Xandra O. Breakefield; Matthew J.A. Wood

Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, are released by almost all cell types, including tumour cells. Through transfer of their molecular contents, EVs are capable of altering the function of recipient cells. Increasing evidence suggests a key role for EV mediated intercellular communication in a variety of cellular processes involved in tumour development and progression, including immune suppression, angiogenesis, and metastasis. Aspects of EV biogenesis or function are therefore increasingly being considered as targets for anticancer therapy. Here, we summarise the current knowledge on the contributions of EVs to cancer pathogenesis and discuss novel therapeutic strategies to target EVs to prevent tumour growth and spread.


Journal of Controlled Release | 2014

Extracellular vesicles as drug delivery systems: Lessons from the liposome field

Roy van der Meel; Marcel H.A.M. Fens; Pieter Vader; Wouter W. van Solinge; Omolola Eniola-Adefeso; Raymond M. Schiffelers

Extracellular vesicles (EVs) are membrane-derived particles surrounded by a (phospho)lipid bilayer that are released by cells in the human body. In addition to direct cell-to-cell contact and the secretion of soluble factors, EVs function as another mechanism of intercellular communication. These vesicles are able to efficiently deliver their parental cell-derived molecular cargo to recipient cells, which can result in structural changes at an RNA, protein, or even phenotypic level. For this reason, EVs have recently gained much interest for drug delivery purposes. In contrast to these natural delivery systems, synthetic (phospho)lipid vesicles, or liposomes, have been employed as drug carriers for decades, resulting in several approved liposomal nanomedicines used in the clinic. This review discusses the similarities and differences between EVs and liposomes with the focus on features that are relevant for drug delivery purposes such as circulation time, biodistribution, cellular interactions and cargo loading. By applying beneficial features of EVs to liposomes and vice versa, improved drug carriers can be developed which will advance the field of nanomedicines and ultimately improve patient outcomes. While the application of EVs for therapeutic drug delivery is still in its infancy, issues regarding the understanding of EV biogenesis, large-scale production and in vivo interactions need to be addressed in order to develop successful and cost-effective EV-based drug delivery systems.


Nanomedicine: Nanotechnology, Biology and Medicine | 2015

Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties

Joel Z. Nordin; Yi Lee; Pieter Vader; Imre Mäger; H. Johansson; Wolf Heusermann; Oscar P. B. Wiklander; Mattias Hällbrink; Yiqi Seow; Jarred J. Bultema; Jonathan Gilthorpe; Tim Davies; Paul J. Fairchild; Susanne Gabrielsson; Nicole Meisner-Kober; Janne Lehtiö; C. I. Edvard Smith; Matthew J.A. Wood; Samir El Andaloussi

UNLABELLEDnExtracellular vesicles (EVs) are natural nanoparticles that mediate intercellular transfer of RNA and proteins and are of great medical interest; serving as novel biomarkers and potential therapeutic agents. However, there is little consensus on the most appropriate method to isolate high-yield and high-purity EVs from various biological fluids. Here, we describe a systematic comparison between two protocols for EV purification: ultrafiltration with subsequent liquid chromatography (UF-LC) and differential ultracentrifugation (UC). A significantly higher EV yield resulted from UF-LC as compared to UC, without affecting vesicle protein composition. Importantly, we provide novel evidence that, in contrast to UC-purified EVs, the biophysical properties of UF-LC-purified EVs are preserved, leading to a different in vivo biodistribution, with less accumulation in lungs. Finally, we show that UF-LC is scalable and adaptable for EV isolation from complex media types such as stem cell media, which is of huge significance for future clinical applications involving EVs.nnnFROM THE CLINICAL EDITORnRecent evidence suggests extracellular vesicles (EVs) as another route of cellular communication. These EVs may be utilized for future therapeutics. In this article, the authors compared ultrafiltration with size-exclusion liquid chromatography (UF-LC) and ultra-centrifugation (UC) for EV recovery.


Molecular Therapy | 2017

Functional Delivery of Lipid-Conjugated siRNA by Extracellular Vesicles

Aisling J. O’Loughlin; Imre Mäger; Olivier G. de Jong; Miguel A. Varela; Raymond M. Schiffelers; Samir El Andaloussi; Matthew J.A. Wood; Pieter Vader

Extracellular vesicles (EVs) are cell-derived, membranous nanoparticles that mediate intercellular communication by transferring biomolecules, including proteins and RNA, between cells. As a result of their suggested natural capability to functionally deliver RNA, EVs may be harnessed as therapeutic RNA carriers. One major limitation for their translation to therapeutic use is the lack of an efficient, robust, and scalable method to load EVs with RNA molecules ofxa0interest. Here, we evaluated and optimized methods to load EVs with cholesterol-conjugated small interfering RNAs (cc-siRNAs)xa0by systematic evaluation of the influence of key parameters, including incubation time, volume, temperature, and EV:cc-siRNA ratio. EV loading under conditions that resulted in the highest siRNA retention percentage, incubating 15xa0moleculesxa0of cc-siRNA per EV at 37°C for 1xa0hr in 100xa0μL, facilitated concentration-dependent silencing of human antigen R (HuR), a therapeutic target in cancer, in EV-treated cells. These results may accelerate the development of EV-based therapeutics.


Brain | 2017

C9orf72 and RAB7L1 regulate vesicle trafficking in amyotrophic lateral sclerosis and frontotemporal dementia.

Yoshitsugu Aoki; Raquel Manzano; Yi Lee; Ruxandra Dafinca; Misako Aoki; Andrew G.L. Douglas; Miguel A. Varela; Chaitra Sathyaprakash; Jakub Scaber; Paola Barbagallo; Pieter Vader; Imre Mäger; Kariem Ezzat; Martin Turner; Naoki Ito; Samanta Gasco; Norihiko Ohbayashi; Samir El Andaloussi; Shin Takeda; Mitsunori Fukuda; Kevin Talbot; Matthew J.A. Wood

A non-coding hexanucleotide repeat expansion in intron 1 of the C9orf72 gene is the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD), however, the precise molecular mechanism by which the C9orf72 hexanucleotide repeat expansion directs C9ALS/FTD pathogenesis remains unclear. Here, we report a novel disease mechanism arising due to the interaction of C9ORF72 with the RAB7L1 GTPase to regulate vesicle trafficking. Endogenous interaction between C9ORF72 and RAB7L1 was confirmed in human SH-SY5Y neuroblastoma cells. The C9orf72 hexanucleotide repeat expansion led to haploinsufficiency resulting in severely defective intracellular and extracellular vesicle trafficking and a dysfunctional trans-Golgi network phenotype in patient-derived fibroblasts and induced pluripotent stem cell-derived motor neurons. Genetic ablation of RAB7L1or C9orf72 in SH-SY5Y cells recapitulated the findings in C9ALS/FTD fibroblasts and induced pluripotent stem cell neurons. When C9ORF72 was overexpressed or antisense oligonucleotides were targeted to the C9orf72 hexanucleotide repeat expansion to upregulate normal variant 1 transcript levels, the defective vesicle trafficking and dysfunctional trans-Golgi network phenotypes were reversed, suggesting that both loss- and gain-of-function mechanisms play a role in disease pathogenesis. In conclusion, we have identified a novel mechanism for C9ALS/FTD pathogenesis highlighting the molecular regulation of intracellular and extracellular vesicle trafficking as an important pathway in C9ALS/FTD pathogenesis.


Frontiers in Immunology | 2018

Extracellular Vesicle Heterogeneity: Subpopulations, Isolation Techniques, and Diverse Functions in Cancer Progression

Eduard Willms; Carlos Cabañas; Imre Mäger; Matthew J.A. Wood; Pieter Vader

Cells release membrane enclosed nano-sized vesicles termed extracellular vesicles (EVs) that function as mediators of intercellular communication by transferring biological information between cells. Tumor-derived EVs have emerged as important mediators in cancer development and progression, mainly through transfer of their bioactive content which can include oncoproteins, oncogenes, chemokine receptors, as well as soluble factors, transcripts of proteins and miRNAs involved in angiogenesis or inflammation. This transfer has been shown to influence the metastatic behavior of primary tumors. Moreover, tumor-derived EVs have been shown to influence distant cellular niches, establishing favorable microenvironments that support growth of disseminated cancer cells upon their arrival at these pre-metastatic niches. It is generally accepted that cells release a number of major EV populations with distinct biophysical properties and biological functions. Exosomes, microvesicles, and apoptotic bodies are EV populations most widely studied and characterized. They are discriminated based primarily on their intracellular origin. However, increasing evidence suggests that even within these EV populations various subpopulations may exist. This heterogeneity introduces an extra level of complexity in the study of EV biology and function. For example, EV subpopulations could have unique roles in the intricate biological processes underlying cancer biology. Here, we discuss current knowledge regarding the role of subpopulations of EVs in cancer development and progression and highlight the relevance of EV heterogeneity. The position of tetraspanins and integrins therein will be highlighted. Since addressing EV heterogeneity has become essential for the EV field, current and novel techniques for isolating EV subpopulations will also be discussed. Further dissection of EV heterogeneity will advance our understanding of the critical roles of EVs in health and disease.


Methods of Molecular Biology | 2017

Preparation and Isolation of siRNA-Loaded Extracellular Vesicles

Pieter Vader; Imre Mäger; Yi Lee; Joel Z. Nordin; Samir El Andaloussi; Matthew J.A. Wood

RNA interference (RNAi) has tremendous potential for specific silencing of disease-causing genes. Its clinical usage however critically depends on the development of carrier systems that can transport the RNAi-mediating small interfering RNA (siRNA) molecules to the cytosol of target cells. Recent reports have suggested that extracellular vesicles (EVs) form a natural transport system through which biomolecules, including RNA, is exchanged between cells. Therefore, EVs are increasingly being considered as potential therapeutic siRNA delivery systems.In this chapter we describe a method for preparing siRNA-loaded EVs, including a robust, scalable method to isolate them from cell culture supernatants.


European Journal of Cancer | 2014

448 Exposure to EGFR inhibitors influences release of extracellular vesicles by tumor cells

R. van der Meel; S.M. van Dommelen; P. de Corte; Maria Coimbra; W.W. van Solinge; Pieter Vader; R.M. Schiffelers

Background: The identification of novel therapeutic targets to block cancer metastasis is a key step towards the reduction of cancer-related patient deaths. For prostate cancer, the most commonly-diagnosed cancer in men, the 5 year survival rate for those with metastatic disease is less than 10%. We have recently established that intravasation, a critical step of metastasis, is dependent upon production cell migration. In this study, we have utilized a quantitative in vivo screening approach that relies on the assessment of in vivo cell migration as a primary readout. Methods: Our screen approach utilizes an ex ovo avian embryo platform combined with real-time, high-resolution 3D visualization of human cancer metastasis. When injected intravenously, fluorescent human cancer cells form hundreds of individual metastatic colonies that can be simultaneously visualized in vivo. A customized Matlab-based analysis software was developed to interpret 3D images from this model to generate a metastatic colony invasiveness index (C.I.). As a proof of principle, we showed that previously described inhibitors of cell migration and metastasis significantly reduces the C.I., which closely correlates with their antimetastatic potential. In the full screen, human head and neck cancer cells (Hep3) and prostate cancer cells (PC3) were transduced with a whole genome shRNA and injected intravenously into the CAM. Metastatic colonies with C.I. below that of control cells were excised and expanded. shRNA identities were determined by high-throughput sequencing. Results: More than fifty novel genetic regulators of metastasis were identified that have not been previously described using other approaches. Of these, there were eleven druggable targets that included cancer cell invasion and cell cycle regulating protein kinases, cell surface and nuclear receptors and cytoskeleton modulating enzymes. shRNA-mediated knockdown of these genes blocked human cancer cell metastasis in both experimental and spontaneous mouse models of metastasis. Lead hit serine-threonine kinase SRPK1 was significantly associated with prostate cancer invasion and metastasis using publicly-available gene expression data. We demonstrate that SRPK1 directly modulates the splicing isoform profile of several key cell adhesion molecules, impacting cell migration in vitro and in vivo. Inhibition of SRPK1 decreases the ability of prostate cancer cells to metastasize in vivo. Conclusion: We developed a novel and robust in vivo screening platform that provides a rapid and quantitative assessment of therapeutic targets of metastasis. Using this platform, we have identified numerous promising therapeutic targets that that are functionally involved in cancer invasion and metastasis.

Collaboration


Dive into the Pieter Vader's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi Lee

University of Oxford

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jakub Scaber

John Radcliffe Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge