1,854
Views
11
CrossRef citations to date
0
Altmetric
REVIEWS: PROGNOSTIC MARKERS

The potential diagnostic and prognostic role of extracellular vesicles in glioma: current status and future perspectives

, , &
Pages 353-362 | Received 31 Aug 2018, Accepted 18 Nov 2018, Published online: 11 Jan 2019

Abstract

Lack of appropriate diagnostic/prognostic tools for glioblastoma (GB) is considered one of the major setbacks in the early diagnosis and treatment of this deadly brain tumor. The current gold standard for its diagnosis and staging still relies on invasive biopsy followed by histological examination as well as molecular profiling. Nevertheless, noninvasive approaches are being explored and one example is through the investigation of extracellular vesicles (EVs) in the biofluids of GB patients. EVs are known to carry molecular cargoes such as DNA, mRNA, miRNA, proteins and lipids in almost every type of body fluids. Thus, molecular signature of GB may be present in the EVs derived from these patients. This review focuses on the diagnostic/prognostic potential of EVs in GB, through presenting recent studies on (i) molecular components of EVs, (ii) links between EVs and GB tumor microenvironment, and (iii) clinical potential of EV biomarkers, together with the technical shortcomings researchers need to consider for future studies.

Introduction

Glioblastoma (GB), the deadliest and most prevalent form of brain tumor, arises from glial cells with an incidence rate of 3.2 per 100,000 populations [Citation1] and the numbers are increasing every year [Citation2]. GB can develop as primary tumor (de novo) without known clinical/histological evidence, or secondarily from low-grade tumors by transformation. Most GB are primary which occur predominantly in older patients and have poorer prognosis [Citation3]. Genetic changes vary in GB, for example, IDH1 mutation is evident in secondary GB only [Citation4] and p53 mutations are more common in secondary GB than primary [Citation5]. Other common mutations in GB include, epidermal growth factor receptor (EGFR) (40–57%) [Citation6], platelet-derived growth factor receptor (60%) [Citation7], mouse double minute homolog 2 (10–15%) [Citation5], the phosphatase and tensin homolog (PTEN) gene (20–34%) [Citation8], NF1 (13.7%) [Citation9], and PIK3CA (12%) [Citation9]. Loss of heterozygosity on the chromosome arm 10q accounts for 60–90% in all GB cases [Citation10]. Based on these genetic variances and their gene expression, GB can be further divided into four subtypes, namely, proneural, neural, classical and mesenchymal [Citation11,Citation12]. Distinct genetic and epigenetic variations are associated with each subtype that may be a result of intra-tumor heterogeneity [Citation13].

GB patients have poor prognosis and short median survival time (only 14.6 months). Current treatment modalities include maximal surgical resurrection (the first line treatment), followed by radiotherapy with complementary chemotherapy using temozolomide [Citation14]. However, these aggressive measures failed to suppress its recurrence in almost every case. This is because of the highly heterogeneous nature of GB tumors [Citation12] and their associated diverse cellular and invasive phenotypes [Citation15–18]. In addition, within the same tumor displaying common gene profile there exists a variety of gene copy number variations and subtype-specific signatures [Citation19]. These patient-specific variations may be useful as biomarkers for prognosis during therapy.

Like other cancer cells, GB cells are capable of communicating with neighboring cells through shedding a range of molecules. Numerous recent studies have shown that molecules secreted by tumor cells are often encapsulated in lipid layer-based structures, known as extracellular vesicles (EVs). These tumor-derived EVs are involved in immune regulation [Citation20–22], angiogenesis [Citation23], tumor progression [Citation24] and intercellular communication by exchanging proteins and RNA [Citation25,Citation26]. Based on recent literature, this review explores the potential roles of extracellular vesicle in GB pathogenesis and discusses their possibility as a source of diagnostic, prognostic and predictive biomarkers for GB.

Extracellular vesicles: definition, biogenesis, release and uptake

According to their origin and/or size, EVs can be classified into three subclasses, which are apoptotic bodies, microvesicles and exosomes [Citation27]. During the programed cell death process, cells release apoptotic bodies as blebs. Their size ranges between 1000 and 5000 nm [Citation28]. Microvesicles, also known as ectosomes, are produced in the plasma membrane by a process of external budding with size ranging from 100 to 1000 nm [Citation29]. The smallest of EVs are the exosomes (30–100 nm), which are produced inside the cell through internal budding of vesicles in the lumen of early endosome [Citation30]. EVs were originally described as a means of removing biological waste from cells [Citation31]. It is now evident that they play a very significant role in intercellular communication, not only for normal body functions but also in disease states such as cancer [Citation32,Citation33]. Furthermore, EVs are also known to determine tissue organization [Citation34], promote sperm egg formation [Citation35], and affects the mating behavior [Citation36]. In addition, EVs are suggested to be involved in structural remodeling of the central nervous system [Citation37,Citation38].

Microvesicle and exosome biogenesis are different. Microvesicles are produced through the outward budding of the plasma membrane while exosomes are generated by fusing the multivesicular bodies to the plasma membrane. We will focus our discussion on the biogenesis, release and uptake mechanism of exosomes. The biogenesis of exosome begins at endosome formation through invagination of the plasma membrane. The endosomes are divided into three different compartment inside cell during the endocytic process and they are early, recycling and late endosomes [Citation39]. Early endosomes are formed after plasma membrane invagination and are able to fuse with endocytic vesicles leading to different cellular fates, e.g., recycling, secretion or degradation [Citation40]. After sorting the recycled amount into recycling endosomes, the remaining early endosomes transform into late endosomes [Citation32]. They accumulate to form intraluminal vesicles through three different pathways: (i) endosomal sorting complexes required for transport (ESCRT) dependent pathway [Citation41,Citation42], (ii) alternative ESCRT pathway [Citation43], and (iii) ESCRT-independent pathway [Citation44]. Cytosolic molecular cargoes including proteins, lipids and nucleic acids are inserted into exosome during intraluminal vesicles formation and the contents differ according to their different biogenesis pathways. Intraluminal vesicles are stored within multivesicular bodies before being degraded by lysosome or released as exosome through fusion with the plasma membrane [Citation39].

Like its biogenesis, exosome is released through different mechanisms. One mechanism is through Rab GTPases and it is evident that RAB11, RAB27A, RAB27B and RAB35 are involved. EVs release via RAB11 and RAB35 are enriched in flotillin, Wnt, transferrin receptor and PLP [Citation45]. But, RAB27A/B-dependent release involves late endosomal proteins, e.g., TSG 101, CD63 and ALIX [Citation46,Citation47]. RAB7 is only involved in exosome release by breast tumor cells [Citation43,Citation47]. Rab GTPases independent mechanisms are also evident in some cases [Citation48–51].

Endocytosis, ligand/receptor interaction and fusion with the plasma membrane are mechanisms described so far for cellular uptake of exosome [Citation52]. Different endocytosis processes such as clathrin-mediated endocytosis, phagocytosis, macropinocytosis are also responsible for exosome internalization. Cell types, physiological states, surface ligands and receptors may affect the uptake mode. For example, in neurons exosome uptake involves phagocytosis or clathrin-dependent endocytosis [Citation53]. Ligands/receptors such as heparin sulfate proteoglycans and scavenger receptor type B-1 are important in exosome uptake because blocking them decreased exosome uptake by cells in vitro [Citation54,Citation55]. Hydrogen ion concentration can affect the plasma membrane fusion process as exosome fusion and the release of its contents into the cytosol can be promoted by pH-sensitive fusogenic peptides [Citation56,Citation57].

The molecular cargoes of extracellular vesicles

EVs contain a variety of proteins, receptors, lipids and nucleic acids in its lipid bilayer or within its aqueous compartment [Citation58]. However, the contents can vary according to their biogenesis and cell of origin. Proteins related to membrane function (e.g., ICAM1, integrins), EV biogenesis (TSG101, ALIX), uptake and release (Annexins, Rab proteins) are commonly found in EVs [Citation59]. In addition, a large array of tetraspanins, e.g., CD9, CD37, CD53, CD63, CD81 and CD82 [Citation59]; proteins related to antigen presentation, e.g. HLA-G, MHC; cytokines, e.g., VEGF-A [Citation60], semaphorin 3A (Sema3A) [Citation61], TGF-beta [Citation62] and EGFRvIII [Citation63] could also be present. The lipid composition of EVs has not been fully investigated but it has been shown that their lipid bilayer is enriched with sphingomyelin, cholesterol, phosphatidylcholine, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine, prostaglandin and ganglioside GM3 [Citation64,Citation65]. EV nucleic acid contents are diverse in nature, and they are usually fragmented [Citation66]. Previous research showed that most of them are small RNAs, especially rRNAs and tRNAs [Citation67], but other small RNAs are also present, e.g. short and long non-coding RNAs, mRNAs, and miRNAs [Citation68–70]. Data on mitochondrial and genomic DNA content in EVs are scarce [Citation71,Citation72].

Extracellular vesicles and the GB tumor microenvironment

It has been postulated that one of the major roles of EVs in the central nervous system is to exchange molecules between different types of cells and help to maintain their normal functions [Citation73]. In pathological conditions such as GB, tumor cells use EVs for their own benefits to promote angiogenesis, clonogenicity, heighten cell proliferation and invasion [Citation74,Citation75]. By transferring non-coding RNAs [Citation76], oncogenic EGFRvIII [Citation77], histones [Citation78], PTEN [Citation79], and pro-migratory factors [Citation80], EVs themselves can influence tumor microenvironment and transform normal cells into malignant cells.

Like other cancers, hypoxia in GB upregulates the production of angiogenic factors [Citation81]. Both in vitro and in vivo experiments confirmed that the level of hypoxia relates to mRNA and protein increase in EVs [Citation82]. A recent study has demonstrated that EVs could transfer hypoxia-dependent miRNAs between CD133+ U87 glioblastoma cells and vascular cells [Citation83]. Invadopodia, a structure formed in highly metastatic cancer cells, can proteolytically degrade extracellular matrix by protruding its actin rich structure into the cell membrane [Citation84]. Functional invadopodia are present in glioma cell lines [Citation85] and increased levels of Tks5, an adaptor protein critical for invadopodia formation, is correlated with poor survival [Citation86]. EV biogenesis can also affect invadopodia formation, stabilization and protein secretion [Citation87]. Recently, it has been reported that five genes, namely, actin-related protein 3, insulin-like growth factor 2 receptor, integrin-β1, annexin A1 and programed cell death 6-interacting protein, related to invadopodia formation are highly expressed in GB tumor-derived EVs [Citation88]. Furthermore, EVs may affect angiogenesis through its action on endothelial cells [Citation89] and through transferring miR-1 [Citation90] and Sema3A [Citation61]. A recent study confirmed that, glioma stem cell derived EVs could alter endothelial cell angiogenesis through miR-21/VEGF/VEGFR2 signaling pathway [Citation91]. In fact, immune activity of cells of monocytic lineage may be altered by up-taking GB-derived EVs and shift their protein expression and cytokine secretion toward a pro-tumoral phenotype [Citation92]. In vivo experiments also showed change in phenotype of brain immune cells that had taken up GB-derived EVs [Citation93]. These observations suggest that EVs can alter tumor microenvironment by exchanging signals between brain cells, which ultimately provide suitable environment for tumor growth.

Diagnostic/prognostic potential of EV biomarkers

The diagnosis of GB is heavily dependent on neuroimaging (e.g., MRI) and tissue biopsies. However, both approaches for predicting response to therapy are somewhat error prone. MRI can only detect established tumors with sufficient mass [Citation94]. It is also difficult to assess the therapeutic response by MRI, because imaging properties of the tumor are often interfered by intervention. Increased or decreased contrast enhancement may have happened due to the interaction of chemo radiation and anti-angiogenic agents with the tumor microenvironment, respectively [Citation95,Citation96]. On the other hand, tumor biopsy is invasive in nature and causes brain swelling and hemorrhage resulting in de-regulation of brain function [Citation97]. It is also impossible to get longitudinal and/or repeated sampling, and to determine the real picture of intra-tumoral heterogeneity [Citation98]. This limits our ability to predict chemotherapeutic resistance, monitor treatment response, clear differentiation between pseudo-progression and real progression [Citation99].

Recently, EVs emerge as a promising source of biomarkers for diagnostic and prognostic purposes. They can be noninvasively collected for longitudinal sampling [Citation97]; their large array of molecules allows characterization of the global tumor genome and transcriptome [Citation100]; their short half-life enables detection of rapid changes in the tumor milieu [Citation101]. Besides, its inherent stability and capability to maintain the integrity of its contents allow researchers to analyze DNA, RNA and proteins from solid tumors. It also provides a potential link between tumor drug-resistance and metastasis. Skog et al. first suggested the potential diagnostic role of EVs in brain cancer in 2008 [Citation102]. His group concluded that EVs isolated from serum could be used to detect the evolving genetic changes relative to tumor progression at any given time. In line with this idea, Noerholm et al. showed that distinct RNA expression pattern is present in serum EVs of GB patients compared with controls [Citation103]. The miR-301a level in EVs is significantly increased and correlated with overall survival [Citation104]. In fact, nucleic acid variations could be detected in EVs collected from GB patients, e.g., IDH1 [Citation105], EGFRvIII [Citation63], miR-21 [Citation106], miR-1587 [Citation107] and EPHA2 [Citation108]. EV nucleic acids can also serve as a source of biomarkers that depicts chemotherapeutic resistance in GB patients. For example, O6-methylguanine-DNA-methyltransferase mRNA expression level was found to be increased in EVs collected from GB patients resistant to temozolomide [Citation109]. Other nucleic acid biomarkers like miR-181d and miR-603 [Citation110], alkylpurine-DNA-N-glycosylase [Citation108], and miR-221 [Citation111] may serve similar purposes. EV protein content may also serve as biomarkers. Pinet et al. highlight that TrkB expression is increased in EVs isolated from plasma of GB patients [Citation112]. Another study showed that Sema3A is expressed on the surface of EVs released by patient-derived glioblastoma cells [Citation61].

Temozolomide (TMZ) is an alkylating agent commonly used in GB treatment together with radiation therapy after surgical resection of the tumor [Citation113]. It is now proven that TMZ affects EVs secretion [Citation114] and could confer drug resistance to recipient cells by transferring molecular cargos through EVs [Citation115]. Mass spectrometry-based analysis reveals that protein levels related to cell adhesion, e.g. β1-integrin, are increased in EVs after TMZ treatment [Citation114]. Whereas TMZ resistant cell-derived EVs containing miR-151a are able to generate drug resistance in recipient cells [Citation115]. It is, therefore, possible to monitor TMZ failure by analyzing the molecular components of GB tumor-derived EVs [Citation116]. Other EV surface proteins such as CD44 and CD133 [Citation117] may serve as biomarkers for chemoresistant GB patients. It is because glioblastoma stem-like cells responsible for chemoresistance and tumor recurrence also express the same cell surface markers [Citation118]. Selected EV-derived molecules and their roles in glioblastoma are summarized in .

Table 1. Selected EV-derived molecules and their roles in glioma.

Although EVs have the potential for GB diagnosis, most research conducted to date are proof-of-principle studies highlighting their roles in tumor biology. Nevertheless, several EV components such as EGFR mutation, mutant IDH1 and miR-21 have gained more attention recently. Molecular methods such as qRT-PCR for the detection of EGFRvIII in EVs derived from serum and cerebrospinal fluid (CSF) have shown high specificity (>95%). Unfortunately, the sensitivity is rather low (∼60%) [Citation102,Citation119]. Similarly, IDH1 mutant detection using the more sensitive BEAMing ddPCR technique was positive only in CSF EVs but not those from serum [Citation105]. Comparable findings are also reported in EV-associated miRNAs detection [Citation120]. In contrast, more promising results are obtained in solid tumors including those of prostate, lung, esophagus and ovary [Citation121–125]. In particular, using EVs for early detection of pancreatic cancer have been studied extensively [Citation126–129].

Challenges and future perspectives

The current techniques used for EV isolation have their own advantages and disadvantages, as described in . The most challenging task regarding utilization of EVs and their components in clinical settings is to isolate pure EV populations without contamination with similar sized particles [Citation130]. Some unwanted biomolecules may nonspecifically attach to the surface of EVs and act as confounding variables. There is also a lack of knowledge on whether compositional difference in EVs from different origins reflect their functional aspects or not. Other challenges include the standardization of the quantitation and characterization methods, as well as downstream analysis of EV contents. Electron microscopy is currently used for characterizing EVs along with flow cytometry, but both techniques require expensive equipment and may not be suitable for routine use [Citation131,Citation132].

Table 2. Overview of extracellular vesicles isolation.

EVs are present in a wide array of biofluids but it is still uncertain which source is the most suitable in terms of their isolation and compositional representation [Citation133]. The concentration of DNA and RNA in EVs differs significantly [Citation134]. Their nucleic acid content varies greatly from patient to patient, and different isolation methods can give very different results [Citation135]. EVs purified from serum, plasma, CSF, cell culture media and other sources vary greatly, in terms of yield, and the amounts of nucleic acid recovered [Citation136,Citation137]. For example, a recent study showed that miRNA species and their abundance are highly variable among EVs isolated from plasma and cell culture media [Citation120].

For glioma diagnosis, EVs from CSF is preferred since their source is confined to the central nervous system. Whereas EVs from blood is contaminated by those derived other cell types in the circulation and thus reduce the abundance of glioma-specific EVs [Citation138]. Physical characteristics of blood (e.g., viscosity and density) and high abundance of lipoproteins also limit the availability of target EVs. High- and low-density lipoproteins can transport miRNA that may be co-precipitated with those derived from EVs [Citation139]. Furthermore, small lipoproteins such as chylomicrons, which have similar sizes as EVs, might interfere with their isolation [Citation140]. Apart from relevance to brain tumor, CSF-derived EVs exhibit higher reproducibility and accuracy in determining miRNA compared with blood [Citation141,Citation142]. In spite of this advantage, CSF is a poor source for detecting circulating tumor cells [Citation143].

Other than the source of EVs, their collection, isolation and storage could also pose problems for downstream analyses [Citation144]. Time of collection and physical activities can influence the level of circulating EVs [Citation145]. The latter is suggested to help release more EVs in the circulation [Citation146]. Regarding isolation methods (summarized in ), none of them could easily be translated into routine clinical use in terms of cost, time, user friendliness and reproducibility. The tube quality and preservatives used may cause artifactual vesicles formation, and some cells are capable of producing EVs on storage [Citation147].

There are a number of existing techniques available for analyzing the molecules present in EVs, including RNAseq, droplet digital PCR, microarrays, and real time PCR. Each technique has its own sensitivity and specificity. Their properties are summarized in . In summary, the major bottleneck for EVs to be useful in clinical diagnosis is the standardization of their sources for different disease, and the collection and isolation methods to be used (Figure S1).

Table 3. Techniques currently used for extracellular vesicles research with their test indexes.

Conclusion

EVs hold great promises as a noninvasive source of biomarkers for clinical applications. Their ease of collection and the stability of their contents provide a window into the pathological status of the patient. They could even be used for early diagnosis of disease like GB in which the clinical symptoms are non-specific! A major hurdle though is in the purification of EVs without contamination by particles of similar sizes. Furthermore, the profile of biomarkers in the EVs must be clearly defined and correlated to the disease state. With more interdisciplinary efforts, these hurdles will be overcome leading to the concrete use of EVs for diagnosis and prognosis in GB, and many other types of diseases.

Supplemental material

Supplemental Material

Download PDF (230.9 KB)

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • Rice T, Lachance DH, Molinaro AM, et al. Understanding inherited genetic risk of adult glioma–a review. Neuro-Oncol Pract. 2016;3:10–16.
  • Abrams DA, Hanson JA, Brown JM, et al. Timing of surgery and bevacizumab therapy in neurosurgical patients with recurrent high grade glioma. J Clin Neurosci. 2015;22:35–39.
  • Wilson TA, Karajannis MA, Harter DH. Glioblastoma multiforme: state of the art and future therapeutics. Surg Neurol Int. 2014;5:64.
  • Ohgaki H, Kleihues P. The definition of primary and secondary glioblastoma. Clin Cancer Res. 2013;19:764–772.
  • Costa B, Bendinelli S, Gabelloni P, et al. Human glioblastoma multiforme: p53 reactivation by a novel MDM2 inhibitor. PloS One. 2013;8:e72281.
  • Desai R, Suryadevara CM, Batich KA, et al. Emerging immunotherapies for glioblastoma. Expert Opin Emerg Drugs. 2016;21:133–145.
  • Ozawa T, Brennan CW, Wang L, et al. PDGFRA gene rearrangements are frequent genetic events in PDGFRA-amplified glioblastomas. Genes Develop. 2010;24:2205–2218.
  • Han F, Hu R, Yang H, et al. PTEN gene mutations correlate to poor prognosis in glioma patients: a meta-analysis. Onco Targets Ther. 2016;9:3485–3492.
  • Chin L, Meyerson M, Aldape K, et al. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068.
  • Waugh MG. Chromosomal instability and phosphoinositide pathway gene signatures in glioblastoma multiforme. Mol Neurobiol. 2016;53:621–630.
  • Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016;131:803–820.
  • Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110.
  • Inda MM, Bonavia R, Seoane J. Glioblastoma multiforme: a look inside its heterogeneous nature. Cancers (Basel). 2014;6:226–239.
  • Ammirati M, Chotai S, Newton H, et al. Hypofractionated intensity modulated radiotherapy with temozolomide in newly diagnosed glioblastoma multiforme. J Clin Neurosci. 2014;21:633–637.
  • Cheng L, Wu Q, Guryanova OA, et al. Elevated invasive potential of glioblastoma stem cells. Biochem Biophys Res Commun. 2011;406:643–648.
  • Yang L, Lin C, Wang L, et al. Hypoxia and hypoxia-inducible factors in glioblastoma multiforme progression and therapeutic implications. Exp Cell Res. 2012;318:2417–2426.
  • Urbanska K, Sokolowska J, Szmidt M, et al. Glioblastoma multiforme – an overview. Contemporary Oncol (Poznan, Poland). 2014;18:307–312.
  • Mammoto T, Jiang A, Jiang E, et al. Role of collagen matrix in tumor angiogenesis and glioblastoma multiforme progression. Am J Pathol. 2013;183:1293–1305.
  • Sottoriva A, Spiteri I, Piccirillo SG, et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci USA. 2013;110:4009–4014.
  • Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14:195–208.
  • Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J Clin Investig. 2016;126:1173–1180.
  • Grange C, Camussi G. Immunosuppressive role of extracellular vesicles: HLA-G, an important player. Ann Transl Med. 2017;5:223.
  • Todorova D, Simoncini S, Lacroix R, et al. Extracellular vesicles in angiogenesis. Circ Res. 2017;120:1658–1673.
  • Zappulli V, Friis KP, Fitzpatrick Z, et al. Extracellular vesicles and intercellular communication within the nervous system. J Clin Investig. 2016;126:1198–1207.
  • Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164:1226–1232.
  • Yamada NO. Extracellular vesicles: emerging mediators of intercellular communication and tumor angiogenesis. Ann Transl Med. 2017;5:59.
  • Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–383.
  • Ban LA, Shackel NA, McLennan SV. Extracellular vesicles: a new frontier in biomarker discovery for non-alcoholic fatty liver disease. Int J Mol Sci. 2016;17:376.
  • Kalamvoki M, Deschamps T. Extracellular vesicles during Herpes Simplex Virus type 1 infection: an inquire. Virol J. 2016;13:63.
  • Xu R, Greening DW, Zhu HJ, et al. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Investig. 2016;126:1152–1162.
  • Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell. 1983;33:967–978.
  • Vlassov AV, Magdaleno S, Setterquist R, et al. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta. 2012;1820:940–948.
  • Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016;36:301–312.
  • Yanez-Mo M, Siljander PR, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066.
  • Miyado K, Yoshida K, Yamagata K, et al. The fusing ability of sperm is bestowed by CD9-containing vesicles released from eggs in mice. Proc Natl Acad Sci USA. 2008;105:12921–12926.
  • Corrigan L, Redhai S, Leiblich A, et al. BMP-regulated exosomes from Drosophila male reproductive glands reprogram female behavior. J Cell Biol. 2014;206:671–688.
  • Fruhbeis C, Frohlich D, Kuo WP, et al. Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 2013;11:e1001604.
  • Frohlich D, Kuo WP, Fruhbeis C, et al. Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation. Philosophical transactions of the Royal Society of London Series B. Biol Sci 2014;369:20130510.
  • Grant BD, Donaldson JG. Pathways and mechanisms of endocytic recycling. Nat Rev Mol Cell Biol. 2009;10:597–608.
  • Iaea DB, Maxfield FR. Membrane order in the plasma membrane and endocytic recycling compartment. PloS One. 2017;12:e0188041.
  • Wollert T, Hurley JH. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature. 2010;464:864–869.
  • Colombo M, Moita C, van Niel G, et al. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 2013;126:5553–5565.
  • Baietti MF, Zhang Z, Mortier E, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14:677–685.
  • Stuffers S, Sem Wegner C, Stenmark H, et al. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic (Copenhagen, Denmark). 2009;10:925–937.
  • Laulagnier K, Grand D, Dujardin A, et al. PLD2 is enriched on exosomes and its activity is correlated to the release of exosomes. FEBS Lett. 2004;572:11–14.
  • Stenmark H. Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol. 2009;10:513–525.
  • Ostrowski M, Carmo NB, Krumeich S, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 2010;12:19–30.
  • Alonso R, Mazzeo C, Merida I, et al. A new role of diacylglycerol kinase alpha on the secretion of lethal exosomes bearing Fas ligand during activation-induced cell death of T lymphocytes. Biochimie. 2007;89:213–221.
  • Fader CM, Sanchez DG, Mestre MB, et al. TI-VAMP/VAMP7 and VAMP3/cellubrevin: two v-SNARE proteins involved in specific steps of the autophagy/multivesicular body pathways. Biochim Biophys Acta. 2009;1793:1901–1916.
  • Puri N, Roche PA. Mast cells possess distinct secretory granule subsets whose exocytosis is regulated by different SNARE isoforms. Proc Natl Acad Sci USA. 2008;105:2580–2585.
  • Tiwari N, Wang CC, Brochetta C, et al. VAMP-8 segregates mast cell-preformed mediator exocytosis from cytokine trafficking pathways. Blood. 2008;111:3665–3674.
  • Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:24641.
  • Svensson KJ, Christianson HC, Wittrup A, et al. Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1. J Biol Chem. 2013;288:17713–17724.
  • Atai NA, Balaj L, van Veen H, et al. Heparin blocks transfer of extracellular vesicles between donor and recipient cells. J Neurooncol. 2013;115:343–351.
  • Plebanek MP, Mutharasan RK, Volpert O, et al. Nanoparticle targeting and cholesterol flux through scavenger receptor type B-1 inhibits cellular exosome uptake. Sci Rep. 2015;5:15724.
  • Parolini I, Federici C, Raggi C, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J Biol Chem. 2009;284:34211–34222.
  • Nakase I, Futaki S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci Rep. 2015;5:10112.
  • M HR, Bayraktar E, G KH, et al. Exosomes: from garbage bins to promising therapeutic targets. Int J Mol Sci. 2017;18:538.
  • Lo Cicero A, Stahl PD, Raposo G. Extracellular vesicles shuffling intercellular messages: for good or for bad. Curr Opin Cell Biol. 2015;35:69–77.
  • Feng Q, Zhang C, Lum D, et al. A class of extracellular vesicles from breast cancer cells activates VEGF receptors and tumour angiogenesis. Nat Comms. 2017;8:14450.
  • Treps L, Edmond S, Harford-Wright E, et al. Extracellular vesicle-transported Semaphorin3A promotes vascular permeability in glioblastoma. Oncogene. 2016;35:2615–2623.
  • Languino LR, Singh A, Prisco M, et al. Exosome-mediated transfer from the tumor microenvironment increases TGFbeta signaling in squamous cell carcinoma. Am J Transl Res. 2016;8:2432–2437.
  • Manda SV, Kataria Y, Tatireddy BR, et al. Exosomes as a biomarker platform for detecting epidermal growth factor receptor-positive high-grade gliomas. J Neurosurg. 2018;128:1091–1101.
  • Subra C, Grand D, Laulagnier K, et al. Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res. 2010;51:2105–2120.
  • Skotland T, Sandvig K, Llorente A. Lipids in exosomes: current knowledge and the way forward. Prog Lipid Res. 2017;66:30–41.
  • Batagov AO, Kurochkin IV. Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3'-untranslated regions. Biol Direct. 2013;8:12.
  • Crescitelli R, Lasser C, Szabo TG, et al. Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J Extracell Vesicles. 2013;2:10.
  • Cheng L, Sun X, Scicluna BJ, et al. Characterization and deep sequencing analysis of exosomal and non-exosomal miRNA in human urine. Kidney Int. 2014;86:433–444.
  • Huang X, Yuan T, Tschannen M, et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genomics. 2013;14:319.
  • Ogawa Y, Taketomi Y, Murakami M, et al. Small RNA transcriptomes of two types of exosomes in human whole saliva determined by next generation sequencing. Biol Pharm Bull. 2013;36:66–75.
  • Waldenstrom A, Genneback N, Hellman U, et al. Cardiomyocyte microvesicles contain DNA/RNA and convey biological messages to target cells. PloS One. 2012;7:e34653.
  • Balaj L, Lessard R, Dai L, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180.
  • Budnik V, Ruiz-Cañada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17:160–172.
  • Liu ZM, Wang YB, Yuan XH. Exosomes from murine-derived GL26 cells promote glioblastoma tumor growth by reducing number and function of CD8 + T cells. Asian Pacific J Cancer Prevent. 2013;14:309–314.
  • Kahlert C, Kalluri R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med. 2013;91:431–437.
  • Li CC, Eaton SA, Young PE, et al. Glioma microvesicles carry selectively packaged coding and non-coding RNAs which alter gene expression in recipient cells. RNA Biol. 2013;10:1333–1344.
  • O'Rourke DM, Nasrallah MP, Desai A, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017;9:399.
  • Schiera G, Di Liegro CM, Saladino P, et al. Oligodendroglioma cells synthesize the differentiation-specific linker histone H1 and release it into the extracellular environment through shed vesicles. Int J Oncol. 2013;43:1771–1776.
  • Putz U, Howitt J, Doan A, et al. The tumor suppressor PTEN is exported in exosomes and has phosphatase activity in recipient cells. Sci Signal. 2012;5:ra70.
  • Arscott WT, Tandle AT, Zhao S, et al. Ionizing radiation and glioblastoma exosomes: implications in tumor biology and cell migration. Transl Oncol. 2013;6:638–648.
  • Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharm Sci. 2012;33:207–214.
  • Kucharzewska P, Christianson HC, Welch JE, et al. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci. 2013;110;7312–7317.
  • Zhang G, Zhang Y, Cheng S, et al. CD133 positive U87 glioblastoma cells-derived exosomal microRNAs in hypoxia- versus normoxia-microenviroment. J Neurooncol. 2017;135:37–46.
  • Paz H, Pathak N, Yang J. Invading one step at a time: the role of invadopodia in tumor metastasis. Oncogene. 2014;33:4193–4202.
  • Leong HS, Robertson AE, Stoletov K, et al. Invadopodia are required for cancer cell extravasation and are a therapeutic target for metastasis. Cell Rep. 2014;8:1558–1570.
  • Stylli SS, I ST, Kaye AH, et al. Prognostic significance of Tks5 expression in gliomas. J Clin Neurosci. 2012;19:436–442.
  • Hoshino D, Kirkbride KC, Costello K, et al. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 2013;5:1159–1168.
  • Mallawaaratchy DM, Hallal S, Russell B, et al. Comprehensive proteome profiling of glioblastoma-derived extracellular vesicles identifies markers for more aggressive disease. J Neurooncol. 2017;131:233–244.
  • Ricklefs F, Mineo M, Rooj AK, et al. Extracellular vesicles from high-grade glioma exchange diverse pro-oncogenic signals that maintain intratumoral heterogeneity. Cancer Res. 2016;76:2876–2881.
  • Bronisz A, Wang Y, Nowicki MO, et al. Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res. 2014;74:738–750.
  • Sun X, Ma X, Wang J, et al. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget. 2017;8:36137–36148.
  • de Vrij J, Maas SL, Kwappenberg KM, et al. Glioblastoma-derived extracellular vesicles modify the phenotype of monocytic cells. Int J Cancer. 2015;137:1630–1642.
  • van der Vos KE, Abels ER, Zhang X, et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 2016;18:58–69.
  • Sosnowski R, Zagrodzka M, Borkowski T. The limitations of multiparametric magnetic resonance imaging also must be borne in mind. Central Eur J Urol. 2016;69:22–23.
  • Peca C, Pacelli R, Elefante A, et al. Early clinical and neuroradiological worsening after radiotherapy and concomitant temozolomide in patients with glioblastoma: tumour progression or radionecrosis? Clin Neurol Neurosurg. 2009;111:331–334.
  • Neagu MR, Huang RY, Reardon DA, et al. How treatment monitoring is influencing treatment decisions in glioblastomas. Curr Treat Options Neurol. 2015;17:343.
  • Ilie M, Hofman P. Pros: can tissue biopsy be replaced by liquid biopsy? Transl Lung Cancer Res. 2016;5:420–423.
  • Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science (New York, NY). 2014;344:1396–1401.
  • Yip S, Miao J, Cahill DP, et al. MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin Cancer Res. 2009;15:4622–4629.
  • Valadi H, Ekstrom K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659.
  • Morishita M, Takahashi Y, Nishikawa M, et al. Quantitative analysis of tissue distribution of the B16BL6-derived exosomes using a streptavidin-lactadherin fusion protein and iodine-125-labeled biotin derivative after intravenous injection in mice. J Pharm Sci. 2015;104:705–713.
  • Skog J, Wurdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–1476.
  • Noerholm M, Balaj L, Limperg T, et al. RNA expression patterns in serum microvesicles from patients with glioblastoma multiforme and controls. BMC Cancer. 2012;12:22.
  • Lan F, Qing Q, Pan Q, et al. Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol. 2018;41:25–33.
  • Chen WW, Balaj L, Liau LM, et al. BEAMing and Droplet Digital PCR analysis of mutant IDH1 mRNA in glioma patient serum and cerebrospinal fluid extracellular vesicles. Mol Ther Nucleic Acids. 2013;2:e109.
  • Akers JC, Hua W, Li H, et al. A cerebrospinal fluid microRNA signature as biomarker for glioblastoma. Oncotarget. 2017;8:68769–68779.
  • Figueroa J, Phillips LM, Shahar T, et al. Exosomes from glioma-associated mesenchymal stem cells increase the tumorigenicity of glioma stem-like cells via transfer of miR-1587. Cancer Res. 2017;77:5808–5819.
  • Shao H, Chung J, Lee K, et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun. 2015;6:6999.
  • Ramakrishnan V, Kushwaha D, Koay DC, et al. Post-transcriptional regulation of O(6)-methylguanine-DNA methyltransferase MGMT in glioblastomas. Cancer Biomark. 2011;10:185–193.
  • Kushwaha D, Ramakrishnan V, Ng K, et al. A genome-wide miRNA screen revealed miR-603 as a MGMT-regulating miRNA in glioblastomas. Oncotarget. 2014;5:4026–4039.
  • Yang JK, Yang JP, Tong J, et al. Exosomal miR-221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma. J Neurooncol. 2017;131:255–265.
  • Pinet S, Bessette B, Vedrenne N, et al. TrkB-containing exosomes promote the transfer of glioblastoma aggressiveness to YKL-40-inactivated glioblastoma cells. Oncotarget. 2016;7:50349–50364.
  • Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–996.
  • Andre-Gregoire G, Bidere N, Gavard J. Temozolomide affects extracellular vesicles released by glioblastoma cells. Biochimie 2018;155:11–15.
  • Zeng A, Wei Z, Yan W, et al. Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma. Cancer Lett. 2018;436:10–21.
  • Garnier D, Meehan B, Kislinger T, et al. Divergent evolution of temozolomide resistance in glioblastoma stem cells is reflected in extracellular vesicles and coupled with radiosensitization. Neuro-oncology. 2018;20:236–248.
  • Uribe D, Torres A, Rocha JD, et al. Multidrug resistance in glioblastoma stem-like cells: role of the hypoxic microenvironment and adenosine signaling. Mol Aspects Med. 2017;55:140–151.
  • Redzic JS, Ung TH, Graner MW. Glioblastoma extracellular vesicles: reservoirs of potential biomarkers. Pharm Personalized Med. 2014;7:65–77.
  • Figueroa JM, Skog J, Akers J, et al. Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients. Neuro-oncology. 2017;19:1494–1502.
  • Akers JC, Ramakrishnan V, Kim R, et al. miRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients. J Neurooncol. 2015;123:205–216.
  • Vlaeminck-Guillem V. Extracellular vesicles in prostate cancer carcinogenesis, diagnosis, and management. Front Oncol. 2018;8:222.
  • Zheng H, Zhan Y, Liu S, et al. The roles of tumor-derived exosomes in non-small cell lung cancer and their clinical implications. J Exp Clin Cancer Res. 2018;37:226.
  • Zhang YG, Zhou MW, Bai L, et al. Extracellular vesicles promote esophageal cancer progression by delivering lncZEB1-AS1 between cells. Eur Rev Med Pharm Sci. 2018;22:2662–2670.
  • Liu MX, Liao J, Xie M, et al. miR-93-5p Transferred by exosomes promotes the proliferation of esophageal cancer cells via intercellular communication by targeting PTEN. Biomed Environ Sci. 2018;31:171–185.
  • Sharma S, Zuniga F, Rice GE, et al. Tumor-derived exosomes in ovarian cancer – liquid biopsies for early detection and real-time monitoring of cancer progression. Oncotarget. 2017;8:104687–104703.
  • Armstrong EA, Beal EW, Chakedis J, et al. Exosomes in pancreatic cancer: from early detection to treatment. J Gastrointest Surg. 2018;22:737–750.
  • Zhao C, Gao F, Weng S, et al. Pancreatic cancer and associated exosomes. Cancer Biomark. 2017;20:357–367.
  • Yan Y, Fu G, Ming L. Role of exosomes in pancreatic cancer. Oncol Lett. 2018;15:7479–7488.
  • Kitagawa T, Taniuchi K, Tsuboi M, et al. Circulating pancreatic cancer exosomal RNAs for detection of pancreatic cancer. Mol Oncol. 2018. DOI: 10.1002/1878-0261.12398. [Epub ahead of print]
  • Tauro BJ, Greening DW, Mathias RA, et al. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods (San Diego, Calif). 2012;56:293–304.
  • van der Pol E, Boing AN, Harrison P, et al. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev. 2012;64:676–705.
  • Zhang HG, Grizzle WE. Exosomes: a novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am J Pathol. 2014;184:28–41.
  • Webber J, Clayton A. How pure are your vesicles? J Extracell Vesicles. 2013;2:19861.
  • Bryzgunova OE, Zaripov MM, Skvortsova TE, et al. Comparative Study of Extracellular Vesicles from the Urine of Healthy Individuals and Prostate Cancer Patients. PloS One. 2016;11:e0157566.
  • Saugstad JA, Lusardi TA, Van Keuren-Jensen KR, et al. Analysis of extracellular RNA in cerebrospinal fluid. J Extracell Vesicles. 2017;6:1317577.
  • Li M, Zeringer E, Barta T, et al. Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130502.
  • Moret I, Sanchez-Izquierdo D, Iborra M, et al. Assessing an improved protocol for plasma microRNA extraction. PloS One. 2013;8:e82753.
  • Dean WL, Lee MJ, Cummins TD, et al. Proteomic and functional characterisation of platelet microparticle size classes. Thromb Haemost. 2009;102:711–718.
  • Vickers KC, Palmisano BT, Shoucri BM, et al. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13:423–433.
  • Sodar BW, Kittel A, Paloczi K, et al. Low-density lipoprotein mimics blood plasma-derived exosomes and microvesicles during isolation and detection. Sci Rep 2016;6:24316.
  • Teplyuk NM, Mollenhauer B, Gabriely G, et al. MicroRNAs in cerebrospinal fluid identify glioblastoma and metastatic brain cancers and reflect disease activity. Neuro-oncology 2012;14:689–700.
  • Kang JH, Mollenhauer B, Coffey CS, et al. CSF biomarkers associated with disease heterogeneity in early Parkinson's disease: the Parkinson's Progression Markers Initiative study. Acta Neuropathol. 2016;131:935–949.
  • Weston CL, Glantz MJ, Connor JR. Detection of cancer cells in the cerebrospinal fluid: current methods and future directions. Fluids Barriers CNS. 2011;8:14.
  • Mora E, Álvarez-Cubela S, Oltra E. Biobanking of exosomes in the era of precision medicine: are we there yet? Int J Mol Sci. 2015;17:13.
  • Madden LA, Vince RV, Sandstrom ME, et al. Microparticle-associated vascular adhesion molecule-1 and tissue factor follow a circadian rhythm in healthy human subjects. Thromb Haemost. 2008;99:909–915.
  • Fruhbeis C, Helmig S, Tug S, et al. Physical exercise induces rapid release of small extracellular vesicles into the circulation. J Extracell Vesicles. 2015;4:28239.
  • Gyorgy B, Paloczi K, Kovacs A, et al. Improved circulating microparticle analysis in acid-citrate dextrose (ACD) anticoagulant tube. Thromb Res. 2014;133:285–292.
  • Lang FM, Hossain A, Gumin J, et al. Mesenchymal stem cells as natural biofactories for exosomes carrying miR-124a in the treatment of gliomas. Neuro-oncology. 2018;20:380–390.
  • Pavlyukov MS, Yu H, Bastola S, et al. Apoptotic Cell-Derived Extracellular Vesicles Promote Malignancy of Glioblastoma Via Intercellular Transfer of Splicing Factors. Cancer Cell. 2018;34:119–135.e10.
  • Kore RA, Abraham EC. Inflammatory cytokines, interleukin-1 beta and tumor necrosis factor-alpha, upregulated in glioblastoma multiforme, raise the levels of CRYAB in exosomes secreted by U373 glioma cells. Biochem Biophys Res Commun. 2014;453:326–331.
  • Manterola L, Guruceaga E, Gallego Perez-Larraya J, et al. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro-oncology. 2014;16:520–527.
  • Cai Q, Zhu A, Gong L. Exosomes of glioma cells deliver miR-148a to promote proliferation and metastasis of glioblastoma via targeting CADM1. Bull Cancer. 2018;105:643–651.
  • Huang K, Fang C, Yi K, et al. The role of PTRF/Cavin1 as a biomarker in both glioma and serum exosomes. Theranostics. 2018;8:1540–1557.
  • Shi R, Wang PY, Li XY, et al. Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget. 2015;6:26971–26981.
  • Zheng J, Liu X, Xue Y, et al. TTBK2 circular RNA promotes glioma malignancy by regulating miR-217/HNF1β/Derlin-1 pathway. J Hematol Oncol. 2017;10:52.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.