CHAPTER
Exosome nanocarriers: a natural, novel, and perspective approach in drug delivery system
7
Jasvinder Singh Bhatti1, Rajesh Vijayvergiya2, Bhupinder Singh3 and Gurjit Kaur Bhatti3,4 1
Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India 2Department of Cardiology, Post Graduate Institute of Medical Education and Research, Chandigarh, India 3 UGC Centre of Excellence in Nano Applications, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India 4University Institute of Pharmacy Sciences, Chandigarh University, Mohali, Punjab, India
ABBREVIATIONS miRNA ILVs MVE EVs RLPs MHC mRNA ESCRT IL DCs APCs CD NF-κB TNF-α PD AD DNA RNA T2DM BBB siRNA EFGR
micro RNA intraluminal vesicles multivesicular endosomes extracellular vesicles retrovirus-like particles major histocompatibility molecules messenger RNA endosomal sorting complexes required for transport interleukins dendritic cells antigen presenting cells cluster of differentiation nuclear factor kappa-light-chain-enhancer of activated B-cells tumor necrosis factor-α Parkinson’s disease Alzheimer’s disease deoxyribonucleic acid ribonucleic acid type 2 diabetes mellitus blood brain barrier small interfering ribonucleic acid epidermal growth factor receptor
Nanoarchitectonics in Biomedicine. DOI: https://doi.org/10.1016/B978-0-12-816200-2.00008-6 © 2019 Elsevier Inc. All rights reserved.
189
190
CHAPTER 7 Exosome as nanocarriers in drug delivery system
7.1 INTRODUCTION The extracellular vesicles (EVs) are naturally present in many biological fluidsand are classified by their functionality, morphological properties, and origin/biogenesis (Street et al., 2012; Bobrie et al., 2011; Buzas et al., 2014). EVs are mainly classified into four broad types according to their biogenesis as exosomes, microvesicles (MVs), apoptotic bodies, and retrovirus-like particles (RLPs) (EL Andaloussi et al., 2013; van der Pol et al., 2012). The origin of MVs, apoptotic bodies, and RLPs occurs by direct budding from the plasma membrane. However, exosomes originate by the process of exocytosis of the multivesicular bodies (MVBs). Recent studies demonstrated that MVs are bigger than exosomes (Akers et al., 2013). MVs bear different nucleic acids and proteins such as actin, tubulin, β1 integrins, and vesicle-associated membrane protein 3 (Buzas et al., 2014). The main feature of all these vesicles is the endosomal biogenesis as intraluminal vesicles (ILVs) within multivesicular endosomes (MVEs), which is different from the origin of other vesicles shedding from the plasma membrane (Kalra et al., 2012; Raposo and Stoorvogel, 2013). Inward invagination of the membrane results in the continuous addition of vesicles in the lumen of MVE into the extracellular space and was first observed in rat reticulocytes (Harding et al., 2013). It has been recognized that after the cells undergo apoptosis they form small vesicles known as apoptotic bodies (Emanueli et al., 2015). In fact, these small vesicles were identified as debris and considered as a means of wrapping the leftovers of the dead cells following an approach that there should not be any collateral damage to the cells in surrounding area. On the other hand, evidence has already materialized the fact that the EVs, in the size range of 30 nm to 1 μm, are not released during cell death but necessarily have their own a biological function (Emanueli et al., 2015). Exosomes are tiny-sized (ranging in 30 100 nm in diameter) membranous vesicles secreted by a variety of cell types in the body through the endosomal pathway. They were first reported in 1981 as exfoliation from monolayer cultures and subsequently demonstrated by Johnstone and his team (Johnstone et al., 1991; Trams et al., 1981). They are known to be associated with diverse pathological conditions and represent a variety of functions like delivering biomolecules and genetic material to recipient cells. Exosomes are enriched with a bundle of specific molecules which are different from proteins normally located in the plasma membrane including tetraspanins (CD9, CD63, CD81, and CD82), heat-shock proteins (HSP60, HSP70, HSPA5, CCT2, and HSP90), major histocompatibility molecules (MHC-I and MHC-II), cytoskeleton proteins (actin, tubulin, cofilin, myosin, vimentin, fibronectin, advillin, talin, CAP1, keratins, and meosin), microRNA (miRNA), messenger RNA (mRNA), Annexins (I, II, IV, V, VI, VII, and X1), apoptotic bodies, and many more (Akers et al., 2013; Keller et al., 2006; Hristov et al., 2004).
7.1 Introduction
Exosomes also contain a domain of lipids including cholesterol, sphingomyelin, ceramide, and ganglioside called membrane rafts (Fig. 7.1). There is another class of cytosolic proteins found in the exosomes includes Rabs, a family of GTPases involved in vesicular transport and protein complexes that regulate membrane fusion and docking. About 40 Rab proteins have been identified in the exosomes RAB27A, RAB27B, RAB11, and RAB35 (Bobrie et al., 2011; Ostrowski et al., 2010). Exosomes are special in their way as they possess particular cell surface markers as a legacy from the cell of their origin. These cell surface
FIGURE 7.1 Structural components of the exosome. Exosomes are composed of a bundle of specific molecules including tetraspanins (CD9, CD63, CD81, CD82), HSPs (HSP60, HSP70, HSPA5, CCT2, and HSP90), MHCs (MHC-I and -II), cytoskeleton proteins (actin, tubulin, cofilin, myosin, vimentin, fibronectin, advillin, talin, CAP1, keratins, meosin), microRNA and mRNA, Annexins I, II, IV, V, VI, VII and X1, and apoptotic bodies. Exosomes also have a domain of lipids including cholesterol, sphingomyelin, ceramide, and ganglioside called membrane rafts. There is another class of cytosolic proteins found in exosomes including Rabs, a family of GTPases involved in vesicular transport and protein complexes that regulate membrane fusion and docking. HSP, Heat-shock protein; MHC, major histocompatibility molecule; mRNA, messenger RNA.
191
192
CHAPTER 7 Exosome as nanocarriers in drug delivery system
markers provide the exosomes the means of interaction with neighboring and distant cells (Skokos et al., 2003). Possession of these qualities has made exosomes a valuable therapeutic tool with inherent qualities in the biomedicine research area for targeted drug delivery to counter deadly diseases. In addition to lipids, proteins, and mRNA, exosomes also transfer miRNA to recipient cells which can alter the pathologic genetic expression (Valadi et al., 2007). It has been a long journey to come to understand the actual morphology and functions of exosomes concerning the dynamic role in intercellular communication by transporting a broad range of bioactive molecules between different cells and tissues (Tkach and Thery, 2016). This function allows exosomes to regulate many physiological activities, including the immune response.
7.2 EXOSOME BIOGENESIS, RELEASE, AND UPTAKE The secretion of exosomes in the cells occurs by fusion of the MVB and the cell membrane’s most vibrant compartments which are actively involved in internalization of extracellular ligands, post membranous invagination, and release of various kinds of endocytic vesicles. During these processes, some membrane proteins get transferred to these vesicles. Exosomes are formed by the inward budding of the endocytic vesicles and contain cytoplasmic components. Continuous formation of exosomes leads to the development of MVBs. The membrane of MVBs is semipermeable and ensures that cargoes are selectively loaded into the exosomes. Exosomes can have similar kinds of membrane constituents even if they have originated from different types of cells (Bartel, 2004). Exosomes keep the basic topology of the membrane intact; phosphatidylserine on the exterior while the cytosolic side of lipid bilayer remains inside. This arrangement causes the isolation of signal domains of the membrane receptors, thereby restricting their functionality of dampening the signal transduction. On the other hand, the luminal part remains exposed. Fig. 7.2 shows the exosome biogenesis and release of exosomes from the endocytic compartment of live cells. The preliminary steps involved in sorting the proteins undergoing endocytosis are the inward budding processes on the cell surface, which can be clathrindependent and independent ways. The endocytotic vesicles are transferred to endosomes which have an internal acidic environment. The acidic pH gets reduced by some modification in the protein composition in the late endosomes which shed into the endosomal lumen to form MVBs and further leads to the formation of ILVs. MVBs are abundant in cholesterol which can have dire consequences for cell survival, so they get fused with the plasma membrane and release exosomes but MVBs with a low amount of cholesterol fuse with the lysosomes and are involved in proteolysis. There are mainly two mechanisms involved in the formation of exosomes; ESCRT (endosomal sorting complexes required for transport) dependent (which comprises four proteins O, I, II, III, and lipid-dependent)
7.2 Exosome Biogenesis, Release, and Uptake
FIGURE 7.2 Mechanism of exosome biogenesis. Early endosomes arose through clathrin-mediated endocytosis at the plasma membrane and converted into late endosomes by changing the protein content and acidification. Then, MVBs are derived from late endosomes budding into their endosomal lumen, and cholesterol-rich MVBs interact with the plasma membrane to secrete exosomes. Exosomes are formed by invagination MVBs that fuse with the plasma membrane. MVB, Multivesicular body.
and ESCRT independent. ESCRT-O separates ubiquitinated proteins, I and II complexes help in budding, and III completes the process and leads to the formation of ILVs in the MVBs rich population. Exosomes firstly originate as ILVs then get matured to MVEs (Huotari and Helenius, 2011; Hanson and Cashikar, 2012). Tetraspanins such as CD81, CD9, and CD63 play a fundamental role in the composition of ESCRT-independent loading into exosomes by organizing the membrane into tetraspanin-enriched domains. The ESCRT complex mechanism, both dependent and independent, for packing of protein cargo into exosomes is a sign of the existence of different MVB and exosome populations. Different types of cancerous cells secrete different types of exosomes which vary in morphology and miRNA composition (Palma et al., 2012; Penfornis et al., 2016), but some secret exosomes which get differentiated by surface antigens having different protein content with various functions. The exosomes released from apical and basolateral cell membranes may have the heterogeneous population of vesicles, but the existence of controlled and specialized mechanisms control the selective, specific sorting of protein and miRNA cargo into these vesicles. While numerous researchers explained the biogenesis of exosomes, most of these mechanisms are still unclear.
193
194
CHAPTER 7 Exosome as nanocarriers in drug delivery system
7.3 EXOSOMES IN BIOLOGICAL FLUIDS Exosomes play a vital role in intercellular communication or as disease biomarkers. An attribute that is independently measured and evaluated as an indicator of a biologic, pathogenic process or pharmacologic response to a therapeutic intervention is referred to a biomarker (Biomarkers Definitions Working, 2001). Indeed, exosomes are secreted by a variety of biological fluids and cells including extracellular fluids, amniotic fluid, cultured cells, cerebrospinal fluid, synovial fluid, bronchoalveolar lavage fluid, blood, breast milk, saliva, urine, semen, bile, and nasal mucus (Johnstone et al., 1987; Michael et al., 2010; Admyre et al., 2007; Keller et al., 2011; Vella et al., 2008; Skriner et al., 2006). Particularly, exosomes released from serum or plasma fluids are more accurate for the development of biomarkers. These blood-based biomarkers can predict patients’ biochemical parameters more accurately and further predict the response to therapeutic intervention helping in earlier diagnosis and inhibiting the development of severe complications. Exosomes from various tissues of the body released into the blood stream can be distributed to distance tissues, and in the specified target tissue they regulate gene transcription, posttranslational modification, and signal transduction. Assorted cellular cargoes released by exosomes become a source of blood-based biomarkers and these biomarkers point toward the pathophysiological mechanisms which could be extremely useful in identifying cardiovascular complications associated with diabetes. That is the reason that plasma- and serum-derived exosomes act as a specific molecular marker for various tissues and determine the pathophysiological condition if any. Exosomes released from the other fluids like milk, saliva, and cerebrospinal fluid can be tissue specific. Although the exosomes exist in almost every biological body fluid, their existence in the serum and plasma is known to be of particular importance and can be exploited as biomarkers. The most important step in the isolation of exosomes and their development into biomarkers is to make sure of the exosomes’ enrichment in the extracellular milieu, which is quite complicated. Plasma and serum are the biological fluids which represent the extracellular environment of almost every organ, thereby; the efficacy of exosomes is entirely dependent on the matrix of the sample. The procedure of isolation of exosomes from biological fluids is a tedious method. Therefore many methods for isolation of exosomes have been standardized but a single optimized method has not been established till date. Out of available methods, the most widely used is differential centrifugation. Different sizes of vesicles get separated after the subsequent rounds of centrifugation, that is, large vesicles (2000 g, 10 minutes), MVs greater than 150 nm (10,000 g, 30 minutes), and exosomes (100,000 g, 70 minutes) (Schulte et al., 2016). However, this method also requires further optimization for the duration of each centrifugation according to the rotor type used. Sedimentation efficiency and density of EVs of varying sizes are dependent on the sedimentation path length and the duration of centrifugation,
7.4 Role of Exosomes in the Pathogenesis
which should be calculated for every individual rotor to minimize the presence of nonexosomal vesicles and MVs (Livshits et al., 2016; Cocucci et al., 2007; Eken et al., 2008). The size-based characterization of the isolated exosomes is done with at least three exosomal markers, for example, tetraspanins, TSG101, and Alix. Nonexosomal markers including Grp94, GM130, and cytochrome C are used for confirming the absence/low levels of contamination (Lotvall et al., 2014). Moreover, morphological characters are also analyzed by electron microscopy. During ultracentrifugation, the membrane of the exosomes may get disrupted and lead to the formation of aggregates of different shapes and sizes which may pose difficulties in the interpretation of the biomarkers (Linares et al., 2015). Size exclusion chromatography is also one of the methods used to isolate exosomes by size and chances of getting vesicles of regular and homogeneous shape with negligible damage are more when using this method. Currently, standard kits are commercially available in the market to isolate exosomes from the lesser volume of biological fluids.
7.4 ROLE OF EXOSOMES IN THE PATHOGENESIS, DIAGNOSTICS, AND THERAPEUTICS Exosomes display a wide variety of functions in the cell including cell cell communication, diagnostic markers, and potential roles in therapeutics. Furthermore, recent studies demonstrated the possible role of exosomes as a biomarker for early diagnosis of diseases (Simpson et al., 2009; Pisitkun et al., 2006). In the past few years more attention has been focused on the role of exosomes in the pathophysiology of a variety of diseases including cancer, metabolic disorders, cardiovascular diseases, immune diseases, and neurodegenerative diseases.
7.4.1 EXOSOMES IN INFLAMMATORY DISEASES Exosomes are known to play a vital role in treating various inflammatory reactions and autoimmune diseases as potential therapeutic agents. On the one hand, exosomes released from infected cells demonstrate a variety of functions. Further, the exosomes released from pathogen-stimulated brain cells have miRNAs which could interfere with the gene expression of various other cells (Gupta and Pulliam, 2014). In asthmatic patients, the release of exosomes from the bronchial epithelial cells in the lungs is enhanced and might be responsible for the increase in intercellular signaling. Exosomes released from the epithelial cells treated with interleukins 13 (IL-13) stimulate the inflammation along with the cell proliferation, but some specific drugs that result in the inhibition of exosomes production can lead to diminishing the inflammation (Kulshreshtha et al., 2013). Exosomes from Mycobacterium tuberculosis infected macrophages have mycobacteria-derived antigens and cause the induction of inflammatory immune
195
196
CHAPTER 7 Exosome as nanocarriers in drug delivery system
responses while inhibiting IFN-γ dependent activation of macrophages (Bhatnagar et al., 2007; Singh et al., 2011). In a similar way, exosomes released from Mycobacterium smegmatis or Mycobacterium avium infected macrophages will have great Hsp70 expression; however, the stimulation of the nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) pathway helps in the production of high levels of tumor necrosis factor-α (TNF-α). Exosomes with Hsp70 expression can produce a proinflammatory response with the release of TNF-κB by macrophages and promotion of NK cell activity (Gastpar et al., 2005). Curcumin loaded exosomes have been used to activate myeloid cells, which are involved in control and/or development of autoimmune diseases associated with inflammation. These hydrophobic compounds loaded onto exosomes were readily internalized by mouse lymphoma cells (EL-4) (Sun et al., 2010). Exosomal curcumin helps in decreasing the number of CD11b 1 Gr-1 1 cells which are responsible for causing acute lung inflammation by increasing the solubility, stability, and bioavailability of curcumin after the encapsulation into the lipid bilayer of exosomes. Nevertheless, it becomes easier to deliver the encapsulated curcumin to the brain through intranasal administration. Recently, an in vivo study explained the efficacy and effectiveness of curcumin-loaded exosomes in the treatment of neuroinflammatory diseases (Zhuang et al., 2011). Exosomes released from the dendritic cells (DCs) have received much attention lately. Pretreated DCs with adenovirus express IL-10 and exhibiting an antiinflammatory response. Conversely, exosomes released from DCs with the expression of IL-10 were established as immunosuppressive and help in suppressing T-cell proliferation and the delayed type of hypersensitivity response in collagen-induced arthritis (Kim et al., 2005). Exosomes released from the DCs produced much improved therapeutic results in comparison to the parental DCs (Kim et al., 2007). Intravenous administration of 10 μg of TGF-β/exosomes may have prevented weight loss with reduced intestinal bleeding in mice with inflammatory bowled disease. The suppression of the symptoms might be due to stable TGF-β in exosomes following intravenous administration and, hence, increased activity (Cai et al., 2012b). These results indicated that exosomes were effective nanocarriers for the delivery of biological agents for treating inflammatory diseases. Previous in vitro studies demonstrated that the exosomes released from the Epstein Barr virus cause some changes in the B-cells, and present MHC class II molecules lying on the membrane to CD4 1 T-cells (Raposo et al., 1996; Admyre et al., 2006; Liu et al., 2006). Other reports demonstrated that exosomes released from DCs stimulate CD8 1 T-cell-dependent antitumor immune response in vivo experiments (Zitvogel et al., 1998). Some research reports indicate that exosomes released from the antigen presenting cells (APCs) have the ability to stimulate the T-cells, but other reports suggest that APCs need exosomes beforehand in the case of T-cell stimulation (Admyre et al., 2006). However, it was revealed that MHC molecules fostered with exosomes have the capability of coming together with the T-cell receptors and can initiate
7.4 Role of Exosomes in the Pathogenesis
the production of CD4 1 and CD8 1 cells. Some reports came forward with the fact that MHC peptides loaded with exosomes, when transferred to DC, can stimulate naı¨ve T-cells (Thery et al., 2002a). T-cell stimulation by means of exosomes is recognized to be dependent on the physiological conditions of the origin cell, but matured DC-derived exosomes trigger T-cell activation in a much more efficient manner compared to immature ones (Segura et al., 2005; Montecalvo et al., 2008). Exosomes also carry antigenic properties so they can be degraded in that way too. Exosomes derived from pathogen-infected cells need to carry some specific antigenic properties which may be inherited from that particular pathogen and can induce CD4 1 and CD8 1 T-cell proliferation (Montecalvo et al., 2008; Bhatnagar and Schorey, 2007). Previous studies have revealed that exosomes secreted from cancerous cells release certain cytokines into the microenvironment which are attributed with triggering the immune response against tumorigenic tissue (Wolfers et al., 2001; Dai et al., 2006). On the contrary, exosomes derived from tumor tissue demonstrate exactly the opposite immune response in a manner of bearing different immunosuppressive molecules. This happens because of either decline in CD4 1 , CD8 1 , and natural killer cell stimulation or amplification in the differentiation of the immunosuppressive cells (Bobrie et al., 2011). Injecting exosomes derived from tumor cells supported metastasis by decreasing natural killer cells activation, but increase the differentiation of myeloid cells and lead to the development of melanoma and carcinoma (Liu et al., 2006). Till date, there is no relevant explanation for these controversial results reported by different scientists, but it could be speculated to be due to the heterogeneous population of exosomes.
7.4.2 EXOSOMES IN CANCER Cancer is the leading cause of mortality worldwide. Current treatment methods include surgery, chemotherapy, and radiation which further raises toxicity concerns. Since these methods are unable to cure metastasis of the cancerous cells there is a great need to develop more effective and less toxic therapies. Immunotherapy is a much-explored way of treatment for many types of cancers. Antibodies have exhibited a clinical accomplishment for cancer immunotherapy as well (Scott et al., 2012). Recent research reports show the potential role of exosomes in cancer treatment as well as diagnosis and prognosis (Salido-Guadarrama et al., 2014; Mahaweni et al., 2013; Chaudhuri et al., 2011; Gross et al., 2012; Lai et al., 2013; Zhang et al., 2016; Chaput et al., 2003; Basu and Ludlow, 2016; Viaud et al., 2008; Cho et al., 2012; Soekmadji et al., 2013). The proteins and RNA content from tumor-derived exosomes are involved in disseminating malignancy to recipient cells. This may provide diagnostic information and aid in therapeutic selection for cancer patients. The most important fact about exosomes is their ability to stimulate the immune response that can be exploited for the development of cancer vaccines (Tan et al., 2010).
197
198
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Many studies have demonstrated the therapeutic potential of DC-derived exosomes in cancer physiology (Chaput et al., 2006; Munich et al., 2012; Pitt et al., 2014, 2016; Viaud et al., 2010; Delcayre et al., 2005; Romagnoli et al., 2014; Zitvogel et al., 1999). Currently, much attention is being given toward exosomes and how to use them as gene delivery systems to specifically target tumor cells (Shtam et al., 2013). Exosomes also have the ability to act as a shuttle for antigen presentation and could be used as cancer vaccines in the near future (Natasha et al., 2014). Exosomes released by immune-suppressive DCs can suppress inflammatory responses and this potential could be used in treating autoimmune diseases (Kim et al., 2005; Yang and Robbins, 2012). Exosomes released from DCs carrying tumor-specific peptides can provoke cytotoxic T lymphocytes to respond and, consequently, inhibit tumor growth (Wolfers et al., 2001). Exosomes released from APCs contain tumor antigens which have been shown to encourage CD4 1 and CD8 1 T-cells and finally results in suppressing tumor growth (Zitvogel et al., 1998). Exosomes can initiate apoptosis by the activation of PTEN and GSK-3β that leads to the inactivation of PI3K/AKT which, in turn, plays a major role in tumor growth (Ristorcelli et al., 2008). Cancer cells are efficient in modulating the normal functioning of immune cells of the body’s defense mechanism and become malignant. The macrophages uptake of exosomes released by cancerous cells leads to the activation of NF-κB and, as a result, there are upregulation inflammatory cytokines (IL-6, TNF-α, GCSF, and CCL2) in the macrophages. Toll-like receptor 2 expressions on the surface of macrophages of these exosomes regulates the immune regulation which is mediated by the presence of palmitoylated protein ligands on the surface of tumor-derived exosomes (Chow et al., 2014).
7.4.3 EXOSOMES IN NEURODEGENERATIVE DISEASES Exosomes are involved in a variety of neurological disorders including Parkinson’s disease (PD) and Alzheimer’s disease (AD) (Haney et al., 2015; Wu et al., 2016; Russo et al., 2012; Rajendran et al., 2006; Malm et al., 2016; Vella et al., 2016). They are released by numerous cell types, whether normal or pathogenic, into the extracellular environment. Particularly, exosomes released by tumor tissues initiate the metastasis and highlight the fact that exosomes have the ability to spread diseases within the body (Costa-Silva et al., 2015; Hoshino et al., 2015). This ability of exosomes to spread the diseases is considered to be responsible for neurodegenerative disorders. The cause of neurodegenerative diseases is mainly the deposition of certain misfolded and aggregated forms of protein molecules in brain tissue. These misfolded proteins further spread to different parts of the body as the disease progresses (Braak et al., 2003). Biochemical and molecular analysis of cerebral spinal fluid and blood of patients suffering from several neurodegenerative diseases illustrated the presence of many misfolded and aggregated proteins which are carried by exosomes. Exosomes contain many of the proteins which are prone to aggregate and are responsible for causing PD, AD,
7.4 Role of Exosomes in the Pathogenesis
Creutzfeldt Jakob disease, and Amyotrophic lateral sclerosis (Brettschneider et al., 2015). Proteins like Aβ, tau, and α-synuclein have been reported to show prion-like activity in causing the pathological conversion of normal conformers of these proteins to those associated with disease. The aggregated nature and stable conformation of these proteins are resistant to proteolytic digestion. Many studies using cells and rat models came forward with evidence that the proteins causing neurodegeneration could be transmissible (Prusiner et al., 2015; Aguzzi and Rajendran, 2009). Although the accurate mechanism of transmission is yet to be discovered, various studies have revealed that it could be either by cell to cell communication or via tunneling nanotubes (Kanu et al., 2002; Gousset et al., 2009). A number of research studies have illustrated the involvement of exosomes in the transmission as neurodegenerative proteins were found to be secreted from the cell in association with exosomes (Fevrier et al., 2004). Due to their small size and relative stability, exosomes can cross the blood brain barrier (BBB) and, therefore, reveal bright perspectives toward diagnosis and as therapeutic tools for future neuro medicine (Aryani and Denecke, 2016; Jarmalaviciute and Pivoriunas, 2016; Howitt and Hill, 2016; Vella et al., 2016; Tsilioni et al., 2014; Wu et al., 2016; Lai and Breakefield, 2012). An exosomal biomarker signature has also recently been established to detect AD (Lugli et al., 2015; Cheng et al., 2015).
7.4.4 EXOSOMES IN METABOLIC DISORDERS The burden of metabolic disorders including type 2 diabetes, dyslipidemia, insulin resistance, and cardiovascular disease is exponentially increasing worldwide (Raj et al., 2014; Bhatti et al., 2016). Recent in vivo and in vitro studies demonstrated that exosomes secreted by a variety of cells play an important role in the regulation of the metabolic functions and may be responsible for the progression of some diseases (Lee et al., 2016). Exosomes are found to be rich in various biomolecules like proteins, mRNA, miRNA, noncoding RNA, and lipids. Recent studies have shown that exosomes isolated from biological fluids can be implemented as biomarkers for early diagnosis of metabolic diseases and can give an idea about the progression of the disease. Exosomes are capable of transferring DNA, RNA, and protein molecules which can regulate the metabolic functioning by interacting directly by transmembrane contact with the cell surface receptors of the nearby cells in remote areas (Aoki et al., 2007; Conde-Vancells et al., 2008; EL Andaloussi et al., 2013). Exosomes have the ability to perform vital functions on various platforms, including having a significant role in tumor progression (Rak and Guha, 2012), tissue regeneration (Timmers et al., 2011), enhancing the immune escape way and immune responses (Cai et al., 2012a; Simhadri et al., 2008; Chivet et al., 2012), transmission of neurological proteins (Bellingham et al., 2012; Emmanouilidou et al., 2010), and metabolic dysfunctions (Aswad et al., 2014).
199
200
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Amplification in the number of exosomes can lead to obesity and inflammatory complications (Feng et al., 2010; Kim et al., 2012). Exosomes released from the surface molecules of skeletal muscle, platelets, and T-cells contribute in metabolic dysfunctions and atherogenic tendencies (Aswad et al., 2014; Diamant et al., 2002). Previous studies linked the functioning of the exosomes with their number that was assessed to be higher in the plasma of type 2 diabetes mellitus (T2DM) patients having insulin resistance. Exosomes have the ability to organize insulin signaling which has become an emerging concept in the exosome-mediated regulation of metabolism in T2DM (Aswad et al., 2014; Safdar et al., 2016; Jalabert et al., 2016; Lee et al., 2015). Diverse complications associated with the T2DM represent miscellaneous patterns of exosomal biomolecules signifying exosomes that may contribute to the pathophysiology of type 2 diabetes and its complications (Lawson et al., 2016; Lee et al., 2016). A study reported that Exosomes-like vesicles released by adipose tissue can act as a mode of communication between adipose tissues and macrophages and subsequent development of insulin resistance in a mouse model (Deng et al., 2009). Moreover, visceral adipose tissue (VAT) derived exosomes are also functional in the intercellular transmission of signals as they possess adipokines (Lee et al., 2015). Skeletal muscle regulates the metabolism of the whole body and the exosomes released from it can facilitate the amendments in muscle homeostasis (Aswad et al., 2014). Keeping in view the merits of exosomes, efforts are being made to flourish them as novel prognostic, and even diagnostic, biomarkers, especially for the exosomes released from biological fluids. The presence of miRNA in blood, specific for the disease and condition/stage of illness, can act as a biomarker for prediction and diagnosing the complications associated with that particular disease (Jansen et al., 2013; Wang et al., 2014; Schulte et al., 2016; O’Neill et al., 2016). Exosomes secreted by various cells and tissues emerged as a new therapeutic strategy in cardiovascular diseases (Sahoo and Losordo, 2014; Davidson et al., 2016; Ibrahim et al., 2014; Ong and Wu, 2015; Cervio et al., 2015; Huber and Holvoet, 2015; Yellon and Davidson, 2014; Emanueli et al., 2015; Zhang et al., 2017; Zhao et al., 2015; Huang et al., 2015; Lawson et al., 2016; Suzuki et al., 2016; Xitong and Xiaorong, 2016). It has been reported that after myocardial infarction, the cardiac tissue discharges many soluble chemokines, cytokines, and growth factors; induce inflammatory responses; and recruit stem and progenitor cells to accelerate the repair process (Liu et al., 2016). Exosomes are reported to be involved in cardiac repair and regeneration by communication at intercellular and tissue levels (Sahoo and Losordo, 2014; Yuan et al., 2016). The exosomes from cardiomyocyte can transfect endothelial cells, stem cells, fibroblasts, and smooth muscle cells to induce cellular changes. Exosomes derived from DCs improve cardiac function via activation of CD4 1 T lymphocytes after myocardial infarction (Liu et al., 2016). A recent study demonstrated exosomes as potential alternatives to stem cell therapy in mediating cardiac regeneration (Ong and Wu, 2015).
7.6 Exosomes in Drug and Gene Delivery
7.5 EXOSOMES AS NANOCARRIERS Nanoparticle drug delivery systems offer many advantages like their ability to load high quantities (Natasha et al., 2014), sustained release of the loaded drug, ability to load both drug along with the adjuvant for the induction of costimulation within a particular nanocarrier (Amoozgar and Goldberg, 2015; Hamdy et al., 2011). Although there has been rigorous and thorough experimentation, partial accomplishments have been developed in clinical trials owing to deficiency in safety and efficiency of the target drug to the desired cells. Different types of nanocarriers have been investigated at length for the targeted drug delivery of drugs/genes to a specific site of cancerous tissue. Nanocarriers are most-exploited in this field because they are more permeable along with the ability to accumulate into tumors passively as well as having the capability of longer retention, for example, DCs. Macrophages and the primary cells are the natural phagocytotic involved in antitumor immunity and inflammatory response. Nano-drug delivery systems are promising platforms to deliver immunomodulatory compounds to these cells (Amoozgar and Goldberg, 2015). Exosomes, also called biological nanocarriers, are becoming visible as a promising alternative to this new concept of drug delivery (Ha et al., 2016; Aryani and Denecke, 2016). Exosomes are secreted by many types of cells, under both normal and pathological conditions. The matter of great interest is that exosomes are secreted by tumor cells and are much more in number compared with the normal cells (Thery et al., 2002b). RNA carried by exosomes could be exploited as drug and gene delivery systems; however, donor cells should not release exosomes with any immunomodulation ability and should be stable enough for the sufficient period to carry the cargo.
7.6 EXOSOMES IN DRUG AND GENE DELIVERY About 98% of drugs cannot cross the BBB. Even the nano-drugs that can solve this problem to some extent have associated issues of nanotoxicity and rapid drug clearance by the mononuclear phagocytic system (MPS) (Pardridge, 2012). Polyethylene glycol is used to solve this problem, but decreases the drug distribution to the brain and the interaction between target cells (Peng et al., 2013). These types of complications can be handled by using exosomes as a drug delivery system as it is a body’s natural product and can be personalized to cross the BBB and increasing the drug distribution to the brain by decreasing the MPS drug clearance (Yoshida et al., 1992). The morphology of the exosomes can be exploited in a manner to facilitate drug delivery as they can be used as vehicles because of their many advantages over other drug delivery systems in practice like liposomal and nano-particulate drug delivery systems (Ferguson and Nguyen, 2016). The phenomenon of communication between cells is an integral part of the multicellular organisms for the maintenance of homeostasis. Gap junctions
201
202
CHAPTER 7 Exosome as nanocarriers in drug delivery system
connect the nearby cells and distant intercellular communication can be facilitated via hormones by sending signals through circulatory systems. EVs can provide many advantages in distant cell communication as EVs can carry a cellular cargo of lipids, proteins, receptors, and active molecules to recipient cells (Lai et al., 2013).
7.6.1 DRUG DELIVERY VEHICLES FOR SMALL MOLECULES Liposomal and the polymeric nanoparticle systems are preferred systems for drug delivery (Sercombe et al., 2015). Liposomes, a synthetic vesicle of the phospholipidic membrane, have the capacity of self-assembling in various sizes and shapes in an aqueous medium whereas polymeric nanoparticles can entrap, encapsulate, or attach drug molecules (Agrawal et al., 2014; Raemdonck et al., 2014). Both systems have been used for delivering anticancer, antifungal, and analgesic drugs. The main advantage of liposomes used as the delivery system is their stability in circulation for a longer duration without causing toxicity and, moreover, their capability of evading the immune system (Li et al., 2015; Aryani and Denecke, 2016). Polymeric nanoparticles have better stability compared to liposomes, but biocompatibility and toxicity issues are of major concern. Fig. 7.3 shows the role of exosomes as nanocarriers for the delivery of small molecules including miRNA, small interference RNA (siRNA), peptides, and drugs. Above all, exosomes come with all the features of longer duration in the circulatory system, the ability to target the specific tissue, enhanced biocompatibility
FIGURE 7.3 Exosomes as nanocarriers for drugs, siRNA, or peptide delivery. Exosomes are nanosized and stable molecules capable of targeted delivery of nucleic acids, proteins, and drugs and, therefore, can be exploited for gene therapy and drug delivery systems. siRNA, Small interference RNA.
7.6 Exosomes in Drug and Gene Delivery
along with the minimal inherent toxicity issues. Hence exosomes are preferred over previously used drug delivery systems due to their unbeatable characteristics (Turturici et al., 2014; Rani and Ritter, 2016). Extensive research has been done using exosomes as vehicles for therapeutic drug delivery. Exosome complexes with curcumin enhance efficacy and safety when administered in cancer patients (Dhillon et al., 2008). Experiments on exosomes carrying curcumin demonstrate the increased solubility, stability, bioavailability, and antiinflammatory activity both in vitro and in vivo. In in vitro experiments, when macrophages were treated with exosomal curcumin, the production of the TNF and IL-6 is less in comparison to those treated with curcumin alone, proving the antiinflammatory capability of exosomes. In lipopolysaccharide (LPS) induced a septic shock model in which CD11bþGr-1þ cells increases response to LPS in the lungs and leads to acute lung inflammation. Exosomal curcumin showed significantly longer survival compared with mice treated with curcumin alone, again verifying the fact that exosomes enhance the antiinflammatory characteristics of curcumin. Exosomes illustrate the potential of brain drug delivery across the BBB by carrying paclitaxel and doxorubicin drugs and also demonstrate the transport mechanism in a zebrafish model (Yang et al., 2015). Exosomes were isolated from the glioblastoma astrocytoma U-87 MG, endothelial bEND.3, neuroectodermal tumor PFSK-1, and glioblastoma A-172 loaded with rhodamine 123, paclitaxel, and doxorubicin. Brain tissue was examined for the presence of the rhodamine 123 fluorescence and drug distribution observed in the brain region of the zebrafish embryos suggested the ability of exosomes to deliver the drug across the BBB. Further, in subsequent experiments, the primary brain cancer model was developed and anticancer drugs loaded with or without exosomes were applied. Drugs loaded with the exosomes showed the therapeutic efficacy of the exosomes better than the drug alone. Exosomes have the potential to deliver the drugs to the brain tissue by crossing the BBB and helps tremendously in curing brain cancers and other neurological disorders (Ha et al., 2016).
7.6.2 DRUG DELIVERY VEHICLES FOR PROTEINS Exosomes can also have the ability to deliver molecules bigger in size. Mounting evidence shows that exosomes loaded with the antioxidant protein catalase was successfully delivered across the BBB resulting in an improved disease state in PD (Haney et al., 2015). Patients suffering from PD have lower levels of antioxidant enzymes like catalase and superoxide dismutase levels and cause oxidative stress along with neurodegeneration (Ambani et al., 1975; Riederer et al., 1989; Abraham et al., 2005). Catalase can neutralize one million free radicals per second per molecule in a single catalytic reaction cycle (Haney et al., 2015), but the delivery of catalase across the BBB is a big task. Exosomes have undertaken this tedious task and fulfilled it efficiently, so are a promising option in PD therapy.
203
204
CHAPTER 7 Exosome as nanocarriers in drug delivery system
7.6.3 DRUG DELIVERY VEHICLES FOR NUCLEIC ACIDS Exosomes are exceptionally excellent delivery systems which can also carry DNA and RNA-like big and heavy molecules to the target cells, thereby instigating genetic modification in the biological processes. This strategy can be employed as a promising tool for delivering genetic material and altering the gene expression in certain diseases and help improve gene therapy.
7.6.4 SMALL INTERFERENCE RNA siRNA is a molecule with low stability and it degrades quickly. It can be used to change gene expression and can be very useful in gene therapy to alter pathological symptoms. Exosomes can deliver siRNA to the cell of interest (Alvarez-Erviti et al., 2011; Wahlgren et al., 2016). A recent study explored the ability of exosomes to transfer nucleic acids to cells in a specific manner using elf exosomes loaded with chemically modified siRNAs and displaying specific targeting molecules (Alvarez-Erviti et al., 2011). The DCs were modified to express LAMP-2b fused to the central nervous system-specific RVG peptide that specifically binds to the acetylcholine receptor. Exosomes were loaded with exogenous siRNA by electroporation and, after in vitro experiments, were injected into mice together with a series of control exosome preparations. The injection of RVG exosomes resulted in a significant knockdown of GAPDH mRNA in the brain regions expressing the target of the RVG ligand-nicotinic acetylcholine receptors. These results not only showed the therapeutic potential of RVG exosome technology for new therapeutic approaches against neurodegenerative diseases, but also strongly supported the use of implemented and innovative exosome technologies for targeting therapies for a number of diseases. The siRNA delivery through exosomes has numeral benefits over typical viruses, lipid nanocarriers, and polycationic delivery mediators currently in practice (van den Boorn et al., 2011). Most importantly, exosomes deliver their load straight into the cytosol, evading the requirement for endosomal escape, while their inertness circumvents clearance in the extracellular environment (van den Boorn et al., 2013). Many studies reported exosomes as an efficient therapeutic vehicle for exogenous and to T-cells and monocytes (Wahlgren et al., 2012). There is no other vehicle available for delivering genetic material, but exosomes proved to be safe, efficient, and target specific. Electroporation was found to be the best method for successful introduction of siRNA (Wahlgren et al., 2016). Exosomes with siRNA in recipient cells showed a decrease in MAPK1 expression indicating downregulation of the specific gene and successful gene silencing. Exosomes delivered siRNA against RAD51, a eukaryotic gene protein which assists in DNA repair and stops the proliferation of cancer cells (Shtam et al., 2013). Recent studies indicated that exosomes could deliver the exogenous molecules to the cells with all the functions still intact (Banizs et al., 2014).
7.6 Exosomes in Drug and Gene Delivery
7.6.5 MICRORNA miRNAs are noncoding RNAs with nonprotein nucleotides and are present in eukaryotic cells and considered as key functional elements. The exosomes are natural carriers of miRNA that could be exploited as an RNA drug delivery system. miRNA-containing exosomes are found in a variety of body fluids and cell regulate gene expression in the target cells (Kosaka et al., 2010; Thery, 2011). miRNA bind to the complementary sequences on the mRNA and function to control the posttranslational gene expression (Bartel, 2004, 2009). A recent study showed that exosomes can efficiently deliver miRNA to epidermal growth factor receptor (EGFR)-expressing breast cancer cells (Ohno et al., 2013). The epidermal growth factor and EGFR-specific peptide (GE11) were incorporated onto the surfaces of exosomes that carried let-7a in order to deliver let-7a to EGFRexpressing cancer tissue. Mounting evidence suggests that miRNA, like let-7a, functions as a tumor suppressor and prevents the development of cancer by reducing RAS and high-mobility group AT-hook protein (HMGA2) expression (Ohno et al., 2013). In this study, modified exosomes with GE11 peptide or EGF on their surfaces delivered miRNA to EGFR-expressing cancer tissues; intravenously injected exosomes targeting EGFR delivered let-7a specifically to xenograft breast cancer cells in RAG2 2 / 2 mice. These data indicate that exosomes targeted to EGFR-expressing cells may provide a platform for miRNA replacement therapies in the treatment of various cancers. Numerous human tumors of epithelial origin display elevated EGFR expression, suggesting that EGFR could serve as a receptor target in cancer drug delivery systems (Gazdar, 2010; HarichandHerdt and Ramalingam, 2008; Huang and Harari, 1999; Woodburn, 1999). Table 7.1 shows the therapeutic cargo molecules loaded into exosomes in cancer (Johnsen et al., 2014). Recent studies also suggested exosomal miRNA as a biomarker of AD (Lugli et al., 2015; Cheng et al., 2015) and a variety of cancers including acute myeloid leukemia, colorectal cancer, and hepatocellular carcinoma (Hornick et al., 2015; Matsumura et al., 2015; Ogata-Kawata et al., 2014; Sugimachi et al., 2015). Among proteins, lipids, DNA, and RNA, the miRNA is found in higher concentrations (Goldie et al., 2014) and could be transferred between cells via exosomes (Valadi et al., 2007). Exosomes carrying miRNA released from the cells circulate and can transfer it to distant cells where the miRNA becomes functional in the recipient cells. Some studies done in both immune cells and other cell types demonstrated that transferred miRNAs inhibit target mRNAs in recipient cells (Montecalvo et al., 2012; Kosaka et al., 2010; Mittelbrunn et al., 2011; Katakowski et al., 2010; Alexander et al., 2015). A decade of research has identified many exosomal miRNAs which are involved in the diagnosis, therapeutics, and pathophysiology of many diseases including cancer, neurodegenerative diseases, metabolic disorders, cardiovascular diseases, and immune disorders.
205
206
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Table 7.1 Various Therapeutic Cargo Molecules Loaded into Exosomes for Targeted Cancer Therapy Study
Cargo
Interfering RNAs Shtam et al. (2013) Wahlgren et al. (2012) Alvarez-Erviti et al. (2011) Pan et al. (2012) Chen et al. (2014) Bryniarski et al. (2013) Zhang et al. (2010) Katakowski et al. (2013) Kosaka et al. (2012) Pan et al. (2012) Xin et al. (2012) Ohno et al. (2013) Munoz et al. (2013)
siRNA against RAD51 and RAD52 MAPK1 siRNA GAPDH siRNA and BACE1 siRNA shNS5b, shCD81 miR-214 miR-150 miR-150 miR-146b miR-143 miR-122 miR-133b Let-7a Cy5-anti-miR-9
Other Types of Therapeutic Cargo Tian et al. (2014) Jang et al. (2013) Hood et al. (2014) Mizrak et al. (2013) Maguire et al. (2012) Zhuang et al. (2011) Sun et al. (2010)
Doxorubicin Doxorubicin SPION5 CD fused with UPRT and EGFP Adeno-associated viral vector Curcumin and JSI-124 Curcumin
CD, Cytosine deaminase; EGFP, enhanced green fluorescence protein; siRNA, small interference RNA; SPION, superparamagnetic iron oxide nanoparticles; UPRT, uracil phosphoribosyltransferase.
7.7 CONCLUDING REMARKS AND FUTURE PERSPECTIVES Exosomes are nanosized vesicles secreted by a variety of cells and have ideal features as nanocarriers for drug and gene delivery. Exosomes are secreted by a variety of cells and body fluids including serum, plasma, urine, semen, breast milk, sweat, synovial fluids, etc. They are composed of a lipid bilayer, proteins, DNA, RNA, enzymes, and many other essential molecules. Due to their normal biological functioning in the cell, exosomes help cells to regulate other cells at a distance and communicate with them. Exosomes are known to be associated with diverse pathologies and represent a variety of functions like delivering biomolecules such as mRNAs, miRNAs, siRNA, proteins, and drugs to the recipient cells by communication at intercellular and tissue levels. Exosomes display a wide variety of functions in the cell including cell cell communication, diagnostic markers, and their potential role in therapeutics. Some exosomes possess
References
immunostimulatory or immunosuppressive effects which can be used as immunotherapies for cancer or autoimmune diseases. Primary studies have also reported the successful delivery of chemotherapeutic drug-loaded exosome mimetic MVs to tumor tissue in vivo and in vitro. Furthermore, exosomes are involved in various pathologic conditions including AD, PD, and metabolic disorders. Emerging evidence has revealed the promising therapeutic potential of exosomes as effective nano-vaccines and immunological treatment for inflammatory diseases. Exosomes are also known to play important role as diagnostic biomarkers of various diseases. In addition, they are considered in many clinical studies as a delivery system to transport RNA, protein, and drug molecules as they can easily cross the BBB. However, developing a strategy to produce exosomes with desirable components and less undesirable effects may have great potential for clinical application. More research is required to fully illucidate the use of exosomes in diagnostic and therapeutic measures.
CONFLICT OF INTEREST None declared.
ACKNOWLEDGMENTS JSB and GKB were financially supported by University Grant Commission, New Delhi, India under Raman Post-Doctoral Research Fellowship in United States [F.No. 5-82/2016 (IC)] and Rajiv Gandhi Post Doc Fellowship (PDFSS-2011-12-SC-CHA-3405), respectively.
REFERENCES Abraham, S., Soundararajan, C.C., Vivekanandhan, S., Behari, M., 2005. Erythrocyte antioxidant enzymes in Parkinson’s disease. Indian J. Med. Res. 121, 111 115. Admyre, C., Johansson, S.M., Paulie, S., Gabrielsson, S., 2006. Direct exosome stimulation of peripheral human T cells detected by ELISPOT. Eur. J. Immunol. 36, 1772 1781. Admyre, C., Johansson, S.M., Qazi, K.R., Filen, J.J., Lahesmaa, R., Norman, M., et al., 2007. Exosomes with immune modulatory features are present in human breast milk. J. Immunol. 179, 1969 1978. Agrawal, U., Sharma, R., Gupta, M., Vyas, S.P., 2014. Is nanotechnology a boon for oral drug delivery? Drug Discov. Today 19, 1530 1546. Aguzzi, A., Rajendran, L., 2009. The transcellular spread of cytosolic amyloids, prions, and prionoids. Neuron 64, 783 790. Akers, J.C., Gonda, D., Kim, R., Carter, B.S., Chen, C.C., 2013. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neurooncol. 113, 1 11.
207
208
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Alexander, M., Hu, R., Runtsch, M.C., Kagele, D.A., Mosbruger, T.L., Tolmachova, T., et al., 2015. Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat. Commun. 6, 7321. Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., Wood, M.J., 2011. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 29, 341 345. Ambani, L.M., Van woert, M.H., Murphy, S., 1975. Brain peroxidase and catalase in Parkinson disease. Arch. Neurol. 32, 114 118. Amoozgar, Z., Goldberg, M.S., 2015. Targeting myeloid cells using nanoparticles to improve cancer immunotherapy. Adv. Drug Deliv. Rev. 91, 38 51. Aoki, N., Jin-No, S., Nakagawa, Y., Asai, N., Arakawa, E., Tamura, N., et al., 2007. Identification and characterization of microvesicles secreted by 3T3-L1 adipocytes: redox- and hormone-dependent induction of milk fat globule-epidermal growth factor 8-associated microvesicles. Endocrinology 148, 3850 3862. Aryani, A., Denecke, B., 2016. Exosomes as a nanodelivery system: a key to the future of neuromedicine? Mol. Neurobiol. 53, 818 834. Aswad, H., Forterre, A., Wiklander, O.P., Vial, G., Danty-Berger, E., Jalabert, A., et al., 2014. Exosomes participate in the alteration of muscle homeostasis during lipidinduced insulin resistance in mice. Diabetologia 57, 2155 2164. Banizs, A.B., Huang, T., Dryden, K., Berr, S.S., Stone, J.R., Nakamoto, R.K., et al., 2014. In vitro evaluation of endothelial exosomes as carriers for small interfering ribonucleic acid delivery. Int. J. Nanomed. 9, 4223 4230. Bartel, D.P., 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281 297. Bartel, D.P., 2009. MicroRNAs: target recognition and regulatory functions. Cell 136, 215 233. Basu, J., Ludlow, J.W., 2016. Exosomes for repair, regeneration and rejuvenation. Expert Opin. Biol. Ther. 16, 489 506. Bellingham, S.A., Guo, B.B., Coleman, B.M., Hill, A.F., 2012. Exosomes: vehicles for the transfer of toxic proteins associated with neurodegenerative diseases? Front. Physiol. 3, 124. Bhatnagar, S., Schorey, J.S., 2007. Exosomes released from infected macrophages contain Mycobacterium avium glycopeptidolipids and are proinflammatory. J. Biol. Chem. 282, 25779 25789. Bhatnagar, S., Shinagawa, K., Castellino, F.J., Schorey, J.S., 2007. Exosomes released from macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro and in vivo. Blood 110, 3234 3244. Bhatti, G.K., Bhadada, S.K., Vijayvergiya, R., Mastana, S.S., Bhatti, J.S., 2016. Metabolic syndrome and risk of major coronary events among the urban diabetic patients: North Indian Diabetes and Cardiovascular Disease Study-NIDCVD-2. J. Diabet. Complications 30, 72 78. Biomarkers Definitions working, G, 2001. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 69, 89 95. Bobrie, A., Colombo, M., Raposo, G., Thery, C., 2011. Exosome secretion: molecular mechanisms and roles in immune responses. Traffic 12, 1659 1668. Braak, H., Rub, U., Gai, W.P., Del tredici, K., 2003. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm. (Vienna) 110, 517 536.
References
Brettschneider, J., Del tredici, K., Lee, V.M., Trojanowski, J.Q., 2015. Spreading of pathology in neurodegenerative diseases: a focus on human studies. Nat. Rev. Neurosci. 16, 109 120. Bryniarski, K., Ptak, W., Jayakumar, A., Pullmann, K., Caplan, M.J., Chairoungdua, A., et al., 2013. Antigen-specific, antibody-coated, exosome-like nanovesicles deliver suppressor T-cell microRNA-150 to effector T cells to inhibit contact sensitivity. J. Allergy Clin. Immunol. 132, 170 181. Buzas, E.I., Gyorgy, B., Nagy, G., Falus, A., Gay, S., 2014. Emerging role of extracellular vesicles in inflammatory diseases. Nat. Rev. Rheumatol. 10, 356 364. Cai, Z., Yang, F., Yu, L., Yu, Z., Jiang, L., Wang, Q., et al., 2012a. Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J. Immunol. 188, 5954 5961. Cai, Z., Zhang, W., Yang, F., Yu, L., Yu, Z., Pan, J., et al., 2012b. Immunosuppressive exosomes from TGF-beta1 gene-modified dendritic cells attenuate Th17-mediated inflammatory autoimmune disease by inducing regulatory T cells. Cell Res. 22, 607 610. Cervio, E., Barile, L., Moccetti, T., Vassalli, G., 2015. Exosomes for intramyocardial intercellular communication. Stem Cells Int. 2015, 482171. Chaput, N., Schartz, N.E., Andre, F., Zitvogel, L., 2003. Exosomes for immunotherapy of cancer. Adv. Exp. Med. Biol. 532, 215 221. Chaput, N., Flament, C., Viaud, S., Taieb, J., Roux, S., Spatz, A., et al., 2006. Dendritic cell derived-exosomes: biology and clinical implementations. J. Leukoc. Biol. 80, 471 478. Chaudhuri, A.A., So, A.Y., Sinha, N., Gibson, W.S., Taganov, K.D., O’connell, R.M., et al., 2011. MicroRNA-125b potentiates macrophage activation. J. Immunol. 187, 5062 5068. Chen, L., Charrier, A., Zhou, Y., Chen, R., Yu, B., Agarwal, K., et al., 2014. Epigenetic regulation of connective tissue growth factor by microRNA-214 delivery in exosomes from mouse or human hepatic stellate cells. Hepatology 59, 1118 1129. Cheng, L., Doecke, J.D., Sharples, R.A., Villemagne, V.L., Fowler, C.J., Rembach, A., et al., 2015. Prognostic serum miRNA biomarkers associated with Alzheimer’s disease shows concordance with neuropsychological and neuroimaging assessment. Mol. Psychiatry 20, 1188 1196. Chivet, M., Hemming, F., Pernet-Gallay, K., Fraboulet, S., Sadoul, R., 2012. Emerging role of neuronal exosomes in the central nervous system. Front. Physiol. 3, 145. Cho, J.A., Park, H., Lim, E.H., Lee, K.W., 2012. Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int. J. Oncol. 40, 130 138. Chow, A., Zhou, W., Liu, L., Fong, M.Y., Champer, J., Van haute, D., et al., 2014. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-kappaB. Sci. Rep. 4, 5750. Cocucci, E., Racchetti, G., Podini, P., Meldolesi, J., 2007. Enlargeosome traffic: exocytosis triggered by various signals is followed by endocytosis, membrane shedding or both. Traffic 8, 742 757. Conde-Vancells, J., Rodriguez-Suarez, E., Embade, N., Gil, D., Matthiesen, R., Valle, M., et al., 2008. Characterization and comprehensive proteome profiling of exosomes secreted by hepatocytes. J. Proteome Res. 7, 5157 5166.
209
210
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Costa-Silva, B., Aiello, N.M., Ocean, A.J., Singh, S., Zhang, H., Thakur, B.K., et al., 2015. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17, 816 826. Dai, S., Zhou, X., Wang, B., Wang, Q., Fu, Y., Chen, T., et al., 2006. Enhanced induction of dendritic cell maturation and HLA-A 0201-restricted CEA-specific CD8(1) CTL response by exosomes derived from IL-18 gene-modified CEA-positive tumor cells. J. Mol. Med. (Berl.) 84, 1067 1076. Davidson, S.M., Takov, K., Yellon, D.M., 2016. Exosomes and cardiovascular protection. Cardiovasc. Drugs Ther. Delcayre, A., Shu, H., Le pecq, J.B., 2005. Dendritic cell-derived exosomes in cancer immunotherapy: exploiting nature’s antigen delivery pathway. Expert Rev. Anticancer Ther. 5, 537 547. Deng, Z.B., Poliakov, A., Hardy, R.W., Clements, R., Liu, C., Liu, Y., et al., 2009. Adipose tissue exosome-like vesicles mediate activation of macrophage-induced insulin resistance. Diabetes 58, 2498 2505. Dhillon, N., Aggarwal, B.B., Newman, R.A., Wolff, R.A., Kunnumakkara, A.B., Abbruzzese, J.L., et al., 2008. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin. Cancer Res. 14, 4491 4499. Diamant, M., Nieuwland, R., Pablo, R.F., Sturk, A., Smit, J.W., Radder, J.K., 2002. Elevated numbers of tissue-factor exposing microparticles correlate with components of the metabolic syndrome in uncomplicated type 2 diabetes mellitus. Circulation 106, 2442 2447. Eken, C., Gasser, O., Zenhaeusern, G., Oehri, I., Hess, C., Schifferli, J.A., 2008. Polymorphonuclear neutrophil-derived ectosomes interfere with the maturation of monocyte-derived dendritic cells. J. Immunol. 180, 817 824. EL Andaloussi, S., Mager, I., Breakefield, X.O., Wood, M.J., 2013. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 347 357. Emanueli, C., Shearn, A.I., Angelini, G.D., Sahoo, S., 2015. Exosomes and exosomal miRNAs in cardiovascular protection and repair. Vascul. Pharmacol. 71, 24 30. Emmanouilidou, E., Melachroinou, K., Roumeliotis, T., Garbis, S.D., Ntzouni, M., Margaritis, L.H., et al., 2010. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 30, 6838 6851. Feng, B., Chen, Y., Luo, Y., Chen, M., Li, X., Ni, Y., 2010. Circulating level of microparticles and their correlation with arterial elasticity and endothelium-dependent dilation in patients with type 2 diabetes mellitus. Atherosclerosis 208, 264 269. Ferguson, S.W., Nguyen, J., 2016. Exosomes as therapeutics: the implications of molecular composition and exosomal heterogeneity. J. Controlled Release 228, 179 190. Fevrier, B., Vilette, D., Archer, F., Loew, D., Faigle, W., Vidal, M., et al., 2004. Cells release prions in association with exosomes. Proc. Natl. Acad. Sci. U.S.A. 101, 9683 9688. Gastpar, R., Gehrmann, M., Bausero, M.A., Asea, A., Gross, C., Schroeder, J.A., et al., 2005. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Cancer Res. 65, 5238 5247. Gazdar, A.F., 2010. Epidermal growth factor receptor inhibition in lung cancer: the evolving role of individualized therapy. Cancer Metastasis Rev. 29, 37 48. Goldie, B.J., Dun, M.D., Lin, M., Smith, N.D., Verrills, N.M., Dayas, C.V., et al., 2014. Activity-associated miRNA are packaged in Map1b-enriched exosomes released from depolarized neurons. Nucleic Acids Res. 42, 9195 9208. Gousset, K., Schiff, E., Langevin, C., Marijanovic, Z., Caputo, A., Browman, D.T., et al., 2009. Prions hijack tunnelling nanotubes for intercellular spread. Nat. Cell Biol. 11, 328 336.
References
Gross, J.C., Chaudhary, V., Bartscherer, K., Boutros, M., 2012. Active Wnt proteins are secreted on exosomes. Nat. Cell Biol. 14, 1036 1045. Gupta, A., Pulliam, L., 2014. Exosomes as mediators of neuroinflammation. J. Neuroinflammation 11, 68. Ha, D., Yang, N., Nadithe, V., 2016. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm. Sin. B 6, 287 296. Hamdy, S., Haddadi, A., Hung, R.W., Lavasanifar, A., 2011. Targeting dendritic cells with nano-particulate PLGA cancer vaccine formulations. Adv. Drug Deliv. Rev. 63, 943 955. Haney, M.J., Klyachko, N.L., Zhao, Y., Gupta, R., Plotnikova, E.G., He, Z., et al., 2015. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Controlled Release 207, 18 30. Hanson, P.I., Cashikar, A., 2012. Multivesicular body morphogenesis. Annu. Rev. Cell Dev. Biol. 28, 337 362. Harding, C.V., Heuser, J.E., Stahl, P.D., 2013. Exosomes: looking back three decades and into the future. J. Cell Biol. 200, 367 371. Harichand-Herdt, S., Ramalingam, S.S., 2008. Targeted therapy for the treatment of non-small cell lung cancer: focus on inhibition of epidermal growth factor receptor. Semin. Thorac. Cardiovasc. Surg. 20, 217 223. Hood, J.L., Scott, M.J., Wickline, S.A., 2014. Maximizing exosome colloidal stability following electroporation. Anal. Biochem. 448, 41 49. Hornick, N.I., Huan, J., Doron, B., Goloviznina, N.A., Lapidus, J., Chang, B.H., et al., 2015. Serum exosome microRNA as a minimally-invasive early biomarker of AML. Sci. Rep. 5, 11295. Hoshino, A., Costa-Silva, B., Shen, T.L., Rodrigues, G., Hashimoto, A., Tesic mark, M., et al., 2015. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329 335. Howitt, J., Hill, A.F., 2016. Exosomes in the pathology of neurodegenerative diseases. J. Biol. Chem. 291, 26589 26597. Hristov, M., Erl, W., Linder, S., Weber, P.C., 2004. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood 104, 2761 2766. Huang, L., Ma, W., Ma, Y., Feng, D., Chen, H., Cai, B., 2015. Exosomes in mesenchymal stem cells, a new therapeutic strategy for cardiovascular diseases? Int. J. Biol. Sci. 11, 238 245. Huang, S.M., Harari, P.M., 1999. Epidermal growth factor receptor inhibition in cancer therapy: biology, rationale and preliminary clinical results. Invest. New Drugs 17, 259 269. Huber, H.J., Holvoet, P., 2015. Exosomes: emerging roles in communication between blood cells and vascular tissues during atherosclerosis. Curr. Opin. Lipidol. 26, 412 419. Huotari, J., Helenius, A., 2011. Endosome maturation. EMBO J. 30, 3481 3500. Ibrahim, A.G., Cheng, K., Marban, E., 2014. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep. 2, 606 619. Jalabert, A., Vial, G., Guay, C., Wiklander, O.P., Nordin, J.Z., Aswad, H., et al., 2016. Exosome-like vesicles released from lipid-induced insulin-resistant muscles modulate gene expression and proliferation of beta recipient cells in mice. Diabetologia 59, 1049 1058.
211
212
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Jang, S.C., Kim, O.Y., Yoon, C.M., Choi, D.S., Roh, T.Y., Park, J., et al., 2013. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano 7, 7698 7710. Jansen, F., Yang, X., Hoelscher, M., Cattelan, A., Schmitz, T., Proebsting, S., et al., 2013. Endothelial microparticle-mediated transfer of microRNA-126 promotes vascular endothelial cell repair via SPRED1 and is abrogated in glucose-damaged endothelial microparticles. Circulation 128, 2026 2038. Jarmalaviciute, A., Pivoriunas, A., 2016. Exosomes as a potential novel therapeutic tools against neurodegenerative diseases. Pharmacol. Res. 113, 816 822. Johnsen, K.B., Gudbergsson, J.M., Skov, M.N., Pilgaard, L., Moos, T., Duroux, M., 2014. A comprehensive overview of exosomes as drug delivery vehicles—endogenous nanocarriers for targeted cancer therapy. Biochim. Biophys. Acta 1846, 75 87. Johnstone, R.M., Adam, M., Hammond, J.R., Orr, L., Turbide, C., 1987. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 262, 9412 9420. Johnstone, R.M., Mathew, A., Mason, A.B., Teng, K., 1991. Exosome formation during maturation of mammalian and avian reticulocytes: evidence that exosome release is a major route for externalization of obsolete membrane proteins. J. Cell Physiol. 147, 27 36. Kalra, H., Simpson, R.J., Ji, Hong, Aikawa, E., Altevogt, P., Askenase, P., et al., 2012. Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation. PLoS Biol. 10, e1001450. Kanu, N., Imokawa, Y., Drechsel, D.N., Williamson, R.A., Birkett, C.R., Bostock, C.J., et al., 2002. Transfer of scrapie prion infectivity by cell contact in culture. Curr. Biol. 12, 523 530. Katakowski, M., Buller, B., Wang, X., Rogers, T., Chopp, M., 2010. Functional microRNA is transferred between glioma cells. Cancer Res. 70, 8259 8263. Katakowski, M., Buller, B., Zheng, X., Lu, Y., Rogers, T., Osobamiro, O., et al., 2013. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 335, 201 204. Keller, S., Sanderson, M.P., Stoeck, A., Altevogt, P., 2006. Exosomes: from biogenesis and secretion to biological function. Immunol. Lett. 107, 102 108. Keller, S., Ridinger, J., Rupp, A.K., Janssen, J.W., Altevogt, P., 2011. Body fluid derived exosomes as a novel template for clinical diagnostics. J. Transl. Med. 9, 86. Kim, S.H., Lechman, E.R., Bianco, N., Menon, R., Keravala, A., Nash, J., et al., 2005. Exosomes derived from IL-10-treated dendritic cells can suppress inflammation and collagen-induced arthritis. J. Immunol. 174, 6440 6448. Kim, S.H., Bianco, N.R., Shufesky, W.J., Morelli, A.E., Robbins, P.D., 2007. Effective treatment of inflammatory disease models with exosomes derived from dendritic cells genetically modified to express IL-4. J. Immunol. 179, 2242 2249. Kim, S.J., Moon, G.J., Cho, Y.H., Kang, H.Y., Hyung, N.K., Kim, D., et al., 2012. Circulating mesenchymal stem cells microparticles in patients with cerebrovascular disease. PLoS One 7, e37036. Kosaka, N., Iguchi, H., Yoshioka, Y., Takeshita, F., Matsuki, Y., Ochiya, T., 2010. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J. Biol. Chem. 285, 17442 17452. Kosaka, N., Iguchi, H., Yoshioka, Y., Hagiwara, K., Takeshita, F., Ochiya, T., 2012. Competitive interactions of cancer cells and normal cells via secretory microRNAs. J. Biol. Chem. 287, 1397 1405.
References
Kulshreshtha, A., Ahmad, T., Agrawal, A., Ghosh, B., 2013. Proinflammatory role of epithelial cell-derived exosomes in allergic airway inflammation. J. Allergy Clin. Immunol. 131, 1194 1203. 1203 e1 14. Lai, C.P., Breakefield, X.O., 2012. Role of exosomes/microvesicles in the nervous system and use in emerging therapies. Front. Physiol. 3, 228. Lai, R.C., Yeo, R.W., Tan, K.H., Lim, S.K., 2013. Exosomes for drug delivery—a novel application for the mesenchymal stem cell. Biotechnol. Adv. 31, 543 551. Lawson, C., Vicencio, J.M., Yellon, D.M., Davidson, S.M., 2016. Microvesicles and exosomes: new players in metabolic and cardiovascular disease. J. Endocrinol. 228, R57 R71. Lee, J.E., Moon, P.G., Lee, I.K., Baek, M.C., 2015. Proteomic analysis of extracellular vesicles released by adipocytes of Otsuka Long-Evans Tokushima fatty (OLETF) rats. Protein J. 34, 220 235. Lee, M.J., Park, D.H., Kang, J.H., 2016. Exosomes as the source of biomarkers of metabolic diseases. Ann. Pediatr. Endocrinol. Metab. 21, 119 125. Li, C., Zhang, J., Zu, Y.J., Nie, S.F., Cao, J., Wang, Q., et al., 2015. Biocompatible and biodegradable nanoparticles for enhancement of anti-cancer activities of phytochemicals. Chin. J. Nat. Med. 13, 641 652. Linares, R., Tan, S., Gounou, C., Arraud, N., Brisson, A.R., 2015. High-speed centrifugation induces aggregation of extracellular vesicles. J. Extracell. Vesicles 4, 29509. Liu, C., Yu, S., Zinn, K., Wang, J., Zhang, L., Jia, Y., et al., 2006. Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function. J. Immunol. 176, 1375 1385. Liu, H., Gao, W., Yuan, J., Wu, C., Yao, K., Zhang, L., et al., 2016. Exosomes derived from dendritic cells improve cardiac function via activation of CD4(1) T lymphocytes after myocardial infarction. J. Mol. Cell. Cardiol. 91, 123 133. Livshits, M.A., Khomyakova, E., Evtushenko, E.G., Lazarev, V.N., Kulemin, N.A., Semina, S.E., et al., 2016. Corrigendum: isolation of exosomes by differential centrifugation: theoretical analysis of a commonly used protocol. Sci. Rep. 6, 21447. Lotvall, J., Hill, A.F., Hochberg, F., Buzas, E.I., Di vizio, D., Gardiner, C., et al., 2014. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 3, 26913. Lugli, G., Cohen, A.M., Bennett, D.A., Shah, R.C., Fields, C.J., Hernandez, A.G., et al., 2015. Plasma exosomal miRNAs in persons with and without Alzheimer disease: altered expression and prospects for biomarkers. PLoS One 10, e0139233. Maguire, C.A., Balaj, L., Sivaraman, S., Crommentuijn, M.H., Ericsson, M., MinchevaNilsson, L., et al., 2012. Microvesicle-associated AAV vector as a novel gene delivery system. Mol. Ther. 20, 960 971. Mahaweni, N.M., Kaijen-Lambers, M.E., Dekkers, J., Aerts, J.G., Hegmans, J.P., 2013. Tumour-derived exosomes as antigen delivery carriers in dendritic cell-based immunotherapy for malignant mesothelioma. J. Extracell. Vesicles 2. Malm, T., Loppi, S., Kanninen, K.M., 2016. Exosomes in Alzheimer’s disease. Neurochem. Int. 97, 193 199. Matsumura, T., Sugimachi, K., Iinuma, H., Takahashi, Y., Kurashige, J., Sawada, G., et al., 2015. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br. J. Cancer 113, 275 281.
213
214
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Michael, A., Bajracharya, S.D., Yuen, P.S., Zhou, H., Star, R.A., Illei, G.G., et al., 2010. Exosomes from human saliva as a source of microRNA biomarkers. Oral Dis. 16, 34 38. Mittelbrunn, M., Gutierrez-Vazquez, C., Villarroya-Beltri, C., Gonzalez, S., Sanchez-Cabo, F., Gonzalez, M.A., et al., 2011. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat. Commun. 2, 282. Mizrak, A., Bolukbasi, M.F., Ozdener, G.B., Brenner, G.J., Madlener, S., Erkan, E.P., et al., 2013. Genetically engineered microvesicles carrying suicide mRNA/protein inhibit schwannoma tumor growth. Mol. Ther. 21, 101 108. Montecalvo, A., Shufesky, W.J., Stolz, D.B., Sullivan, M.G., Wang, Z., Divito, S.J., et al., 2008. Exosomes as a short-range mechanism to spread alloantigen between dendritic cells during T cell allorecognition. J. Immunol. 180, 3081 3090. Montecalvo, A., Larregina, A.T., Shufesky, W.J., Stolz, D.B., Sullivan, M.L., Karlsson, J. M., et al., 2012. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood 119, 756 766. Munich, S., Sobo-Vujanovic, A., Buchser, W.J., Beer-Stolz, D., Vujanovic, N.L., 2012. Dendritic cell exosomes directly kill tumor cells and activate natural killer cells via TNF superfamily ligands. Oncoimmunology 1, 1074 1083. Munoz, J.L., Bliss, S.A., Greco, S.J., Ramkissoon, S.H., Ligon, K.L., Rameshwar, P., 2013. Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol. Ther. Nucleic Acids 2, e126. Natasha, G., Gundogan, B., Tan, A., Farhatnia, Y., Wu, W., Rajadas, J., et al., 2014. Exosomes as immunotheranostic nanoparticles. Clin. Ther. 36, 820 829. O’neill, S., Bohl, M., Gregersen, S., Hermansen, K., O’driscoll, L., 2016. Blood-based biomarkers for metabolic syndrome. Trends Endocrinol. Metab. 27, 363 374. Ogata-Kawata, H., Izumiya, M., Kurioka, D., Honma, Y., Yamada, Y., Furuta, K., et al., 2014. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One 9, e92921. Ohno, S., Takanashi, M., Sudo, K., Ueda, S., Ishikawa, A., Matsuyama, N., et al., 2013. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21, 185 191. Ong, S.G., Wu, J.C., 2015. Exosomes as potential alternatives to stem cell therapy in mediating cardiac regeneration. Circ. Res. 117, 7 9. Ostrowski, M., Carmo, N.B., Krumeich, S., Fanget, I., Raposo, G., Savina, A., et al., 2010. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 12, 19 30. Suppl. 1 13. Palma, J., Yaddanapudi, S.C., Pigati, L., Havens, M.A., Jeong, S., Weiner, G.A., et al., 2012. MicroRNAs are exported from malignant cells in customized particles. Nucleic Acids Res. 40, 9125 9138. Pan, Q., Ramakrishnaiah, V., Henry, S., Fouraschen, S., De ruiter, P.E., Kwekkeboom, J., et al., 2012. Hepatic cell-to-cell transmission of small silencing RNA can extend the therapeutic reach of RNA interference (RNAi). Gut 61, 1330 1339. Pardridge, W.M., 2012. Drug transport across the blood brain barrier. J. Cereb. Blood Flow Metab. 32, 1959 1972. Penfornis, P., Vallabhaneni, K.C., Whitt, J., Pochampally, R., 2016. Extracellular vesicles as carriers of microRNA, proteins and lipids in tumor microenvironment. Int. J. Cancer 138, 14 21.
References
Peng, Q., Zhang, S., Yang, Q., Zhang, T., Wei, X.Q., Jiang, L., et al., 2013. Preformed albumin corona, a protective coating for nanoparticles based drug delivery system. Biomaterials 34, 8521 8530. Pisitkun, T., Johnstone, R., Knepper, M.A., 2006. Discovery of urinary biomarkers. Mol. Cell Proteomics 5, 1760 1771. Pitt, J.M., Charrier, M., Viaud, S., Andre, F., Besse, B., Chaput, N., et al., 2014. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J. Immunol. 193, 1006 1011. Pitt, J.M., Andre, F., Amigorena, S., Soria, J.C., Eggermont, A., Kroemer, G., et al., 2016. Dendritic cell-derived exosomes for cancer therapy. J. Clin. Invest. 126, 1224 1232. Prusiner, S.B., Woerman, A.L., Mordes, D.A., Watts, J.C., Rampersaud, R., Berry, D.B., et al., 2015. Evidence for alpha-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proc. Natl. Acad. Sci. U.S.A. 112, E5308 E5317. Raemdonck, K., Braeckmans, K., Demeester, J., De smedt, S.C., 2014. Merging the best of both worlds: hybrid lipid-enveloped matrix nanocomposites in drug delivery. Chem. Soc. Rev. 43, 444 472. Raj, R., Bhatti, J.S., Badada, S.K., Ramteke, P.W., 2014. Genetic basis of dyslipidemia in disease precipitation of coronary artery disease (CAD) associated type 2 diabetes mellitus (T2DM). Diabetes Metab. Res. Rev. Rajendran, L., Honsho, M., Zahn, T.R., Keller, P., Geiger, K.D., Verkade, P., et al., 2006. Alzheimer’s disease beta-amyloid peptides are released in association with exosomes. Proc. Natl. Acad. Sci. U.S.A. 103, 11172 11177. Rak, J., Guha, A., 2012. Extracellular vesicles—vehicles that spread cancer genes. Bioessays 34, 489 497. Rani, S., Ritter, T., 2016. The exosome—a naturally secreted nanoparticle and its application to wound healing. Adv. Mater. 28, 5542 5552. Raposo, G., Stoorvogel, W., 2013. Extracellular vesicles: exosomes, microvesicles, and friends. J. Cell Biol. 200, 373 383. Raposo, G., Nijman, H.W., Stoorvogel, W., Liejendekker, R., Harding, C.V., Melief, C.J., et al., 1996. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 183, 1161 1172. Riederer, P., Sofic, E., Rausch, W.D., Schmidt, B., Reynolds, G.P., Jellinger, K., et al., 1989. Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J. Neurochem. 52, 515 520. Ristorcelli, E., Beraud, E., Verrando, P., Villard, C., Lafitte, D., Sbarra, V., et al., 2008. Human tumor nanoparticles induce apoptosis of pancreatic cancer cells. FASEB J. 22, 3358 3369. Romagnoli, G.G., Zelante, B.B., Toniolo, P.A., Migliori, I.K., Barbuto, J.A., 2014. Dendritic cell-derived exosomes may be a tool for cancer immunotherapy by converting tumor cells into immunogenic targets. Front. Immunol. 5, 692. Russo, I., Bubacco, L., Greggio, E., 2012. Exosomes-associated neurodegeneration and progression of Parkinson’s disease. Am. J. Neurodegener. Dis. 1, 217 225. Safdar, A., Saleem, A., Tarnopolsky, M.A., 2016. The potential of endurance exercisederived exosomes to treat metabolic diseases. Nat. Rev. Endocrinol. 12, 504 517. Sahoo, S., Losordo, D.W., 2014. Exosomes and cardiac repair after myocardial infarction. Circ. Res. 114, 333 344. Salido-Guadarrama, I., Romero-Cordoba, S., Peralta-Zaragoza, O., Hidalgo-Miranda, A., Rodriguez-Dorantes, M., 2014. MicroRNAs transported by exosomes in body fluids as mediators of intercellular communication in cancer. Onco Targets Ther. 7, 1327 1338.
215
216
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Schulte, E.C., Altmaier, E., Berger, H.S., Do, K.T., Kastenmuller, G., Wahl, S., et al., 2016. Alterations in lipid and inositol metabolisms in two dopaminergic disorders. PLoS One 11, e0147129. Scott, A.M., Wolchok, J.D., Old, L.J., 2012. Antibody therapy of cancer. Nat. Rev. Cancer 12, 278 287. Segura, E., Nicco, C., Lombard, B., Veron, P., Raposo, G., Batteux, F., et al., 2005. ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming. Blood 106, 216 223. Sercombe, L., Veerati, T., Moheimani, F., Wu, S.Y., Sood, A.K., Hua, S., 2015. Advances and challenges of liposome assisted drug delivery. Front. Pharmacol. 6, 286. Shtam, T.A., Kovalev, R.A., Varfolomeeva, E.Y., Makarov, E.M., Kil, Y.V., Filatov, M.V., 2013. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun. Signal 11, 88. Simhadri, V.R., Reiners, K.S., Hansen, H.P., Topolar, D., Simhadri, V.L., Nohroudi, K., et al., 2008. Dendritic cells release HLA-B-associated transcript-3 positive exosomes to regulate natural killer function. PLoS One 3, e3377. Simpson, R.J., Lim, J.W., Moritz, R.L., Mathivanan, S., 2009. Exosomes: proteomic insights and diagnostic potential. Expert Rev. Proteomics 6, 267 283. Singh, P.P., Lemaire, C., Tan, J.C., Zeng, E., Schorey, J.S., 2011. Exosomes released from M. tuberculosis infected cells can suppress IFN-gamma mediated activation of naive macrophages. PLoS One 6, e18564. Skokos, D., Botros, H.G., Demeure, C., Morin, J., Peronet, R., Birkenmeier, G., et al., 2003. Mast cell-derived exosomes induce phenotypic and functional maturation of dendritic cells and elicit specific immune responses in vivo. J. Immunol. 170, 3037 3045. Skriner, K., Adolph, K., Jungblut, P.R., Burmester, G.R., 2006. Association of citrullinated proteins with synovial exosomes. Arthritis Rheum. 54, 3809 3814. Soekmadji, C., Russell, P.J., Nelson, C.C., 2013. Exosomes in prostate cancer: putting together the pieces of a puzzle. Cancers (Basel) 5, 1522 1544. Street, J.M., Barran, P.E., Mackay, C.L., Weidt, S., Balmforth, C., Walsh, T.S., et al., 2012. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J. Transl. Med. 10, 5. Sugimachi, K., Matsumura, T., Hirata, H., Uchi, R., Ueda, M., Ueo, H., et al., 2015. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br. J. Cancer 112, 532 538. Sun, D., Zhuang, X., Xiang, X., Liu, Y., Zhang, S., Liu, C., et al., 2010. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. 18, 1606 1614. Suzuki, E., Fujita, D., Takahashi, M., Oba, S., Nishimatsu, H., 2016. Stem cell-derived exosomes as a therapeutic tool for cardiovascular disease. World J. Stem Cells 8, 297 305. Tan, A., De Lapena, H., Seifalian, A.M., 2010. The application of exosomes as a nanoscale cancer vaccine. Int. J. Nanomed. 5, 889 900. Thery, C., 2011. Exosomes: secreted vesicles and intercellular communications. F1000 Biol. Rep. 3, 15. Thery, C., Duban, L., Segura, E., Veron, P., Lantz, O., Amigorena, S., 2002a. Indirect activation of naive CD4 1 T cells by dendritic cell-derived exosomes. Nat. Immunol. 3, 1156 1162.
References
Thery, C., Zitvogel, L., Amigorena, S., 2002b. Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569 579. Tian, Y., Li, S., Song, J., Ji, T., Zhu, M., Anderson, G.J., et al., 2014. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 35, 2383 2390. Timmers, L., Lim, S.K., Hoefer, I.E., Arslan, F., Lai, R.C., Van oorschot, A.A., et al., 2011. Human mesenchymal stem cell-conditioned medium improves cardiac function following myocardial infarction. Stem Cell Res. 6, 206 214. Tkach, M., Thery, C., 2016. Communication by extracellular vesicles: where we are and where we need to go. Cell 164, 1226 1232. Trams, E.G., Lauter, C.J., Salem Jr., N., Heine, U., 1981. Exfoliation of membrane ectoenzymes in the form of micro-vesicles. Biochim. Biophys. Acta 645, 63 70. Tsilioni, I., Panagiotidou, S., Theoharides, T.C., 2014. Exosomes in neurologic and psychiatric disorders. Clin. Ther. 36, 882 888. Turturici, G., Tinnirello, R., Sconzo, G., Geraci, F., 2014. Extracellular membrane vesicles as a mechanism of cell-to-cell communication: advantages and disadvantages. Am. J. Physiol. Cell Physiol. 306, C621 C633. Valadi, H., Ekstrom, K., Bossios, A., Sjostrand, M., Lee, J.J., Lotvall, J.O., 2007. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654 659. van den boorn, J.G., Schlee, M., Coch, C., Hartmann, G., 2011. SiRNA delivery with exosome nanoparticles. Nat. Biotechnol. 29, 325 326. van der pol, E., Boing, A.N., Harrison, P., Sturk, A., Nieuwland, R., 2012. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol. Rev. 64, 676 705. van den boorn, J.G., Dassler, J., Coch, C., Schlee, M., Hartmann, G., 2013. Exosomes as nucleic acid nanocarriers. Adv. Drug Deliv. Rev. 65, 331 335. Vella, L.J., Greenwood, D.L., Cappai, R., Scheerlinck, J.P., Hill, A.F., 2008. Enrichment of prion protein in exosomes derived from ovine cerebral spinal fluid. Vet. Immunol. Immunopathol. 124, 385 393. Vella, L.J., Hill, A.F., Cheng, L., 2016. Focus on extracellular vesicles: exosomes and their role in protein trafficking and biomarker potential in Alzheimer’s and Parkinson’s disease. Int. J. Mol. Sci. 17, 173. Viaud, S., Thery, C., Ploix, S., Tursz, T., Lapierre, V., Lantz, O., et al., 2010. Dendritic cell-derived exosomes for cancer immunotherapy: what’s next? Cancer Res. 70, 1281 1285. Viaud, S., Ullrich, E., Zitvogel, L., Chaput, N., 2008. Exosomes for the treatment of human malignancies. Horm. Metab. Res. 40, 82 88. Wahlgren, J., De, L.K.T., Brisslert, M., Vaziri sani, F., Telemo, E., Sunnerhagen, P., et al., 2012. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 40, e130. Wahlgren, J., Statello, L., Skogberg, G., Telemo, E., Valadi, H., 2016. Delivery of small interfering RNAs to cells via exosomes. Methods Mol. Biol. 1364, 105 125. Wang, X., Huang, W., Liu, G., Cai, W., Millard, R.W., Wang, Y., et al., 2014. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells, J. Mol. Cell. Cardiol., 74. pp. 139 150. Wolfers, J., Lozier, A., Raposo, G., Regnault, A., Thery, C., Masurier, C., et al., 2001. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 7, 297 303.
217
218
CHAPTER 7 Exosome as nanocarriers in drug delivery system
Woodburn, J.R., 1999. The epidermal growth factor receptor and its inhibition in cancer therapy. Pharmacol. Ther. 82, 241 250. Wu, X., Zheng, T., Zhang, B., 2016. Exosomes in Parkinson’s disease. Neurosci. Bull. Xin, H., Li, Y., Buller, B., Katakowski, M., Zhang, Y., Wang, X., et al., 2012. Exosomemediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 30, 1556 1564. Xitong, D., Xiaorong, Z., 2016. Targeted therapeutic delivery using engineered exosomes and its applications in cardiovascular diseases. Gene 575, 377 384. Yang, C., Robbins, P.D., 2012. Immunosuppressive exosomes: a new approach for treating arthritis. Int. J. Rheumatol. 2012, 573528. Yang, T., Martin, P., Fogarty, B., Brown, A., Schurman, K., Phipps, R., et al., 2015. Exosome delivered anticancer drugs across the blood brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 32, 2003 2014. Yellon, D.M., Davidson, S.M., 2014. Exosomes: nanoparticles involved in cardioprotection? Circ. Res. 114, 325 332. Yoshida, K., Burton, G.F., Mckinney, J.S., Young, H., Ellis, E.F., 1992. Brain and tissue distribution of polyethylene glycol-conjugated superoxide dismutase in rats. Stroke 23, 865 869. Yuan, M.J., Maghsoudi, T., Wang, T., 2016. Exosomes mediate the intercellular communication after myocardial infarction. Int. J. Med. Sci. 13, 113 116. Zhang, Y., Liu, D., Chen, X., Li, J., Li, L., Bian, Z., et al., 2010. Secreted monocytic miR150 enhances targeted endothelial cell migration. Mol. Cell 39, 133 144. Zhang, X., Pei, Z., Chen, J., Ji, C., Xu, J., Zhang, X., et al., 2016. Exosomes for immunoregulation and therapeutic intervention in cancer. J. Cancer 7, 1081 1087. Zhang, Y., Hu, Y.W., Zheng, L., Wang, Q., 2017. Characteristics and roles of exosomes in cardiovascular disease. DNA Cell Biol. Zhao, W., Zheng, X.L., Zhao, S.P., 2015. Exosome and its roles in cardiovascular diseases. Heart Fail Rev. 20, 337 348. Zhuang, X., Xiang, X., Grizzle, W., Sun, D., Zhang, S., Axtell, R.C., et al., 2011. Treatment of brain inflammatory diseases by delivering exosome encapsulated antiinflammatory drugs from the nasal region to the brain. Mol. Ther. 19, 1769 1779. Zitvogel, L., Regnault, A., Lozier, A., Wolfers, J., Flament, C., Tenza, D., et al., 1998. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cellderived exosomes. Nat. Med. 4, 594 600. Zitvogel, L., Fernandez, N., Lozier, A., Wolfers, J., Regnault, A., Raposo, G., et al., 1999. Dendritic cells or their exosomes are effective biotherapies of cancer. Eur. J. Cancer 35 (Suppl. 3), S36 S38.