Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2020 Sep 12;85(2):e13329. doi: 10.1111/aji.13329

The emerging role of exosomes as novel therapeutics: Biology, technologies, clinical applications, and the next

Yonghee Song 1, Youngeun Kim 1, Sunhyung Ha 1, Samantha Sheller‐Miller 2, Jaekwang Yoo 1, Chulhee Choi 1,3,, Cheol Hyoung Park 1,
PMCID: PMC7900947  PMID: 32846024

Abstract

The extracellular vesicles (EVs) research area has grown rapidly because of their pivotal roles in intercellular communications and maintaining homeostasis of individual organism. As a subtype of EVs, exosomes are made via unique biogenesis pathway and exhibit disparate functional and phenotypic characteristics. Functionally, exosomes transfer biological messages from donor cell to recipient cell, which makes exosomes as a novel therapeutic platform delivering therapeutic materials to the target tissue/cell. Currently, both academia and industry try to develop exosome platform‐based therapeutics for disease management, some of which are already in clinical trials. In this review, we will discuss focusing on therapeutic values of exosomes, recent advances in therapeutic exosome platform development, and late development of exosome therapeutics in diverse therapeutic areas.

Keywords: clinical trial, exosomes, inflammatory diseases, therapeutic platform

1. INTRODUCTION

For maintaining homeostasis of organisms, cell‐to‐cell communication controls multiple biological processes such as cellular proliferation, differentiation, apoptosis, biological mediator secretion, and so on. Cellular communication is mediated largely by either direct cell‐to‐cell contact or soluble factors including cytokines/chemokines, hormones, neurotransmitters, lipid mediators, and extracellular vesicles (EVs). 1 , 2 , 3 Of these, EVs are emerging as novel intercellular communication mediators in both normal and pathophysiological conditions.

EVs can be classified into three major subtypes including microvesicles, exosomes, and apoptotic bodies which show different characteristics in size (microvesicles:50‐1,000 nm, exosomes:30‐200 nm, and apoptotic bodies:1‐5um), content, morphology, and biogenesis mechasnism. 1 , 4 Especially, both microvesicles and exosomes act as safe containers of diverse biological molecules for intercellular communication and play a crucial role in cell‐cell communication via delivery of functional proteins, nucleic acids, and lipids. In this review, we will provide overview focusing on exosome biology, therapeutic values of exosomes, recent advances in exosome therapeutics development.

1.1. Exosome biogenesis

The biogenesis of exosomes has been proposed via either endosomal sorting complex required for transport (ESCRT)‐dependent or ESCRT‐independent pathway. ESCRT consists of about 20 different kinds of proteins that can be grouped into four complexes (ie, ESCRT‐0, ESCRT‐I, ESCRT‐II, and ESCRT‐III), and each of complexes has its own unique role during exosome biogenesis. 5 In ESCRT‐dependent pathway, ESCRT‐0 initiates the pathway by engaging ubiquitinated cargo proteins which are sequestered in the pre‐MVB endosomes (intra‐vesicle containing endosome). Both ESCRT‐I and ESCRT‐II bind ubiquitinated proteins to sort the proteins into the bud of endosome. ESCRT‐II coordinates assembly of ESCRT‐III, vesicle scission complex, and subsequently, ESCRT‐III cleaves fully established vesicles from the membrane. 6 , 7 , 8 , 9 At the end of the pathway, these ESCRT complexes are disassembled and recycled for next round of exosome biogenesis. In the ESCRT‐independent pathway, membrane lipid components including sphingomyelinases and ceramide induce spontaneous budding of intra‐luminal vesicles (ILVs) into MVBs. 10 , 11 The interactome study of tetraspanin‐enriched microdomains (TEMs) by mass spectrometry revealed that tetraspanins are involved in exosome cargo sorting processes in human lymphoblast derived exosomes. 12 Finally, in both exosome biogenesis pathways, via actin and microtubule filament‐mediated transport system exosomes in MVBs are secreted out extracellularly. 13 , 14 During this trafficking process, molecules including cortactin and Rab GTPases play a pivotal role in MVB and plasma membrane docking for exosome secretion. 15 , 16

Exosomes secreted via biogenesis pathways show heterogeneity in terms of size and composition of biological cargos even generated from same producing cells. 17 , 18 On the contrary, exosome biomarkers which are universally contained in the exosomes have been identified 10 : Proteins including heat‐shock proteins (eg, HSP70 and HSP 90), MVB biogenesis proteins (eg, Alix and TSG101), and tetraspanins (eg, CD9, CD63, and CD81) are commonly loaded in/onto exosome and used as exosome biomarkers. 10 , 19 Although exact biological function is not fully uncovered, it has been known that various forms of nucleic acids including fragmented DNA, mRNA, miRNA, piRNA, snoRNA, snRNA, rRNA, tRNA, Y‐RNA, and scRNA can be loaded into exosomes. 20 Exosomes also contain lipids including cholesterol, phosphatidylserine (PS), sphingomyelin, phosphatidic acid, sphingolipid, and ceramide, of which cholesterol, PS, and sphingolipid are mostly found in the exosomal membrane. 21

1.2. Clinical applications of exosomes

As of August 12, 2020, a key word search in the US‐NIH clinical trial database with “exosomes” gives 182 clinical studies search result. 22 In most of these studies, exosomes are being used as diagnosis biomarkers from body fluids such as blood and urine based on the findings that in vivo circulating exosomes carry biological materials (eg, proteins and nucleic acid) representing current physiological conditions. For example, miRNA expressions such as miR17‐92, miR‐15, and miR‐1246 are highly associated with multiple types of cancer, which are readily detected by analyzing circulating exosomes from the patients. Therefore, miRNA analysis of circulating exosomes can be used as non‐invasive method for cancer diagnostsis. 23 , 24

Now, the therapeutic potential of exosomes is also being investigated. 17 , 25 As it has been well known that exosomes inherit many physiological characteristics of originated cells, exosomes can be used as cell‐free therapeutics with comparable potency but better safety profile compared to original cell therapy. Moreover, therapeutic potential of exosome as therapeutic vehicles is also being investigated by loading active pharmaceutical ingredients (API) into exosomes to deliver APIs safely and specifically to the target tissues/cells. Now, the exosome therapeutics field has been hot up in the past few years, and as described below, a number of companies are actively developing exosome‐based therapeutics in various disease areas (Table 1).

TABLE 1.

Biotech companies preparing for exosome human clinical trials

Product Development stages
Type of exosome Pre‐clinical Phase I and IIa
ExoCoBio Aegle Therapeutics
Naïve exosome Capricor Exopharm
AgeX Therapeutics United Therapeutics
ReNeuron
Avalon Codiak (Not yet recruiting)
Evox
ILIAS
Carmine Therapeutics
Engineered exosome Aruna Bio
Anjarium
Adipomics
Brainstorm Cell Therapeutics
Exocure Biosciences
TriArm Therapeutics

Naïve exosomes are naturally produced by cells. Engineered exosomes use exosomes loaded up with other material such as proteins, nucleic acids, or other biomolecules.

So far, there are couple of companies in clinical stage of exosome therapeutics development: Codiak Biosciences is one of the major biotech start‐up companies developing exosome therapeutics and is dedicated to develop therapeutic exosomes to deliver siRNA API targeting KRAS (G12D) mutation in pancreatic cancer. 26 Recently, they got investigational new drug (IND) approval for Phase Ia/b study in pancreatic cancer patients from US FDA (NCT03608631). The company has also teamed up with Sarepta Therapeutics, the leader in precision genetic medicine for rare disease, to develop engineered exosome therapeutics to deliver RNA for neuromuscular diseases. Avalon Globocare is developing engineered exosome therapeutics, and with its lead pipeline, AVA‐201 (miR‐185 loaded exosomes produced from mesenchymal stem cells) in Leucoplakia, the company plans to have its first clinical study in United States in the year 2020. 27 In the same year, Aegle Therapeutics also plans to have a Phase I/IIa clinical study in dystrophic epidermolysis bullosa patients with their naïve regenerative exosomes, AGLE‐102, isolated from bone marrow mesenchymal stem cells (NCT04173650). Exopharm has started its PLEXOVAL Phase I study using exosomes isolated from human platelets for wound healing (NCT number, undisclosed). United Therapeutics has IND approval for a Phase I trial (NCT03857841) of UNEX‐42, EVs secreted from human bone marrow‐derived mesenchymal stem cells, against bronchopulmonary dysplasia, a condition common in preterm infants that receive assisted ventilation and supplemental oxygen. There are multiple other exosome therapeutics companies in pre‐clinical stage of their pipelines: Evox Therapeutics has developed technologies which allow API molecules (eg, proteins or nucleic acids) to be actively loaded into the exosomes as membrane anchored molecules. The company’s lead pipeline targets a lysosomal storage disorder called Niemann‐Pick Disease type C. Recently, Evox Therapeutics has licensed‐out its exosome‐based RNA interference/antisense RNA delivery technology to US big pharma Eli Lilly for neurological disease. ILIAS Biologics is also developing exosome therapeutics. The company has unique patented technology which allows active loading of large size proteins into luminal side of exosomes. With its current lead therapeutic candidate targeting inflammatory diseases, ILIAS Biologics plans to submit the IND application to the US FDA in the year 2021. ILIAS Biologics has other therapeutics pipelines in various disease indications which are mostly at the pre‐clinical stage of development. Compared to cell therapies, exosomes might be able to overcome safety issues surrounding persistent cell proliferation, while having the same therapeutic efficacy. Capricor Therapeutics, a company originally developing stem cell therapies using cardiospheres heart stem cells, is now evaluating therapeutic value of exosomes derived from cardiospheres to treat cardiac and inflammatory conditions. 28 ReNeuron, a CTX stem cells therapeutics company, also is investigating the possibility to use CTX stem cells derived exosomes as drug‐ and gene‐delivery vehicles in stroke to promote post‐stroke rehabilitation. 29

Currently, there are about 20 biotech companies around the world developing exosome therapeutics with various therapeutic exosome platform technologies. 30 , 31 Some of these companies already moved on to more promising stage of clinical development with their own exosome engineering technologies improving efficacy and safety profiles. Next, we will overview currently available exosome therapeutic platforms and their potential use in multiple therapeutics areas.

1.3. Therapeutic exosome platforms and their potential use in clinic

Exosomes are increasingly being recognized as novel modulators for different therapeutic purposes, including vaccination, biological targeting agents, and drug delivery tools. 32 , 33 , 34 Thus, several applications for exosome‐mediated therapy have been proposed. Herein, we will focus on the application of exosomes as drug delivery systems. Therapeutic exosome platforms can be largely sub‐grouped into either naïve exosomes or artificial exosomes: Naive exosomes are therapeutics exosomes derived from non‐engineered producing cells. Artificial exosomes are therapeutics exosomes either derived from engineered producing cells or made via in vitro engineering post‐isolation. More details of each therapeutic exosome platform will be discussed as following.

1.3.1. Naïve/natural exosomes

As natural cell‐derived exosomes, known as naïve exosomes, by carrying not fully identified functional molecules inherit functional characteristics from their parent cell (Figure 1A), they have been effectively employed to provide an alternative way of developing strategies to cure or modify disease processes. 35 , 36 , 37 , 38 Recent studies have suggested that mesenchymal stem cell (MSC)‐derived exosomes might represent as novel therapeutics with advantages over parent MSCs such as lower immunogenicity and tumorogenicity. 37 , 38 Phase I clinical trials evaluating therapeutic potential of MSC‐derived naïve exosomes have been either completed or are underway in multiple disease areas (NCT04173650 and NCT03857841). 22 Although too early for clinical use, these clinical studies together with pre‐clinical observation suggest that MSC‐derived naïve exosomes may hold promise for a cell‐free therapeutic strategy.

FIGURE 1.

FIGURE 1

Exosome engineering methods for encapsulation of therapeutic agents. The techniques of loading cargo into exosomes can be divided into four approaches. A, Naïve exosomes are relatively simple technique, but the bioactive molecules contained in them are unknown. B, Passive methods are based on the use of sonication, Freezing/Thawing cycles, or electroporation to destabilize exosome membrane integrity, allowing drugs to be introduced into exosomes. C and D, Active methods spontaneously load interested molecules by hijacking the exosome biogenesis pathway. These techniques are divided into two approaches depending on whether the loaded cargo can reside as free form in the exosome

1.3.2. Artificial exosomes with engineering technologies

To improve API loading efficacy and in vivo target specificity, multiple exosome engineering technologies are being used. Especially, related to API loading into exosomes, currently there are two major technical approaches are being used including passive or active API loading into exosomes.

The passive cargo‐loading methods involve an incubation procedure that recruit bioactive agents into exosomes through sonication, electroporation, freeze‐thaw cycles, and extrusion (Figure 1B). 39 , 40 , 41 , 42 , 43 , 44 However, a major drawback of this method is that during the membrane disruption process, there is high potential of damaging exosome structure with compromised targeting properties. 45 , 46 , 47 The active cargo‐loading methods provide cells with the means to incorporate API proteins or nucleic acids into exosomes during their biogenesis process. The cargo can be endogenously loaded by genetically modifying the parental cells to overexpress desired API proteins or nucleic acids of interest (with or without additional modification to promote cargo packaging), which is then naturally incorporated into the exosomes. One of active cargo‐loading technologies is anchoring cargo on exosome inner surfaces via transgene expression in parent cells (Figure 1C): System Biosciences, an exosome biotech company, developed specific peptide sequences which target interior exosomal membrane proteins, allowing the fusion protein to be packaged into exosomes for secretion. 48 Evox Therapeutics also developed another API loading techniques by simply making fusion proteins between protein API and exosomal membrane proteins such as CD63, CD81, and syntenin. 49 However, the caveat this approach is that the API remains attached to the exosomal membrane in the target cells, which may significantly limit their biological function in the target cells. Alternatively, a novel technology called “EXosomes for Protein Loading via Optically Reversible protein‐protein interaction (EXPLOR)” has been developed by ILIAS Biologics to overcome this limitation. 50 EXPLOR technology makes it possible for getting high API loading yield into the exosomes with controllable way that in the target cells protein APIs can be delivered as free form to show unlimited biological function (Figure 1D). The next question is how we can achieve target‐specific delivery of loaded API without premature immune‐mediated clearance. By applying various exosome tissue targeting technology to their own API loading platforms, exosome therapeutics companies try to provide solutions for this question: System Biosciences employed exosome surface engineering post exosome isolation which decorates the outer membrane of exosomes with streptavidin to bind to biotinylated targeting moiety such as target‐specific single‐chain variable fragment (ScFv). This technology enables exosome‐based targeted delivery of API to HIV‐infected target cells in vivo. Codiak Biosciences uses Prostaglandin F2 Receptor Inhibitor (PTGFRN) and Brain Abundant Membrane Attached Signal Protein 1 (BASP‐1) proteins as scaffolds for anchoring of direct targeting ligands and therapeutic molecules to the surface and the lumen of exosomes, respectively. However, these engineered exosomes need to be considered for potential drawback, including induction of immunogenicity by exogenous modification, unlike naïve exosomes. Although several studies showed that the engineered exosomes can deliver therapeutic RNAi safely into target tissues/cells with minimal immunogenicity even after repeated doses, 26 , 41 , 51 still it is not known whether the cargo has no immunogenic activity or not.

Next, we will overview more details about this EXPLOR technology and discuss the clinical potential of “Exo‐target,” therapeutic exosomes with EXPLOR technology, in multiple disease areas.

1.3.3. Artificial exosomes with EXPLOR technology (Exo‐target)

EXPLOR technology utilizes photo‐reactive binding modules, cryptochrome2 (CRY2) and N‐terminal of CRY‐interacting basic‐helix‐loop helix 1 (CIBN) isolated from Arabidopsis thaliana. In Exo‐target producing donor cells, CRY2 protein is fused to a cargo protein API, and CIBN is conjugated with a representative marker of exosomes, CD9 protein. Blue‐light illumination induces the reversible protein‐protein interaction (PPI) between CIBN and CRY2 fusion proteins. 52 , 53 With continuous blue‐light irradiation, the cargo proteins are guided to the inner surface of the cell membrane or the surface of early endosomes. Mature MVBs then readily secrete cargo protein‐carrying exosomes, Exo‐target, from the cells by membrane fusion with the plasma membrane. After exocytosis, Exo‐target can be easily isolated and purified from producing cell culture supernatant with blue‐light removal which allows the exosome loaded cargo protein APIs present as free form in the luminal side of Exo‐target. Purified Exo‐target can be used for cargo protein APIs delivery into the cytosolic compartment of target cells via membrane fusion or endocytosis processes (Figure 2).

FIGURE 2.

FIGURE 2

Scheme of EXPLOR technology. The cargo proteins are actively loaded into exosomes under light irradiation. The interaction is reversible; thus, the cargo proteins can be detached from exosome membrane as free forms in the lumen of exosomes

Recently, Exo‐srIκB, Exo‐target loaded with super‐repressor IκB (srIκB) by EXPLOR, was proposed as a potential medicine in inflammatory disease such as sepsis and preterm birth (PTB). 54 , 55 The srIκB indicates a degradation‐resistant IκB mutant in which both Ser32 and Ser36 are replaced to Ala. It blocks nuclear translocation of NF‐κB even in the presence of pro‐inflammatory stimuli and thus inhibits the expression of specific NF‐κB target genes. As 293T cell‐derived exosomes are one of the most widely used as tools for drug delivery, 56 , 57 , 58 Exo‐srIκB was generated from HEK293T cells. The role and potential of Exo‐srIκB in sepsis and PTB are as follows.

Sepsis is a syndrome associated with severe infection and is one of the leading causes of deaths world‐wide. Yet there are no specific therapies available and mortality continues to be high especially in low‐income countries. Based on findings from pre‐clinical studies, there is growing interest on therapeutic intervention for nuclear factor (NF)‐κB pathway to limit inflammatory injury during sepsis. 59 , 60 To date, numerous small molecule inhibitors for NF‐κB pathway have been developed, but none of them have been clinically approved. 61 Recently, ILIAS Biologics generated Exo‐srIκB and clearly verified its therapeutic potential in sepsis pre‐clinical models. 50 , 54 The data from Exo‐srIκB in vivo pre‐clinical studies showed the srIκB pharmacology, driving significantly greater anti‐inflammatory activity in septic animal models: Exo‐srIκB were administered intraperitoneally (i.p.) by either single or multiple dose (four times every 6 hours, 10 9 particles/dose) in lipopolysaccharide (LPS)‐induced or cecum ligation and puncture (CLP)‐induced septic mouse model, respectively. In these septic mouse models, Exo‐srIκB treatment ameliorates systemic inflammation associated mouse mortality and morbidity compared to naïve exosome treatment (ie, Exo‐naïve) generated from same parent cells. 54 Neutrophils and macrophages are major components of the innate immune system and play critical roles in orchestrating host immune response during sepsis. 62 At the early phase of sepsis, they secrete an increased level of pro‐inflammatory factors, which exacerbate the inflammatory response and subsequently contribute to disease mortality and morbidity. 63 In septic animals, administered Exo‐srIkB via intravenous (iv) injection are targeted to these innate immune cells within 30 min and inhibit overwhelming pro‐inflammatory response (ie, TNF‐α, IL‐1β, and IL‐6), which subsequently ameliorates tissue/organ damages including kidney and liver. Taken together, these findings suggest therapeutic potential of Exo‐srIkB in treating hyper‐immunity of sepsis which has significant medical unmet need (Figure 3A).

FIGURE 3.

FIGURE 3

The role and therapeutic potential of Exo‐srIκB in inflammatory disease. A, Exo‐srIκB is successfully delivered to target neutrophil and macropage in sepsis model. B, Unlike small molecules, Exo‐srIκB can inhibit/delay preterm birth by penetrating the placental layer and delivering drug substances to fetus

Although the causes of PTB are numerous, it is well known that inflammation represents a highly significant risk factor in PTB. 64 , 65 , 66 Current PTB prevention strategies are that minimizing contractions of maternal uterine tissues to prolong labor or administering corticosteroids to provide enough time for fetal lung maturation. Multiple drugs (eg, tocolytics and progesterone) have been developed; however, none have significantly reduced the risk of PTB in nulliparous subjects over the past 30 years. 67 The potential therapeutics for PTB include cytokine‐suppressive anti‐inflammatory drugs, 68 , 69 , 70 cell‐penetrating peptides and small molecule inhibitors, 71 , 72 , 73 non‐specific inhibitors of NF‐κB pathway, 74 , 75 , 76 , 77 and flavonoids. 78 , 79 However, very few of these drugs have been in clinical trials and none are in‐use clinically due to key issues such as placental permeability, in vivo half‐life and stability, effectiveness in reducing the fetal inflammatory response, and their toxicity.

Recently, University of Texas Medical Branch and ILIAS Biologics reported fetal‐maternal communication during pregnancy is mediated by exosomes. 55 They used mT/mG, a cell membrane targeted and two‐color fluorescent Cre reporter mouse. Prior to Cre‐mediated excision, membrane‐localized tdTomato (mT) expression is widespread in cells/tissues. Cre recombinase expressing cells express membrane‐localized EGFP (mG) fluorescence replacing the mT fluorescence to identify placental permeability of exosomes, Exo‐Cre (Cre‐loaded exosomes by EXPLOR) were injected intraperitoneally into pregnant mT/mG mice. The maternally administered Exo‐Cre was transferred fetal tissues across placental barriers, inducing expression of mG instead of mT. These results propose that exosomes are able to have a therapeutic effect on PTB by passing through the placenta. NF‐κB is a well‐known mechanistic mediator in infection‐associated PTB. 80 , 81 , 82 , 83 , 84 In infection‐induced PTB pre‐clinical model, Exo‐srIκB treatment delayed PTB by 24‐hours, improved pup viability, and significantly reduced histologic chorioamnionitis in fetal, not maternal (unpublished data). Taken together, these findings suggest Exo‐srIκB may provide a potential intervention for reducing the host inflammatory response associated with PTB (Figure 3B).

1.4. The challenges for bringing exosomes into the clinic

Up to now, there are enough scientific evidence accumulated to support therapeutic value of exosomes in various human diseases. For successful exosome therapeutics development, the CMC (ie, chemistry, manufacturing, and control) development for good manufacturing practice (GMP) grade therapeutic exosome production needs to be established. The CMC development for exosome therapeutics covers multiple area including establishing master cell bank (MCB), process development for large quantity of exosome production/isolation and quality control (QC)/analytical method development for therapeutic exosome production. 85 , 86 , 87 , 88 As exosome is a novel therapeutic platform, in the field so far there is no gold standard process established for the CMC of exosome therapeutics. Currently, most of exosome therapeutics companies are working on developing CMC process suitable for their own exosome therapeutic platforms. 89 Here, we will briefly overview the biggest hurdles we are facing to solve for successful exosome therapeutics development and current status of CMC development in the field.

As exosomes are products secreted from cells, cell selection is crucial first step for generating exosomes without changing the composition and function of the exosomes. In order to produce exosomes in small or large scale, several types of cells including human cardiac progenitor cells, bone marrow mesenchymal stem cells, adipose tissue‐derived stem cells, monocyte‐derived dendritic cells, and HEK293 cells have been cultured in fixed or moving culture systems such as the flask and bioreactor. 89 , 90 , 91 , 92 Further studies are focusing on developing exosome producing stable cell‐lines which can produce API loaded exosomes with better yield and less variability. In parallel, scalable culture system in optimal serum‐free culture condition using well‐established bioreactors is desired to manufacture clinical grade exosomes.

Next, establishing a robust exosome purification process is also crucial to obtain high yield and purity therapeutic exosomes. 93 During exosome therapeutics production, impurities such as host cell proteins, DNA, cellular debris, and non‐exosomal EVs (eg, apoptotic bodies and microvesicles) should be controlled and removed as thoroughly as possible. 94 , 95 To increase production yield, diverse methods for exosome separation and concentration from the cell culture medium have been applied: ultracentrifugation using centrifugal force and weight, size‐based separation methods using column filled with porous polymeric beads and tangential flow filtration using ultrafiltration membranes, precipitation‐based separation methods using polymer and protein organic solvent, immunoaffinity‐based separation methods using specific bindings between exosome membrane‐bound antigens and immobilized antibodies, ion‐exchanged chromatography based on electrostatic interaction, and microfluidics‐based separation using microfluidic control technology. 96 , 97 , 98 , 99 , 100 , 101 Still, there are significant needs for technical improvement for these methods to improve exosome separation capability, scalability, yield, and cost for commercialization.

QC/analytical method development for exosome therapeutic product is essential for successful exosome therapeutics development. Certainly, current limitation in exosome analytical technologies is one of biggest huddles for large‐scale exosome manufacturing. 87 , 88 , 102 Currently, exosomes are being defined and identified according to the proteomic identification criteria including transmembrane proteins (eg, tetraspanins) and cytosolic proteins (eg, TSG101, Rab proteins, or annexins), established by the International Society of Extracellular Vesicles (ISEV). 103 Although electron microscopy and particle size distribution analysis methods are generally accepted in the field, more advanced and accurate methods to evaluate morphology and size of exosomes are also required. Additional more reliable in vitro or in vivo exosome QC and characterization methods are needed, which can possibility be achieved with advanced technologies using chromatography, nanoFACS, microfluidics analysis, etc 87 , 102

None the less, by overcoming these technical difficulties of manufacturing, several leading exosome biotech companies successfully already entered or are entering into clinical trials, albeit in an early stage or MSC‐derived naïve exosomes. Exopharm (Australia)’s purification technology called Ligand‐based Exosome Affinity Purification (LEAP) was applied to Plexaris, which is being developed as a wound treatment at Phase I clinical stage in Australia. The company demonstrates that LEAP technology enables production and purification of clinical grade and scale exosome, and takes exosome‐based therapeutics to clinical trials successfully. 104 Kalluri’s group in University of Texas MD Anderson Cancer Center reported a large‐scale production of GMP‐compliant exosomes from bone marrow–derived MSCs loaded with siRNA to target Kras G12D available for clinical use for the first time in the world. 26 Aegle Therapeutics developed a new technology for the isolation and purification of extracellular vesicle including exosomes with high purity and therapeutic grade. The company demonstrates the method offers a simple large‐scale production and purification of extracellular vesicles without complicated equipment at GMP condition. However, the details of the techniques listed above about how their exosomes are produced and isolated have not been released. ILIAS Biologics 105 employs engineered suspension cells to produce therapeutic exosomes by the EXPLORs technology in single‐use bioreactors. Master and work cell banks for non‐clinical and clinical production were successfully established from stabilized cells, based on productivity, viability, and characteristics. The company optimized cell culture scale and manufacturing process to produce large quantities of a variety of homogeneous engineered exosomes in the high yield and purity. To isolate highly pure exosomes, culture media harvested from the bioreactors is processed through centrifugation, filtration, chromatography, and concentration. Purity, contents, and potency in the final products are analyzed by size‐exclusion chromatography, immunoblotting, nanoparticle tracking analysis, protein quantification, and cell‐based activity assay. With active pre‐clinical pharmacological and toxicological studies for first‐in‐human study, ILIAS Biologics is currently attempting to develop robust GMP production process that allows exosome therapeutics to translate into clinic.

2. SUMMARY AND PERSPECTIVE

In both normal and pathophysiological conditions, exosomes play pivotal roles as messenger delivering biological messages between tissues/cells. Due to their biological function and characteristics, exosomes can be utilized as novel biological platform for disease diagnose and therapeutics. With non‐invasive liquid biopsies, circulating exosomes can be analyzed to determine the presence of cancer biomarkers including oncogenic proteins and nucleic acids (eg, miRNAs, mRNAs, and DNAs). Therapeutically, due to their unique characteristics as EVs transmigrating tissue barriers (eg, blood‐brain barrier and placenta), high yield capacity for intercellular cargo delivery, high biocompatibility and low immunogenicity, exosomes are actively being studied as therapeutic vehicles by loading many different forms of therapeutic APIs into exosomes via multiple exosome engineering technologies. With demonstrated therapeutics potentials in pre‐clinical studies, there are exosome biotech companies entering into clinical development including ILIAS Biologics. Although there are still significant technological huddles present for large‐scale manufacturing, these clinical stage leading companies are paving the way for successful exosome therapeutic development by overcoming scientific and technological challenges, which can give clear guidance for other following exosome therapeutics companies. Finally, with scientific and technological innovation in exosome biology, we expect to have successful novel therapeutic platform development to treat the various diseases having significant medical unmet needs in the near future.

CONFLICT OF INTEREST

C.C is an inventor of a patent related to this work filed by ILIAS Biologics Inc (no. KR 10‐1877010 and US 10,702,581), the founder and shareholder of ILIAS Biologics Inc YS, YK, JY, and CHP are minor shareholders of ILIAS Biologics Inc SS‐M. is a paid consultant for ILIAS Therapeutics. All other authors declare that they have no conflict of interests.

Song Y, Kim Y, Ha S, et al. The emerging role of exosomes as novel therapeutics: Biology, technologies, clinical applications, and the next. Am J Reprod Immunol.2021;85:e13329 10.1111/aji.13329

Yonghee Song and Youngeun Kim are the authors contributed equally to this work.

Contributor Information

Chulhee Choi, Email: cchoi@iliasbio.com.

Cheol Hyoung Park, Email: spc@iliasbio.com.

REFERENCES

  • 1. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373‐383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Fradelizi D. Cytokines: soluble factors in intercellular communication. Med Trop (Mars). 1998;58(4 Suppl):427‐432. [PubMed] [Google Scholar]
  • 3. Lauder JM. Neurotransmitters as growth regulatory signals: role of receptors and second messengers. Trends Neurosci. 1993;16(6):233‐240. [DOI] [PubMed] [Google Scholar]
  • 4. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569‐579. [DOI] [PubMed] [Google Scholar]
  • 5. 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(Pt 24):5553‐5565. [DOI] [PubMed] [Google Scholar]
  • 6. Katzmann DJ, Babst M, Emr SD. Ubiquitin‐dependent sorting into the multivesicular body pathway requires the function of a conserved endosomal protein sorting complex, ESCRT‐I. Cell. 2001;106(2):145‐155. [DOI] [PubMed] [Google Scholar]
  • 7. Katzmann DJ, Stefan CJ, Babst M, Emr SD. Vps27 recruits ESCRT machinery to endosomes during MVB sorting. J Cell Biol. 2003;162(3):413‐423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Bilodeau PS, Winistorfer SC, Kearney WR, Robertson AD, Piper RC. Vps27‐Hse1 and ESCRT‐I complexes cooperate to increase efficiency of sorting ubiquitinated proteins at the endosome. J Cell Biol. 2003;163(2):237‐243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Lu Q, Hope LW, Brasch M, Reinhard C, Cohen SN. TSG101 interaction with HRS mediates endosomal trafficking and receptor down‐regulation. Proc Natl Acad Sci USA. 2003;100(13):7626‐7631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 2019;9:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Babst M. MVB vesicle formation: ESCRT‐dependent, ESCRT‐independent and everything in between. Curr Opin Cell Biol. 2011;23(4):452‐457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Perez‐Hernandez D, Gutiérrez‐Vázquez C, Jorge I, et al. The intracellular interactome of tetraspanin‐enriched microdomains reveals their function as sorting machineries toward exosomes. J Biol Chem. 2013;288(17):11649‐11661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10(7):925‐937. [DOI] [PubMed] [Google Scholar]
  • 14. Escola JM, Kleijmeer MJ, Stoorvogel W, Griffith JM, Yoshie O, Geuze HJ. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B‐lymphocytes. J Biol Chem. 1998;273(32):20121‐20127. [DOI] [PubMed] [Google Scholar]
  • 15. Sinha S, Hoshino D, Hong NH, et al. Cortactin promotes exosome secretion by controlling branched actin dynamics. J Cell Biol. 2016;214(2):197‐213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Stenmark H. Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol. 2009;10(8):513‐525. [DOI] [PubMed] [Google Scholar]
  • 17. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Wen SW, Lima LG, Lobb RJ, et al. Breast cancer‐derived exosomes reflect the cell‐of‐origin phenotype. Proteomics. 2019;19(8):e1800180. [DOI] [PubMed] [Google Scholar]
  • 19. Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019;8(7):727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Turchinovich A, Drapkina O, Tonevitsky A. Transcriptome of extracellular vesicles: state‐of‐the‐art. Front Immunol. 2019;10:202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Skotland T, Hessvik NP, Sandvig K, Llorente A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J Lipid Res. 2019;60(1):9‐18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. ClinicalTrials.gov . https://clinicaltrials.gov/. Accessed July 6, 2020.
  • 23. Choi D‐S, Choi D‐Y, Hong BokSil, et al. Quantitative proteomics of extracellular vesicles derived from human primary and metastatic colorectal cancer cells. J Extracell Vesicles. 2012;1(1):18704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Moon P‐G, Lee J‐E, You S, et al. Proteomic analysis of urinary exosomes from patients of early IgA nephropathy and thin basement membrane nephropathy. Proteomics. 2011;11(12):2459‐2475. [DOI] [PubMed] [Google Scholar]
  • 25. Kim YK, Choi Y, Nam GH, Kim IS. Functionalized exosome harboring bioactive molecules for cancer therapy. Cancer Lett. 2020;489:155‐162. [DOI] [PubMed] [Google Scholar]
  • 26. Mendt M, Kamerkar S, Sugimoto H, et al. Generation and testing of clinical‐grade exosomes for pancreatic cancer. JCI Insight. 2018;3(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Wang L, Yin P, Wang J, et al. Delivery of mesenchymal stem cells‐derived extracellular vesicles with enriched miR‐185 inhibits progression of OPMD. Artif Cells Nanomed Biotechnol. 2019;47(1):2481‐2491. [DOI] [PubMed] [Google Scholar]
  • 28. Gallet R, Dawkins J, Valle J, et al. Exosomes secreted by cardiosphere‐derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2017;38(3):201‐211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Stevanato L, Thanabalasundaram L, Vysokov N, Sinden JD. Investigation of content, stoichiometry and transfer of miRNA from human neural stem cell line derived exosomes. PLoS One. 2016;11(1):e0146353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Zipkin M. Exosome redux. Nat Biotechnol. 2019;37(12):1395‐1400. [DOI] [PubMed] [Google Scholar]
  • 31. Jafari D, Shajari S, Jafari R, et al. Designer exosomes: a new platform for biotechnology therapeutics. BioDrugs. 2020. 10.1007/s40259-020-00434-x. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Xiao L, Erb U, Zhao K, Hackert T, Zoller M. Efficacy of vaccination with tumor‐exosome loaded dendritic cells combined with cytotoxic drug treatment in pancreatic cancer. Oncoimmunology. 2017;6(6):e1319044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Wahlund CJE, Gucluler G, Hiltbrunner S, Veerman RE, Naslund TI, Gabrielsson S. Exosomes from antigen‐pulsed dendritic cells induce stronger antigen‐specific immune responses than microvesicles in vivo. Sci Rep. 2017;7(1):17095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. André F, Chaput N, Schartz NEC, et al. Exosomes as potent cell‐free peptide‐based vaccine. I. Dendritic cell‐derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. J Immunol. 2004;172(4):2126‐2136. [DOI] [PubMed] [Google Scholar]
  • 35. Escudier B, Dorval T, Chaput N, et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived‐exosomes: results of thefirst phase I clinical trial. J Transl Med. 2005;3(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Zhu L, Kalimuthu S, Gangadaran P, et al. exosomes derived from natural killer cells exert therapeutic effect in melanoma. Theranostics. 2017;7(10):2732‐2745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214‐222. [DOI] [PubMed] [Google Scholar]
  • 38. Lai RC, Arslan F, Tan SS, et al. Derivation and characterization of human fetal MSCs: an alternative cell source for large‐scale production of cardioprotective microparticles. J Mol Cell Cardiol. 2010;48(6):1215‐1224. [DOI] [PubMed] [Google Scholar]
  • 39. Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle drug delivery system: the anti‐inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18(9):1606‐1614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Kim MS, Haney MJ, Zhao Y, et al. Development of exosome‐encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine. 2016;12(3):655‐664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Alvarez‐Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341‐345. [DOI] [PubMed] [Google Scholar]
  • 42. Lamichhane TN, Jeyaram A, Patel DB, et al. Oncogene knockdown via active loading of small RNAs into extracellular vesicles by sonication. Cell Mol Bioeng. 2016;9(3):315‐324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Hood JL. Post isolation modification of exosomes for nanomedicine applications. Nanomedicine (Lond). 2016;11(13):1745‐1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Kamerkar S, LeBleu VS, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546(7659):498‐503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for Parkinson's disease therapy. J Control Release. 2015;207:18‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Hood JL, Scott MJ, Wickline SA. Maximizing exosome colloidal stability following electroporation. Anal Biochem. 2014;448:41‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol Sin. 2017;38(6):754‐763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. System biosciences . XPack™ Exosome Protein Engineering. Available at: https://systembio.com/shop/xpack‐mscv‐xp‐mcs‐ef1%CE%B1‐puro‐cloning‐lentivector/. Accessed June 1, 2020
  • 49. Evox Therapeutics . https://www.evoxtherapeutics.com. Accessed June 1, 2020
  • 50. Yim N, Ryu S‐W, Choi K, et al. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein‐protein interaction module. Nat Commun. 2016;7:12277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Zhu X, Badawi M, Pomeroy S, et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from HEK293T cells. J Extracell Vesicles. 2017;6(1):1324730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Kennedy MJ, Hughes RM, Peteya LA, Schwartz JW, Ehlers MD, Tucker CL. Rapid blue‐light‐mediated induction of protein interactions in living cells. Nat Methods. 2010;7(12):973‐975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Shalitin D, Yang H, Mockler TC, et al. Regulation of arabidopsis cryptochrome 2 by blue‐light‐dependent phosphorylation. Nature. 2002;417(6890):763‐767. [DOI] [PubMed] [Google Scholar]
  • 54. Choi H, Kim Y, Mirzaaghasi A, et al. Exosome‐based delivery of super‐repressor IkappaBalpha relieves sepsis‐associated organ damage and mortality. Sci Adv. 2020;6(15):eaaz6980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Sheller‐Miller S, Choi K, Choi C, Menon R. Cyclic‐recombinase‐reporter mouse model to determine exosome communication and function during pregnancy. Am J Obstet Gynecol. 2019;221(5):502 e501‐502;e512. [DOI] [PubMed] [Google Scholar]
  • 56. Shurtleff MJ, Temoche‐Diaz MM, Karfilis KV, Ri S, Schekman R. Y‐box protein 1 is required to sort microRNAs into exosomes in cells and in a cell‐free reaction. Elife. 2016;5:e19276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Ohno S‐I, Takanashi M, Sudo K, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther. 2013;21(1):185‐191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Kanada M, Bachmann MH, Hardy JW, et al. Differential fates of biomolecules delivered to target cells via extracellular vesicles. Proc Natl Acad Sci USA. 2015;112(12):E1433‐E1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Altavilla D, Squadrito G, Minutoli L, et al. Inhibition of nuclear factor‐kappaB activation by IRFI 042, protects against endotoxin‐induced shock. Cardiovasc Res. 2002;54(3):684‐693. [DOI] [PubMed] [Google Scholar]
  • 60. Choi JH, Park SH, Jung JK, et al. Caffeic acid cyclohexylamide rescues lethal inflammation in septic mice through inhibition of IkappaB Kinase in innate immune process. Sci Rep. 2017;7:41180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Gupta SC, Sundaram C, Reuter S, Aggarwal BB. Inhibiting NF‐kappaB activation by small molecules as a therapeutic strategy. Biochim Biophys Acta. 2010;1799(10‐12):775‐787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Parihar A, Eubank TD, Doseff AI. Monocytes and macrophages regulate immunity through dynamic networks of survival and cell death. J Innate Immun. 2010;2(3):204‐215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Hamidzadeh K, Christensen SM, Dalby E, Chandrasekaran P, Mosser DM. Macrophages and the recovery from acute and chronic inflammation. Annu Rev Physiol. 2017;79:567‐592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Romero R, Dey SK, Fisher SJ. Preterm labor: one syndrome, many causes. Science. 2014;345(6198):760‐765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Xu YI, Romero R, Miller D, et al. Innate lymphoid cells at the human maternal‐fetal interface in spontaneous preterm labor. Am J Reprod Immunol. 2018;79(6):e12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Romero R, Espinoza J, Kusanovic JP, et al. The preterm parturition syndrome. BJOG. 2006;113(Suppl 3):17‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Beck S, Wojdyla D, Say L, et al. The worldwide incidence of preterm birth: a systematic review of maternal mortality and morbidity. Bull World Health Organ. 2010;88(1):31‐38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Stinson LF, Ireland DJ, Kemp MW, et al. Effects of cytokine‐suppressive anti‐inflammatory drugs on inflammatory activation in ex vivo human and ovine fetal membranes. Reproduction. 2014;147(3):313‐320. [DOI] [PubMed] [Google Scholar]
  • 69. Ireland DJ, Kemp MW, Miura Y, Saito M, Newnham JP, Keelan JA. Intra‐amniotic pharmacological blockade of inflammatory signalling pathways in an ovine chorioamnionitis model. Mol Hum Reprod. 2015;21(5):479‐489. [DOI] [PubMed] [Google Scholar]
  • 70. Ireland DJ, Nathan EA, Li S, et al. Preclinical evaluation of drugs to block inflammation‐driven preterm birth. Innate Immun. 2017;23(1):20‐33. [DOI] [PubMed] [Google Scholar]
  • 71. Gurney LRI, Taggart J, Tong WC, Jones AT, Robson SC, Taggart MJ. Inhibition of inflammatory changes in human myometrial cells by cell penetrating peptide and small molecule inhibitors of NFkappaB. Front Immunol. 2018;9:2966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Pekson R, Poltoratsky V, Gorasiya S, et al. N, N‐dimethylacetamide significantly attenuates LPS‐ and TNFalpha‐induced proinflammatory responses via inhibition of the nuclear factor Kappa B pathway. Mol Med. 2016;22:747‐758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Sundaram S, Ashby CR, Pekson R, et al. N, N‐dimethylacetamide regulates the proinflammatory response associated with endotoxin and prevents preterm birth. Am J Pathol. 2013;183(2):422‐430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Buhimschi IA, Buhimschi CS, Weiner CP. Protective effect of N‐acetylcysteine against fetal death and preterm labor induced by maternal inflammation. Am J Obstet Gynecol. 2003;188(1):203‐208. [DOI] [PubMed] [Google Scholar]
  • 75. Lappas M, Yee K, Permezel M, Rice GE. Sulfasalazine and BAY 11–7082 interfere with the nuclear factor‐kappa B and I kappa B kinase pathway to regulate the release of proinflammatory cytokines from human adipose tissue and skeletal muscle in vitro. Endocrinology. 2005;146(3):1491‐1497. [DOI] [PubMed] [Google Scholar]
  • 76. Keelan JA, Khan S, Yosaatmadja F, Mitchell MD. Prevention of inflammatory activation of human gestational membranes in an ex vivo model using a pharmacological NF‐kappaB inhibitor. J Immunol. 2009;183(8):5270‐5278. [DOI] [PubMed] [Google Scholar]
  • 77. Jiang L, Yan Q, Liu RH, Zhang L. Preventive and therapeutic effect of N‐Acetyl‐l‐cysteine on infection‐associated preterm labor in mice. Asian Pac J Trop Med. 2016;9(2):197‐200. [DOI] [PubMed] [Google Scholar]
  • 78. Wall C, Lim R, Poljak M, Lappas M. Dietary flavonoids as therapeutics for preterm birth: luteolin and kaempferol suppress inflammation in human gestational tissues in vitro. Oxid Med Cell Longev. 2013;2013:485201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Morwood CJ, Lappas M. The citrus flavone nobiletin reduces pro‐inflammatory and pro‐labour mediators in fetal membranes and myometrium: implications for preterm birth. PLoS One. 2014;9(9):e108390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Filipovich Y, Klein J, Zhou Y, Hirsch E. Maternal and fetal roles in bacterially induced preterm labor in the mouse. Am J Obstet Gynecol. 2016;214(3):386;e381‐389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Behnia F, Sheller S, Menon R. Mechanistic differences leading to infectious and sterile inflammation. Am J Reprod Immunol. 2016;75(5):505‐518. [DOI] [PubMed] [Google Scholar]
  • 82. Lindstrom TM, Bennett PR. The role of nuclear factor kappa B in human labour. Reproduction. 2005;130(5):569‐581. [DOI] [PubMed] [Google Scholar]
  • 83. Filipovich Y, Lu SJ, Akira S, Hirsch E. The adaptor protein MyD88 is essential for E coli‐induced preterm delivery in mice. Am J Obstet Gynecol. 2009;200(1):93;e91‐98. [DOI] [PubMed] [Google Scholar]
  • 84. Elovitz MA. Anti‐inflammatory interventions in pregnancy: now and the future. Semin Fetal Neonatal Med. 2006;11(5):327‐332. [DOI] [PubMed] [Google Scholar]
  • 85. Ng KS, Smith JA, McAteer MP, et al. Bioprocess decision support tool for scalable manufacture of extracellular vesicles. Biotechnol Bioeng. 2019;116(2):307‐319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Burnouf T, Agrahari V, Agrahari V. Extracellular vesicles as nanomedicine: hopes and hurdles in clinical translation. Int J Nanomedicine. 2019;14:8847‐8859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Wu Y, Deng W, Klinke DJ 2nd. Exosomes: improved methods to characterize their morphology, RNA content, and surface protein biomarkers. Analyst. 2015;140(19):6631‐6642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Bachurski D, Schuldner M, Nguyen P‐H, et al. Extracellular vesicle measurements with nanoparticle tracking analysis ‐ an accuracy and repeatability comparison between NanoSight NS300 and ZetaView. J Extracell Vesicles. 2019;8(1):1596016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Watson DC, Yung BC, Bergamaschi C, et al. Scalable, cGMP‐compatible purification of extracellular vesicles carrying bioactive human heterodimeric IL‐15/lactadherin complexes. J Extracell Vesicles. 2018;7(1):1442088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Lamparski HG, Metha‐Damani A, Yao J‐Y, et al. Production and characterization of clinical grade exosomes derived from dendritic cells. J Immunol Methods. 2002;270(2):211‐226. [DOI] [PubMed] [Google Scholar]
  • 91. Pachler K, Lener T, Streif D, et al. A good manufacturing practice‐grade standard protocol for exclusively human mesenchymal stromal cell‐derived extracellular vesicles. Cytotherapy. 2017;19(4):458‐472. [DOI] [PubMed] [Google Scholar]
  • 92. Bari E, Perteghella S, Catenacci L, et al. Freeze‐dried and GMP‐compliant pharmaceuticals containing exosomes for acellular mesenchymal stromal cell immunomodulant therapy. Nanomedicine (Lond). 2019;14(6):753‐765. [DOI] [PubMed] [Google Scholar]
  • 93. Kowal J, Arras G, Colombo M, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci USA. 2016;113(8):E968‐E977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Huang T, Banizs AB, Shi W, Klibanov AL, He J. Size exclusion HPLC detection of small‐size impurities as a complementary means for quality analysis of extracellular vesicles. J Circ Biomark. 2015;4:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Wang X, Hunter AK, Mozier NM. Host cell proteins in biologics development: Identification, quantitation and risk assessment. Biotechnol Bioeng. 2009;103(3):446‐458. [DOI] [PubMed] [Google Scholar]
  • 96. Greening DW, Xu R, Ji H, Tauro BJ, Simpson RJ. A protocol for exosome isolation and characterization: evaluation of ultracentrifugation, density‐gradient separation, and immunoaffinity capture methods. Methods Mol Biol. 2015;1295:179‐209. [DOI] [PubMed] [Google Scholar]
  • 97. Koh YQ, Almughlliq FB, Vaswani K, Peiris HN, Mitchell MD. Exosome enrichment by ultracentrifugation and size exclusion chromatography. Front Biosci (Landmark Ed). 2018;23:865‐874. [DOI] [PubMed] [Google Scholar]
  • 98. Kosanovic M, Milutinovic B, Goc S, Mitic N, Jankovic M. Ion‐exchange chromatography purification of extracellular vesicles. Biotechniques. 2017;63(2):65‐71. [DOI] [PubMed] [Google Scholar]
  • 99. Heath N, Grant L, De Oliveira TM, et al. Rapid isolation and enrichment of extracellular vesicle preparations using anion exchange chromatography. Sci Rep. 2018;8(1):5730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Busatto S, Vilanilam G, Ticer T, et al. Tangential flow filtration for highly efficient concentration of extracellular vesicles from large volumes of fluid. Cells. 2018;7(12):273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Contreras‐Naranjo JC, Wu HJ, Ugaz VM. Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. Lab Chip. 2017;17(21):3558‐3577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Koritzinsky EH, Street JM, Star RA, Yuen PS. Quantification of Exosomes. J Cell Physiol. 2017;232(7):1587‐1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Lötvall J, Hill AF, Hochberg F, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Exopharm . https://exopharm.com/the‐power‐of‐exopharms‐leap‐technology. Accessed June 1, 2020.
  • 105. ILIAS Biologics . http://www.iliasbio.com. Accessed June 1, 2020

Articles from American Journal of Reproductive Immunology are provided here courtesy of Wiley

RESOURCES