Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2022

Open Access 01.12.2022 | Review

Emerging role of exosomes in cancer progression and tumor microenvironment remodeling

verfasst von: Mahshid Deldar Abad Paskeh, Maliheh Entezari, Sepideh Mirzaei, Amirhossein Zabolian, Hossein Saleki, Mohamad Javad Naghdi, Sina Sabet, Mohammad Amin Khoshbakht, Mehrdad Hashemi, Kiavash Hushmandi, Gautam Sethi, Ali Zarrabi, Alan Prem Kumar, Shing Cheng Tan, Marios Papadakis, Athanasios Alexiou, Md Asiful Islam, Ebrahim Mostafavi, Milad Ashrafizadeh

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2022

Abstract

Cancer is one of the leading causes of death worldwide, and the factors responsible for its progression need to be elucidated. Exosomes are structures with an average size of 100 nm that can transport proteins, lipids, and nucleic acids. This review focuses on the role of exosomes in cancer progression and therapy. We discuss how exosomes are able to modulate components of the tumor microenvironment and influence proliferation and migration rates of cancer cells. We also highlight that, depending on their cargo, exosomes can suppress or promote tumor cell progression and can enhance or reduce cancer cell response to radio- and chemo-therapies. In addition, we describe how exosomes can trigger chronic inflammation and lead to immune evasion and tumor progression by focusing on their ability to transfer non-coding RNAs between cells and modulate other molecular signaling pathways such as PTEN and PI3K/Akt in cancer. Subsequently, we discuss the use of exosomes as carriers of anti-tumor agents and genetic tools to control cancer progression. We then discuss the role of tumor-derived exosomes in carcinogenesis. Finally, we devote a section to the study of exosomes as diagnostic and prognostic tools in clinical courses that is important for the treatment of cancer patients. This review provides a comprehensive understanding of the role of exosomes in cancer therapy, focusing on their therapeutic value in cancer progression and remodeling of the tumor microenvironment.

Graphical Abstract

Hinweise
Mahshid Deldar Abad Paskeh, Maliheh Entezari, and Sepideh Mirzaei contributed equally to this paper and are considered co-first authors.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CSC
Cancer stem cell
EVs
Extracellular vesicles
TME
Tumor microenvironment
ncRNAs
Non-coding RNAs
miRNA
MicroRNA
lncRNA
Long non-coding RNA
circRNA
Circular RNA
MVBs
Multi-vesicular bodies
ILVs
Intraluminal vesicles
ESCRT
Endosomal sorting complex required for transport
VTA1
Vesicle trafficking 1
VPS4
Vacuolar protein sorting-associated protein 4
TSG101
Tumor susceptibility gene 101 protein
HSP60
Heat shock protein 60
Rab
Ras-associated binding
SNARE
Soluble NSF-attachment protein receptor
STAT3
Signal transducer and activator of transcription 3
IL-6
Interleukin-6
MMP-9
Matrix metalloproteinase-9
ROS
Reactive oxygen species
BBB
Blood–brain barrier
ERK
Extracellular-signal-regulated kinase
SIRT2
Sirtuin 2
TFEB
Transcription factor EB
EphA2
Eph receptor A2
EMT
Epithelial-to-mesenchymal transition
EMT-TFs
EMT-inducing transcription factors
MMPs
Matrix metalloproteinases
TFF3
Trefoil factor 3
VEGF
Vascular endothelial growth factor
VEGFR2
VEGF receptor 2
NRP-1
Neuropilin-1
OSCC
Oral squamous cell carcinoma
NPS
Nasopharyngeal carcinoma
ANGPT2
Angiopoietin-2
HCC
Hepatocellular carcinoma
HIF-1α
Hypoxia inducible factor-1α
TEX
Tumor-derived exosomes
5-FU
5-Fluorouracil
CP
Cisplatin
TGF-β
Transforming growth factor-beta
Treg cells
T regulatory cells
PD-1
Programmed death-1
IDO
Indoleamine 2:3-dioxygenase
3/-UTR
3/-Untranslated region
PTEN
Phosphatase and tensin homolog
PTX
Paclitaxel
AA-PEG
Aminoethylanisamide-polyethylene glycol
RISC
RNA-induced silencing complex
HGF
Hepatocyte growth factor
PLK1
Polo-like kinase 1
EGFR
Epidermal growth factor receptor
shRNA
Short hairpin RNA
TA2
Tafazzin
MZF1
Myeloid zinc finger 1
DKK1
Dickkopf1

Introduction

Cancer remains one of the diseases that threaten the lives of many people around the world. It is the second leading cause of death worldwide after cardiovascular diseases. Cancer cells possess unique features such as high proliferation rate, self-renewal ability, cancer stem cell (CSC) characteristics, metastasis, and the ability to switch between different molecular pathways to develop drug resistance [14]. Based on these properties, novel therapeutics, including nucleic acid drugs and anti-cancer agents, have been developed to target cancer cells and suppress their progression [59]. In addition, novel methods such as the use of nanoparticles have been employed for targeted delivery of therapeutics to cancer cells [10].
Recently, attention has focused on the role of a new type of structure, called extracellular vesicles (EVs) in cancer [11, 12]. EVs originate from the cell membrane and are considered micro- or nanovesicles. These structures can be secreted by all prokaryotic and eukaryotic cells in an evolutionarily conserved manner [13, 14]. Initially, EVs were thought to be waste products of cells or entities formed by cellular damage [15]. However, further studies on EVs have shown that they have vital biological functions and are important cellular components [16, 17]. There are several types of EVs that are categorized based on their size, origin, and localization [1820]. The best known EVs include exosomes, microparticles, shedding vesicles, apoptotic bodies, tolerosomes, proteasomes, and prominosomes [21, 22]. There are two different mechanisms for the formation of EVs. In the first mechanism, EVs arise directly from cell membrane budding [15]. In the second mechanism, EVs arise during exocytosis of multivesicular bodies as part of the endocytosis system [23]. EVs are involved in biological functions in cells and play an important role in pathological conditions. They can transfer various molecules between cells and are a means of communication [24]. Therefore, special attention should be paid to their role in diseases, especially cancer [2530].
The present review focuses on the role of exosomes in cancer. This comprehensive review first provides an overview of the discovery of exosomes, their composition, and the pathway of their biogenesis, which are of important for understanding these structures. Then, we focus specifically on the role of exosomes in cancer by introducing a section on exosomes in tumor microenvironment (TME) remodeling and how they influence various cancer hallmarks, including proliferation, migration, and therapy response. Next, we discuss exosomal non-coding RNAs (ncRNAs) and how they can affect cancer cell progression. We then turn our attention to exosomes and the key molecular signaling pathways that regulate cancer progression. Finally, we provide insight into tumor-derived exosomes and the clinical applications of exosomes relevant to the treatment of cancer patients.

Exosome structure, isolation and dosing

Exosomes are double-membraned vesicles (30–150 nm in size; average particle size: 100 nm) secreted by different types of cells. Their specific functions depend on their origin. For example, exosomes originating from tumor cells provide cell-to-cell communication and are mainly involved in migration and invasion [31]. In the phospholipid membrane of exosomes originating from the parent cell, there are a variety of proteins and lipids [32, 33]. Among the lipid molecules, phosphatidylcholine, phosphatidylethanolamines, phosphatidylinositol, phosphatidylserine, and sphingomyelin are present in the exosome membrane. The composition and levels of these lipid molecules mainly influence the properties of exosomes. For example, the high stability of exosomes in body fluids and at different pH values is due to the high levels of sphingomyelin and phosphatidylinositol in their membrane. Therefore, these lipid molecules protect exosomes from degradation by proteolytic or lipolytic enzymes [34]. The phospholipid membrane of exosomes has lipid rafts containing proteins such as tyrosine kinase Src and glycosylphosphatidylinositol-containing proteins [35]. The presence of proteins in exosomes is a bit complex. Exosomes are thought to contain both general and specific proteins. The general or nonspecific proteins are present in all cell types, including CD63, tetraspanins, CD81, and CD9, whereas specific proteins include MHC II found in exosomes from dendritic cells and B lymphocytes, HER2 in exosomes from breast cancer, and EGFR in exosomes from gliomas [36]. It is worth noting that nonspecific proteins are critical for exosome function. Tetraspanins, for example, are nonspecific proteins that can interact with integrin or MHC molecules and form complexes. In addition to proteins, exosomes may also contain ncRNAs including microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) [3739].
Since exosomes are present in various body fluids, they can be considered as novel biomarkers for the detection and diagnosis of various diseases. Therefore, it is important to develop methods for their isolation. A total of six strategies have been developed for the isolation of exosomes, including ultracentrifugation, ultrafiltration, size exclusion chromatography, precipitation, immunoaffinity-based capture, and microfluidics. Each method has its own advantages and problems that should be addressed [4045]. Ultracentrifugation is capable of detecting exosomes based on their density, size, and shape, and its advantages include affordability, large sample capacity, and ability to isolate high concentrations of exosomes. The disadvantages of ultracentrifugation are the time-consuming process, the risk of exosome damage to exosomes from high-speed centrifugation, and the need for complex equipment [4648]. Ultrafiltration isolates exosomes based on size differences from other particles. This strategy is fast and portable but has drawbacks such as low purity, shear stress, exosome loss, and clogging [4951]. Size exclusion chromatography also uses size differences and has the advantage that it can accurately separate exosomes and isolate the intact exosomes without damaging them. Its disadvantage is the time-consuming process, which needs further advancement and development [5254]. The precipitation method is based on changing the solubility of exosomes, and its advantages include the ease of performance, applicability to large sample volumes, and little damage to exosomes. Its disadvantages include the time-consuming process and the possibility of precipitating other particles such as polymeric materials and proteins [55, 56]. Immunoaffinity-based capture is based on the interaction between antibodies and antigens. Advantages of this method include high purity and the possibility of subtyping, whereas problems include high cost, low yield, risk of antigen blockade, and loss of exosome functionality [57]. The final technique for exosome isolation is the microfluidic strategy, which has the advantage of being inexpensive, time-saving, and requiring only a small amount of sample, but it has low sensitivity [5862]. Further information on exosome isolation techniques has been reviewed elsewhere [63, 64].
The dosing of exosome has been the subject of debate and investigation in recent years. Three different methods have been used to determine exosome dosage, including cell equivalents, protein concentration, and/or specific quantitative analytical measurements using tools, with each with its own advantages and disadvantages. However, there is still a need to develop a standardized method for exosome dosing and currently available technologies suffer from accurate and precise assessment of exosomes at the level of individual vesicles. To improve the accuracy in exosome dosing, it is proposed to use multiple methods. For example, although the protein method that assesses total protein levels is fast and inexpensive, it may also assess proteins that are not exosome-related and may not indicate bioactive ingredients. TRPS, NTA, ELISA, cell equivalents, and flow cytometry are other methods for exosome dosing. A review by Willis and colleagues provides more details on techniques related to exosome dosing [65]. With regard to the use of exosomes in clinical trials, good manufacturing practices (GMPs) are important. Indeed, exosomes used in clinical trials should comply with GMPs. GMPs for exosomes consider three major factors, including upstream cell cultivation, downstream purification process, and exosome quality control [66].

Biogenesis route of exosomes

Exosomes are formed by the endocytic pathway after passing through several steps [67]. In the first step, invaginations of the cytoplasmic membrane generate an early secretory endosome. Then, biogenesis of multivesicular bodies (MVBs) occurs by inward sprouting, generating intraluminal vesicles (ILVs) surrounded by endosomes. Acidification is then required for maturation of the late endosomes. In the final step, the ILVs fuse with the cell membrane and the exosomes are released [68]. MVBs have a size of 250–100 nm and therefore multiple ILVs with a particle size of 30–150 nm can be formed within the MVBs [69]. A number of proteins are involved in the formation of ILVs and MVBs, and in cargo selection [70]. The best known proteins for exosome biogenesis are the endosomal sorting complexes required for transport (ESCRT), which consist of four members, including ESCRT-0, -I, -II, and -III, that play a special role in membrane formation and cargo sorting [71]. Association of ubiquitylated cargoes with lipid microdomains is performed by ESCRT-0 and ESCRT-I. Then, ESCRT-II and -III are involved in invagination and formation of MVBs and ILVs. ALIX (Apoptosis-linked gene 2-interacting protein X, encoded by PDCD6IP), VTA1 (Vesicle Trafficking 1), VPS4 (Vacuolar protein sorting-associated protein 4), and TSG101 (Tumor susceptibility gene 101 protein) are other proteins that help the ESCRT machinery in exosome biogenesis.
Of note, there is another pathway for exosome biogenesis that is independent of ESCRT. In this ESCRT-independent pathway, heat shock protein-60 (HSP60), HSP70, and HSP90 act as chaperones, and CD63, CD81, CD82, CD37, and CD9 act as tetraspanins, which play important roles in membrane formation and cargo binding to lipid microdomains. Therefore, the mechanisms of exosome biogenesis are divided into two categories: ESCRT-dependent and -independent mechanisms [13, 7275]. Among the tetraspanins, CD63 and CD81 are the most abundant proteins in the membranes of ILVs and are considered to be markers for exosomes [75, 76].
The preferred mechanism of exosome biogenesis (either ESCRT-dependent or -independent pathway) is determined by cargo and the specific cell type [74]. Two major proteins play notable roles in the transfer and fusion of MVBs: the Ras-associated binding (Rab) family of GTPases and the soluble NSF attachment protein receptor (SNARE) [72, 77, 78]. It is worth noting that some of the MVBs are not fused to cell membranes and are transferred to lysosomes for degradation [72, 77]. The ILVs secreted from MVBs are known as exosomes. There are some limitations to exosome biogenesis and related mechanisms that may be considered in future studies. The underlying mechanism of differentiation of MVBs destined for degradation or fusion with the cell membrane is not known and needs to be studied in detail. Another limitation is the mechanism by which the endocytic system regulates the percentage of MVBs destined for fusion with the cell membrane. In addition, how the sorting of materials from ILVs to MVBs is regulated is still unknown [79]. Figure 1 shows a schematic representation of the biogenesis of exosomes.

Exosomes and the tumor microenvironment

Most of the tumor mass is occupied by the TME, which comprises the stroma of the tumor [80]. Low oxygen levels, high lactate levels, extracellular acidosis, and poor nutrient content are prominent features of the TME [81, 82]. A variety of cells, including mesenchymal stem cells, fibroblasts, endothelial cells, and immune cells, are present in the TME and can secrete cytokines and growth factors [83]. Cancer-associated fibroblasts are one of the most abundant cells in the TME, creating conditions for tumor growth and progression [84, 85]. The interactions that occur in the TME and the activation/inhibition of signaling networks may determine tumor progression. Therefore, much attention has been devoted to understanding the interactions and developing targeted therapies for the TME [8688]. This section summarizes the role of exosomes in influencing TME components.
Macrophages are abundant in the TME and have two distinct phenotypes, including M1- and M2-polarized macrophages [89]. Changing the polarization of macrophages toward the M2 phenotype leads to tumor progression and an event that mediates therapy resistance [90, 91]. One of the molecular signaling pathways shown to play an oncogenic role is the signal transducer and activator of transcription 3 (STAT3) pathway [9296]. A recent experiment attempted to establish a link between STAT3, exosomes, and macrophage polarization in gliomas. The hypoxic state leads to the secretion of exosomes from glioma cells, which subsequently promote cancer progression by inducing M2 polarization of macrophages by triggering autophagy. Exosomes contain high levels of interleukin-6 (IL-6) and miRNA-155-3p. Activation of STAT3 occurs through IL-6, which in turn enhances the expression of miRNA-155-3p to induce autophagy. Due to a positive feedback loop, induced autophagy enhances STAT3 phosphorylation and thus tumorigenesis. Exosome-induced autophagy leads to M2 polarization of macrophages and paves the way for enhanced glioma progression [97]. Similar to glioma, the presence of hypoxia leads to the secretion of exosomes in the TME of colorectal carcinoma. These exosomes contain high levels of miRNA-210-3p, which inhibit apoptosis and promote the transition from G1 to S cycle by downregulating the expression of CELF2. Clinical investigation has also revealed that exosomes containing miRNA-210-3p have high levels in colorectal cancer patients and are correlated with an unfavorable prognosis [98]. Therefore, the signaling networks affected by exosomes may determine tumor progression by influencing TME [99, 100].
Now, the question arises: how can macrophages promote cancer cell migration and invasion? Polarized M2 macrophages are capable of secreting exosomes that promote hepatocellular carcinoma cell metastasis. M2 macrophages-derived exosomes transfer CD11b/CD18 to hepatocellular carcinoma cells. Subsequently, matrix metalloproteinase-9 (MMP-9) is activated, which significantly promotes cancer migration and metastasis [101]. Considering this important role of macrophages in cancer progression, exosomes targeting the TME have been developed. Galectin-9 siRNA was loaded into exosomes and then oxaliplatin was embedded as an antitumor agent and trigger of immunogenic cell death. Exosome-delivered galectin-9 siRNA suppressed M2 polarization of macrophages and oxaliplatin inhibited pancreatic cancer progression [102]. This study demonstrates how exosomes can reprogram the TME in favor of anticancer activity.
Because of the potential of exosomes to affect the TME, efforts have been made to develop exosomes that target the TME and regulate cancer progression. In a recent experiment, exosomes were loaded with manganese carbonyl to mediate their delivery to the TME. This resulted in increased formation of reactive oxygen species (ROS) and was able to reduce tumor proliferation by up to 90% during low-dose radiotherapy [103]. In addition, exosomes that are responsive to inflammatory TME were developed and, because of their ability to cross the blood–brain barrier (BBB), effectively transport doxorubicin into the TME and suppress glioma progression [104]. Therefore, exosomes may be considered promising candidates for targeting the TME and influencing cancer progression. When exosomes are present in the TME, a number of agents such as cytokines can alter their surface. For example, a recent experiment has shown that the surface of exosomes is modified by the CCL2 cytokine via binding to glycosaminoglycan side chains of proteoglycans, altering their cellular uptake and tropism toward certain cells and tissues [105]. Thus, if exosomes are to be manipulated, their interaction with components of the TME and the modification of their cellular uptake should be emphasized. Overall, exosomes exhibit interactions with the TME [106] and further experimentation is needed in basic research, in the development of exosomes for targeting the TME, and also in the introduction of these concepts into clinical courses (Fig. 2).

Exosomes and tumor angiogenesis

Angiogenesis and vasculogenesis are considered the two most important mechanisms for the formation of new vessels [107110]. However, there are major differences between angiogenesis and vasculogenesis. Vasculogenesis is involved in the formation of a whole vessel during embryonic development and is responsible for the development of the cardiovascular system. Thus, the endoderm releases vascular endothelial growth factor (VEGF), which induces VEGF receptor 2 (VEGFR2) on mesodermal cells in a paracrine manner [111]. Subsequently, mesodermal cells are converted into angioblasts or endothelial progenitor cells in the periphery of blood islands, whose fusion leads to the formation of primitive capillary networks [112]. Whereas vasculogenesis refers to the formation of new vessels, angiogenesis is the process of vessel formation from preexisting vessels [113]. The process of angiogenesis is inactive in adults and can be observed in physiological processes such as placental angiogenesis and embryo implantation [114116]. Both angiogenesis and vasculogenesis are critical to the process of wound healing and facilitate this process [117]. Recently, attention has focused on the role of angiogenesis in cancer. Tumor cells should induce angiogenesis to ensure their survival, grow, and spread to different parts of the body. It has been reported that cancer cells cannot grow beyond a size of 1–2 mm if angiogenesis does not occur. Therefore, a promising strategy in cancer treatment could be the inhibition of angiogenesis. The best known factor responsible for the induction of angiogenesis is VEGF, a cytokine involved in cancer progression [118]. The activity of VEGF in neovascularization is related to its binding to receptors such as VEGFR1 and VEGFR2. In addition, VEGF has an affinity for binding to cofactors such as neuropilin-1 (NRP-1) and NRP-2. VEGFR2 expression is mainly observed in endothelial cells, whereas VEGFR1 is found on macrophages, cancer cells, and fibroblasts. The use of monoclonal antibodies is of interest for inhibition of VEGF or VEGFR and suppression of angiogenesis [119].
Since induction of angiogenesis promotes cancer progression, tumor cells secrete exosomes to trigger this mechanism. In this case, multiple molecular signaling pathways are involved that ultimately induce angiogenesis. Oral squamous cell carcinoma (OSCC) cells are able to secrete exosomes containing miRNA-210-3p. Upregulation of miRNA-210-3p occurs in OSCC cells and acts as a tumor-promoting factor by increasing microvessel density (MD) and tumor grade. Mechanistically, exosomal miRNA-210-3p reduces ephrin A3 expression to stimulate the PI3K/Akt axis, trigger angiogenesis, and promote OSCC progression [120]. Indeed, exosomes function as tools of cell–cell communication and can influence the conditions that promote cancer progression. Nasopharyngeal carcinoma (NPC) cells have a high migratory capacity that has been linked to their ability to trigger angiogenesis. Exosomal miRNA-23a binds to the 3’-UTR of TSGA10 and reduces its expression, leading to angiogenesis and increased metastasis of NPC cells [121]. The question now arises: since exosomes are able to regulate angiogenesis, can we isolate exosomes that suppress angiogenesis and thereby impair cancer progression? The answer is affirmative, and such a strategy has already been used in the treatment of lung cancer. It has been reported that exosomes derived from Plasmodium-infected mice inhibit angiogenesis. To test this hypothesis, an animal model of Plasmodium infection was developed in an experiment and then exosomes were isolated for the treatment of lung cancer. These exosomes contained high levels of miRNA-16, -322, -497, and -17, and when injected into a mouse model of lung cancer, there was a significant reduction in the expression of VEGFR2, resulting in inhibition of angiogenesis and reduced tumor progression [122]. This experiment clearly indicates that more studies should be conducted on exosomes and their role in affecting angiogenesis. By developing isolation methods, such exosomes can be obtained and their potential for cancer treatment can be revealed.
In addition to miRNAs, exosomes may also contain lncRNAs involved in the regulation of angiogenesis. In this case, the induction or inhibition of angiogenesis depends on the role of lncRNA as a tumor-suppressor or tumor-promoter. The lncRNA GAS5 is thought to suppress lung cancer progression. Exosomes containing high levels of GAS5 stimulate apoptosis in lung cancer and impair its growth. To this end, exosomal lncRNA GAS5 reduces miRNA-29-3p expression to increase PTEN expression. Subsequently, activated PTEN signaling suppresses PI3K/Akt phosphorylation to inhibit angiogenesis [123]. The role of exosomal ncRNAs in cancer progression will be discussed mechanistically in the next sections. However, it is clear that one way to modulate cancer progression is to influence angiogenesis through exosomes.
Angiopoietin-2 (ANGPT2) is thought to mediate resistance to antiangiogenic therapy by destroying vascular stability and promoting angiogenesis [124]. Suppression of the ANGPT2/Tie2 axis is a promising target [125, 126] because studies have shown the role of this factor in angiogenesis of cancer angiogenesis and in inflammation [127, 128]. Hepatocellular carcinoma (HCC) cells are capable of secreting ANGPT2-containing exosomes. These exosomes are introduced into HUVECs by endocytosis, and increased expression of ANGPT2 induces angiogenesis that promotes cancer progression [129]. As more experiments are performed, the novel signaling networks involved in angiogenesis are revealed. Hypoxia is a common feature of the TME. Recent experiments have shown that hypoxia can induce the secretion of exosomes from tumor cells, which increases their stemness and proliferation rate [130, 131]. A similar phenomenon occurs in colorectal cancer, where hypoxia leads to the secretion of exosomes, which in turn promote both growth and migration of tumor cells. Inhibition of exosome secretion by silencing RAB27a impairs proliferation and growth of colorectal tumors. Under hypoxic conditions, hypoxia-inducible factor-1α (HIF-1α) induces the secretion of exosomes containing Wnt4a. Subsequently, the β-catenin signaling pathway is activated and the nucleus is translocated, leading to angiogenesis and colorectal cancer progression [132].
In the previous sections, we have shown that exosomes affect the TME. The interaction of exosomes with the components of the TME may influence angiogenesis and thus cancer progression. Several experiments have shown that macrophages can induce angiogenesis. Recruitment of macrophages can induce angiogenesis to enhance nerve regeneration [133]. In addition, reduction or depletion of macrophages suppresses angiogenesis [134]. Tumor-derived exosomes (TEX) are capable of carrying CD39/CD73 and adenosine, which are enzymatically active. The TEX leads to polarization of macrophages into the M2 phenotype via A2BR. Subsequently, M2 macrophages secrete angiogenic factors (ANGPT2, IL-8, MMP9, PF4, and TIMP-1) that induce angiogenesis and promote cancer progression [135]. Overall, the studies are consistent with the fact that angiogenic factors are strongly regulated by exosomes. Depending on the cargo of exosomes, they can act as tumor suppressive or tumor promoting factors to influence angiogenesis in cancer cells. Exosomes can affect various molecular signaling pathways such as MAPK, YAP, VEGF, and miRNAs in modulating angiogenesis in cancer cells (Table 1) [136141]. Figure 3 illustrates the role of exosomes in regulating angiogenesis in cancer cells.
Table 1
Exosomes and their association with angiogenesis in different cancers
Cancer type
In vitro/In vivo
Cell line/animal model
Signaling network
Remarks
Refs
Thyroid cancer
In vitro
In vivo
Nthy-ori-3–1 cells
miRNA-21-5p/TGFBI
miRNA-21-5p/COL4A1
miRNA-21-5p in exosomes is upregulated under hypoxic conditions
Angiogenesis is induced
TGFBI and COL4A1 are inhibited by miRNA-21-5p to promote angiogenesis and cancer progression
[217]
Esophageal squamous cell carcinoma
In vitro
In vivo
ECA109, KYSE410 and HET-1A cell lines
Nude mice
Angiogenesis is promoted by the increased levels of exosomes under hypoxic conditions
[218]
Head and neck squamous cell carcinoma
In vitro
In vivo
PCI-13 (HPV) and UMSCC47 (HPV+) cell lines
Mouse model
Functional reprogramming and phenotypic modulation are observed in endothelial cells
Vascular structure formation is increased
Proliferation and invasion are promoted
Angiogenesis is induced by exosomes carrying angiogenic proteins
[219]
Gastric cancer
In vitro
In vivo
SGC7901 cells
Xenograft model
miRNA-155/FOXO3a
FOXO3a is inhibited by miRNA-155 in exosomes to induce angiogenesis in gastric to drive cancer progression
[220]
Gastric cancer
In vitro
HUVECs
YB-1/VEGF
Exosomes derived from gastric cancer cells have high levels of YB-1
Apoptosis is inhibited, and metastasis and angiogenesis are enhanced
Protein and mRNA levels of VEGF are increased
[221]
Gastric cancer
In vitro
SGC7901 and MGC803 cells
miRNA-6785-5p/INHBA
INHBA expression is reduced by exosomes containing miRNA-6785-5p to impair migration and angiogenesis of cancer cells
[222]
Gastric tumor
In vitro
SGC7901 cells
miRNA-135b/FOXO1
FOXO1 expression is decreased by the overexpression of miRNA-135b in exosomes to induce angiogenesis and exert tumor-promoting effects
[223]
Endometrial cancer
In vitro
SPEC2 and ISK cells
LGALS3BP/PI3K/Akt/VEGFA
Cancer progression is enhanced by exosomes containing LGALS3BP
Associated with unfavorable prognosis
VEGFA expression is induced by triggering the PI3K/Akt axis
Angiogenesis is promoted
[224]
Breast cancer
In vitro
MDA-MB-231, MCF-7 and T47D cells
miRNA-100/mTOR/HIF-1α/VEGF
miRNA-100 is delivered by exosomes in breast cancer therapy
Angiogenesis is suppressed to impair cancer progression
VEGF expression is downregulated in a time-dependent manner
The mTOR/HIF-1α axis is suppressed
[225]
Breast cancer
In vitro
4T1 cells
miRNA-16/VEGF
VEGF is downregulated by exosomes containing miRNA-16
Angiogenesis is suppressed
Cancer progression is impaired
[226]
Ovarian cancer
In vitro
A2780 and HO-8910 cells
PKR1/STAT3
Ovarian cancer migration is promoted by exosomes containing PKR1 through induction of angiogenesis
Phosphorylation level of STAT3 is increased by PKR1
[227]
Ovarian cancer
In vitro
SKOV3 cells
miRNA-130a
miRNA-130a is delivered by exosomes
Angiogenesis is increased to promote cancer progression and trigger drug resistance
[228]
Small-cell lung cancer
In vitro
In vivo
H446 cells
Xenograft model
Profilin 2
Migration and tube formation capacity of endothelial cells are enhanced
Smad2/3 is stimulated by profilin 2 in H446 cells
Cancer development and metastasis are enhanced by exosomes
[229]
Colon cancer
In vitro
In vivo
HCT-15 cells
Nude mice
GDF15/Smad/periostin
Angiogenesis is enhanced by exosomes derived from cancer cells
Smad is inhibited by GDF15 to enhance periostin expression to promote angiogenesis
[229]
Colorectal cancer
In vitro
In vivo
LoVo and HT29 cells
miRNA-135b-5p
Angiogenesis is triggered, and proliferation and migration are enhanced
[230]
Renal cancer
In vitro
789-0 cells
hepaCAM/VEGF
VEGF is downregulated and angiogenesis is inhibited by exosomes enriched with hepaCAM
[231]
Renal cancer
In vitro
In vivo
786-0 cells
Nude mice
miRNA-27a/SFRP1/VEGF
SFRP1 is downregulated and VEGF expression is increased by miRNA-27a delivered by exosomes to trigger angiogenesis and promote cancer progression
[232]

Exosomes and tumor growth

Proliferation of cancer cells increases abnormally and is one of the factors by which cancer cells differ from normal cells. Increased cell cycle progression, inhibition of apoptosis, and glycolysis are considered to be major main factors in cancer growth [142146]. The aim of this section is to evaluate the role of exosomes in modulating proliferation of cancer cells. Rapidly dividing cancer cells require high levels of energy to sustain their proliferation. Oxidative phosphorylation is a slow process and cannot provide tumor cells with energy to meet their needs. Therefore, a switch in metabolism from oxidative phosphorylation to glycolysis is initiated. Therefore, suppression of glycolysis can be considered a promising strategy in cancer therapy [147150]. Exosomes have been shown to increase the growth rate of lung cancer cells via the induction of glycolysis. To this end, exosomes were isolated from irradiated lung cancer cells. They exhibited high levels of ALDOA and ALDH3A1, which stimulate glycolysis to increase lung cancer proliferation [151]. With regard to the close link between drug resistance and glycolysis, studies have attempted to elucidate a link between exosomes, glycolysis, and chemoresistance. Macrophages are capable of secreting exosomes that play a tumor-promoting role. A recent experiment has shown that macrophage-derived exosomes enhance glycolysis, mediating lung cancer cell resistance to cisplatin chemotherapy. Molecular pathway study shows that these exosomes have high levels of miRNA-3679-5p and decrease the expression of NEDD4L to promote the stability of c-Myc, leading to lung cancer growth, induction of glycolysis, and mediation of cisplatin resistance [151]. Therefore, exosomes can induce glycolysis in favor of cancer growth and induce drug resistance [152].
STAT3 signaling is an oncogenic pathway that inhibits apoptosis and cell cycle arrest and promotes growth and metastasis [153]. STAT3 induces EMT and promotes cancer invasion. Overexpression of STAT3 is associated with poor prognosis and triggers chemoresistance [154158]. Exosomes alter the polarization of macrophages and transform them into cancer-associated macrophages. The exosomes are enriched in gp130 and induce STAT3 signaling via IL-6 upregulation [159]. STAT3-containing exosomes are able to promote ovarian cancer progression by inducing an imbalance between T cells and tumor-associated macrophages in favor of immunosuppression [160]. Cyclin D1, MMP-2, and MMP-9 are upregulated by STAT3-containing exosomes and promote proliferation and invasion of breast cancer cells [161]. Similarly, hypoxic conditions in the TME enhance the ability of colon cancer cells to self-proliferate by upregulating STAT3 expression [162]. Overall, several molecular signaling pathways are affected by exosomes, and understanding their interaction may pave the way for the development of novel therapeutics [163165].
Apoptosis is an important signaling pathway regulated by exosomes in tumors. Inhibition of apoptosis may pave the way for tumor progression and resistance to therapy [166]. A recent experiment has shown that cancer-associated fibroblasts secrete exosomes containing miRNA-92a-3p, which act as a tumor-promoting factor and induce the Wnt/β-catenin axis, leading to inhibition of mitochondrial apoptosis and inducing resistance of colorectal cancer cells to 5-fluorouracil [167]. The ROS can induce apoptosis in cancer cells. It has been reported that modulation of the levels of ROS may be important for the response of cancer cells to therapy [168171]. In pancreatic cancer, exosomes containing miRNA-155 reduce the expression of DCK, an enzyme involved in the metabolism of gemcitabine. This is followed by an increase in superoxide dismutase and catalase, leading to a reduction in ROS and subsequent growth of cancer cells and mediating their resistance to chemotherapy [172, 173]. Interestingly, not only can apoptosis in cancer cells reduce their proliferation, but apoptosis in immune cells can also affect cancer progression. A recent experiment has shown that pancreatic cancer-derived exosomes are taken up by lymphocytes to induce p38 MAPK signaling and mediate apoptosis triggered by endoplasmic reticulum stress apoptosis to stimulate immunosuppression and pave the way for cancer progression [174]. On the other hand, exosomes derived from colorectal cancer cells stimulate extracellular signal-regulated kinase (ERK) to suppress apoptosis and promote growth [175]. Apoptosis as a mechanism of programmed cell death is closely related to autophagy. In general, autophagy is involved in cell homeostasis by degrading aged and toxic organelles and macromolecules. However, the activation of autophagy in cancer cells is controversial and requires further clarification because it plays both tumor-promoting and tumor-suppressive roles [176179]. A recent experiment has shown that sirtuin 2 (SIRT2) increases the mRNA stability of transcription factor EB (TFEB) and induces the release of exosomes to trigger autophagy and decrease apoptosis in non-small cell lung cancer cells [180]. Therefore, special attention should be paid to autophagy in cancer progression when studying apoptosis regulation by exosomes. Overall, studies support the fact that exosomes can either increase or decrease cancer cell proliferation [181185].

Exosomes and tumor metastasis

Cancer cell invasion threatens the lives of many cancer patients around the world by enabling the spread of tumor cells to various organs and tissues of the body and mediating their malignancy [186188]. Therefore, the factors involved in cancer metastasis should be highlighted to direct future experiments to target them [189192]. Exosomes have been shown to be critical regulators of cancer metastasis. RelA and RelB are able to decrease the levels of MCAM and CD146 adhesion molecules in the release of EVs, leading to breast cancer metastasis. Silencing of RelA and RelB decreases the organotropic ability of exosomes in vivo and significantly reduces their ability to promote breast cancer migration and invasion [193]. It appears that exosomes containing Eph receptor A2 (EphA2) are able to transfer metastatic potential to pancreatic cancer cells and promote their invasion [194]. In contrast, there are exosomes capable of suppressing the cancer cell metastasis. For example, migration and invasion of non-small cell lung cancer cells were significantly decreased by miRNA-let7e-containing exosomes. LSD1 is upregulated in lung cancer and reduces E-cadherin levels to promote migration. Exosomes containing miRNA-let7e are able to increase CDH1 expression via LSD1 down-regulation to impair lung cancer metastasis [195]. Therefore, exosomes are important modulators of cancer migration and invasion.
The molecular mechanisms responsible for cancer migration and invasion are influenced by exosomes. The epithelial-to-mesenchymal transition (EMT) is among the best known mechanisms involved in cancer migration and invasion [196]. The decrease in E-cadherin, and the increase in N-cadherin and vimentin mediate EMT-induced metastasis in cancer cells [197, 198]. There are a number of factors known as EMT-inducing transcription factors (EMT-TFs), including ZEB1/2, TGF-β, Snail, Slug, and Twist, which can stimulate EMT in cancer cells and promote tumor invasion [199, 200]. A recent experiment has shown that exosomes containing the integrin alpha 2 subunit (ITAG2) are able to induce EMT and enhance prostate cancer cell metastasis [201]. On the other hand, exosomes containing miRNA-204 exhibit anti-tumor activity and reduce lung tumor cell invasion and migration by inhibiting EMT. To this end, exosomal miRNA-204 reduces the expression of KLF7 to inhibit the Akt/HIF-1α axis, resulting in a reduction of lung cancer migration and invasion by inhibiting EMT [202]. The ascites of ovarian cancer secretes exosomes containing miRNA-6780b-5p, which increase cancer migration and invasion in patients. It appears that exosomes containing miRNA-6780b-5p induce EMT to promote ovarian cancer metastasis [203]. The ability of exosomes to inhibit or induce EMT depends on their cargo. For example, miRNA-381-3p plays a tumor-suppressive role and exosomes containing this miRNA suppress EMT-mediated metastasis of breast cancer cells [204]. Therefore, there is increasing evidence for the role of exosomes in regulating metastasis by targeting the EMT mechanism [205, 206].
In addition to EMT, matrix metalloproteinases (MMPs) are also involved in increasing cancer metastasis [207211], and recent experiments have confirmed this. MMP-2 induces EMT to increase squamous cell carcinoma metastasis, and it may act as an independent factor in patient prognosis [212]. A clinical experiment demonstrated overexpression of MMP-7 in bladder cancer, which is associated with unfavorable prognosis and shortened overall survival of patients [213]. Moreover, MMP-3 is involved in the induction of angiogenesis, which promotes cancer progression [214]. Therefore, suppression of MMP activity may be of interest to inhibit cancer metastasis. Overexpression of trefoil factor 3 (TFF3) leads to upregulation of MMP-2 and MMP-9, enhancing prostate cancer cell invasion. Mesenchymal stroma cell-derived exosomes containing miRNA-143 exhibit anti-tumor activity and inhibit TFF3 to downregulate MMP-2 and MMP-9, leading to suppression of metastasis [215]. In contrast, exosomes derived from renal cancer cells are able to increase the expression of MMP-9 to promote invasion [216]. Although some studies have focused on the interaction between exosomes and MMP, there is still a long way to go to uncover the signaling networks involved (Fig. 4).

Exosomes and cancer resistance

Drug resistance

In the field of cancer therapy, a variety of antitumor agents have been developed, including cisplatin, 5-fluorouracil (5-FU), sorafenib, and oxaliplatin [233]. However, long-term use of these chemotherapeutic agents leads to drug resistance and an unfavorable prognosis for cancer patients. A specific mechanism is responsible for chemoresistance. Among others, drug efflux, upregulation of anti-apoptotic factors, DNA damage repair, epigenetic changes, and the TME may influence drug resistance [234239]. The current section focuses on the potential role of exosomes in drug resistance of cancer cells.
A growing body of evidence suggests that exosomes are capable of influencing the response of cancer cells to chemotherapy [240]. The ability of exosomes to transport cargoes has made them promising agents in cancer chemotherapy. As nanostructures, exosomes can mediate the co-delivery of a miRNA-21 inhibitor and 5-FU in colon tumor chemotherapy. The 5-FU and miRNA-21 inhibitor were loaded into exosomes via electroporation. Systematic administration of exosomes containing the miRNA-21 inhibitor and 5-FU suppressed tumor growth in mice. Exosomes administration enhances cellular uptake and reduces miRNA-21 expression in favor of colon cancer suppression. Moreover, miRNA-21 inhibitor and exosomes loaded with 5-FU induce cell cycle arrest and apoptosis. These anti-tumor activities are mediated via the upregulation of PTEN and hMSH2 as tumor suppressor factors in colon cancer [241]. The process of exosome secretion, cargo transport, and involvement in drug resistance are complex and should be elucidated. The epithelial ovarian cancer cells are able to recruit macrophages and stimulate their tumor-associated phenotype. Hypoxia in the TME leads to the secretion of exosomes from macrophages containing high levels of miRNA-223 as a tumor-promoting factor. The process of mediating drug resistance is that miRNA-223 delivered by exosomes reduces PTEN expression to induce PI3K/Akt signaling. To establish a link between hypoxia and exosome secretion, patients with ovarian cancer were studied. It was found that overexpression of HIF-1α, a hypoxia marker, occurs in ovarian cancer patients and is associated with upregulation of miRNA-223. Therefore, complicated molecular pathways and mechanisms are involved in the secretion of exosomes and the triggering of chemoresistance [242]. Another experiment demonstrates the potential role of macrophage-derived exosomes in triggering drug resistance in pancreatic cancer. An interesting point is that exosomes may be involved in the inactivation of chemotherapeutic agents in triggering drug resistance. Macrophage-derived exosomes contain miRNA-365 as a tumor-promoting factor and are able to induce gemcitabine resistance in pancreatic cancer. To this end, exosome-derived miRNA-365 stimulates the cytidine deaminase enzyme to inactivate gemcitabine, leading to chemoresistance in pancreatic cancer [243].
In addition to inactivating chemotherapeutic agents, exosomes can direct cancer cells toward cell death. It has been reported that exosomes can be obtained from CSCs in pancreatic cancer. These exosomes contain miRNA-210, which can induce gemcitabine resistance via inducing mTOR signaling. Moreover, these exosomes suppress gemcitabine-mediated apoptosis and cell cycle arrest [244]. Consequently, various signaling networks are affected by exosomes in triggering chemoresistance. In addition, the accumulation of chemotherapeutic agents in tumor cells is impaired. Exosomes are able to induce efflux of cisplatin from ovarian cancer cells under hypoxic conditions, demonstrating that they can prevent internalization of chemotherapeutic agents. Furthermore, STAT3 plays an important role in this case. Overexpression of STAT3 in hypoxic condition is crucial for exosome release and triggering cisplatin resistance in ovarian cancer. Suppression of STAT3 signaling alters the levels of Rab7 and Rab27a proteins, preventing the secretion of exosomes [245].
Tumor cells exhibiting features of drug resistance are able to secrete exosomes that accelerates chemoresistance. Such a strategy has been studied in lung cancer. Exosomes derived from cisplatin-resistant lung cancer cells have high levels of miRNA-100-5p, which decrease the expression of mTOR, leading to cisplatin resistance [246]. In addition, exosomes may act as a means of communication between normal and cancer cells in inducing drug resistance. Endothelial cells are able to secrete exosomes with a particle size of 40–100 nm, which trigger EMT-mediated metastasis in nasopharyngeal carcinomas and mediate their resistance to chemotherapy [247]. Exosomes can be used to suppress chemoresistance. In one experiment, exosomes were used to deliver anti-miRNA-214 to gastric cancer cells to induce apoptosis and decrease proliferation and invasion, leading to drug sensitivity [248]. Overall, the studies are consistent with the fact that exosomes can affect the growth and invasion of cancer cells to influence their response to chemotherapy. They contain various cargoes and can modulate molecular signaling pathways in favor of chemoresistance or chemosensitivity. Such exosomes and associated signaling networks should be elucidated to prevent chemoresistance in cancer cells [249259]. Table 2 provides an overview of exosomes and their association with drug resistance in cancer. Figure 5 shows a schematic representation of exosomes in regulating drug action.
Table 2
Exosomes and their function in mediating drug resistance/sensitivity in cancer
Cancer type
Chemotherapeutic agent
Signaling network
Remark
Refs
Breast cancer
Adriamycin
Drug resistance is induced by the transfer of P-gp and UCH-L1 proteins through exosomes into the extracellular microenvironment
[260]
Breast cancer
Anthracycline and taxane agents
Chemoresistance is observed in breast cancer patients who had high levels of GSTP1-containing exosomes
[261]
Breast cancer
Adriamycin
MDR1
P-glycoprotein
Drug resistance is induced by exosomes by enhancing the expression of MDR1 and P-glycoprotein
Chemoresistance is inhibited by suppression of exosome formation and secretion by psoralen
[262]
Breast cancer
Gemcitabine
Autophagy
EMT/HIF-α
Autophagy is inhibited by exosomes containing siMTA1
EMT is suppressed
Tumor growth in vitro and in vivo is retarded
[263]
Liver cancer
Sorafenib
Selectivity of exosomes against cancer cells is increased by modifying the surface of exosomes
Drug resistance is suppressed by synergistic cancer chemotherapy with sgIQ 1.1 plasmid-loaded exosomes
[263]
Leukemia
Etoposide
Bax
Bcl-2
PARP
Caspase-3
Drug resistance is induced by exosomes derived from bone mesenchymal stem cells by increasing the expression of Bcl-2 and decreasing the expression of Bax, caspase-3, and PARP
[264]
Leukemia
Imatinib
Bax
Bcl-2
Caspase-3
Caspase-9
Apoptosis is prevented by exosomes derived from mesenchymal stromal cells, and leukemia cell survival is increased
The expression of Bax, caspase-3 and caspase-9 is downregulated, and the expression of Bcl-2 is increased
[265]
Leukemia
Imatinib
miRNA-328/ABCG2
Drug sensitivity is increased by decreasing ABCG2 expression through miRNA-328 in exosomes
[266]
Glioblastoma
Temozolomide
PD-L1/AMPK/ULK1/autophagy
Autophagy is induced by the exosomes containing PD-L1 through stimulation of the AMPK/ULK1 axis, which mediates drug resistance
[267]
Glioblastoma
Temozolomide
STAT3/miRNA-21/PDCD4
STAT3 is downregulated by a combination of temozolomide and pacritinib
miRNA-21 expression is reduced to upregulate the PDCD4 tumor suppressor
M2 polarization of macrophages is inhibited
Glioblastoma tumorigenesis is prevented
[268]
Non-small cell lung cancer
Cisplatin
miRNA-146a-5p
Low levels of miRNA-146a-5p are observed in cisplatin-resistant A549 cells and can be used to predict cancer recurrence
[269]
Oral cancer
Cisplatin
miRNA-155/FOXO3a
FOXO3a expression is enhanced by exosomes containing the miRNA-155-inhibitor
Mesenchymal-to-epithelial transition is triggered to suppress cancer cell migration and invasion
[270]
Hepatocellular carcinoma
Oxaliplatin
miRNA-214/P-gp
miRNA-214/SF3B3
P-gp and SF3B3 expression is decreased by exosomal miRNA-214
Drug sensitivity is increased
[271]
Hepatocellular carcinoma
Cisplatin
miRNA-199a-3p
Drug resistance is suppressed by apoptosis induction through the increased expression of miRNA-199a-3p delivered by exosomes
[272]
Prostate cancer
Docetaxel
CD44v8-10 mRNA
Drug resistance is mediated by the presence of CD44v8-10-containing exosomes in the serum of prostate cancer patients
[273]

Radio-resistance

Radiotherapy is another cancer treatment option that uses radiation to inhibit cancer progression and induce cell death [274]. However, due to specific conditions in the TME such as hypoxia, cancer cells could develop resistance to radiotherapy, and the factors involved in this phenomenon should be elucidated [275, 276].
Most experiments have focused on the relationship between exosomes and drug resistance. However, there are also a few studies examining the role of exosomes in radioresistance. For example, a recent experiment has shown that cancer-associated fibroblasts are able to secrete exosomes to promote stemness of colorectal tumors and trigger their clonogenicity and radioresistance. Mechanistically, these exosomes induce transforming growth factor-beta (TGF-β) to mediate radioresistance. When this signaling pathway is suppressed using antibodies, colorectal tumor progression is impaired and sensitivity to radiotherapy is increased [277]. In contrast, there are exosomes capable of suppressing radioresistance. Exosomes containing miRNA-34c suppress proliferation, invasion, and EMT in nasopharyngeal carcinomas. In addition, miRNA-34c-loaded exosomes induce apoptosis and mediate radiosensitivity. Molecular pathway study shows that miRNA-34c-loaded exosomes suppress the β-catenin signaling pathway, thereby increasing the sensitivity of nasopharyngeal carcinoma cells to radiotherapy [278]. In the previous section, it was shown that chemotherapy of cancer cells induces the secretion of exosomes. Moreover, chemoresistant cancer cells are capable of secreting exosomes, which favors their progression and promotes drug resistance [279, 280]. A recent experiment has shown that exosomes can be obtained from irradiated gastric cancer cells [281]. However, further studies are needed to determine whether exosomes are involved in the development of radioresistance.

Immune evasion and inflammation

Although few experiments have investigated the role of exosomes in immune resistance and evasion, these studies show that exosomes are promising candidates in this case because of their modulatory effect on immune cells. The T-regulatory cells (Treg cells) are well known because of their immunosuppressive effects. In breast cancer, the CD73 + Treg cells are able to facilitate immune evasion by producing adenosine. The exosomes containing the lncRNA SNHG16 increase the expression level of CD73 on Treg cells. To this end, exosomal SNHG16 decreases the expression of miRNA-16-5p via sponging to induce the TGF-β/SMAD5 axis, resulting in overexpression of CD73 on Treg cells. Therefore, the ability of exosomes to transmit SNHG16 may mediate overexpression of CD73 on Treg cells and lead to immunosuppression in breast cancer [282]. Programmed death-1 (PD-1) is a molecular pathway that causes T cell exhaustion and prevents their proliferation. Moreover, PD-1 induces apoptosis in cytotoxic T cells and inhibits their anti-tumor activity to mediate immune evasion. Binding of PD-L1 to PD-1 triggers this pathway [283]. A recent experiment has shown that exosomes derived from cancer-associated fibroblasts contain high levels of miRNA-92 as a tumor-promoting factor. Exosomal miRNA-92 mediates the interaction between LATS2 and YAP1 in breast cancer cells. Subsequently, YAP1 translocates to the nucleus and binds to the PD-L1 promoter to enhance its expression, leading to the apoptosis of T cells and a decrease in proliferation of these cytotoxic cells [284]. Exosomes can not only evade immune defences but also influence immune cells to promote cancer progression. Indeed, interactions between exosomes and immune cells can create optimal conditions for increased cancer growth and invasion. NF-κB signaling is related to the inflammatory process and may promote cancer progression. NF-κB expression in cancer is regulated by other molecular signaling pathways, of which miRNAs are the best known [285]. On the other hand, there is growing evidence that chronic inflammation and pro-inflammatory cytokines promote cancer progression [286288]. A recent experiment has shown that exosomes derived from breast cancer cells have high levels of miRNA-183-5p and are able to decrease the expression of PPP2CA. Decreased expression of PPP2CA paves the way for triggering NF-κB signaling and mediating chronic inflammation. In addition, this signaling network increases the levels of pro-inflammatory cytokines such as IL-1β, IL-6 and TNF-α. Therefore, the transmission of miRNA-183-5p by exosomes and its effect on inflammation may promote the proliferation and invasion of breast cancer cells [289].
TGF-β mediates immune evasion of breast cancer cells. To this end, TGF-β increases the levels of PD-L1 in exosomes and stimulates the dysfunction of cytotoxic CD8 T cells [290]. Another experiment shows that exosomes derived from multiple myeloma suppress apoptosis and increase the growth rate of Treg cells, triggering immune dysfunction [291]. In addition, exosomes are able to promote the progression of gastric cancer by suppressing the maturation of dendritic cells [292]. Exosomes containing indoleamine 2,3-dioxygenase (IDO) may induce T-cell dysfunction via triggering the IL-6/TNF-α axis [293]. Future experiments may focus on targeting exosomes in preventing immune evasion and suppressing inflammation to impair cancer progression [294, 295].

Exosomal miRNAs

miRNAs are endogenous, short noncoding RNAs with a length of 19–24 nucleotides that can regulate gene expression at the posttranscriptional level by binding to the 3’-untranslated region (3’-UTR) of target mRNAs [296, 297]. Recent experiments have shed light on the role of miRNAs in cancer. For example, hypoxic conditions enhance lung cancer progression by decreasing the expression of miRNA-495 and miRNA-5688 and subsequently increasing IL-11 levels [298]. Moreover, decreased expression of miRNA-100 and miRNA-125b paves the way for overexpression of IGF2 and subsequent cancer stem cell features in hepatocellular carcinoma [299]. Further studies have shown that miRNAs can be considered as reliable biomarkers for cancer diagnosis [300, 301]. Because exosomes are capable of transmitting miRNAs, we dedicated this section to the study of exosomal miRNAs in the regulation of cancer progression.
In the previous sections, the role of exosomes in cancer progression has been clearly demonstrated as they affect the TME and the therapeutic response of cancer cells. It has been discussed that exosomes may contain various genes that influence cancer progression. The current section focuses specifically on exosomal miRNAs and how they may modulate cancer progression. In a recent experiment, exosomes were isolated exosomes by centrifugation from colorectal cancer cells infected with Fusobacterium nucleatum and transferred to uninfected cancer cells. The exosomes were found to contain high levels of miRNA-1264, miRNA-92b-3p, and miRNA-27a-3p, which are able to enhance metastasis and tumor stage of colorectal cancer [302]. There is increasing evidence that hypoxic conditions in the TME significantly promote carcinogenesis in gastric cancer [303, 304]. It appears that hypoxia induces the secretion of exosomes from gastric cancer cells. These exosomes promote both growth and migration of gastric cancer. These exosomes contain miRNA-301a-3p, which acts as a tumor-promoting factor and increases the stability of HIF-1α and inhibits its degradation by targeting PDH3 and hydroxylating HIF-1α subunits. Moreover, there is a positive feedback loop between HIF-1α and miRNA-301a-3p in enhancing the proliferation and invasion of gastric cancer cells. Clinical investigation shows that exosomal miRNA-301a-3p is upregulated in gastric cancer patients and mediates peritoneal metastasis [305].
The PI3K/Akt axis is a trigger of cancer progression and its induction promotes cancer cell proliferation and invasion [306308]. In addition, the PI3K/Akt axis enhances the aggressive behavior of cancer and is associated with drug resistance. PTEN is the negative regulator of the PI3K/Akt axis and increasing its expression is a promising strategy to interrupt cancer progression [309311]. A recent experiment shows that exosomes containing miRNA-22-3p have a tumor suppressive effect and prevent colorectal cancer progression by downregulating PI3K/Akt [312]. In addition to increased proliferation, exosomes may facilitate the transfer of apolipoprotein E between cells to induce the PI3K/Akt axis, which mediates cytoskeletal remodeling and promotes gastric cancer cell migration and invasion [313]. Therefore, the PI3K/Akt axis is strongly regulated by exosomes [314]. The question now arises: is there a link between exosomes and PTEN as upstream mediators of the PI3K/Akt axis? The answer is positive, and this potential has been confirmed in several experiments. In non-small cell lung cancer, exosomes containing miRNA-126 are able to enhance PTEN expression in suppressing the PI3K/Akt axis and impair metastasis. The in vivo experiment showed that miRNA-126 reduced lung cancer metastasis by modulating the PTEN/PI3K/Akt axis [315]. On the other hand, hypoxia leads to the secretion of exosomes from colorectal cancer cells. The exosomes contain high levels of miRNA-410-3p, which decrease PTEN expression to induce the PI3K/Akt axis and promote cancer cell invasion. Moreover, exosomal miRNA-410-3p is associated with unfavorable prognosis of colorectal cancer patients [316]. The growth rate of esophageal cancer cells is significantly increased by the transfer of miRNA-93-5p through exosomes and the downregulation of PTEN [317].
Another molecular signaling pathway involved in cancer progression is the Wnt/β-catenin axis. Nuclear translocation of β-catenin promotes cancer growth and invasion [318320]. Activation of Wnt signaling is associated with poor prognosis. In addition, the Wnt/β-catenin axis can mediate features of drug resistance in cancer cells [321325]. Exosomes are able to induce Wnt5b signaling, thereby increasing the progression of lung cancer cells [326]. The exosomes containing miRNA-320a act as tumor-promoting factors and reduce the expression of SOX4. As a result, Wnt/β-catenin activation occurs, which significantly promotes the growth and metastasis of lung cancer cells [327]. Another experiment shows that exosomes from breast cancer cells contain high levels of miRNA-146a, which reduces the expression of TXNIP to induce the Wnt/β-catenin axis, leading to activation of cancer-associated fibroblasts in the TME and promoting breast cancer progression [328]. Therefore, regulation of Wnt signaling by exosomes modulates cancer progression [329].
One of the increasing challenges in breast cancer is bone metastasis, which is associated with pain, decreased overall patient survival, and an unfavorable prognosis. Therefore, efforts have been made to uncover the role of exosomal miRNAs in bone metastasis of breast cancer cells in order to target them in future experiments. A clinical study has shown that serum exosomes containing miRNA-21 promote bone metastasis in breast cancer patients [330]. Although these studies demonstrate the tumor-promoting role of exosomal miRNAs, there are also experiments showing that exosomal miRNAs can suppress cancer progression. In pancreatic cancer cells, exosomal miRNA-34a can effectively enter the cell membrane and decrease the expression of Bcl-2 to induce apoptosis and reduce growth and viability. The in vivo experiment on nude mouse xenografts also demonstrated the role of exosomal miRNA-34a in retarding tumor growth [331]. In addition to apoptotic factors, other signaling networks responsible for cancer progression may also be influenced by exosomal miRNAs. It is suggested that exosomal miRNA-210 is a tumor-promoting factor in lung cancer. Secretion of exosomal miRNA-210 by cancer-associated fibroblasts significantly promotes lung cancer migration and invasion. Molecular pathway study reveals that exosomal miRNA-210 induces the PI3K/Akt axis via PTEN down-regulation to induce EMT and enhance lung cancer cell metastasis [332]. UbiA prenyltransferase domain-containing protein 1 (UBIAD1) is downregulated by exosomal miRNA-4644 via binding to its 3’-UTR to enhance bladder cancer cell invasion [333]. Thus, exosomal miRNAs influence a variety of molecular pathways in regulating cancer progression [334337].

Exosomal long noncoding RNAs

Recently, lncRNAs have attracted considerable attention because of their potential role in modulating of various molecular signaling pathways in cancer therapy [338341]. Briefly, lncRNAs are RNA molecules longer than 200 nucleotides and their function differs depending on their localization in the nucleus or cytoplasm [342, 343]. There are five types of lncRNAs and they are able to affect proteins and genes under physiological and pathological conditions [93, 344347]. The lncRNA DILA1 functions as a tumor-promoting factor and increases the stability of cyclin D1 to promote breast cancer progression and mediate resistance to tamoxifen [348]. Upregulation of lncRNA ENO1-IT1 by the gut microbiota mediates glycolysis and increases the proliferation rate of colorectal cancer cells [349]. Similar to miRNAs, lncRNAs may function as diagnostic and prognostic tools in cancer [350].
Similar to miRNAs, lncRNAs can also be transferred between cells via exosomes. Depending on the function of lncRNAs, they can reduce or promote cancer progression. The lncRNA H19 is considered a tumor-promoting factor because its upregulation induces drug resistance and promote both proliferation and invasion of cancer cells [351]. Exosomes transfer lncRNA H19 to non-small cell lung cancer cells to inhibit apoptosis and induce resistance to gefitinib chemotherapy [352]. Cancer cell migration is also regulated by exosomal lncRNAs. The lncRNA linc-ROR can be transferred into the TME to promote distant metastasis through EMT induction [353]. The exosomal lncRNAs are able to modulate the expression level of miRNAs to target other molecular pathways. The exosomal lncRNA CASC15 is overexpressed in osteosarcomas and increases growth and metastasis. Silencing of CASC15 impairs progression of osteosarcoma cells. Further studies show that exosomal CASC15 decreased the expression of miRNA-338-3p by sponging and increases the expression of RAB14 in osteosarcomas [354]. Delivery of lncRNAs through exosomes is a challenge for the treatment of some kinds of tumor types, particularly brain tumors. The blood–brain barrier (BBB) is an obstacle that prevents antitumor drugs from entering the brain and limits our ability to target brain tumors [355]. However, exosomes are able to disrupt BBB when transporting lncRNAs into the brain. A recent experiment has shown that exosomes are capable of crossing the BBB and transport the lncRNA GS1-6000G8.5 into the brain and mediate metastasis of breast cancer cells to the brain [356].
Due to the interaction between lncRNAs and miRNAs, downregulation of tumor-promoting lncRNAs may pave the way for upregulation of miRNAs with anti-tumor activity. It has been reported that downregulation of the exosomal lncRNA SBF2-AS1 in polarized M2 macrophages leads to the expression of miRNA-122-5p, a tumor suppressor factor. Subsequently, upregulated miRNA-122-5p suppresses pancreatic cancer progression via inhibition of XIAP [357]. As for the ability of exosomal lncRNAs to regulate apoptosis pathways, they can modulate the therapeutic response of cancer cells. The exosomal lncRNA UCA1 shows overexpression in breast cancer cells (MCF-7) and suppresses apoptosis via downregulation of caspase-3 to mediate tamoxifen resistance [358]. In addition to chemotherapy, exosomal lncRNAs regulate the response of cancer cells to radiotherapy. Because of the tumor-promoting role of the lncRNA HOTAIR, its transfer to laryngeal cancer cells via exosomes induces the expression of E2F2 via downregulation of miRNA-454-3p. This accelerates the progression of laryngeal cancer and reduces their sensitivity to radiotherapy [359]. Therefore, the identification of exosomal lncRNAs may increase our understanding of the factors involved in cancer progression and develop novel therapeutics in the near future [360363].

Exosomal circular RNAs

CircRNAs are another subset of noncoding RNAs that have a covalently closed loop structure and exhibit vital functions under physiological and pathological conditions [364367]. Aberrant expression of circRNAs is observed in various cancers. The hsa-circRNA-000166 increases the progression of colorectal cancer by downregulating miRNA-326 and subsequently overexpressing LASP1 [368]. Downregulation of miRNA-665 by circ-100876 occurs in gastric cancer, which triggers EMT via upregulation of YAP1 [369]. Experiments have shown that circRNAs affect cancer growth and metastasis mainly by regulating the expression of miRNAs [369, 370].
The circRNA IARS (circ-IARS) is thought to promote cancer metastasis. This circRNA is located in exosomes and enters HUVECs to increase cancer metastasis. Exosomal circ-IARS decreases overall survival and increases metastasis and TNM stage. Mechanistically, exosomal circ-IARS decreases the levels of miRNA-122 and ZO-1, whereas it increases the levels of RhoA and RhoA-GTP and increases the permeability of endothelial monolayers. Moreover, exosomal circ-IARS enhances F-actin expression and focal adhesion to promote invasion and metastasis [371]. The Wnt signaling pathway is related to cancer proliferation and metastasis. In the context of Wnt pathway, β-catenin translocates to the nucleus to promote cancer progression [372]. The exosomal circ-ABCC1 is overexpressed in colorectal cancer and promotes stemness and invasion. Mechanistically, circ-ABCC1 induces β-catenin to enhance colorectal cancer progression [373]. Similar to lncRNAs, exosomal circRNAs can regulate the response of cancer cells to chemotherapy. Exosomal circ-0002130 shows overexpression in lung cancer and mediates osimertinib resistance. To this end, exosomal circ-0002130 reduces miRNA-498 expression via sponging to enhance GLUT1, HK2, and LDHA expression, leading to lung cancer progression and drug resistance [374]. Another experiment shows how exosomal circRNAs can regulate drug sensitivity via affecting HK2. A recent experiment has shown that exosomal circ-0008928 can increase lung cancer progression and glycolysis. Indeed, exosomal circ-0008928 increases the proliferation rate of lung cancer cells via inducing glycolysis and then, decreases their sensitivity to cisplatin. Molecular pathway study shows that exosomal circ-0008928 enhances HK2 expression in triggering glycolysis and mediating drug resistance in lung cancer [375].
Exosomal circRNAs can also be considered as diagnostic and prognostic tools. For example, the expression levels of circ_0047921, circ_0056285, and circ_0007761 can be used to diagnose non-small cell lung cancer in Chinese. In addition, circ-0056285 is positively associated with the clinical stage and may increase lymph node metastasis [376]. The potential of exosomes as diagnostic and prognostic tools will be specifically discussed in the next sections. However, exosomal circRNAs can be used independently in this case [377380]. All in all, exosomal ncRNAs regulate proliferation, invasion, immune response and drug sensitivity of cancer cells and can be considered as diagnostic and prognostic factors in cancer (Fig. 6, Table 3) [381389].
Table 3
The exosomal ncRNAs in cancer cells
Exosomal ncRNA
Signaling network
Cancer type
Remarks
Refs
miRNA-34a
Breast cancer
Proliferation of cancer cells is suppressed
[390]
miRNA-145
MMP-9
TP53
Breast cancer
Apoptosis is induced
Metastasis is inhibited
[391]
miRNA-21-5p
ZNF367
Breast cancer
Cancer cell invasion is suppressed by downregulation of ZNF367
[392]
miRNA-5100
CXC12/CXCR4/EMT
Breast cancer
CXC12/CXCR4 axis is suppressed by miRNA-5100, which acts as a tumor suppressor
EMT is inhibited, and cancer cell invasion and migration are decreased
[393]
miRNA-3613-3p
SOCS2
Breast cancer
Cancer cell proliferation and metastasis are enhanced
SOCS2 is downregulated
[394]
miRNA-423-5p
Breast cancer
The sensitivity of breast cancer cells to cisplatin is reduced
[395]
miRNA-19b-3p
PTEN/EMT
Esophageal cancer
miRNA-19b-3p is upregulated
EMT is induced by exosomal miRNA-19b-3p by downregulating PTEN
Apoptosis is inhibited
Growth and metastasis of cancer cells are enhanced
[396]
miRNA-124
EZH2
Pancreatic cancer
Exosomal miRNA-124 is downregulated
Apoptosis is induced, EMT is inhibited and cancer cell migration is decreased by miRNA-124
EZH2 is downregulated by miRNA-124
[397]
miRNA-21-5p
miRNA-155-5p
BRG1
Colon cancer
miRNA-21-5p and miRNA-155-5p are transferred from exosomes
BRG1 expression is reduced
M2 polarization of cancer cells is induced
Cancer metastasis is enhanced
[398]
miRNA-34c-3p
Integrin α2β1
Non-small cell lung cancer
Metastasis and invasion of A549 cells are promoted by increased expression of integrins
[399]
miRNA-7
YAP
Lung cancer
YAP expression is inhibited and cancer cell sensitivity to gefitinib is increased
[400]
miRNA-126a
Lung cancer
Secretion of exosomes by lung cancer cells is induced by exposure to doxorubicin
Cancer cell migration and invasion are increased by exosomal miRNA-126a
[401]
miRNA-122
Hepatocellular carcinoma
Sensitivity of cancer cells to chemotherapy is increased by exosomal miRNA-122
[402]
miRNA-302b
ERK1/2
MMP-9
TGFβRII
Lung cancer
Cancer cell growth and invasion are inhibited
ERK1/2, MMP-9, and TGFβRII are downregulated
[403]
miRNA-21
PDCD4
Lung cancer
Lung cancer proliferation is increased
Anti-tumor immunity is suppressed by the proliferation of myeloid-derived suppressor cells
PDCD4 is downregulated
[404]
miRNA-375
ENAH
Esophageal cancer
Cancer progression is suppressed by decreasing the expression of ENAH
[405]
miRNA-146b
miRNA-222
Papillary thyroid cancer
Proliferation of cancer cells is increased
[406]
miRNA-200b
KLF6
Ovarian cancer
KLF6 is downregulated by miRNA-200b
M2 polarization of macrophages is induced
[407]
miRNA-92b-3p
SOX4
Ovarian cancer
Cancer progression is suppressed by inhibiting angiogenesis
SOX4 is downregulated
[408]
miRNA-224-5p
Renal cancer
Invasion and growth of cancer cells are suppressed
[407]
miRNA-1228
MMP-14
Gastric cancer
Cancer progression is suppressed by downregulation of MMP-14
[408]
lncRNA ZFAS1
Gastric cancer
Association with lymph node metastasis and TNM stage is observed
EMT is induced
Apoptosis is inhibited
[409]
lncRNA KCNQ1OT1
Colorectal cancer
miRNA-30a-5p/USP22/PD-L1
Immune evasion is induced
CD8 + T cell response is suppressed
Expression of miRNA-30a-5p is decreased by acting as ceRNA
USPP22 expression is upregulated to prevent PD-L1 ubiquitination
PD-L1 expression is enhanced
[410]
lncRNA HOTAIR
Breast cancer
ErB2
A positive association is observed between HOTAIR and ErB2
HOTAIR expression is increased by ErB2 in a MAPK-dependent manner
[411]
LINC01133
Bladder cancer
Wnt
Low levels of LINC01133 in exosomes from bladder cancer cells are observed
Wnt signaling is suppressed to impair cancer cell growth and metastasis
[412]
Circ-ABCC1
Colorectal cancer
Wnt/β-catenin
Cancer cell progression is enhanced by circ-ABCC1 via induction of β-catenin signaling
[376]
Circ-0002130
Non-small cell lung cancer
miRNA-498/HK2-GLUT1-LDHA
Cancer cell proliferation and invasion are increased in vitro and in vivo
Osimertinib resistance is observed
miRNA-498 is downregulated via sponging
Expression of HK2, GLUT1, and LDHA is increased
[374]
Circ-0008928
Non-small cell lung cancer
miRNA-488/HK2
Glycolysis, proliferation and cisplatin resistance of cancer cells are induced
Expression of miRNA-488 is decreased to induce HK2 expression
(375)

Exosomes as carrier systems

Anti-tumor agents

The previous sections have obviously shown that exosomes can affect cancer progression in several ways and are able to modulate the TME. These effects are based on exosome cargo. In this section, we discuss how exosomes can be used to deliver anti-tumor agents in cancer therapy. Remarkably, exosomes can deliver both synthetic and natural agents. In a recent experiment, exosomes with triptolide were used in the treatment of ovarian cancer. The exosomes showed high encapsulation efficiency and were able to slow tumor growth in vivo. Triptolide-loaded exosomes induce apoptosis in ovarian cancer cells and suppress their proliferation and viability [413]. Paclitaxel (PTX) is an anticancer agent that arrests the cell cycle by disrupting microtubule polymerization. Some cancer cells have developed resistance to PTX chemotherapy. Various techniques including nanoscale delivery systems have been developed to suppress chemoresistance. In one study, exosomes were used as delivery vehicles for PTX in lung cancer therapy. Exosomes were derived from macrophages and then modified with aminoethylanisamide-polyethylene glycol (AA-PEG) to selectively target sigma receptors that are upregulated on the surface of lung cancer cells. These exosomes are preferentially internalized into lung cancer cells and release PTX to suppress lung cancer cell progression [414]. One of the advantages of exosomes is their biocompatibility. In addition, they can deliver drugs as well as act as and imaging agents, which is referred to as theranostics. In a recent experiment, exosomes were isolated from cancer cells (e.g., HeLa cells) and then loaded with doxorubicin. In addition, silver nanoclusters were loaded into doxorubicin-coated exosomes. These exosomes enable imaging while delivering doxorubicin to suppress cancer progression, while exhibiting high biocompatibility and safety profile [415]. In the same study, exosomes were also used to deliver geldanamycin as an HSP90 inhibitor to affect the growth rate of cancer cells [416]. Drug-loaded exosomes can also regulate the TME in favor of cancer therapy. It has been reported that exosomes derived from M1-polarized macrophages can be loaded with PTX. PTX-loaded exosomes induced a pro-inflammatory environment and enhanced inflammation, which promoted the upregulation of caspase-3 expression, triggered apoptosis, and the subsequent enhancement of the anti-tumor activity of PTX [417]. Overall, these studies suggest that exosomes are promising candidates for drug delivery. Further experiments should be performed to elucidate their role in drug delivery, their encapsulation efficiency, and how their surface can be modified to increase their selectivity toward cancer cells [418421].

Genetic tools

Small interfering RNAs (siRNAs) are double-stranded RNA molecules of up to 25 nucleotides in length. They are produced from mRNA and lncRNAs via the function of the RNase III enzyme Dicer [422]. The actual function of siRNA is achieved it is incorporated into the RNA-induced silencing complex (RISC) to direct the RNAi machinery to target mRNA for degradation after complementary sequences are found. Recently, siRNA has paved the way to treat various diseases in preclinical and clinical research, such as viral infections, neurological disorders, ocular diseases, autoimmune diseases, and cancer [421, 423]. Although siRNA has shown great capacity in suppressing gene expression and subsequently treating disease, naked siRNA appears to require modification in alleviating disease. Degradation of siRNA by RNase enzymes, tumor barriers, and off-targeting are drawbacks of siRNA that can be solved using delivery systems [424, 425]. Another genetic tool used in cancer therapy is the CRISPR/Cas system. The CRISPR/Cas9 system is the best known type of CRISPR system that has recently been used in the treatment of diseases [426]. The CRISPR/Cas9 system was discovered in prokaryotes and its main function is adaptive immunity [427]. The CRISPR/Cas9 system consists of three main components, including Cas9, sgRNA, and tracrRNA. The specificity, efficiency, and accuracy of the CRISPR/Cas9 system are provided by sgRNA. Cas9 acts as a scissor and is responsible for the destruction of double-stranded DNA [428]. Various experiments have been conducted on the use of CRISPR/Cas9 system in cancer therapy. CRISPR/Cas9 is able to target fusion oncogenes or transcription factors to suppress cancer progression and reduce growth and mortality [429]. Downregulation of ZEB1 and ZEB2 by the CRISPR/Cas9 system significantly reduces lung cancer cell migration and invasion [430]. The present section addresses the role of exosomes in the delivery of siRNA and the CRISPR/Cas system in cancer therapy.

siRNA

In one experiment, exosomes were isolated from embryonic kidney cells (HEK-293 cells) by ultracentrifugation and loaded with siRNA. The resulting exosomes had a diameter of 107 nm and an encapsulation efficiency of 10–20%. The exosomes efficiently transported siRNA to PANC-1 cells, demonstrating their potential as a transport system [431]. Induction of apoptosis is an ideal strategy to suppress cancer proliferation. To this end, Bcl-2-siRNA was loaded into exosomes and its anti-tumor activity against digestive system tumors was evaluated. Bcl-2-siRNA-loaded exosomes penetrated the cell membrane and delivered siRNA, resulting in apoptosis induction and reduced tumor growth. The anti-tumor activity of these exosomes was confirmed in both in vitro and in vivo experiments [432].
Hepatocyte growth factor (HGF) was first identified in mouse liver and is considered a cytokine with physiological functions in cell proliferation, survival, and migration [433, 434]. Recent experiments have revealed the tumor-promoting role of HGF in cancer. Overexpression of HGF enhances the growth and invasion of cervical cancer cells via affecting c-Met [435]. By inducing the c-Met/PI3K/Akt axis, HGF induces EMT and mediates drug resistance in pancreatic cancer [436]. HGF-siRNA-loaded exosomes may serve as nanostructures for cargo transport in gastric cancer treatment. These exosomes effectively transport siRNA to gastric cancer cells, leading to a significant reduction in their growth and migration and inhibition of angiogenesis [437]. Polo-like kinase 1 (PLK1) is another tumor-promoting factor in cancer. Overexpression of PLK1 inhibits autophagic cell death in prostate cancer [438]. In addition, Silencing of PLK1 suppresses breast cancer cell migration and invasion and promotes their sensitivity to drugs [439]. PLK1-siRNA was introduced into exosomes by electroporation, and exposure of bladder cancer cells to these exosomes resulted in a significant decrease in PLK1 mRNA levels and subsequent cancer eradication [440].
Efforts are underway to engineer exosomes to increase their selectivity toward cancer cells. One of the promising methods is to modify exosomes with ligands. The exosomes carrying DARPin G3 on their surface can bind to HER2/Neu on breast cancer cells. These targeted exosomes deliver TPD52-siRNA and reduce the expression of HER2/Neu by up to 70% [441]. Future experiments may therefore focus on the development of engineered and surface-modified exosomes for cancer therapy. Another ligand that can be used for surface modification of exosomes is the tLyp-1 peptide with the amino acid sequence CGNKRTR. The tLyp-1 peptide is able to bind to receptors such as neuropilin-1 (NRP1) and neuropilin-2 (NRP2), which are overexpressed on the surface of lung cancer cells [442444]. Recently, tLyp-1 exosomes were used for siRNA delivery. These nanostructures were 100 nm in diameter and, thanks due to their selectivity toward lung cancer cells, they efficiently delivered siRNA and significantly reduced cancer stemness [445].
The factors involved in cancer metabolism and growth can be targeted by siRNA to increase drug sensitivity. Recently, CPT1A-siRNA was loaded into exosomes and surface modification with iRGD was performed to promote their selectivity toward colon cancer cells. These exosomes increased the sensitivity of colon cancer cells to oxaliplatin by downregulating CPT1A. Simultaneous administration of these exosomes and oxaliplatin induced apoptosis in colon cancer cells and suppressed their proliferation [446]. In another experiment, exosomes were modified with an epidermal growth factor receptor (EGFR) aptamer and then loaded with survivin-siRNA. Since survivin is involved in cancer progression and functions as an anti-apoptotic factor, its downregulation by exosomes sensitizes lung cancer cells to apoptosis. Exosomes provide endosomal escape of siRNA, which is important for increasing its efficiency in anticancer activity [447]. Overall, it is evident that siRNA is an efficient tool in cancer therapy and exosomes enhance its potential to suppress cancer and promote chemosensitivity [448]. Short hairpin RNA (shRNA) is another genetic tool that has a similar function to siRNA and can be used to regulate gene expression in cancer therapy. However, there is no study evaluating the potential of exosomes in shRNA, and further experiments could focus on this aspect.

CRISPR/Cas9 system

To date, only two studies have focused on CRISPR/Cas9 transfer through exosomes. In one experiment, cancer-derived exosomes were used to transfer the CRISPR/Cas9 system in the treatment of ovarian cancer. It seems that cancer-derived exosomes have high selectivity toward ovarian cancer cells due to cell tropism. CRISPR/Cas9-loaded exosomes significantly stimulated apoptosis by downregulating PARP-1. In addition, CRISPR/Cas9-loaded exosomes are able to increase the cytotoxicity of cisplatin (CP) against ovarian cancer cells [449]. Limitations of the CRISPR/Cas9 system include its difficulty in specifically targeting all cancer cells and its low efficacy in vivo. Therefore, it seems crucial to use exosomes for the delivery of the CRISPR/Cas9 system in cancer therapy. Exosomes can induce necroptosis in cancer cells through the CRISPR/Cas9 system (Fig. 7) [450]. However, few studies have investigated this potential of exosomes, and the development of engineered and surface-modified exosomes is encouraged.

Tumor-derived exosomes

Brain tumors

The exosomes can be derived from glioblastoma cells. These exosomes mediate immune evasion of cancer cells via induction of PD-L1 expression and transfer of STAT3. In addition, exosomes induce M2 polarization of macrophages, which promotes glioblastoma progression [451]. When glioblastoma cells are exposed to hypoxia, the secretion of exosomes is triggered. These exosomes contain miRNA-1246, which is a tumor-promoting factor to promote cancer progression by upregulating STAT3 expression, suppressing NF-κB signaling, and mediating M2 polarization of macrophages [452]. Glioblastoma-derived exosomes contain high levels of VEGF-C, which inhibits Hippo signaling and enhances tafazzin (TAZ) expression, leading to angiogenesis [453]. Proteomics can be used to identify proteins embedded in exosomes and use them as biomarkers [454]. It appears that glioblastoma-derived exosomes have immunomodulatory effects. As mentioned previously, these exosomes are able to mediate M2 polarization of macrophages, which is attributed to the induction of NF-κB signaling. Then, macrophages secrete factors responsible for cancer progression. Moreover, these exosomes suppress the activity of cytotoxic CD4 + T cells to evade the immune response in glioblastomas [455]. Furthermore, exosomes from glioblastomas promote cancer stemness by transferring Notch1 protein [456].
A recent experiment has shown that exosomes derived from glioma stem cells contain high levels of Linc01060, which acts as a tumor-promoting factor and promotes cancer progression. The exosomal Linc01060 increases the stability of myeloid zinc finger 1 (MZF1) as a transcription factor and induces its nuclear translocation. Then, MZF1 induces HIF-1α via upregulation of c-Myc to enhance glioma progression [457]. Glioma-derived exosomes contain high levels of miRNA-10a and miRNA-21, which regulate PTEN and RORA, leading to activation of myeloid-derived suppressor cells and impairing immune function [458]. miRNA-1246 and mIRNA-10b-5p are other miRNAs found in glioma-derived exosomes that promote cancer cell metastasis [459]. Furthermore, glioma cells secrete exosomes containing the lncRNA CCAT2 to stimulate angiogenesis and inhibit apoptosis, setting the stage for tumor progression [460]. To trigger angiogenesis, glioma-derived exosomes may deliver miRNA-21, which upregulates VEGF expression [461]. Overall, these studies are consistent with the fact that glioma-derived exosomes modulate proliferation and migration by transporting various cargoes [462464].

Thoracic and breast tumors

Breast cancer-derived exosomes are capable of suppressing immune function to enhance tumor progression. Injection of exosomes into naïve mice leads to accumulation of myeloid-derived suppressor cells in the lungs and the liver. Breast cancer-derived exosomes prevent T cell proliferation and suppress natural killer cell cytotoxicity to mediate immune evasion [465]. In enhancing cancer progression, breast cancer-derived exosomes transfer miRNA-155 to induce cachexia via downregulation of PARP-1 expression. Further studies revealed that these exosomes can also induce EMT-mediated metastasis in breast cancer by triggering catabolism and release of metabolites in adipocytes and muscle cells [466]. Metastatic breast cancer cells secrete exosomes containing miRNA-21 and miRNA-200c, which can be detected in patients and are used as diagnostic and prognostic factors [467]. The protein content of exosomes can be analyzed to distinguish breast cancer subtypes [468]. The presence of CD44 in breast cancer-derived exosomes leads to doxorubicin resistance [469]. Moreover, activation of fibroblasts by exosomes containing survivin can promote both growth and metastasis of breast cancer cells [470]. By inducing M2 polarization of macrophages, breast cancer-derived exosomes enhance lymph node metastasis of breast cancer cells [471]. Therefore, breast cancer-derived exosomes may modulate the progression of these tumor cells [472].
Exosomes derived from lung cancer cells, on the other hand, may act as triggers of EMT via upregulation of vimentin [473]. Exosomes derived from gemcitabine-resistant cancer cells may promote the progression of non-small cell lung cancer cells, mediate their drug resistance, and enhance their malignant phenotype through the transmission of miRNA-222-3p [474]. Exosomes derived from lung cancer cells are able to induce the Wnt3a/β-catenin axis to promote growth and survival [475]. Irradiation stimulates the release of exosomes from non-small cell lung cancer and induces Akt, STAT3, and ERK signaling pathways that mediate resistance to kinase inhibitors [476]. In additions, some of the proteins, such as MUC1, are enriched in exosomes to determine their localization and biological function [477].

Gastrointestinal tumors

Exosomes derived from gastric cancer enhance peritoneal metastasis and disrupt the mesothelial barrier [478]. These exosomes induce NF-κB signaling in macrophages to mediate secretion of pro-inflammatory factors and promote gastric cancer progression [479]. Induction of NF-κB signaling by gastric cancer-derived exosomes maintains inflammatory conditions in the TME that promote gastric cancer progression [480]. Gastric cancer-derived exosomes are able to stimulate PI3K/Akt and MERK/ERK signaling pathways. Moreover, inhibition of BMP prevents the potential of exosomes to transform pericytes into cancer-associated fibroblasts [481]. Exposure of gastric cancer cells to various antitumor agents may affect their ability to secrete exosomes. Pyrotinib, for example, induces the release of exosomes from gastric cancer cells. The secreted exosomes enhance migration, and the use of apatinib as a VEGFR inhibitor, suppresses this condition [482].
Hepatocellular carcinoma is another gastrointestinal tumor. Exosomes derived from hepatocellular carcinoma cells can induce ERK signaling to mediate EMT via upregulation of ZEB1/2, leading to cancer metastasis [483]. In addition to metastasis, exosomes derived from hepatocellular carcinomas also mediate cancer recurrence and can be used for early diagnosis of this malignancy [484]. By triggering chaperone-mediated autophagy, hepatocellular carcinoma-derived exosomes induce drug resistance and inhibit apoptosis [485]. Exosomes are also known to transfer Linc-ROR to liver cancer cells to increase their growth rate and inhibit their apoptosis [485].
Pancreatic cancer cells may also secrete exosomes. A recent experiment has shown that Dickkopf1 (DKK1)-dependent endocytosis is involved in the biogenesis of exosomes. Pancreatic cancer cell-derived exosomes have high levels of CKAP4 and are associated with poor prognosis in patients [486]. Moreover, pancreatic cancer-derived exosomes mediate M2 polarization of macrophages to suppress immune function against cancer cells [487]. To demonstrate the potential of exosomes in cancer migration, an experiment isolated serum exosomes from pancreatic cancer patients and showed that they can induce EMT and promote metastasis [488]. To enhance invasion and migration ability, pancreatic cancer cell-derived exosomes recruit cancer-associated fibroblasts and transfer Lin28B to reduce let-7 expression, leading to upregulation of HMGA2 and subsequent overexpression of PDGFB [489]. All in all, exosomes derived from pancreatic cancer cells regulate cancer progression, and their isolation and targeting may be important for cancer therapy [489494].

Reproductive tumors

Most experiments on reproductive tumor-derived exosomes have focused on ovarian cancer. Proteomic and lipidomic analysis of exosomes can be used in the early diagnosis of ovarian cancer [495]. Ovarian cancer cell-derived exosomes may be involved in the development of malignant TME by promoting fibroblast migration [496]. They can be considered as potential therapeutic targets, as their modulation can suppress growth and invasion of ovarian cancer cells [497]. Moreover, ovarian cancer cell-derived exosomes can transport miRNAs into the TME and induce M2 polarization of macrophages that promote cancer progression [498]. Exosomal miRNA-940 stimulates ovarian cancer progression by inducing polarization of macrophages to the M2 phenotype [499]. Ovarian cancer-cell derived exosomes can induce angiogenesis and migration via upregulation of VEGF [500]. The same phenomenon occurs in cervical cancer. It has been reported that cervical cancer cell-derived exosomes can promote angiogenesis via Hedgehog-GLI signaling and enhancement of VEGF-A, VEGFR2, and angiopoietin-2 expression [501]. Loading dendritic cells with exosomes derived from HeLa cells stimulates anti-tumor immunity by increasing T-cell cytotoxicity [502]. Overall, these experiments highlight the role of ovarian and cervical cancer cell-derived exosomes in modulating migration, growth, TME, anti-tumor immunity, and angiogenesis. An experiment was conducted to investigate the role of prostate cancer-derived exosomes in immunomodulation. These exosomes impair dendritic cell function and suppress CD8 + T cell activity. The exosomes mediate the expression of CD73 on dendritic cells, which subsequently upregulate the expression of CD39, resulting in ATP-dependent inhibition of TNF-α and IL-12 production. In addition, exosomes have been found to contain prostaglandin E2, which enhances CD73 expression (Fig. 8) [503].

Clinical application and role of exosomes as biomarkers

According to the role of exosomes in influencing cancer progression, a significant attempt has been made to reveal the role of exosomes in clinical background [504508]. The genes contained in exosomes can be considered as biomarkers for the diagnosis of lung cancer. A recent experiment has shown that exosomes derived from non-small cell lung cancer cells have Hippo, Rap1, and Wnt as important signaling networks and can be considered as prognostic tools [509]. Another study shows that exosomes derived from non-small cell lung cancer contain high levels of PRPS2 and can mediate cisplatin resistance. In addition, PRPS2-containing exosomes are capable of inducing M2 polarization of macrophages and are associated with an unfavorable prognosis [510]. Exosomes containing high levels of miRNA-3362, miRNA-146a, and miRNA-1290 are observed in breast cancer patients and mediate lymph node metastasis and clinical stage [511]. In colorectal cancer patients, the abundance of QSOX1 in plasma exosomes decreases and can be considered as a diagnostic factor [511]. These studies demonstrate how exosomes can be analyzed to determine the prognosis of cancer patients (Table 4).
Table 4
Clinical trials on the use of exosomes in cancer patients
Status
Remarks
Reference
Completed
Use of exosomes as reliable biomarkers for the diagnosis of men with prostate cancer
NCT02702856
Unknown
Combination of computed tomography and exosomes for diagnosis of early stage lung cancer
NCT03542253
Active, not recruiting
Use of exosomes present in blood plasma to diagnose lung cancer in patients
NCT04529915
Unknown
Use of circulating exosomes for diagnosis of advanced gastric cancer
NCT01779583
Recruiting
New diagnostic method for colorectal cancer using exosomes
NCT04394572
Recruiting
Presence of exosomes in tumor-draining vein and their molecular profiling
NCT04939324
Recruiting
Use of plant exosome for delivery of curcumin in the treatment of colon cancer
NCT01294072

Conclusion and future perspectives

Thanks to the attempts made in recent years to uncover the factors involved in cancer progression, it is now clear that each factor has a unique fingerprint in cancer pathogenesis. If we know exactly how these factors interact in cancer, we can develop novel and effective therapeutics. Exosomes are minute structures that are involved in the regulation of biological processes through their cargo, which can be proteins, lipids, or nucleic acids. Genetic tools and anti-tumor agents can also be loaded into exosomes. Therefore, they provide intercellular communication and their involvement in cancer progression or inhibition is inevitable. Depending on the cargo, the effect of exosomes on the target cell may be different. In addition, normal cells such as macrophages and mesenchymal stem cells are capable of secreting exosomes to affect cancer cell progression. Therefore, exosomes can mediate both normal cell-cancer cell and cancer cell-cancer cell interactions.
Growth and invasion are the two most important aspects of tumor cells. When exosomes contain tumor-promoting substances, they can promote cell cycle progression and glycolysis and inhibit apoptosis. The role of autophagy is a bit confusing. It has been mentioned that induction of autophagy by exosomes can prevent apoptosis in cancer cells. Therefore, further experiments should be performed to reveal the interaction between apoptosis and autophagy in cancer cells affected by exosomes. Similar to proliferation, the cargo of exosomes determines the function of these structures in increasing or decreasing cancer migration and invasion. EMT and MMPs are strongly influenced by exosomes in regulating cancer progression. However, most studies have focused on the EMT mechanism, and it is proposed to uncover the signaling networks affected by exosomes in targeting MMPs and modulating cancer metastasis, because of the important role of MMPs in this case. Cancer cell proliferation and invasion rates determine response to therapy. If cancer cells have a high capacity to migrate and grow, they may develop resistance to therapy. Therefore, by targeting exosomes, proliferation and invasion of cancer cells can be modulated and their response to therapy can be predicted. The aggressive behavior of cancer cells depends mainly on interactions in the TME. The best known interaction in the TME is macrophage polarization mediated by exosomes. Exosomes can induce M2 polarization of macrophages, promoting cancer cell progression.
Since response to therapy is a major concern for physicians treating cancer patients, we have provided a section specifically addressing the role of exosomes in this case. The sensitivity of cancer cells to chemotherapy-mediated apoptosis can be reduced by exosomes. Because of the potential of exosomes to transfer various genes, they can influence the progression of cancer cells and determine their response to therapy. In addition to drug resistance, exosomes may also be involved in triggering radioresistance. In addition, exosomes can induce immune cell exhaustion, decrease T cell cytotoxicity, and mediate immune evasion. By triggering chronic inflammation, exosomes promote cancer progression. When new therapeutics are to be introduced into clinics for the treatment of cancer patients, they can focus on these aspects.
For internalization into cells, exosomes can follow different pathways. Exosomes are able to bind to receptors on the surface of cells, and can be internalized by binding to integrins, tetraspanins and intercellular adhesion molecules. Clathrin- and caveolin-mediated endocytosis, lipid raft uptake, macropinocytosis, phagocytosis and fusion with the plasma membrane [512]. Therefore, if exosomes are to be used for cargo transport in cancer therapy, the method of their internalization should be elucidated and subsequent functionalization should be performed to improve their intracellular accumulation.
Exosomes may contain miRNAs, lncRNAs, circRNAs, and other genes such as PTEN, PI3K/Akt, and STAT3 that affect cancer progression. Indeed, cancer progression is strongly influenced by exosome cargo. Since exosomes are capable of delivering various drugs, they have the potential to be used as delivery systems for anti-tumor agents and genetic tools in cancer therapy. The various anti-cancer agents, including plant-derived natural compounds such as triptolides and synthetic agents such as cisplatin, doxorubicin and paclitaxel, can be transferred by exosomes in cancer therapy. The siRNA and CRISPR/Cas9 are genetic tools embedded in exosomes for cancer therapy. Delivery of therapeutics using exosomes can potentially lead to increased intracellular accumulation and improved therapeutic efficacy. In addition, the surface of exosomes can be modified with ligands to increase their selectivity toward cancer cells. Since exosomes affect various aspects of cancer cells, they can be isolated from the serum of patients and are considered reliable biomarkers for the diagnosis and prognosis of cancer patients. As shown in Table 4, exosomes have been used as biomarkers in various experiments in cancer patients. In addition to diagnosis, exosomes have also been used to increase the accuracy of other methods of detecting cancer patients, such as CT. Of note, a clinical trial is currently underway to deliver curcumin as an anti-cancer agent to treat colon cancer. The results of this clinical trial are of great importance, as they may provide novel insights into the role of exosomes as drug delivery systems in the clinical course and their safety. Furthermore, a number of clinical trials on molecular profiling of exosomes are currently ongoing, which could be useful in the field of precision medicine in the near future.

Acknowledgements

We would like to thank Academic Proofreading (www.​academicproofrea​ding.​uk) for their excellent English language editing services.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Cai W, Xiong Chen Z, Rane G, Satendra Singh S, Choo Z, Wang C, et al. Wanted DEAD/H or alive: helicases winding up in cancers. J Natl Cancer Inst. 2017;109(6):djw278. Cai W, Xiong Chen Z, Rane G, Satendra Singh S, Choo Z, Wang C, et al. Wanted DEAD/H or alive: helicases winding up in cancers. J Natl Cancer Inst. 2017;109(6):djw278.
2.
Zurück zum Zitat Manu KA, Shanmugam MK, Ramachandran L, Li F, Siveen KS, Chinnathambi A, et al. Isorhamnetin augments the anti-tumor effect of capecitabine through the negative regulation of NF-κB signaling cascade in gastric cancer. Cancer Lett. 2015;363(1):28–36.PubMed Manu KA, Shanmugam MK, Ramachandran L, Li F, Siveen KS, Chinnathambi A, et al. Isorhamnetin augments the anti-tumor effect of capecitabine through the negative regulation of NF-κB signaling cascade in gastric cancer. Cancer Lett. 2015;363(1):28–36.PubMed
3.
Zurück zum Zitat Wang C, Kar S, Lai X, Cai W, Arfuso F, Sethi G, et al. Triple negative breast cancer in Asia: an insider’s view. Cancer Treat Rev. 2018;62:29–38.PubMed Wang C, Kar S, Lai X, Cai W, Arfuso F, Sethi G, et al. Triple negative breast cancer in Asia: an insider’s view. Cancer Treat Rev. 2018;62:29–38.PubMed
4.
Zurück zum Zitat Li F, Shanmugam MK, Chen L, Chatterjee S, Basha J, Kumar AP, et al. Garcinol, a polyisoprenylated benzophenone modulates multiple proinflammatory signaling cascades leading to the suppression of growth and survival of head and neck carcinoma. Cancer Prev Res (Phila). 2013;6(8):843–54. Li F, Shanmugam MK, Chen L, Chatterjee S, Basha J, Kumar AP, et al. Garcinol, a polyisoprenylated benzophenone modulates multiple proinflammatory signaling cascades leading to the suppression of growth and survival of head and neck carcinoma. Cancer Prev Res (Phila). 2013;6(8):843–54.
5.
Zurück zum Zitat Ang HL, Yuan Y, Lai X, Tan TZ, Wang L, Huang BB, et al. Putting the BRK on breast cancer: From molecular target to therapeutics. Theranostics. 2021;11(3):1115–28.PubMedPubMedCentral Ang HL, Yuan Y, Lai X, Tan TZ, Wang L, Huang BB, et al. Putting the BRK on breast cancer: From molecular target to therapeutics. Theranostics. 2021;11(3):1115–28.PubMedPubMedCentral
6.
Zurück zum Zitat Sethi G, Shanmugam MK, Warrier S, Merarchi M, Arfuso F, Kumar AP, et al. Pro-apoptotic and anti-cancer properties of diosgenin: a comprehensive and critical review. Nutrients. 2018;10(5):645.PubMedCentral Sethi G, Shanmugam MK, Warrier S, Merarchi M, Arfuso F, Kumar AP, et al. Pro-apoptotic and anti-cancer properties of diosgenin: a comprehensive and critical review. Nutrients. 2018;10(5):645.PubMedCentral
7.
Zurück zum Zitat Patel SM, Nagulapalli Venkata KC, Bhattacharyya P, Sethi G, Bishayee A. Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic diseases. Semin Cancer Biol. 2016;40–41:100–15.PubMed Patel SM, Nagulapalli Venkata KC, Bhattacharyya P, Sethi G, Bishayee A. Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic diseases. Semin Cancer Biol. 2016;40–41:100–15.PubMed
8.
Zurück zum Zitat Chopra P, Sethi G, Dastidar SG, Ray A. Polo-like kinase inhibitors: an emerging opportunity for cancer therapeutics. Expert Opin Investig Drugs. 2010;19(1):27–43.PubMed Chopra P, Sethi G, Dastidar SG, Ray A. Polo-like kinase inhibitors: an emerging opportunity for cancer therapeutics. Expert Opin Investig Drugs. 2010;19(1):27–43.PubMed
9.
Zurück zum Zitat Warrier S, Patil M, Bhansali S, Varier L, Sethi G. Designing precision medicine panels for drug refractory cancers targeting cancer stemness traits. Biochim Biophys Acta. 2021;1875(1):188475. Warrier S, Patil M, Bhansali S, Varier L, Sethi G. Designing precision medicine panels for drug refractory cancers targeting cancer stemness traits. Biochim Biophys Acta. 2021;1875(1):188475.
10.
Zurück zum Zitat Kashyap D, Tuli HS, Yerer MB, Sharma A, Sak K, Srivastava S, et al. Natural product-based nanoformulations for cancer therapy: opportunities and challenges. Semin Cancer Biol. 2021;69:5–23.PubMed Kashyap D, Tuli HS, Yerer MB, Sharma A, Sak K, Srivastava S, et al. Natural product-based nanoformulations for cancer therapy: opportunities and challenges. Semin Cancer Biol. 2021;69:5–23.PubMed
11.
Zurück zum Zitat Ailuno G, Baldassari S, Lai F, Florio T, Caviglioli GJC. Exosomes and extracellular vesicles as emerging theranostic platforms in cancer research cells. Semin Cancer Biol. 2020;9(12):2569. Ailuno G, Baldassari S, Lai F, Florio T, Caviglioli GJC. Exosomes and extracellular vesicles as emerging theranostic platforms in cancer research cells. Semin Cancer Biol. 2020;9(12):2569.
13.
Zurück zum Zitat Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMed Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMed
15.
Zurück zum Zitat Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19(2):43–51.PubMed Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19(2):43–51.PubMed
16.
Zurück zum Zitat Hristov M, Erl W, Linder S, Weber PC. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood. 2004;104(9):2761–6.PubMed Hristov M, Erl W, Linder S, Weber PC. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood. 2004;104(9):2761–6.PubMed
17.
Zurück zum Zitat Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–83.PubMedPubMedCentral Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–83.PubMedPubMedCentral
18.
Zurück zum Zitat Wu P, Zhang B, Ocansey DKW, Xu W, Qian H. Extracellular vesicles: a bright star of nanomedicine. Biomaterials. 2021;269:120467.PubMed Wu P, Zhang B, Ocansey DKW, Xu W, Qian H. Extracellular vesicles: a bright star of nanomedicine. Biomaterials. 2021;269:120467.PubMed
19.
Zurück zum Zitat Möller A, Lobb RJ. The evolving translational potential of small extracellular vesicles in cancer. Nat Rev Cancer. 2020;20(12):697–709.PubMed Möller A, Lobb RJ. The evolving translational potential of small extracellular vesicles in cancer. Nat Rev Cancer. 2020;20(12):697–709.PubMed
20.
Zurück zum Zitat Mathew M, Zade M, Mezghani N, Patel R, Wang Y, Momen-Heravi F. Extracellular vesicles as biomarkers in cancer immunotherapy. Cancers. 2020;12(10):2825.PubMedCentral Mathew M, Zade M, Mezghani N, Patel R, Wang Y, Momen-Heravi F. Extracellular vesicles as biomarkers in cancer immunotherapy. Cancers. 2020;12(10):2825.PubMedCentral
21.
Zurück zum Zitat Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochem Biophys Acta. 2012;1820(7):940–8.PubMed Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochem Biophys Acta. 2012;1820(7):940–8.PubMed
22.
Zurück zum Zitat Wee I, Syn N, Sethi G, Goh BC, Wang L. Role of tumor-derived exosomes in cancer metastasis. Biochim Biophys Acta. 2019;1871(1):12–9. Wee I, Syn N, Sethi G, Goh BC, Wang L. Role of tumor-derived exosomes in cancer metastasis. Biochim Biophys Acta. 2019;1871(1):12–9.
23.
Zurück zum Zitat Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci. 2000;113(Pt 19):3365–74.PubMed Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci. 2000;113(Pt 19):3365–74.PubMed
25.
Zurück zum Zitat Hu T, Wolfram J, Srivastava S. Extracellular vesicles in cancer detection: hopes and hypes. Trends Cancer. 2021;7(2):122–33.PubMed Hu T, Wolfram J, Srivastava S. Extracellular vesicles in cancer detection: hopes and hypes. Trends Cancer. 2021;7(2):122–33.PubMed
26.
Zurück zum Zitat Shoucair I, Weber Mello F, Jabalee J, Maleki S, Garnis C. The role of cancer-associated fibroblasts and extracellular vesicles in tumorigenesis. Int J Mol Sci. 2020;21(18):6837.PubMedCentral Shoucair I, Weber Mello F, Jabalee J, Maleki S, Garnis C. The role of cancer-associated fibroblasts and extracellular vesicles in tumorigenesis. Int J Mol Sci. 2020;21(18):6837.PubMedCentral
27.
Zurück zum Zitat Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Mol Oncol. 2021;15(1):3–26.PubMed Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Mol Oncol. 2021;15(1):3–26.PubMed
28.
Zurück zum Zitat Palazzolo S, Memeo L, Hadla M, Duzagac F, Steffan A, Perin T, et al. Cancer extracellular vesicles: next-generation diagnostic and drug delivery nanotools. Cancers. 2020;12(11):3165.PubMedCentral Palazzolo S, Memeo L, Hadla M, Duzagac F, Steffan A, Perin T, et al. Cancer extracellular vesicles: next-generation diagnostic and drug delivery nanotools. Cancers. 2020;12(11):3165.PubMedCentral
29.
Zurück zum Zitat Sun H, Burrola S, Wu J, Ding WQ. Extracellular vesicles in the development of cancer therapeutics. Int J Mol Sci. 2020;21(17):6097.PubMedCentral Sun H, Burrola S, Wu J, Ding WQ. Extracellular vesicles in the development of cancer therapeutics. Int J Mol Sci. 2020;21(17):6097.PubMedCentral
30.
Zurück zum Zitat Huyan T, Li H, Peng H, Chen J, Yang R, Zhang W, et al. Extracellular vesicles - advanced nanocarriers in cancer therapy: progress and achievements. Int J Nanomed. 2020;15:6485–502. Huyan T, Li H, Peng H, Chen J, Yang R, Zhang W, et al. Extracellular vesicles - advanced nanocarriers in cancer therapy: progress and achievements. Int J Nanomed. 2020;15:6485–502.
31.
Zurück zum Zitat Deb A, Gupta S, Mazumder PJ. Exosomes: a new horizon in modern medicine. Life Sci. 2020;264:118623.PubMed Deb A, Gupta S, Mazumder PJ. Exosomes: a new horizon in modern medicine. Life Sci. 2020;264:118623.PubMed
32.
Zurück zum Zitat Théry C, Zitvogel L, Amigorena SJ. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.PubMed Théry C, Zitvogel L, Amigorena SJ. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.PubMed
33.
Zurück zum Zitat Syn N, Wang L, Sethi G, Thiery JP, Goh BC. Exosome-mediated metastasis: from epithelial-mesenchymal transition to escape from immunosurveillance. Trends Pharmacol Sci. 2016;37(7):606–17.PubMed Syn N, Wang L, Sethi G, Thiery JP, Goh BC. Exosome-mediated metastasis: from epithelial-mesenchymal transition to escape from immunosurveillance. Trends Pharmacol Sci. 2016;37(7):606–17.PubMed
34.
Zurück zum Zitat Laulagnier K, Motta C, Hamdi S, Roy S, Fauvelle F, Pageaux J-F, et al. Mast cell-and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem J. 2004;380(1):161–71.PubMedPubMedCentral Laulagnier K, Motta C, Hamdi S, Roy S, Fauvelle F, Pageaux J-F, et al. Mast cell-and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem J. 2004;380(1):161–71.PubMedPubMedCentral
35.
Zurück zum Zitat Staubach S, Razawi H, Hanisch FGJ. Proteomics of MUC1-containing lipid rafts from plasma membranes and exosomes of human breast carcinoma cells MCF-7. Proteomics. 2009;9(10):2820–35.PubMed Staubach S, Razawi H, Hanisch FGJ. Proteomics of MUC1-containing lipid rafts from plasma membranes and exosomes of human breast carcinoma cells MCF-7. Proteomics. 2009;9(10):2820–35.PubMed
36.
Zurück zum Zitat Tamkovich S, Tutanov O, Laktionov P. Exosomes: generation, structure, transport, biological activity, and diagnostic application. Biochem Moscow Suppl Ser A. 2016;10(3):163–73. Tamkovich S, Tutanov O, Laktionov P. Exosomes: generation, structure, transport, biological activity, and diagnostic application. Biochem Moscow Suppl Ser A. 2016;10(3):163–73.
37.
Zurück zum Zitat Lu GD, Cheng P, Liu T, Wang Z. BMSC-derived exosomal miR-29a promotes angiogenesis and osteogenesis. Front Cell Dev Biol. 2020;8:608521.PubMedPubMedCentral Lu GD, Cheng P, Liu T, Wang Z. BMSC-derived exosomal miR-29a promotes angiogenesis and osteogenesis. Front Cell Dev Biol. 2020;8:608521.PubMedPubMedCentral
38.
Zurück zum Zitat Jia J, Guo S, Zhang D, Tian X, Xie X. Exosomal-lncRNA DLEU1 Accelerates the Proliferation, Migration, and Invasion of Endometrial Carcinoma Cells by Regulating microRNA-E2F3. Onco Targets Ther. 2020;13:8651–63.PubMedPubMedCentral Jia J, Guo S, Zhang D, Tian X, Xie X. Exosomal-lncRNA DLEU1 Accelerates the Proliferation, Migration, and Invasion of Endometrial Carcinoma Cells by Regulating microRNA-E2F3. Onco Targets Ther. 2020;13:8651–63.PubMedPubMedCentral
39.
Zurück zum Zitat Tian C, Liu J, Di X, Cong S, Zhao M, Wang K. Exosomal hsa_circRNA_104484 and hsa_circRNA_104670 may serve as potential novel biomarkers and therapeutic targets for sepsis. Sci Rep. 2021;11(1):14141.PubMedPubMedCentral Tian C, Liu J, Di X, Cong S, Zhao M, Wang K. Exosomal hsa_circRNA_104484 and hsa_circRNA_104670 may serve as potential novel biomarkers and therapeutic targets for sepsis. Sci Rep. 2021;11(1):14141.PubMedPubMedCentral
40.
Zurück zum Zitat Romano E, Netti PA, Torino EJP. Exosomes in gliomas: biogenesis, isolation, and preliminary applications in nanomedicine. Pharmaceuticals. 2020;13(10):319.PubMedCentral Romano E, Netti PA, Torino EJP. Exosomes in gliomas: biogenesis, isolation, and preliminary applications in nanomedicine. Pharmaceuticals. 2020;13(10):319.PubMedCentral
41.
Zurück zum Zitat Burkova EE, Sedykh SE, Nevinsky GA. Human placenta exosomes: Biogenesis, isolation, composition, and prospects for use in diagnostics. Int J Mol Sci. 2021;22(4):2158.PubMedPubMedCentral Burkova EE, Sedykh SE, Nevinsky GA. Human placenta exosomes: Biogenesis, isolation, composition, and prospects for use in diagnostics. Int J Mol Sci. 2021;22(4):2158.PubMedPubMedCentral
42.
Zurück zum Zitat Whiteside TL, Diergaarde B, Hong C-S. Tumor-derived exosomes (TEX) and their role in immuno-oncology. Int J Mol Sci. 2021;22(12):6234.PubMedPubMedCentral Whiteside TL, Diergaarde B, Hong C-S. Tumor-derived exosomes (TEX) and their role in immuno-oncology. Int J Mol Sci. 2021;22(12):6234.PubMedPubMedCentral
44.
Zurück zum Zitat Jiang Z, Liu G, Li J. Recent progress on the isolation and detection methods of exosomes. Chem Asian J. 2020;15(23):3973–82.PubMed Jiang Z, Liu G, Li J. Recent progress on the isolation and detection methods of exosomes. Chem Asian J. 2020;15(23):3973–82.PubMed
46.
Zurück zum Zitat An M, Wu J, Zhu J, Lubman DM. Comparison of an optimized ultracentrifugation method versus size-exclusion chromatography for isolation of exosomes from human serum. J Proteome Res. 2018;17(10):3599–605.PubMedPubMedCentral An M, Wu J, Zhu J, Lubman DM. Comparison of an optimized ultracentrifugation method versus size-exclusion chromatography for isolation of exosomes from human serum. J Proteome Res. 2018;17(10):3599–605.PubMedPubMedCentral
47.
Zurück zum Zitat 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 (Clifton, NJ). 2015;1295:179–209. 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 (Clifton, NJ). 2015;1295:179–209.
48.
Zurück zum Zitat Momen-Heravi F. Isolation of extracellular vesicles by ultracentrifugation. Methods Mol Biol (Clifton, NJ). 2017;1660:25–32. Momen-Heravi F. Isolation of extracellular vesicles by ultracentrifugation. Methods Mol Biol (Clifton, NJ). 2017;1660:25–32.
49.
Zurück zum Zitat Barreiro K, Huber TB, Holthofer H. Isolating urinary extracellular vesicles as biomarkers for diabetic disease. Methods Mol Biol (Clifton, NJ). 2020;2067:175–88. Barreiro K, Huber TB, Holthofer H. Isolating urinary extracellular vesicles as biomarkers for diabetic disease. Methods Mol Biol (Clifton, NJ). 2020;2067:175–88.
50.
Zurück zum Zitat Lobb RJ, Becker M, Wen SW, Wong CS, Wiegmans AP, Leimgruber A, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015;4:27031.PubMed Lobb RJ, Becker M, Wen SW, Wong CS, Wiegmans AP, Leimgruber A, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015;4:27031.PubMed
51.
Zurück zum Zitat Shu S, Yang Y, Allen CL, Hurley E, Tung KH, Minderman H, et al. Purity and yield of melanoma exosomes are dependent on isolation method. J Extracell Vesicles. 2020;9(1):1692401.PubMed Shu S, Yang Y, Allen CL, Hurley E, Tung KH, Minderman H, et al. Purity and yield of melanoma exosomes are dependent on isolation method. J Extracell Vesicles. 2020;9(1):1692401.PubMed
52.
Zurück zum Zitat Feng Y, Huang W, Wani M, Yu X, Ashraf M. Ischemic preconditioning potentiates the protective effect of stem cells through secretion of exosomes by targeting Mecp2 via miR-22. PLoS One. 2014;9(2):e88685.PubMedPubMedCentral Feng Y, Huang W, Wani M, Yu X, Ashraf M. Ischemic preconditioning potentiates the protective effect of stem cells through secretion of exosomes by targeting Mecp2 via miR-22. PLoS One. 2014;9(2):e88685.PubMedPubMedCentral
53.
Zurück zum Zitat Takov K, Yellon DM, Davidson SM. Comparison of small extracellular vesicles isolated from plasma by ultracentrifugation or size-exclusion chromatography: yield, purity and functional potential. J Extracell Vesicles. 2019;8(1):1560809.PubMed Takov K, Yellon DM, Davidson SM. Comparison of small extracellular vesicles isolated from plasma by ultracentrifugation or size-exclusion chromatography: yield, purity and functional potential. J Extracell Vesicles. 2019;8(1):1560809.PubMed
54.
Zurück zum Zitat Buschmann D, Kirchner B, Hermann S, Märte M, Wurmser C, Brandes F, et al. Evaluation of serum extracellular vesicle isolation methods for profiling miRNAs by next-generation sequencing. J Extracell Vesicles. 2018;7(1):1481321.PubMedPubMedCentral Buschmann D, Kirchner B, Hermann S, Märte M, Wurmser C, Brandes F, et al. Evaluation of serum extracellular vesicle isolation methods for profiling miRNAs by next-generation sequencing. J Extracell Vesicles. 2018;7(1):1481321.PubMedPubMedCentral
55.
Zurück zum Zitat Rider MA, Hurwitz SN, Meckes DG Jr. ExtraPEG: a polyethylene glycol-based method for enrichment of extracellular vesicles. Sci Rep. 2016;6:23978.PubMedPubMedCentral Rider MA, Hurwitz SN, Meckes DG Jr. ExtraPEG: a polyethylene glycol-based method for enrichment of extracellular vesicles. Sci Rep. 2016;6:23978.PubMedPubMedCentral
56.
Zurück zum Zitat Alvarez ML, Khosroheidari M, Kanchi Ravi R, DiStefano JK. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney Int. 2012;82(9):1024–32.PubMed Alvarez ML, Khosroheidari M, Kanchi Ravi R, DiStefano JK. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney Int. 2012;82(9):1024–32.PubMed
57.
Zurück zum Zitat Zarovni N, Corrado A, Guazzi P, Zocco D, Lari E, Radano G, et al. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods (San Diego, Calif). 2015;87:46–58. Zarovni N, Corrado A, Guazzi P, Zocco D, Lari E, Radano G, et al. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods (San Diego, Calif). 2015;87:46–58.
58.
Zurück zum Zitat Helwa I, Cai J, Drewry MD, Zimmerman A, Dinkins MB, Khaled ML, et al. A comparative study of serum exosome isolation using differential ultracentrifugation and three commercial reagents. PLoS One. 2017;12(1):e0170628.PubMedPubMedCentral Helwa I, Cai J, Drewry MD, Zimmerman A, Dinkins MB, Khaled ML, et al. A comparative study of serum exosome isolation using differential ultracentrifugation and three commercial reagents. PLoS One. 2017;12(1):e0170628.PubMedPubMedCentral
59.
Zurück zum Zitat Salafi T, Zeming KK, Zhang Y. Advancements in microfluidics for nanoparticle separation. Lab Chip. 2016;17(1):11–33.PubMed Salafi T, Zeming KK, Zhang Y. Advancements in microfluidics for nanoparticle separation. Lab Chip. 2016;17(1):11–33.PubMed
60.
Zurück zum Zitat Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, et al. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small. 2020;16(9):e1903916.PubMed Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, et al. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small. 2020;16(9):e1903916.PubMed
61.
Zurück zum Zitat He M, Crow J, Roth M, Zeng Y, Godwin AK. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip. 2014;14(19):3773–80.PubMedPubMedCentral He M, Crow J, Roth M, Zeng Y, Godwin AK. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip. 2014;14(19):3773–80.PubMedPubMedCentral
62.
Zurück zum Zitat Contreras-Naranjo JC, Wu HJ, Ugaz VM. Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. Lab Chip. 2017;17(21):3558–77.PubMedPubMedCentral Contreras-Naranjo JC, Wu HJ, Ugaz VM. Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. Lab Chip. 2017;17(21):3558–77.PubMedPubMedCentral
63.
Zurück zum Zitat Zhu L, Sun H-T, Wang S, Huang S-L, Zheng Y, Wang C-Q, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):1–24. Zhu L, Sun H-T, Wang S, Huang S-L, Zheng Y, Wang C-Q, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):1–24.
64.
Zurück zum Zitat Kurian TK, Banik S, Gopal D, Chakrabarti S, Mazumder N. Elucidating methods for isolation and quantification of exosomes: a review. Mol Biotechnol. 2021;63:249–66.PubMedPubMedCentral Kurian TK, Banik S, Gopal D, Chakrabarti S, Mazumder N. Elucidating methods for isolation and quantification of exosomes: a review. Mol Biotechnol. 2021;63:249–66.PubMedPubMedCentral
65.
Zurück zum Zitat Willis GR, Kourembanas S, Mitsialis SA. Toward exosome-based therapeutics: isolation, heterogeneity, and fit-for-purpose potency. Front Cardiovasc Med. 2017;4:63.PubMedPubMedCentral Willis GR, Kourembanas S, Mitsialis SA. Toward exosome-based therapeutics: isolation, heterogeneity, and fit-for-purpose potency. Front Cardiovasc Med. 2017;4:63.PubMedPubMedCentral
66.
Zurück zum Zitat Chen Y-S, Lin E-Y, Chiou T-W, Harn H-J. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Tzu-Chi Med J. 2019;32(2):113–20.PubMedCentral Chen Y-S, Lin E-Y, Chiou T-W, Harn H-J. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Tzu-Chi Med J. 2019;32(2):113–20.PubMedCentral
67.
Zurück zum Zitat Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: key players in cancer and potential therapeutic strategy. Sig Transduct Target Ther. 2020;5(1):1–10. Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: key players in cancer and potential therapeutic strategy. Sig Transduct Target Ther. 2020;5(1):1–10.
68.
Zurück zum Zitat Ibrahim A, Marbán E. Exosomes: fundamental biology and roles in cardiovascular physiology. Annu Rev Physiol. 2016;78:67–83.PubMed Ibrahim A, Marbán E. Exosomes: fundamental biology and roles in cardiovascular physiology. Annu Rev Physiol. 2016;78:67–83.PubMed
69.
Zurück zum Zitat Rastogi S, Sharma V, Bharti PS, Rani K, Modi GP, Nikolajeff F, et al. The evolving landscape of exosomes in neurodegenerative diseases: exosomes characteristics and a promising role in early diagnosis. Int J Mol Sci. 2021;22(1):440.PubMedCentral Rastogi S, Sharma V, Bharti PS, Rani K, Modi GP, Nikolajeff F, et al. The evolving landscape of exosomes in neurodegenerative diseases: exosomes characteristics and a promising role in early diagnosis. Int J Mol Sci. 2021;22(1):440.PubMedCentral
70.
Zurück zum Zitat Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood. 1999;94(11):3791–9.PubMed Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood. 1999;94(11):3791–9.PubMed
71.
Zurück zum Zitat Radulovic M, Stenmark H. ESCRTs in membrane sealing. Biochem Soc Trans. 2018;46(4):773–8.PubMed Radulovic M, Stenmark H. ESCRTs in membrane sealing. Biochem Soc Trans. 2018;46(4):773–8.PubMed
72.
Zurück zum Zitat Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic (Copenhagen, Denmark). 2009;10(7):925–37. Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic (Copenhagen, Denmark). 2009;10(7):925–37.
73.
Zurück zum Zitat Henne WM, Buchkovich NJ, Emr SD. The ESCRT pathway. Dev Cell. 2011;21(1):77–91.PubMed Henne WM, Buchkovich NJ, Emr SD. The ESCRT pathway. Dev Cell. 2011;21(1):77–91.PubMed
74.
Zurück zum Zitat Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:24641. Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:24641.
75.
Zurück zum Zitat Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, 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.PubMed Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, 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.PubMed
76.
Zurück zum Zitat Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci USA. 2016;113(8):E968–77.PubMedPubMedCentral Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci USA. 2016;113(8):E968–77.PubMedPubMedCentral
77.
Zurück zum Zitat Klumperman J, Raposo G. The complex ultrastructure of the endolysosomal system. Cold Spring Harb Perspect Biol. 2014;6(10):a016857.PubMedPubMedCentral Klumperman J, Raposo G. The complex ultrastructure of the endolysosomal system. Cold Spring Harb Perspect Biol. 2014;6(10):a016857.PubMedPubMedCentral
78.
Zurück zum Zitat Anand S, Samuel M, Kumar S, Mathivanan S. Ticket to a bubble ride: Cargo sorting into exosomes and extracellular vesicles. Biochim Biophys Acta. 2019;1867(12):140203. Anand S, Samuel M, Kumar S, Mathivanan S. Ticket to a bubble ride: Cargo sorting into exosomes and extracellular vesicles. Biochim Biophys Acta. 2019;1867(12):140203.
79.
Zurück zum Zitat Alenquer M, Amorim MJ. Exosome biogenesis, regulation, and function in viral infection. Viruses. 2015;7(9):5066–83.PubMedPubMedCentral Alenquer M, Amorim MJ. Exosome biogenesis, regulation, and function in viral infection. Viruses. 2015;7(9):5066–83.PubMedPubMedCentral
80.
Zurück zum Zitat Camuzard O, Santucci-Darmanin S, Carle GF, Pierrefite-Carle V. Autophagy in the crosstalk between tumor and microenvironment. Cancer Lett. 2020;490:143–53.PubMed Camuzard O, Santucci-Darmanin S, Carle GF, Pierrefite-Carle V. Autophagy in the crosstalk between tumor and microenvironment. Cancer Lett. 2020;490:143–53.PubMed
81.
Zurück zum Zitat Ma Z, Wang LZ, Cheng JT, Lam WST, Ma X, Xiang X, et al. Targeting hypoxia-inducible factor-1-mediated metastasis for cancer therapy. Antioxid Redox Signal. 2021;34(18):1484–97.PubMed Ma Z, Wang LZ, Cheng JT, Lam WST, Ma X, Xiang X, et al. Targeting hypoxia-inducible factor-1-mediated metastasis for cancer therapy. Antioxid Redox Signal. 2021;34(18):1484–97.PubMed
82.
Zurück zum Zitat Shanmugam MK, Warrier S, Kumar AP, Sethi G, Arfuso F. Potential role of natural compounds as anti-angiogenic agents in cancer. Curr Vasc Pharmacol. 2017;15(6):503–19.PubMed Shanmugam MK, Warrier S, Kumar AP, Sethi G, Arfuso F. Potential role of natural compounds as anti-angiogenic agents in cancer. Curr Vasc Pharmacol. 2017;15(6):503–19.PubMed
83.
Zurück zum Zitat Ye J, Wu D, Wu P, Chen Z, Huang JJ. The cancer stem cell niche: cross talk between cancer stem cells and their microenvironment. Tumor Biol. 2014;35(5):3945–51. Ye J, Wu D, Wu P, Chen Z, Huang JJ. The cancer stem cell niche: cross talk between cancer stem cells and their microenvironment. Tumor Biol. 2014;35(5):3945–51.
84.
Zurück zum Zitat Kalluri R, Zeisberg MJ. Fibroblasts in cancer. Nat Rev Cancer. 2006;6(5):392–401.PubMed Kalluri R, Zeisberg MJ. Fibroblasts in cancer. Nat Rev Cancer. 2006;6(5):392–401.PubMed
85.
Zurück zum Zitat Qian L-W, Mizumoto K, Maehara N, Ohuchida K, Inadome N, Saimura M, et al. Co-cultivation of pancreatic cancer cells with orthotopic tumor-derived fibroblasts: fibroblasts stimulate tumor cell invasion via HGF secretion whereas cancer cells exert a minor regulative effect on fibroblasts HGF production. Cancer Lett. 2003;190(1):105–12.PubMed Qian L-W, Mizumoto K, Maehara N, Ohuchida K, Inadome N, Saimura M, et al. Co-cultivation of pancreatic cancer cells with orthotopic tumor-derived fibroblasts: fibroblasts stimulate tumor cell invasion via HGF secretion whereas cancer cells exert a minor regulative effect on fibroblasts HGF production. Cancer Lett. 2003;190(1):105–12.PubMed
86.
Zurück zum Zitat Jin M-Z, Jin W-L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther. 2020;5(1):166.PubMedPubMedCentral Jin M-Z, Jin W-L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther. 2020;5(1):166.PubMedPubMedCentral
87.
Zurück zum Zitat Datta A, Deng S, Gopal V, Yap KC, Halim CE, Lye ML, et al. Cytoskeletal dynamics in epithelial-mesenchymal transition: insights into therapeutic targets for cancer metastasis. Cancers. 2021;13(8):1882.PubMedPubMedCentral Datta A, Deng S, Gopal V, Yap KC, Halim CE, Lye ML, et al. Cytoskeletal dynamics in epithelial-mesenchymal transition: insights into therapeutic targets for cancer metastasis. Cancers. 2021;13(8):1882.PubMedPubMedCentral
88.
Zurück zum Zitat Cheng JT, Wang L, Wang H, Tang FR, Cai WQ, Sethi G, et al. Insights into biological role of LncRNAs in epithelial-mesenchymal transition. Cells. 2019;8(10):1178.PubMedCentral Cheng JT, Wang L, Wang H, Tang FR, Cai WQ, Sethi G, et al. Insights into biological role of LncRNAs in epithelial-mesenchymal transition. Cells. 2019;8(10):1178.PubMedCentral
89.
Zurück zum Zitat Moraes LA, Kar S, Foo SL, Gu T, Toh YQ, Ampomah PB, et al. Annexin-A1 enhances breast cancer growth and migration by promoting alternative macrophage polarization in the tumour microenvironment. Sci Rep. 2017;7(1):17925.PubMedPubMedCentral Moraes LA, Kar S, Foo SL, Gu T, Toh YQ, Ampomah PB, et al. Annexin-A1 enhances breast cancer growth and migration by promoting alternative macrophage polarization in the tumour microenvironment. Sci Rep. 2017;7(1):17925.PubMedPubMedCentral
90.
Zurück zum Zitat Yang Y, Guo Z, Chen W, Wang X, Cao M, Han X, et al. M2 macrophage-derived exosomes promote angiogenesis and growth of pancreatic ductal adenocarcinoma by targeting E2F2. Mol Ther J Am Soc Gene Ther. 2021;29(3):1226–38. Yang Y, Guo Z, Chen W, Wang X, Cao M, Han X, et al. M2 macrophage-derived exosomes promote angiogenesis and growth of pancreatic ductal adenocarcinoma by targeting E2F2. Mol Ther J Am Soc Gene Ther. 2021;29(3):1226–38.
91.
Zurück zum Zitat Dong F, Ruan S, Wang J, Xia Y, Le K, Xiao X, et al. M2 macrophage-induced lncRNA PCAT6 facilitates tumorigenesis and angiogenesis of triple-negative breast cancer through modulation of VEGFR2. Cell Death Dis. 2020;11(9):728.PubMedPubMedCentral Dong F, Ruan S, Wang J, Xia Y, Le K, Xiao X, et al. M2 macrophage-induced lncRNA PCAT6 facilitates tumorigenesis and angiogenesis of triple-negative breast cancer through modulation of VEGFR2. Cell Death Dis. 2020;11(9):728.PubMedPubMedCentral
92.
Zurück zum Zitat Jung YY, Um J-Y, Nasif O, Alharbi SA, Sethi G, Ahn KS. Blockage of the JAK/STAT3 signaling pathway in multiple myeloma by leelamine. Phytomedicine. 2021;87:153574.PubMed Jung YY, Um J-Y, Nasif O, Alharbi SA, Sethi G, Ahn KS. Blockage of the JAK/STAT3 signaling pathway in multiple myeloma by leelamine. Phytomedicine. 2021;87:153574.PubMed
93.
Zurück zum Zitat Mohan CD, Rangappa S, Nayak SC, Sethi G, Rangappa KS. Paradoxical functions of long noncoding RNAs in modulating STAT3 signaling pathway in hepatocellular carcinoma. Biochim Biophys Acta (BBA) Rev Cancer. 2021;1876:188574. Mohan CD, Rangappa S, Nayak SC, Sethi G, Rangappa KS. Paradoxical functions of long noncoding RNAs in modulating STAT3 signaling pathway in hepatocellular carcinoma. Biochim Biophys Acta (BBA) Rev Cancer. 2021;1876:188574.
94.
Zurück zum Zitat Garg M, Shanmugam MK, Bhardwaj V, Goel A, Gupta R, Sharma A, et al. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Med Res Rev. 2021;41(3):1291–336. Garg M, Shanmugam MK, Bhardwaj V, Goel A, Gupta R, Sharma A, et al. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Med Res Rev. 2021;41(3):1291–336.
96.
Zurück zum Zitat Lee JH, Mohan CD, Deivasigamani A, Jung YY, Rangappa S, Basappa S, et al. Brusatol suppresses STAT3-driven metastasis by downregulating epithelial-mesenchymal transition in hepatocellular carcinoma. J Adv Res. 2020;26:83–94.PubMedPubMedCentral Lee JH, Mohan CD, Deivasigamani A, Jung YY, Rangappa S, Basappa S, et al. Brusatol suppresses STAT3-driven metastasis by downregulating epithelial-mesenchymal transition in hepatocellular carcinoma. J Adv Res. 2020;26:83–94.PubMedPubMedCentral
97.
Zurück zum Zitat Xu J, Zhang J, Zhang Z, Gao Z, Qi Y, Qiu W, et al. Hypoxic glioma-derived exosomes promote M2-like macrophage polarization by enhancing autophagy induction. Cell Death Dis. 2021;12(4):373.PubMedPubMedCentral Xu J, Zhang J, Zhang Z, Gao Z, Qi Y, Qiu W, et al. Hypoxic glioma-derived exosomes promote M2-like macrophage polarization by enhancing autophagy induction. Cell Death Dis. 2021;12(4):373.PubMedPubMedCentral
99.
Zurück zum Zitat Yin H, Qiu X, Shan Y, You B, Xie L, Zhang P, et al. HIF-1α downregulation of miR-433-3p in adipocyte-derived exosomes contributes to NPC progression via targeting SCD1. Cancer Sci. 2021;112(4):1457–70.PubMedPubMedCentral Yin H, Qiu X, Shan Y, You B, Xie L, Zhang P, et al. HIF-1α downregulation of miR-433-3p in adipocyte-derived exosomes contributes to NPC progression via targeting SCD1. Cancer Sci. 2021;112(4):1457–70.PubMedPubMedCentral
100.
Zurück zum Zitat Li Y, Zhang X, Zheng Q, Zhang Y, Ma Y, Zhu C, et al. YAP1 inhibition in HUVECs Is associated with released exosomes and increased hepatocarcinoma invasion and metastasis. Mol Ther Nucleic Acids. 2020;21:86–97.PubMedPubMedCentral Li Y, Zhang X, Zheng Q, Zhang Y, Ma Y, Zhu C, et al. YAP1 inhibition in HUVECs Is associated with released exosomes and increased hepatocarcinoma invasion and metastasis. Mol Ther Nucleic Acids. 2020;21:86–97.PubMedPubMedCentral
101.
Zurück zum Zitat Wu J, Gao W, Tang Q, Yu Y, You W, Wu Z, et al. M2 macrophage-derived exosomes facilitate HCC metastasis by transferring α(M) β(2) integrin to tumor cells. Hepatology (Baltimore, MD). 2021;73(4):1365–80. Wu J, Gao W, Tang Q, Yu Y, You W, Wu Z, et al. M2 macrophage-derived exosomes facilitate HCC metastasis by transferring α(M) β(2) integrin to tumor cells. Hepatology (Baltimore, MD). 2021;73(4):1365–80.
102.
Zurück zum Zitat Zhou W, Zhou Y, Chen X, Ning T, Chen H, Guo Q, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268:120546.PubMed Zhou W, Zhou Y, Chen X, Ning T, Chen H, Guo Q, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268:120546.PubMed
103.
Zurück zum Zitat Zhu D, Liu Z, Li Y, Huang Q, Xia L, Li K. Delivery of manganese carbonyl to the tumor microenvironment using tumor-derived exosomes for cancer gas therapy and low dose radiotherapy. Biomaterials. 2021;274:120894.PubMed Zhu D, Liu Z, Li Y, Huang Q, Xia L, Li K. Delivery of manganese carbonyl to the tumor microenvironment using tumor-derived exosomes for cancer gas therapy and low dose radiotherapy. Biomaterials. 2021;274:120894.PubMed
104.
Zurück zum Zitat Wang J, Tang W, Yang M, Yin Y, Li H, Hu F, et al. Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery system for targeted glioma therapy. Biomaterials. 2021;273:120784.PubMed Wang J, Tang W, Yang M, Yin Y, Li H, Hu F, et al. Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery system for targeted glioma therapy. Biomaterials. 2021;273:120784.PubMed
105.
Zurück zum Zitat Lima LG, Ham S, Shin H, Chai EPZ, Lek ESH, Lobb RJ, et al. Tumor microenvironmental cytokines bound to cancer exosomes determine uptake by cytokine receptor-expressing cells and biodistribution. Nat Commun. 2021;12(1):3543.PubMedPubMedCentral Lima LG, Ham S, Shin H, Chai EPZ, Lek ESH, Lobb RJ, et al. Tumor microenvironmental cytokines bound to cancer exosomes determine uptake by cytokine receptor-expressing cells and biodistribution. Nat Commun. 2021;12(1):3543.PubMedPubMedCentral
106.
Zurück zum Zitat Ferguson Bennit HR, Gonda A, Kabagwira J, Oppegard L, Chi D, Licero Campbell J, et al. Natural killer cell phenotype and functionality affected by exposure to extracellular survivin and lymphoma-derived exosomes. Int J Mol Sci. 2021;22(3):1255.PubMedPubMedCentral Ferguson Bennit HR, Gonda A, Kabagwira J, Oppegard L, Chi D, Licero Campbell J, et al. Natural killer cell phenotype and functionality affected by exposure to extracellular survivin and lymphoma-derived exosomes. Int J Mol Sci. 2021;22(3):1255.PubMedPubMedCentral
107.
Zurück zum Zitat Markiewski MM, Daugherity E, Reese B, Karbowniczek M. The role of complement in angiogenesis. Antibodies. 2020;9(4):67.PubMedCentral Markiewski MM, Daugherity E, Reese B, Karbowniczek M. The role of complement in angiogenesis. Antibodies. 2020;9(4):67.PubMedCentral
108.
Zurück zum Zitat Lee JH, Kim C, Um JY, Sethi G, Ahn KS. Casticin-induced inhibition of cell growth and survival are mediated through the dual modulation of Akt/mTOR signaling cascade. Cancers. 2019;11(2):254.PubMedCentral Lee JH, Kim C, Um JY, Sethi G, Ahn KS. Casticin-induced inhibition of cell growth and survival are mediated through the dual modulation of Akt/mTOR signaling cascade. Cancers. 2019;11(2):254.PubMedCentral
109.
Zurück zum Zitat Padmavathi G, Banik K, Monisha J, Bordoloi D, Shabnam B, Arfuso F, et al. Novel tumor necrosis factor-α induced protein eight (TNFAIP8/TIPE) family: functions and downstream targets involved in cancer progression. Cancer Lett. 2018;432:260–71.PubMed Padmavathi G, Banik K, Monisha J, Bordoloi D, Shabnam B, Arfuso F, et al. Novel tumor necrosis factor-α induced protein eight (TNFAIP8/TIPE) family: functions and downstream targets involved in cancer progression. Cancer Lett. 2018;432:260–71.PubMed
110.
Zurück zum Zitat Siveen KS, Ahn KS, Ong TH, Shanmugam MK, Li F, Yap WN, et al. Y-tocotrienol inhibits angiogenesis-dependent growth of human hepatocellular carcinoma through abrogation of AKT/mTOR pathway in an orthotopic mouse model. Oncotarget. 2014;5(7):1897–911.PubMedPubMedCentral Siveen KS, Ahn KS, Ong TH, Shanmugam MK, Li F, Yap WN, et al. Y-tocotrienol inhibits angiogenesis-dependent growth of human hepatocellular carcinoma through abrogation of AKT/mTOR pathway in an orthotopic mouse model. Oncotarget. 2014;5(7):1897–911.PubMedPubMedCentral
111.
Zurück zum Zitat Flamme I, Breier G, Risau W. Vascular endothelial growth factor (VEGF) and VEGF receptor 2 (flk-1) are expressed during vasculogenesis and vascular differentiation in the quail embryo. Dev Biol. 1995;169(2):699–712.PubMed Flamme I, Breier G, Risau W. Vascular endothelial growth factor (VEGF) and VEGF receptor 2 (flk-1) are expressed during vasculogenesis and vascular differentiation in the quail embryo. Dev Biol. 1995;169(2):699–712.PubMed
112.
Zurück zum Zitat Risau W, Sariola H, Zerwes HG, Sasse J, Ekblom P, Kemler R, et al. Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies. Development (Cambridge, England). 1988;102(3):471–8. Risau W, Sariola H, Zerwes HG, Sasse J, Ekblom P, Kemler R, et al. Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies. Development (Cambridge, England). 1988;102(3):471–8.
113.
Zurück zum Zitat Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285(21):1182–6.PubMed Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285(21):1182–6.PubMed
114.
Zurück zum Zitat Eelen G, Treps L, Li X, Carmeliet P. Basic and therapeutic aspects of angiogenesis updated. Circ Res. 2020;127(2):310–29.PubMed Eelen G, Treps L, Li X, Carmeliet P. Basic and therapeutic aspects of angiogenesis updated. Circ Res. 2020;127(2):310–29.PubMed
115.
Zurück zum Zitat Anbalagan D, Yap G, Yuan Y, Pandey VK, Lau WH, Arora S, et al. Annexin-A1 regulates microRNA-26b* and microRNA-562 to directly target NF-κB and angiogenesis in breast cancer cells. PLoS One. 2014;9(12):e114507.PubMedPubMedCentral Anbalagan D, Yap G, Yuan Y, Pandey VK, Lau WH, Arora S, et al. Annexin-A1 regulates microRNA-26b* and microRNA-562 to directly target NF-κB and angiogenesis in breast cancer cells. PLoS One. 2014;9(12):e114507.PubMedPubMedCentral
116.
Zurück zum Zitat Puar YR, Shanmugam MK, Fan L, Arfuso F, Sethi G, Tergaonkar V. Evidence for the involvement of the master transcription factor NF-κB in cancer initiation and progression. Biomedicines. 2018;6(3):82.PubMedCentral Puar YR, Shanmugam MK, Fan L, Arfuso F, Sethi G, Tergaonkar V. Evidence for the involvement of the master transcription factor NF-κB in cancer initiation and progression. Biomedicines. 2018;6(3):82.PubMedCentral
117.
Zurück zum Zitat Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Medi. 2014;6(265):265sr6. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Medi. 2014;6(265):265sr6.
118.
Zurück zum Zitat Singh N, Badrun D, Ghatage PJ. State of the art and up-and-coming angiogenesis inhibitors for ovarian cancer. Expert Opin Pharmacother. 2020;21(13):1579–90.PubMed Singh N, Badrun D, Ghatage PJ. State of the art and up-and-coming angiogenesis inhibitors for ovarian cancer. Expert Opin Pharmacother. 2020;21(13):1579–90.PubMed
119.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMed Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMed
120.
Zurück zum Zitat Wang H, Wang L, Zhou X, Luo X, Liu K, Jiang E, et al. OSCC exosomes regulate miR-210-3p targeting EFNA3 to promote oral cancer angiogenesis through the PI3K/AKT pathway. Biomed Res Int. 2020;2020:2125656.PubMedPubMedCentral Wang H, Wang L, Zhou X, Luo X, Liu K, Jiang E, et al. OSCC exosomes regulate miR-210-3p targeting EFNA3 to promote oral cancer angiogenesis through the PI3K/AKT pathway. Biomed Res Int. 2020;2020:2125656.PubMedPubMedCentral
121.
Zurück zum Zitat Bao L, You B, Shi S, Shan Y, Zhang Q, Yue H, et al. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene. 2018;37(21):2873–89.PubMedPubMedCentral Bao L, You B, Shi S, Shan Y, Zhang Q, Yue H, et al. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene. 2018;37(21):2873–89.PubMedPubMedCentral
122.
Zurück zum Zitat Yang Y, Liu Q, Lu J, Adah D, Yu S, Zhao S, et al. Exosomes from Plasmodium-infected hosts inhibit tumor angiogenesis in a murine Lewis lung cancer model. Oncogenesis. 2017;6(6):e351.PubMedPubMedCentral Yang Y, Liu Q, Lu J, Adah D, Yu S, Zhao S, et al. Exosomes from Plasmodium-infected hosts inhibit tumor angiogenesis in a murine Lewis lung cancer model. Oncogenesis. 2017;6(6):e351.PubMedPubMedCentral
123.
Zurück zum Zitat Cheng Y, Dai X, Yang T, Zhang N, Liu Z, Jiang Y. Low long noncoding RNA growth arrest-specific transcript 5 expression in the exosomes of lung cancer cells promotes tumor angiogenesis. J Oncol. 2019;2019:2476175.PubMedPubMedCentral Cheng Y, Dai X, Yang T, Zhang N, Liu Z, Jiang Y. Low long noncoding RNA growth arrest-specific transcript 5 expression in the exosomes of lung cancer cells promotes tumor angiogenesis. J Oncol. 2019;2019:2476175.PubMedPubMedCentral
124.
Zurück zum Zitat Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018;15(5):325–40.PubMedPubMedCentral Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018;15(5):325–40.PubMedPubMedCentral
125.
Zurück zum Zitat Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. Lancet (London, England). 2016;388(10043):518–29. Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. Lancet (London, England). 2016;388(10043):518–29.
126.
Zurück zum Zitat Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov. 2017;16(9):635–61.PubMed Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov. 2017;16(9):635–61.PubMed
127.
Zurück zum Zitat Akwii RG, Sajib MS, Zahra FT, Mikelis CM. Role of angiopoietin-2 in vascular physiology and pathophysiology. Cells. 2019;8(5):471.PubMedCentral Akwii RG, Sajib MS, Zahra FT, Mikelis CM. Role of angiopoietin-2 in vascular physiology and pathophysiology. Cells. 2019;8(5):471.PubMedCentral
128.
Zurück zum Zitat Scholz A, Plate KH, Reiss Y. Angiopoietin-2: a multifaceted cytokine that functions in both angiogenesis and inflammation. Ann N Y Acad Sci. 2015;1347:45–51.PubMed Scholz A, Plate KH, Reiss Y. Angiopoietin-2: a multifaceted cytokine that functions in both angiogenesis and inflammation. Ann N Y Acad Sci. 2015;1347:45–51.PubMed
129.
Zurück zum Zitat Xie JY, Wei JX, Lv LH, Han QF, Yang WB, Li GL, et al. Angiopoietin-2 induces angiogenesis via exosomes in human hepatocellular carcinoma. Cell Commun Signal. 2020;18(1):46.PubMedPubMedCentral Xie JY, Wei JX, Lv LH, Han QF, Yang WB, Li GL, et al. Angiopoietin-2 induces angiogenesis via exosomes in human hepatocellular carcinoma. Cell Commun Signal. 2020;18(1):46.PubMedPubMedCentral
130.
Zurück zum Zitat Kling MJ, Chaturvedi NK, Kesherwani V, Coulter DW, McGuire TR, Sharp JG, et al. Exosomes secreted under hypoxia enhance stemness in Ewing’s sarcoma through miR-210 delivery. Oncotarget. 2020;11(40):3633–45.PubMedPubMedCentral Kling MJ, Chaturvedi NK, Kesherwani V, Coulter DW, McGuire TR, Sharp JG, et al. Exosomes secreted under hypoxia enhance stemness in Ewing’s sarcoma through miR-210 delivery. Oncotarget. 2020;11(40):3633–45.PubMedPubMedCentral
132.
Zurück zum Zitat Huang Z, Feng Y. Exosomes derived from hypoxic colorectal cancer cells promote angiogenesis through Wnt4-induced β-catenin signaling in endothelial cells. Oncol Res. 2017;25(5):651–61.PubMedPubMedCentral Huang Z, Feng Y. Exosomes derived from hypoxic colorectal cancer cells promote angiogenesis through Wnt4-induced β-catenin signaling in endothelial cells. Oncol Res. 2017;25(5):651–61.PubMedPubMedCentral
133.
Zurück zum Zitat Pan D, Acevedo-Cintrón JA, Sayanagi J, Snyder-Warwick AK, Mackinnon SE, Wood MD. The CCL2/CCR2 axis is critical to recruiting macrophages into acellular nerve allograft bridging a nerve gap to promote angiogenesis and regeneration. Exp Neurol. 2020;331:113363.PubMedPubMedCentral Pan D, Acevedo-Cintrón JA, Sayanagi J, Snyder-Warwick AK, Mackinnon SE, Wood MD. The CCL2/CCR2 axis is critical to recruiting macrophages into acellular nerve allograft bridging a nerve gap to promote angiogenesis and regeneration. Exp Neurol. 2020;331:113363.PubMedPubMedCentral
134.
Zurück zum Zitat Tay H, Du Cheyne C, Demeyere K, De Craene J, De Bels L, Meyer E, et al. Depletion of embryonic macrophages leads to a reduction in angiogenesis in the ex ovo chick chorioallantoic membrane assay. Cells. 2020;10(1):5.PubMedCentral Tay H, Du Cheyne C, Demeyere K, De Craene J, De Bels L, Meyer E, et al. Depletion of embryonic macrophages leads to a reduction in angiogenesis in the ex ovo chick chorioallantoic membrane assay. Cells. 2020;10(1):5.PubMedCentral
135.
Zurück zum Zitat Ludwig N, Yerneni SS, Azambuja JH, Gillespie DG, Menshikova EV, Jackson EK, et al. Tumor-derived exosomes promote angiogenesis via adenosine A(2B) receptor signaling. Angiogenesis. 2020;23(4):599–610.PubMedPubMedCentral Ludwig N, Yerneni SS, Azambuja JH, Gillespie DG, Menshikova EV, Jackson EK, et al. Tumor-derived exosomes promote angiogenesis via adenosine A(2B) receptor signaling. Angiogenesis. 2020;23(4):599–610.PubMedPubMedCentral
136.
Zurück zum Zitat Mao L, Li X, Gong S, Yuan H, Jiang Y, Huang W, et al. Serum exosomes contain ECRG4 mRNA that suppresses tumor growth via inhibition of genes involved in inflammation, cell proliferation, and angiogenesis. Cancer Gene Ther. 2018;25(9–10):248–59.PubMed Mao L, Li X, Gong S, Yuan H, Jiang Y, Huang W, et al. Serum exosomes contain ECRG4 mRNA that suppresses tumor growth via inhibition of genes involved in inflammation, cell proliferation, and angiogenesis. Cancer Gene Ther. 2018;25(9–10):248–59.PubMed
137.
Zurück zum Zitat Yi H, Ye J, Yang XM, Zhang LW, Zhang ZG, Chen YP. High-grade ovarian cancer secreting effective exosomes in tumor angiogenesis. Int J Clin Exp Pathol. 2015;8(5):5062–70.PubMedPubMedCentral Yi H, Ye J, Yang XM, Zhang LW, Zhang ZG, Chen YP. High-grade ovarian cancer secreting effective exosomes in tumor angiogenesis. Int J Clin Exp Pathol. 2015;8(5):5062–70.PubMedPubMedCentral
138.
Zurück zum Zitat Horie K, Kawakami K, Fujita Y, Sugaya M, Kameyama K, Mizutani K, et al. Exosomes expressing carbonic anhydrase 9 promote angiogenesis. Biochem Biophys Res Commun. 2017;492(3):356–61.PubMed Horie K, Kawakami K, Fujita Y, Sugaya M, Kameyama K, Mizutani K, et al. Exosomes expressing carbonic anhydrase 9 promote angiogenesis. Biochem Biophys Res Commun. 2017;492(3):356–61.PubMed
139.
Zurück zum Zitat Alcayaga-Miranda F, González PL, Lopez-Verrilli A, Varas-Godoy M, Aguila-Díaz C, Contreras L, et al. Prostate tumor-induced angiogenesis is blocked by exosomes derived from menstrual stem cells through the inhibition of reactive oxygen species. Oncotarget. 2016;7(28):44462–77.PubMedPubMedCentral Alcayaga-Miranda F, González PL, Lopez-Verrilli A, Varas-Godoy M, Aguila-Díaz C, Contreras L, et al. Prostate tumor-induced angiogenesis is blocked by exosomes derived from menstrual stem cells through the inhibition of reactive oxygen species. Oncotarget. 2016;7(28):44462–77.PubMedPubMedCentral
140.
Zurück zum Zitat Umezu T, Imanishi S, Azuma K, Kobayashi C, Yoshizawa S, Ohyashiki K, et al. Replenishing exosomes from older bone marrow stromal cells with miR-340 inhibits myeloma-related angiogenesis. Blood Adv. 2017;1(13):812–23.PubMedPubMedCentral Umezu T, Imanishi S, Azuma K, Kobayashi C, Yoshizawa S, Ohyashiki K, et al. Replenishing exosomes from older bone marrow stromal cells with miR-340 inhibits myeloma-related angiogenesis. Blood Adv. 2017;1(13):812–23.PubMedPubMedCentral
141.
Zurück zum Zitat Wang Y, Cen A, Yang Y, Ye H, Li J, Liu S, et al. miR-181a, delivered by hypoxic PTC-secreted exosomes, inhibits DACT2 by downregulating MLL3, leading to YAP-VEGF-mediated angiogenesis. Mol Ther Nucleic Acids. 2021;24:610–21.PubMedPubMedCentral Wang Y, Cen A, Yang Y, Ye H, Li J, Liu S, et al. miR-181a, delivered by hypoxic PTC-secreted exosomes, inhibits DACT2 by downregulating MLL3, leading to YAP-VEGF-mediated angiogenesis. Mol Ther Nucleic Acids. 2021;24:610–21.PubMedPubMedCentral
142.
Zurück zum Zitat Dai X, Wang L, Deivasigamni A, Looi CY, Karthikeyan C, Trivedi P, et al. A novel benzimidazole derivative, MBIC inhibits tumor growth and promotes apoptosis via activation of ROS-dependent JNK signaling pathway in hepatocellular carcinoma. Oncotarget. 2017;8(8):12831–42.PubMedPubMedCentral Dai X, Wang L, Deivasigamni A, Looi CY, Karthikeyan C, Trivedi P, et al. A novel benzimidazole derivative, MBIC inhibits tumor growth and promotes apoptosis via activation of ROS-dependent JNK signaling pathway in hepatocellular carcinoma. Oncotarget. 2017;8(8):12831–42.PubMedPubMedCentral
143.
Zurück zum Zitat Ong PS, Wang LZ, Dai X, Tseng SH, Loo SJ, Sethi G. Judicious toggling of mTOR activity to combat insulin resistance and cancer: current evidence and perspectives. Front Pharmacol. 2016;7:395.PubMedPubMedCentral Ong PS, Wang LZ, Dai X, Tseng SH, Loo SJ, Sethi G. Judicious toggling of mTOR activity to combat insulin resistance and cancer: current evidence and perspectives. Front Pharmacol. 2016;7:395.PubMedPubMedCentral
144.
Zurück zum Zitat Mohan CD, Bharathkumar H, Bulusu KC, Pandey V, Rangappa S, Fuchs JE, et al. Development of a novel azaspirane that targets the Janus kinase-signal transducer and activator of transcription (STAT) pathway in hepatocellular carcinoma in vitro and in vivo. J Biol Chem. 2014;289(49):34296–307.PubMedPubMedCentral Mohan CD, Bharathkumar H, Bulusu KC, Pandey V, Rangappa S, Fuchs JE, et al. Development of a novel azaspirane that targets the Janus kinase-signal transducer and activator of transcription (STAT) pathway in hepatocellular carcinoma in vitro and in vivo. J Biol Chem. 2014;289(49):34296–307.PubMedPubMedCentral
145.
Zurück zum Zitat Manu KA, Shanmugam MK, Li F, Chen L, Siveen KS, Ahn KS, et al. Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. J Mol Med (Berl). 2014;92(3):267–76. Manu KA, Shanmugam MK, Li F, Chen L, Siveen KS, Ahn KS, et al. Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. J Mol Med (Berl). 2014;92(3):267–76.
146.
Zurück zum Zitat Tan SC. Low penetrance genetic polymorphisms as potential biomarkers for colorectal cancer predisposition. J Gene Med. 2018;20(4):e3010.PubMed Tan SC. Low penetrance genetic polymorphisms as potential biomarkers for colorectal cancer predisposition. J Gene Med. 2018;20(4):e3010.PubMed
147.
Zurück zum Zitat Jeong H, Kim S, Hong BJ, Lee CJ, Kim YE, Bok S, et al. Tumor-associated macrophages enhance tumor hypoxia and aerobic glycolysis. Can Res. 2019;79(4):795–806. Jeong H, Kim S, Hong BJ, Lee CJ, Kim YE, Bok S, et al. Tumor-associated macrophages enhance tumor hypoxia and aerobic glycolysis. Can Res. 2019;79(4):795–806.
148.
Zurück zum Zitat Nie H, Ju H, Fan J, Shi X, Cheng Y, Cang X, et al. O-GlcNAcylation of PGK1 coordinates glycolysis and TCA cycle to promote tumor growth. Nat Commun. 2020;11(1):36.PubMedPubMedCentral Nie H, Ju H, Fan J, Shi X, Cheng Y, Cang X, et al. O-GlcNAcylation of PGK1 coordinates glycolysis and TCA cycle to promote tumor growth. Nat Commun. 2020;11(1):36.PubMedPubMedCentral
149.
Zurück zum Zitat Deng F, Zhou R, Lin C, Yang S, Wang H, Li W, et al. Tumor-secreted dickkopf2 accelerates aerobic glycolysis and promotes angiogenesis in colorectal cancer. Theranostics. 2019;9(4):1001–14.PubMedPubMedCentral Deng F, Zhou R, Lin C, Yang S, Wang H, Li W, et al. Tumor-secreted dickkopf2 accelerates aerobic glycolysis and promotes angiogenesis in colorectal cancer. Theranostics. 2019;9(4):1001–14.PubMedPubMedCentral
150.
Zurück zum Zitat Liang Y, Wang H, Chen B, Mao Q, Xia W, Zhang T, et al. circDCUN1D4 suppresses tumor metastasis and glycolysis in lung adenocarcinoma by stabilizing TXNIP expression. Mol Ther Nucleic Acids. 2021;23:355–68.PubMed Liang Y, Wang H, Chen B, Mao Q, Xia W, Zhang T, et al. circDCUN1D4 suppresses tumor metastasis and glycolysis in lung adenocarcinoma by stabilizing TXNIP expression. Mol Ther Nucleic Acids. 2021;23:355–68.PubMed
151.
Zurück zum Zitat Wang H, Wang L, Pan H, Wang Y, Shi M, Yu H, et al. Exosomes derived from macrophages enhance aerobic glycolysis and chemoresistance in lung cancer by stabilizing c-Myc via the inhibition of NEDD4L. Front Cell Dev Biol. 2020;8:620603.PubMed Wang H, Wang L, Pan H, Wang Y, Shi M, Yu H, et al. Exosomes derived from macrophages enhance aerobic glycolysis and chemoresistance in lung cancer by stabilizing c-Myc via the inhibition of NEDD4L. Front Cell Dev Biol. 2020;8:620603.PubMed
152.
Zurück zum Zitat Wang B, Wang X, Hou D, Huang Q, Zhan W, Chen C, et al. Exosomes derived from acute myeloid leukemia cells promote chemoresistance by enhancing glycolysis-mediated vascular remodeling. J Cell Physiol. 2019;234(7):10602–14.PubMed Wang B, Wang X, Hou D, Huang Q, Zhan W, Chen C, et al. Exosomes derived from acute myeloid leukemia cells promote chemoresistance by enhancing glycolysis-mediated vascular remodeling. J Cell Physiol. 2019;234(7):10602–14.PubMed
153.
Zurück zum Zitat Lee JH, Kim C, Baek SH, Ko JH, Lee SG, Yang WM, et al. Capsazepine inhibits JAK/STAT3 signaling, tumor growth, and cell survival in prostate cancer. Oncotarget. 2017;8(11):17700–11.PubMed Lee JH, Kim C, Baek SH, Ko JH, Lee SG, Yang WM, et al. Capsazepine inhibits JAK/STAT3 signaling, tumor growth, and cell survival in prostate cancer. Oncotarget. 2017;8(11):17700–11.PubMed
154.
Zurück zum Zitat Liu Y, Liao S, Bennett S, Tang H, Song D, Wood D, et al. STAT3 and its targeting inhibitors in osteosarcoma. Cell Prolif. 2021;54(2):e12974.PubMed Liu Y, Liao S, Bennett S, Tang H, Song D, Wood D, et al. STAT3 and its targeting inhibitors in osteosarcoma. Cell Prolif. 2021;54(2):e12974.PubMed
155.
Zurück zum Zitat Wang H, Tao Z, Feng M, Li X, Deng Z, Zhao G, et al. Dual PLK1 and STAT3 inhibition promotes glioblastoma cells apoptosis through MYC. Biochem Biophys Res Commun. 2020;533(3):368–75.PubMed Wang H, Tao Z, Feng M, Li X, Deng Z, Zhao G, et al. Dual PLK1 and STAT3 inhibition promotes glioblastoma cells apoptosis through MYC. Biochem Biophys Res Commun. 2020;533(3):368–75.PubMed
156.
Zurück zum Zitat Xu Y, Zhu Y, Yue Y, Pu S, Wu J, Lv Y, et al. Tamoxifen attenuates reactive astrocyte-induced brain metastasis and drug resistance through the IL-6/STAT3 signaling pathway. Acta Biochim Biophys Sin. 2020;52(12):1299–305.PubMed Xu Y, Zhu Y, Yue Y, Pu S, Wu J, Lv Y, et al. Tamoxifen attenuates reactive astrocyte-induced brain metastasis and drug resistance through the IL-6/STAT3 signaling pathway. Acta Biochim Biophys Sin. 2020;52(12):1299–305.PubMed
157.
Zurück zum Zitat Cong Y, Cui Y, Zhu S, Cao J, Zou H, Martin TA, et al. Tim-3 promotes cell aggressiveness and paclitaxel resistance through NF-κB/STAT3 signalling pathway in breast cancer cells. Chin J Cancer Res. 2020;32(5):564–79.PubMedPubMedCentral Cong Y, Cui Y, Zhu S, Cao J, Zou H, Martin TA, et al. Tim-3 promotes cell aggressiveness and paclitaxel resistance through NF-κB/STAT3 signalling pathway in breast cancer cells. Chin J Cancer Res. 2020;32(5):564–79.PubMedPubMedCentral
158.
Zurück zum Zitat Jiang Y, Chen P, Hu K, Dai G, Li J, Zheng D, et al. Inflammatory microenvironment of fibrotic liver promotes hepatocellular carcinoma growth, metastasis and sorafenib resistance through STAT3 activation. J Cell Mol Med. 2021;25(3):1568–82.PubMedPubMedCentral Jiang Y, Chen P, Hu K, Dai G, Li J, Zheng D, et al. Inflammatory microenvironment of fibrotic liver promotes hepatocellular carcinoma growth, metastasis and sorafenib resistance through STAT3 activation. J Cell Mol Med. 2021;25(3):1568–82.PubMedPubMedCentral
159.
Zurück zum Zitat Ham S, Lima LG, Chai EPZ, Muller A, Lobb RJ, Krumeich S, et al. Breast cancer-derived exosomes alter macrophage polarization via gp130/STAT3 signaling. Front Immunol. 2018;9:871.PubMedPubMedCentral Ham S, Lima LG, Chai EPZ, Muller A, Lobb RJ, Krumeich S, et al. Breast cancer-derived exosomes alter macrophage polarization via gp130/STAT3 signaling. Front Immunol. 2018;9:871.PubMedPubMedCentral
160.
Zurück zum Zitat Zhou J, Li X, Wu X, Zhang T, Zhu Q, Wang X, et al. Exosomes released from tumor-associated macrophages transfer miRNAs that induce a Treg/Th17 cell imbalance in epithelial ovarian cancer. Cancer Immunol Res. 2018;6(12):1578–92.PubMed Zhou J, Li X, Wu X, Zhang T, Zhu Q, Wang X, et al. Exosomes released from tumor-associated macrophages transfer miRNAs that induce a Treg/Th17 cell imbalance in epithelial ovarian cancer. Cancer Immunol Res. 2018;6(12):1578–92.PubMed
161.
Zurück zum Zitat Yu X, Zhang Q, Zhang X, Han Q, Li H, Mao Y, et al. Exosomes from macrophages exposed to apoptotic breast cancer cells promote breast cancer proliferation and metastasis. J Cancer. 2019;10(13):2892–906.PubMedPubMedCentral Yu X, Zhang Q, Zhang X, Han Q, Li H, Mao Y, et al. Exosomes from macrophages exposed to apoptotic breast cancer cells promote breast cancer proliferation and metastasis. J Cancer. 2019;10(13):2892–906.PubMedPubMedCentral
162.
Zurück zum Zitat Ren R, Sun H, Ma C, Liu J, Wang H. Colon cancer cells secrete exosomes to promote self-proliferation by shortening mitosis duration and activation of STAT3 in a hypoxic environment. Cell Biosci. 2019;9:62.PubMedPubMedCentral Ren R, Sun H, Ma C, Liu J, Wang H. Colon cancer cells secrete exosomes to promote self-proliferation by shortening mitosis duration and activation of STAT3 in a hypoxic environment. Cell Biosci. 2019;9:62.PubMedPubMedCentral
163.
Zurück zum Zitat He C, Hua W, Liu J, Fan L, Wang H, Sun G. Exosomes derived from endoplasmic reticulum-stressed liver cancer cells enhance the expression of cytokines in macrophages via the STAT3 signaling pathway. Oncol Lett. 2020;20(1):589–600.PubMedPubMedCentral He C, Hua W, Liu J, Fan L, Wang H, Sun G. Exosomes derived from endoplasmic reticulum-stressed liver cancer cells enhance the expression of cytokines in macrophages via the STAT3 signaling pathway. Oncol Lett. 2020;20(1):589–600.PubMedPubMedCentral
164.
Zurück zum Zitat Xing H, Liang C, Xu X, Sun H, Ma X, Jiang Z. Mesenchymal stroma/stem-like cells of GARP knockdown inhibits cell proliferation and invasion of mouse colon cancer cells (MC38) through exosomes. J Cell Mol Med. 2020;24(23):13984–90.PubMedPubMedCentral Xing H, Liang C, Xu X, Sun H, Ma X, Jiang Z. Mesenchymal stroma/stem-like cells of GARP knockdown inhibits cell proliferation and invasion of mouse colon cancer cells (MC38) through exosomes. J Cell Mol Med. 2020;24(23):13984–90.PubMedPubMedCentral
165.
Zurück zum Zitat Dorayappan KDP, Gardner ML, Hisey CL, Zingarelli RA, Smith BQ, Lightfoot MDS, et al. A microfluidic chip enables isolation of exosomes and establishment of their protein profiles and associated signaling pathways in ovarian cancer. Can Res. 2019;79(13):3503–13. Dorayappan KDP, Gardner ML, Hisey CL, Zingarelli RA, Smith BQ, Lightfoot MDS, et al. A microfluidic chip enables isolation of exosomes and establishment of their protein profiles and associated signaling pathways in ovarian cancer. Can Res. 2019;79(13):3503–13.
166.
Zurück zum Zitat Tan SC, Ankathil R. Genetic susceptibility to cervical cancer: role of common polymorphisms in apoptosis-related genes. Tumour Biol J Int Soc Oncodev Biol Med. 2015;36(9):6633–44. Tan SC, Ankathil R. Genetic susceptibility to cervical cancer: role of common polymorphisms in apoptosis-related genes. Tumour Biol J Int Soc Oncodev Biol Med. 2015;36(9):6633–44.
167.
Zurück zum Zitat Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer. 2019;18(1):91.PubMedPubMedCentral Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer. 2019;18(1):91.PubMedPubMedCentral
168.
Zurück zum Zitat Kim C, Lee SG, Yang WM, Arfuso F, Um JY, Kumar AP, et al. Formononetin-induced oxidative stress abrogates the activation of STAT3/5 signaling axis and suppresses the tumor growth in multiple myeloma preclinical model. Cancer Lett. 2018;431:123–41.PubMed Kim C, Lee SG, Yang WM, Arfuso F, Um JY, Kumar AP, et al. Formononetin-induced oxidative stress abrogates the activation of STAT3/5 signaling axis and suppresses the tumor growth in multiple myeloma preclinical model. Cancer Lett. 2018;431:123–41.PubMed
169.
Zurück zum Zitat Kirtonia A, Sethi G, Garg M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci CMLS. 2020;77(22):4459–83.PubMed Kirtonia A, Sethi G, Garg M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci CMLS. 2020;77(22):4459–83.PubMed
170.
Zurück zum Zitat Lee JH, Kim C, Lee SG, Sethi G, Ahn KS. Ophiopogonin D, a steroidal glycoside abrogates STAT3 signaling cascade and exhibits anti-cancer activity by causing GSH/GSSG imbalance in lung carcinoma. Cancers. 2018;10(11):427.PubMedCentral Lee JH, Kim C, Lee SG, Sethi G, Ahn KS. Ophiopogonin D, a steroidal glycoside abrogates STAT3 signaling cascade and exhibits anti-cancer activity by causing GSH/GSSG imbalance in lung carcinoma. Cancers. 2018;10(11):427.PubMedCentral
171.
Zurück zum Zitat Ko JH, Lee JH, Jung SH, Lee SG, Chinnathambi A, Alharbi SA, et al. 2,5-Dihydroxyacetophenone induces apoptosis of multiple myeloma cells by regulating the MAPK activation pathway. Molecules (Basel, Switzerland). 2017;22(7):1157. Ko JH, Lee JH, Jung SH, Lee SG, Chinnathambi A, Alharbi SA, et al. 2,5-Dihydroxyacetophenone induces apoptosis of multiple myeloma cells by regulating the MAPK activation pathway. Molecules (Basel, Switzerland). 2017;22(7):1157.
172.
Zurück zum Zitat Patel GK, Khan MA, Bhardwaj A, Srivastava SK, Zubair H, Patton MC, et al. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme. DCK Br J Cancer. 2017;116(5):609–19.PubMed Patel GK, Khan MA, Bhardwaj A, Srivastava SK, Zubair H, Patton MC, et al. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme. DCK Br J Cancer. 2017;116(5):609–19.PubMed
173.
Zurück zum Zitat Subramaniam A, Loo SY, Rajendran P, Manu KA, Perumal E, Li F, et al. An anthraquinone derivative, emodin sensitizes hepatocellular carcinoma cells to TRAIL induced apoptosis through the induction of death receptors and downregulation of cell survival proteins. Apoptosis Int J Progr Cell Death. 2013;18(10):1175–87. Subramaniam A, Loo SY, Rajendran P, Manu KA, Perumal E, Li F, et al. An anthraquinone derivative, emodin sensitizes hepatocellular carcinoma cells to TRAIL induced apoptosis through the induction of death receptors and downregulation of cell survival proteins. Apoptosis Int J Progr Cell Death. 2013;18(10):1175–87.
174.
Zurück zum Zitat Shen T, Huang Z, Shi C, Pu X, Xu X, Wu Z, et al. Pancreatic cancer-derived exosomes induce apoptosis of T lymphocytes through the p38 MAPK-mediated endoplasmic reticulum stress. FASEB J Off Publ Federation Am Soc Exp Biol. 2020;34(6):8442–58. Shen T, Huang Z, Shi C, Pu X, Xu X, Wu Z, et al. Pancreatic cancer-derived exosomes induce apoptosis of T lymphocytes through the p38 MAPK-mediated endoplasmic reticulum stress. FASEB J Off Publ Federation Am Soc Exp Biol. 2020;34(6):8442–58.
175.
Zurück zum Zitat Wang B, Wang Y, Yan Z, Sun Y, Su C. Colorectal cancer cell-derived exosomes promote proliferation and decrease apoptosis by activating the ERK pathway. Int J Clin Exp Pathol. 2019;12(7):2485–95.PubMedPubMedCentral Wang B, Wang Y, Yan Z, Sun Y, Su C. Colorectal cancer cell-derived exosomes promote proliferation and decrease apoptosis by activating the ERK pathway. Int J Clin Exp Pathol. 2019;12(7):2485–95.PubMedPubMedCentral
176.
Zurück zum Zitat Deng S, Shanmugam MK, Kumar AP, Yap CT, Sethi G, Bishayee A. Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer. 2019;125(8):1228–46.PubMed Deng S, Shanmugam MK, Kumar AP, Yap CT, Sethi G, Bishayee A. Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer. 2019;125(8):1228–46.PubMed
178.
Zurück zum Zitat Hwang ST, Kim C, Lee JH, Chinnathambi A, Alharbi SA, Shair OHM, et al. Cycloastragenol can negate constitutive STAT3 activation and promote paclitaxel-induced apoptosis in human gastric cancer cells. Phytomed Int J Phytother Phytopharmacol. 2019;59:152907. Hwang ST, Kim C, Lee JH, Chinnathambi A, Alharbi SA, Shair OHM, et al. Cycloastragenol can negate constitutive STAT3 activation and promote paclitaxel-induced apoptosis in human gastric cancer cells. Phytomed Int J Phytother Phytopharmacol. 2019;59:152907.
179.
Zurück zum Zitat Praharaj PP, Naik PP, Panigrahi DP, Bhol CS, Mahapatra KK, Patra S, et al. Intricate role of mitochondrial lipid in mitophagy and mitochondrial apoptosis: its implication in cancer therapeutics. Cell Mol Life Sci CMLS. 2019;76(9):1641–52.PubMed Praharaj PP, Naik PP, Panigrahi DP, Bhol CS, Mahapatra KK, Patra S, et al. Intricate role of mitochondrial lipid in mitophagy and mitochondrial apoptosis: its implication in cancer therapeutics. Cell Mol Life Sci CMLS. 2019;76(9):1641–52.PubMed
180.
Zurück zum Zitat Wang L, Xu P, Xie X, Hu F, Jiang L, Hu R, et al. Down regulation of SIRT2 reduced ASS induced NSCLC apoptosis through the release of autophagy components via exosomes. Front Cell Dev Biol. 2020;8:601953.PubMedPubMedCentral Wang L, Xu P, Xie X, Hu F, Jiang L, Hu R, et al. Down regulation of SIRT2 reduced ASS induced NSCLC apoptosis through the release of autophagy components via exosomes. Front Cell Dev Biol. 2020;8:601953.PubMedPubMedCentral
181.
Zurück zum Zitat Zhang X, Shi H, Yuan X, Jiang P, Qian H, Xu W. Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Mol Cancer. 2018;17(1):146.PubMedPubMedCentral Zhang X, Shi H, Yuan X, Jiang P, Qian H, Xu W. Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Mol Cancer. 2018;17(1):146.PubMedPubMedCentral
182.
Zurück zum Zitat Yang P, Cao X, Cai H, Feng P, Chen X, Zhu Y, et al. The exosomes derived from CAR-T cell efficiently target mesothelin and reduce triple-negative breast cancer growth. Cell Immunol. 2021;360:104262.PubMed Yang P, Cao X, Cai H, Feng P, Chen X, Zhu Y, et al. The exosomes derived from CAR-T cell efficiently target mesothelin and reduce triple-negative breast cancer growth. Cell Immunol. 2021;360:104262.PubMed
183.
Zurück zum Zitat Wang S, Su X, Xu M, Xiao X, Li X, Li H, et al. Exosomes secreted by mesenchymal stromal/stem cell-derived adipocytes promote breast cancer cell growth via activation of Hippo signaling pathway. Stem Cell Res Ther. 2019;10(1):117.PubMedPubMedCentral Wang S, Su X, Xu M, Xiao X, Li X, Li H, et al. Exosomes secreted by mesenchymal stromal/stem cell-derived adipocytes promote breast cancer cell growth via activation of Hippo signaling pathway. Stem Cell Res Ther. 2019;10(1):117.PubMedPubMedCentral
184.
Zurück zum Zitat Lu Z, Zuo B, Jing R, Gao X, Rao Q, Liu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMed Lu Z, Zuo B, Jing R, Gao X, Rao Q, Liu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMed
185.
Zurück zum Zitat Li H, Chi X, Li R, Ouyang J, Chen Y. HIV-1-infected cell-derived exosomes promote the growth and progression of cervical cancer. Int J Biol Sci. 2019;15(11):2438–47.PubMedPubMedCentral Li H, Chi X, Li R, Ouyang J, Chen Y. HIV-1-infected cell-derived exosomes promote the growth and progression of cervical cancer. Int J Biol Sci. 2019;15(11):2438–47.PubMedPubMedCentral
186.
Zurück zum Zitat Shanmugam MK, Ahn KS, Hsu A, Woo CC, Yuan Y, Tan KHB, et al. Thymoquinone inhibits bone metastasis of breast cancer cells through abrogation of the CXCR4 signaling axis. Front Pharmacol. 2018;9:1294.PubMedPubMedCentral Shanmugam MK, Ahn KS, Hsu A, Woo CC, Yuan Y, Tan KHB, et al. Thymoquinone inhibits bone metastasis of breast cancer cells through abrogation of the CXCR4 signaling axis. Front Pharmacol. 2018;9:1294.PubMedPubMedCentral
187.
Zurück zum Zitat Chua AW, Hay HS, Rajendran P, Shanmugam MK, Li F, Bist P, et al. Butein downregulates chemokine receptor CXCR4 expression and function through suppression of NF-κB activation in breast and pancreatic tumor cells. Biochem Pharmacol. 2010;80(10):1553–62.PubMed Chua AW, Hay HS, Rajendran P, Shanmugam MK, Li F, Bist P, et al. Butein downregulates chemokine receptor CXCR4 expression and function through suppression of NF-κB activation in breast and pancreatic tumor cells. Biochem Pharmacol. 2010;80(10):1553–62.PubMed
188.
Zurück zum Zitat Shanmugam MK, Ong TH, Kumar AP, Lun CK, Ho PC, Wong PT, et al. Ursolic acid inhibits the initiation, progression of prostate cancer and prolongs the survival of TRAMP mice by modulating pro-inflammatory pathways. PLoS One. 2012;7(3):e32476.PubMedPubMedCentral Shanmugam MK, Ong TH, Kumar AP, Lun CK, Ho PC, Wong PT, et al. Ursolic acid inhibits the initiation, progression of prostate cancer and prolongs the survival of TRAMP mice by modulating pro-inflammatory pathways. PLoS One. 2012;7(3):e32476.PubMedPubMedCentral
189.
Zurück zum Zitat Feng W, Dean DC, Hornicek FJ, Shi H, Duan Z. Exosomes promote pre-metastatic niche formation in ovarian cancer. Mol Cancer. 2019;18(1):124.PubMedPubMedCentral Feng W, Dean DC, Hornicek FJ, Shi H, Duan Z. Exosomes promote pre-metastatic niche formation in ovarian cancer. Mol Cancer. 2019;18(1):124.PubMedPubMedCentral
190.
Zurück zum Zitat Yoshida K, Tsuda M, Matsumoto R, Semba S, Wang L, Sugino H, et al. Exosomes containing ErbB2/CRK induce vascular growth in premetastatic niches and promote metastasis of bladder cancer. Cancer Sci. 2019;110(7):2119–32.PubMedPubMedCentral Yoshida K, Tsuda M, Matsumoto R, Semba S, Wang L, Sugino H, et al. Exosomes containing ErbB2/CRK induce vascular growth in premetastatic niches and promote metastasis of bladder cancer. Cancer Sci. 2019;110(7):2119–32.PubMedPubMedCentral
191.
Zurück zum Zitat Wang H, Wei H, Wang J, Li L, Chen A, Li Z. MicroRNA-181d-5p-containing exosomes derived from CAFs promote EMT by regulating CDX2/HOXA5 in breast cancer. Mol Ther Nucleic Acids. 2020;19:654–67.PubMed Wang H, Wei H, Wang J, Li L, Chen A, Li Z. MicroRNA-181d-5p-containing exosomes derived from CAFs promote EMT by regulating CDX2/HOXA5 in breast cancer. Mol Ther Nucleic Acids. 2020;19:654–67.PubMed
192.
Zurück zum Zitat Maziveyi M, Dong S, Baranwal S, Mehrnezhad A, Rathinam R, Huckaba TM, et al. Exosomes from nischarin-expressing cells reduce breast cancer cell motility and tumor growth. Can Res. 2019;79(9):2152–66. Maziveyi M, Dong S, Baranwal S, Mehrnezhad A, Rathinam R, Huckaba TM, et al. Exosomes from nischarin-expressing cells reduce breast cancer cell motility and tumor growth. Can Res. 2019;79(9):2152–66.
194.
Zurück zum Zitat Wei Q, Wei L, Zhang J, Li Z, Feng H, Ren L. EphA2-enriched exosomes promote cell migration and are a potential diagnostic serum marker in pancreatic cancer. Mol Med Rep. 2020;22(4):2941–7.PubMedPubMedCentral Wei Q, Wei L, Zhang J, Li Z, Feng H, Ren L. EphA2-enriched exosomes promote cell migration and are a potential diagnostic serum marker in pancreatic cancer. Mol Med Rep. 2020;22(4):2941–7.PubMedPubMedCentral
195.
Zurück zum Zitat Xu S, Zheng L, Kang L, Xu H, Gao L. microRNA-let-7e in serum-derived exosomes inhibits the metastasis of non-small-cell lung cancer in a SUV39H2/LSD1/CDH1-dependent manner. Cancer Gene Ther. 2021;28(3–4):250–64.PubMed Xu S, Zheng L, Kang L, Xu H, Gao L. microRNA-let-7e in serum-derived exosomes inhibits the metastasis of non-small-cell lung cancer in a SUV39H2/LSD1/CDH1-dependent manner. Cancer Gene Ther. 2021;28(3–4):250–64.PubMed
196.
Zurück zum Zitat Hwang ST, Yang MH, Kumar AP, Sethi G, Ahn KS. Corilagin represses epithelial to mesenchymal transition process through modulating Wnt/β-catenin signaling cascade. Biomolecules. 2020;10(10):1406.PubMedCentral Hwang ST, Yang MH, Kumar AP, Sethi G, Ahn KS. Corilagin represses epithelial to mesenchymal transition process through modulating Wnt/β-catenin signaling cascade. Biomolecules. 2020;10(10):1406.PubMedCentral
197.
Zurück zum Zitat Yang MH, Lee JH, Ko JH, Jung SH, Sethi G, Ahn KS. Brassinin represses invasive potential of lung carcinoma cells through deactivation of PI3K/Akt/mTOR signaling cascade. Molecules (Basel, Switzerland). 2019;24(8):1584. Yang MH, Lee JH, Ko JH, Jung SH, Sethi G, Ahn KS. Brassinin represses invasive potential of lung carcinoma cells through deactivation of PI3K/Akt/mTOR signaling cascade. Molecules (Basel, Switzerland). 2019;24(8):1584.
198.
Zurück zum Zitat Ko JH, Nam D, Um JY, Jung SH, Sethi G, Ahn KS. Bergamottin suppresses metastasis of lung cancer cells through abrogation of diverse oncogenic signaling cascades and epithelial-to-mesenchymal transition. Molecules (Basel, Switzerland). 2018;23(7):1601. Ko JH, Nam D, Um JY, Jung SH, Sethi G, Ahn KS. Bergamottin suppresses metastasis of lung cancer cells through abrogation of diverse oncogenic signaling cascades and epithelial-to-mesenchymal transition. Molecules (Basel, Switzerland). 2018;23(7):1601.
199.
Zurück zum Zitat Dai X, Ahn KS, Wang LZ, Kim C, Deivasigamni A, Arfuso F, et al. Ascochlorin enhances the sensitivity of doxorubicin leading to the reversal of epithelial-to-mesenchymal transition in hepatocellular carcinoma. Mol Cancer Ther. 2016;15(12):2966–76.PubMed Dai X, Ahn KS, Wang LZ, Kim C, Deivasigamni A, Arfuso F, et al. Ascochlorin enhances the sensitivity of doxorubicin leading to the reversal of epithelial-to-mesenchymal transition in hepatocellular carcinoma. Mol Cancer Ther. 2016;15(12):2966–76.PubMed
200.
Zurück zum Zitat Ong MS, Deng S, Halim CE, Cai W, Tan TZ, Huang RY, et al. Cytoskeletal proteins in cancer and intracellular stress: a therapeutic perspective. Cancers. 2020;12(1):238.PubMedCentral Ong MS, Deng S, Halim CE, Cai W, Tan TZ, Huang RY, et al. Cytoskeletal proteins in cancer and intracellular stress: a therapeutic perspective. Cancers. 2020;12(1):238.PubMedCentral
201.
Zurück zum Zitat Gaballa R, Ali HEA, Mahmoud MO, Rhim JS, Ali HI, Salem HF, et al. Exosomes-mediated transfer of Itga2 promotes migration and invasion of prostate cancer cells by inducing epithelial-mesenchymal transition. Cancers. 2020;12(8):2300.PubMedCentral Gaballa R, Ali HEA, Mahmoud MO, Rhim JS, Ali HI, Salem HF, et al. Exosomes-mediated transfer of Itga2 promotes migration and invasion of prostate cancer cells by inducing epithelial-mesenchymal transition. Cancers. 2020;12(8):2300.PubMedCentral
202.
Zurück zum Zitat Liu XN, Zhang CB, Lin H, Tang XY, Zhou R, Wen HL, et al. microRNA-204 shuttled by mesenchymal stem cell-derived exosomes inhibits the migration and invasion of non-small-cell lung cancer cells via the KLF7/AKT/HIF-1α axis. Neoplasma. 2021;68(4):719–31.PubMed Liu XN, Zhang CB, Lin H, Tang XY, Zhou R, Wen HL, et al. microRNA-204 shuttled by mesenchymal stem cell-derived exosomes inhibits the migration and invasion of non-small-cell lung cancer cells via the KLF7/AKT/HIF-1α axis. Neoplasma. 2021;68(4):719–31.PubMed
203.
Zurück zum Zitat Cai J, Gong L, Li G, Guo J, Yi X, Wang Z. Exosomes in ovarian cancer ascites promote epithelial-mesenchymal transition of ovarian cancer cells by delivery of miR-6780b-5p. Cell Death Dis. 2021;12(2):210.PubMedPubMedCentral Cai J, Gong L, Li G, Guo J, Yi X, Wang Z. Exosomes in ovarian cancer ascites promote epithelial-mesenchymal transition of ovarian cancer cells by delivery of miR-6780b-5p. Cell Death Dis. 2021;12(2):210.PubMedPubMedCentral
204.
Zurück zum Zitat Shojaei S, Hashemi SM, Ghanbarian H, Sharifi K, Salehi M, Mohammadi-Yeganeh S. Delivery of miR-381-3p mimic by mesenchymal stem cell-derived exosomes inhibits triple negative breast cancer aggressiveness; an in vitro study. Stem Cell Rev Rep. 2021;17(3):1027–38.PubMed Shojaei S, Hashemi SM, Ghanbarian H, Sharifi K, Salehi M, Mohammadi-Yeganeh S. Delivery of miR-381-3p mimic by mesenchymal stem cell-derived exosomes inhibits triple negative breast cancer aggressiveness; an in vitro study. Stem Cell Rev Rep. 2021;17(3):1027–38.PubMed
205.
Zurück zum Zitat Scavo MP, Rizzi F, Depalo N, Fanizza E, Ingrosso C, Curri ML, et al. A possible role of FZD10 delivering exosomes derived from colon cancers cell lines in inducing activation of epithelial-mesenchymal transition in normal colon epithelial cell line. Int J Mol Sci. 2020;21(18):6705.PubMedCentral Scavo MP, Rizzi F, Depalo N, Fanizza E, Ingrosso C, Curri ML, et al. A possible role of FZD10 delivering exosomes derived from colon cancers cell lines in inducing activation of epithelial-mesenchymal transition in normal colon epithelial cell line. Int J Mol Sci. 2020;21(18):6705.PubMedCentral
206.
Zurück zum Zitat Zhou J, Wang H, Sun Q, Liu X, Wu Z, Wang X, et al. miR-224-5p-enriched exosomes promote tumorigenesis by directly targeting androgen receptor in non-small cell lung cancer. Mol Ther Nucleic Acids. 2021;23:1217–28.PubMedPubMedCentral Zhou J, Wang H, Sun Q, Liu X, Wu Z, Wang X, et al. miR-224-5p-enriched exosomes promote tumorigenesis by directly targeting androgen receptor in non-small cell lung cancer. Mol Ther Nucleic Acids. 2021;23:1217–28.PubMedPubMedCentral
207.
Zurück zum Zitat Monisha J, Roy NK, Padmavathi G, Banik K, Bordoloi D, Khwairakpam AD, et al. NGAL is downregulated in oral squamous cell carcinoma and leads to increased survival, proliferation, migration and chemoresistance. Cancers. 2018;10(7):228.PubMedCentral Monisha J, Roy NK, Padmavathi G, Banik K, Bordoloi D, Khwairakpam AD, et al. NGAL is downregulated in oral squamous cell carcinoma and leads to increased survival, proliferation, migration and chemoresistance. Cancers. 2018;10(7):228.PubMedCentral
208.
Zurück zum Zitat Ko JH, Um JY, Lee SG, Yang WM, Sethi G, Ahn KS. Conditioned media from adipocytes promote proliferation, migration, and invasion in melanoma and colorectal cancer cells. J Cell Physiol. 2019;234(10):18249–61.PubMed Ko JH, Um JY, Lee SG, Yang WM, Sethi G, Ahn KS. Conditioned media from adipocytes promote proliferation, migration, and invasion in melanoma and colorectal cancer cells. J Cell Physiol. 2019;234(10):18249–61.PubMed
209.
Zurück zum Zitat Kothapalli R, Sivaraman Siveen K, Tan TZ, Thiery JP, Kumar AP, Sethi G, et al. Functional characterization of selective exosite-binding inhibitors of matrix metalloproteinase-13 (MMP-13) - experimental validation in human breast and colon cancer. Biosci Biotechnol Biochem. 2016;80(11):2122–31.PubMed Kothapalli R, Sivaraman Siveen K, Tan TZ, Thiery JP, Kumar AP, Sethi G, et al. Functional characterization of selective exosite-binding inhibitors of matrix metalloproteinase-13 (MMP-13) - experimental validation in human breast and colon cancer. Biosci Biotechnol Biochem. 2016;80(11):2122–31.PubMed
210.
Zurück zum Zitat Lee H, Baek SH, Lee JH, Kim C, Ko JH, Lee SG, et al. Isorhynchophylline, a potent plant alkaloid, induces apoptotic and anti-metastatic effects in human hepatocellular carcinoma cells through the modulation of diverse cell signaling cascades. Int J Mol Sci. 2017;18(5):1095.PubMedCentral Lee H, Baek SH, Lee JH, Kim C, Ko JH, Lee SG, et al. Isorhynchophylline, a potent plant alkaloid, induces apoptotic and anti-metastatic effects in human hepatocellular carcinoma cells through the modulation of diverse cell signaling cascades. Int J Mol Sci. 2017;18(5):1095.PubMedCentral
211.
Zurück zum Zitat Jung YY, Lee JH, Nam D, Narula AS, Namjoshi OA, Blough BE, et al. Anti-myeloma effects of icariin are mediated through the attenuation of JAK/STAT3-dependent signaling cascade. Front Pharmacol. 2018;9:531.PubMedPubMedCentral Jung YY, Lee JH, Nam D, Narula AS, Namjoshi OA, Blough BE, et al. Anti-myeloma effects of icariin are mediated through the attenuation of JAK/STAT3-dependent signaling cascade. Front Pharmacol. 2018;9:531.PubMedPubMedCentral
212.
Zurück zum Zitat Sáenz-de-Santa-María I, Celada L, San José Martínez A, Cubiella T, Chiara MD. Blockage of squamous cancer cell collective invasion by FAK inhibition is released by CAFs and MMP-2. Cancers. 2020;12(12):3708.PubMedCentral Sáenz-de-Santa-María I, Celada L, San José Martínez A, Cubiella T, Chiara MD. Blockage of squamous cancer cell collective invasion by FAK inhibition is released by CAFs and MMP-2. Cancers. 2020;12(12):3708.PubMedCentral
213.
Zurück zum Zitat Szarvas T, Hoffmann MJ, Olah C, Szekely E, Kiss A, Hess J, et al. MMP-7 serum and tissue levels are associated with poor survival in platinum-treated bladder cancer patients. Diagnostics (Basel, Switzerland). 2020;11(1):48. Szarvas T, Hoffmann MJ, Olah C, Szekely E, Kiss A, Hess J, et al. MMP-7 serum and tissue levels are associated with poor survival in platinum-treated bladder cancer patients. Diagnostics (Basel, Switzerland). 2020;11(1):48.
214.
Zurück zum Zitat Frieling JS, Li T, Tauro M, Lynch CC. Prostate cancer-derived MMP-3 controls intrinsic cell growth and extrinsic angiogenesis. Neoplasia (New York, NY). 2020;22(10):511–21. Frieling JS, Li T, Tauro M, Lynch CC. Prostate cancer-derived MMP-3 controls intrinsic cell growth and extrinsic angiogenesis. Neoplasia (New York, NY). 2020;22(10):511–21.
215.
Zurück zum Zitat Che Y, Shi X, Shi Y, Jiang X, Ai Q, Shi Y, et al. Exosomes derived from miR-143-overexpressing MSCs inhibit cell migration and invasion in human prostate cancer by downregulating TFF3. Mol Ther Nucleic Acids. 2019;18:232–44.PubMedPubMedCentral Che Y, Shi X, Shi Y, Jiang X, Ai Q, Shi Y, et al. Exosomes derived from miR-143-overexpressing MSCs inhibit cell migration and invasion in human prostate cancer by downregulating TFF3. Mol Ther Nucleic Acids. 2019;18:232–44.PubMedPubMedCentral
216.
Zurück zum Zitat Chen G, Zhang Y, Wu X. 786–0 renal cancer cell line-derived exosomes promote 786–0 cell migration and invasion in vitro. Oncol Lett. 2014;7(5):1576–80.PubMedPubMedCentral Chen G, Zhang Y, Wu X. 786–0 renal cancer cell line-derived exosomes promote 786–0 cell migration and invasion in vitro. Oncol Lett. 2014;7(5):1576–80.PubMedPubMedCentral
217.
Zurück zum Zitat Wu F, Li F, Lin X, Xu F, Cui RR, Zhong JY, et al. Exosomes increased angiogenesis in papillary thyroid cancer microenvironment. Endocr Relat Cancer. 2019;26(5):525–38.PubMed Wu F, Li F, Lin X, Xu F, Cui RR, Zhong JY, et al. Exosomes increased angiogenesis in papillary thyroid cancer microenvironment. Endocr Relat Cancer. 2019;26(5):525–38.PubMed
218.
Zurück zum Zitat Mao Y, Wang Y, Dong L, Zhang Y, Zhang Y, Wang C, et al. Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells. J Exp Clin Cancer Res CR. 2019;38(1):389.PubMed Mao Y, Wang Y, Dong L, Zhang Y, Zhang Y, Wang C, et al. Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells. J Exp Clin Cancer Res CR. 2019;38(1):389.PubMed
219.
Zurück zum Zitat Ludwig N, Yerneni SS, Razzo BM, Whiteside TL. Exosomes from HNSCC promote angiogenesis through reprogramming of endothelial cells. Mol Cancer Res MCR. 2018;16(11):1798–808.PubMed Ludwig N, Yerneni SS, Razzo BM, Whiteside TL. Exosomes from HNSCC promote angiogenesis through reprogramming of endothelial cells. Mol Cancer Res MCR. 2018;16(11):1798–808.PubMed
220.
Zurück zum Zitat Zhou Z, Zhang H, Deng T, Ning T, Liu R, Liu D, et al. Exosomes carrying MicroRNA-155 target forkhead box O3 of endothelial cells and promote angiogenesis in gastric cancer. Mol Ther Oncolytics. 2019;15:223–33.PubMedPubMedCentral Zhou Z, Zhang H, Deng T, Ning T, Liu R, Liu D, et al. Exosomes carrying MicroRNA-155 target forkhead box O3 of endothelial cells and promote angiogenesis in gastric cancer. Mol Ther Oncolytics. 2019;15:223–33.PubMedPubMedCentral
221.
Zurück zum Zitat Xue X, Huang J, Yu K, Chen X, He Y, Qi D, et al. YB-1 transferred by gastric cancer exosomes promotes angiogenesis via enhancing the expression of angiogenic factors in vascular endothelial cells. BMC Cancer. 2020;20(1):996.PubMedPubMedCentral Xue X, Huang J, Yu K, Chen X, He Y, Qi D, et al. YB-1 transferred by gastric cancer exosomes promotes angiogenesis via enhancing the expression of angiogenic factors in vascular endothelial cells. BMC Cancer. 2020;20(1):996.PubMedPubMedCentral
222.
Zurück zum Zitat Chen Z, Xie Y, Chen W, Li T, Chen X, Liu B. microRNA-6785-5p-loaded human umbilical cord mesenchymal stem cells-derived exosomes suppress angiogenesis and metastasis in gastric cancer via INHBA. Life Sci. 2021;284:119222.PubMed Chen Z, Xie Y, Chen W, Li T, Chen X, Liu B. microRNA-6785-5p-loaded human umbilical cord mesenchymal stem cells-derived exosomes suppress angiogenesis and metastasis in gastric cancer via INHBA. Life Sci. 2021;284:119222.PubMed
223.
Zurück zum Zitat Bai M, Li J, Yang H, Zhang H, Zhou Z, Deng T, et al. miR-135b delivered by gastric tumor exosomes inhibits FOXO1 expression in endothelial cells and promotes angiogenesis. Mol Ther J Am Soc Gene Ther. 2019;27(10):1772–83. Bai M, Li J, Yang H, Zhang H, Zhou Z, Deng T, et al. miR-135b delivered by gastric tumor exosomes inhibits FOXO1 expression in endothelial cells and promotes angiogenesis. Mol Ther J Am Soc Gene Ther. 2019;27(10):1772–83.
224.
Zurück zum Zitat Song Y, Wang M, Tong H, Tan Y, Hu X, Wang K, et al. Plasma exosomes from endometrial cancer patients contain LGALS3BP to promote endometrial cancer progression. Oncogene. 2021;40(3):633–46.PubMed Song Y, Wang M, Tong H, Tan Y, Hu X, Wang K, et al. Plasma exosomes from endometrial cancer patients contain LGALS3BP to promote endometrial cancer progression. Oncogene. 2021;40(3):633–46.PubMed
225.
Zurück zum Zitat Pakravan K, Babashah S, Sadeghizadeh M, Mowla SJ, Mossahebi-Mohammadi M, Ataei F, et al. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol (Dordr). 2017;40(5):457–70. Pakravan K, Babashah S, Sadeghizadeh M, Mowla SJ, Mossahebi-Mohammadi M, Ataei F, et al. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol (Dordr). 2017;40(5):457–70.
226.
Zurück zum Zitat Lee JK, Park SR, Jung BK, Jeon YK, Lee YS, Kim MK, et al. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 2013;8(12):e84256.PubMedPubMedCentral Lee JK, Park SR, Jung BK, Jeon YK, Lee YS, Kim MK, et al. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 2013;8(12):e84256.PubMedPubMedCentral
227.
Zurück zum Zitat Zhang X, Sheng Y, Li B, Wang Q, Liu X, Han J. Ovarian cancer derived PKR1 positive exosomes promote angiogenesis by promoting migration and tube formation in vitro. Cell Biochem Funct. 2021;39(2):308–16.PubMed Zhang X, Sheng Y, Li B, Wang Q, Liu X, Han J. Ovarian cancer derived PKR1 positive exosomes promote angiogenesis by promoting migration and tube formation in vitro. Cell Biochem Funct. 2021;39(2):308–16.PubMed
228.
Zurück zum Zitat Li Z, Yan-Qing W, Xiao Y, Shi-Yi L, Meng-Qin Y, Shu X, et al. Exosomes secreted by chemoresistant ovarian cancer cells promote angiogenesis. J Ovarian Res. 2021;14(1):7.PubMedPubMedCentral Li Z, Yan-Qing W, Xiao Y, Shi-Yi L, Meng-Qin Y, Shu X, et al. Exosomes secreted by chemoresistant ovarian cancer cells promote angiogenesis. J Ovarian Res. 2021;14(1):7.PubMedPubMedCentral
229.
Zurück zum Zitat Zheng X, Ma N, Wang X, Hu J, Ma X, Wang J, et al. Exosomes derived from 5-fluorouracil-resistant colon cancer cells are enriched in GDF15 and can promote angiogenesis. J Cancer. 2020;11(24):7116–26.PubMedPubMedCentral Zheng X, Ma N, Wang X, Hu J, Ma X, Wang J, et al. Exosomes derived from 5-fluorouracil-resistant colon cancer cells are enriched in GDF15 and can promote angiogenesis. J Cancer. 2020;11(24):7116–26.PubMedPubMedCentral
230.
Zurück zum Zitat Yin H, Yu S, Xie Y, Dai X, Dong M, Sheng C, et al. Cancer-associated fibroblasts-derived exosomes upregulate microRNA-135b-5p to promote colorectal cancer cell growth and angiogenesis by inhibiting thioredoxin-interacting protein. Cell Signal. 2021;84:110029.PubMed Yin H, Yu S, Xie Y, Dai X, Dong M, Sheng C, et al. Cancer-associated fibroblasts-derived exosomes upregulate microRNA-135b-5p to promote colorectal cancer cell growth and angiogenesis by inhibiting thioredoxin-interacting protein. Cell Signal. 2021;84:110029.PubMed
231.
Zurück zum Zitat Zhang L, Wu X, Luo C, Chen X, Yang L, Tao J, et al. The 786–0 renal cancer cell-derived exosomes promote angiogenesis by downregulating the expression of hepatocyte cell adhesion molecule. Mol Med Rep. 2013;8(1):272–6.PubMed Zhang L, Wu X, Luo C, Chen X, Yang L, Tao J, et al. The 786–0 renal cancer cell-derived exosomes promote angiogenesis by downregulating the expression of hepatocyte cell adhesion molecule. Mol Med Rep. 2013;8(1):272–6.PubMed
232.
Zurück zum Zitat Hou Y, Fan L, Li H. Oncogenic miR-27a delivered by exosomes binds to SFRP1 and promotes angiogenesis in renal clear cell carcinoma. Mol Ther Nucleic Acids. 2021;24:92–103.PubMed Hou Y, Fan L, Li H. Oncogenic miR-27a delivered by exosomes binds to SFRP1 and promotes angiogenesis in renal clear cell carcinoma. Mol Ther Nucleic Acids. 2021;24:92–103.PubMed
233.
Zurück zum Zitat Song H, Liu D, Dong S, Zeng L, Wu Z, Zhao P, et al. Epitranscriptomics and epiproteomics in cancer drug resistance: therapeutic implications. Signal Transduct Target Ther. 2020;5(1):193.PubMedPubMedCentral Song H, Liu D, Dong S, Zeng L, Wu Z, Zhao P, et al. Epitranscriptomics and epiproteomics in cancer drug resistance: therapeutic implications. Signal Transduct Target Ther. 2020;5(1):193.PubMedPubMedCentral
234.
Zurück zum Zitat Asano T. Drug resistance in cancer therapy and the role of epigenetics. J Nippon Med Scl. 2020;87(5):244–51. Asano T. Drug resistance in cancer therapy and the role of epigenetics. J Nippon Med Scl. 2020;87(5):244–51.
235.
Zurück zum Zitat Haider T, Pandey V, Banjare N, Gupta PN, Soni V. Drug resistance in cancer: mechanisms and tackling strategies. Pharmacol Rep. 2020;72(5):1125–51.PubMed Haider T, Pandey V, Banjare N, Gupta PN, Soni V. Drug resistance in cancer: mechanisms and tackling strategies. Pharmacol Rep. 2020;72(5):1125–51.PubMed
236.
Zurück zum Zitat Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714–26.PubMed Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714–26.PubMed
237.
Zurück zum Zitat Kansara S, Pandey V, Lobie PE, Sethi G, Garg M, Pandey AK. Mechanistic involvement of long non-coding RNAs in oncotherapeutics resistance in triple-negative breast cancer. Cells. 2020;9(6):1511.PubMedCentral Kansara S, Pandey V, Lobie PE, Sethi G, Garg M, Pandey AK. Mechanistic involvement of long non-coding RNAs in oncotherapeutics resistance in triple-negative breast cancer. Cells. 2020;9(6):1511.PubMedCentral
238.
Zurück zum Zitat Li F, Sethi G. Targeting transcription factor NF-kappaB to overcome chemoresistance and radioresistance in cancer therapy. Biochem Biophys Acta. 2010;1805(2):167–80.PubMed Li F, Sethi G. Targeting transcription factor NF-kappaB to overcome chemoresistance and radioresistance in cancer therapy. Biochem Biophys Acta. 2010;1805(2):167–80.PubMed
239.
Zurück zum Zitat Aggarwal V, Tuli HS, Thakral F, Singhal P, Aggarwal D, Srivastava S, et al. Molecular mechanisms of action of hesperidin in cancer: recent trends and advancements. Exp Biol Med (Maywood). 2020;245(5):486–97. Aggarwal V, Tuli HS, Thakral F, Singhal P, Aggarwal D, Srivastava S, et al. Molecular mechanisms of action of hesperidin in cancer: recent trends and advancements. Exp Biol Med (Maywood). 2020;245(5):486–97.
240.
Zurück zum Zitat Hon KW, Ab-Mutalib NS, Abdullah NMA, Jamal R, Abu N. Extracellular vesicle-derived circular RNAs confers chemoresistance in colorectal cancer. Sci Rep. 2019;9(1):16497.PubMedPubMedCentral Hon KW, Ab-Mutalib NS, Abdullah NMA, Jamal R, Abu N. Extracellular vesicle-derived circular RNAs confers chemoresistance in colorectal cancer. Sci Rep. 2019;9(1):16497.PubMedPubMedCentral
241.
Zurück zum Zitat Liang G, Zhu Y, Ali DJ, Tian T, Xu H, Si K, et al. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnol. 2020;18(1):10. Liang G, Zhu Y, Ali DJ, Tian T, Xu H, Si K, et al. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnol. 2020;18(1):10.
242.
Zurück zum Zitat Zhu X, Shen H, Yin X, Yang M, Wei H, Chen Q, et al. Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J Exp Clin Cancer Res. 2019;38(1):81.PubMedPubMedCentral Zhu X, Shen H, Yin X, Yang M, Wei H, Chen Q, et al. Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J Exp Clin Cancer Res. 2019;38(1):81.PubMedPubMedCentral
243.
Zurück zum Zitat Binenbaum Y, Fridman E, Yaari Z, Milman N, Schroeder A, Ben David G, et al. Transfer of miRNA in macrophage-derived exosomes induces drug resistance in pancreatic adenocarcinoma. Can Res. 2018;78(18):5287–99. Binenbaum Y, Fridman E, Yaari Z, Milman N, Schroeder A, Ben David G, et al. Transfer of miRNA in macrophage-derived exosomes induces drug resistance in pancreatic adenocarcinoma. Can Res. 2018;78(18):5287–99.
244.
Zurück zum Zitat Yang Z, Zhao N, Cui J, Wu H, Xiong J, Peng T. Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cell Oncol (Dordr). 2020;43(1):123–36. Yang Z, Zhao N, Cui J, Wu H, Xiong J, Peng T. Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cell Oncol (Dordr). 2020;43(1):123–36.
245.
Zurück zum Zitat Dorayappan KDP, Wanner R, Wallbillich JJ, Saini U, Zingarelli R, Suarez AA, et al. Hypoxia-induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: a novel mechanism linking STAT3/Rab proteins. Oncogene. 2018;37(28):3806–21.PubMedPubMedCentral Dorayappan KDP, Wanner R, Wallbillich JJ, Saini U, Zingarelli R, Suarez AA, et al. Hypoxia-induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: a novel mechanism linking STAT3/Rab proteins. Oncogene. 2018;37(28):3806–21.PubMedPubMedCentral
246.
Zurück zum Zitat Qin X, Yu S, Zhou L, Shi M, Hu Y, Xu X, et al. Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100-5p-dependent manner. Int J Nanomed. 2017;12:3721–33. Qin X, Yu S, Zhou L, Shi M, Hu Y, Xu X, et al. Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100-5p-dependent manner. Int J Nanomed. 2017;12:3721–33.
247.
Zurück zum Zitat Huang L, Hu C, Chao H, Zhang Y, Li Y, Hou J, et al. Drug-resistant endothelial cells facilitate progression, EMT and chemoresistance in nasopharyngeal carcinoma via exosomes. Cell Signal. 2019;63:109385.PubMed Huang L, Hu C, Chao H, Zhang Y, Li Y, Hou J, et al. Drug-resistant endothelial cells facilitate progression, EMT and chemoresistance in nasopharyngeal carcinoma via exosomes. Cell Signal. 2019;63:109385.PubMed
248.
Zurück zum Zitat Wang X, Zhang H, Bai M, Ning T, Ge S, Deng T, et al. Exosomes serve as nanoparticles to deliver anti-miR-214 to reverse chemoresistance to cisplatin in gastric cancer. Mol Ther J Am Soc Gene Ther. 2018;26(3):774–83. Wang X, Zhang H, Bai M, Ning T, Ge S, Deng T, et al. Exosomes serve as nanoparticles to deliver anti-miR-214 to reverse chemoresistance to cisplatin in gastric cancer. Mol Ther J Am Soc Gene Ther. 2018;26(3):774–83.
249.
Zurück zum Zitat Zhang X, Yang Y, Yang Y, Chen H, Tu H, Li J. Exosomes from bone marrow microenvironment-derived mesenchymal stem cells affect CML cells growth and promote drug resistance to tyrosine kinase inhibitors. Stem Cells Int. 2020;2020:8890201.PubMedPubMedCentral Zhang X, Yang Y, Yang Y, Chen H, Tu H, Li J. Exosomes from bone marrow microenvironment-derived mesenchymal stem cells affect CML cells growth and promote drug resistance to tyrosine kinase inhibitors. Stem Cells Int. 2020;2020:8890201.PubMedPubMedCentral
250.
Zurück zum Zitat Sadegh-Nejadi S, Afrisham R, Emamgholipour S, Izadi P, Eivazi N, Tahbazlahafi B, et al. Influence of plasma circulating exosomes obtained from obese women on tumorigenesis and tamoxifen resistance in MCF-7 cells. IUBMB Life. 2020;72(9):1930–40.PubMed Sadegh-Nejadi S, Afrisham R, Emamgholipour S, Izadi P, Eivazi N, Tahbazlahafi B, et al. Influence of plasma circulating exosomes obtained from obese women on tumorigenesis and tamoxifen resistance in MCF-7 cells. IUBMB Life. 2020;72(9):1930–40.PubMed
251.
Zurück zum Zitat Liu J, Zhu S, Tang W, Huang Q, Mei Y, Yang H. Exosomes from tamoxifen-resistant breast cancer cells transmit drug resistance partly by delivering miR-9-5p. Cancer Cell Int. 2021;21(1):55.PubMedPubMedCentral Liu J, Zhu S, Tang W, Huang Q, Mei Y, Yang H. Exosomes from tamoxifen-resistant breast cancer cells transmit drug resistance partly by delivering miR-9-5p. Cancer Cell Int. 2021;21(1):55.PubMedPubMedCentral
252.
Zurück zum Zitat Deng X, Ruan H, Zhang X, Xu X, Zhu Y, Peng H, et al. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer. 2020;146(6):1700–16.PubMed Deng X, Ruan H, Zhang X, Xu X, Zhu Y, Peng H, et al. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer. 2020;146(6):1700–16.PubMed
253.
Zurück zum Zitat Qu Z, Wu J, Wu J, Luo D, Jiang C, Ding Y. Exosomes derived from HCC cells induce sorafenib resistance in hepatocellular carcinoma both in vivo and in vitro. J Exp Clin Cancer Res. 2016;35(1):159.PubMedPubMedCentral Qu Z, Wu J, Wu J, Luo D, Jiang C, Ding Y. Exosomes derived from HCC cells induce sorafenib resistance in hepatocellular carcinoma both in vivo and in vitro. J Exp Clin Cancer Res. 2016;35(1):159.PubMedPubMedCentral
254.
Zurück zum Zitat Liu X, Jiang T, Li X, Zhao C, Li J, Zhou F, et al. Exosomes transmit T790M mutation-induced resistance in EGFR-mutant NSCLC by activating PI3K/AKT signalling pathway. J Cell Mol Med. 2020;24(2):1529–40.PubMedPubMedCentral Liu X, Jiang T, Li X, Zhao C, Li J, Zhou F, et al. Exosomes transmit T790M mutation-induced resistance in EGFR-mutant NSCLC by activating PI3K/AKT signalling pathway. J Cell Mol Med. 2020;24(2):1529–40.PubMedPubMedCentral
255.
Zurück zum Zitat Shi S, Huang X, Ma X, Zhu X, Zhang Q. Research of the mechanism on miRNA193 in exosomes promotes cisplatin resistance in esophageal cancer cells. PLoS One. 2020;15(5):e0225290.PubMedPubMedCentral Shi S, Huang X, Ma X, Zhu X, Zhang Q. Research of the mechanism on miRNA193 in exosomes promotes cisplatin resistance in esophageal cancer cells. PLoS One. 2020;15(5):e0225290.PubMedPubMedCentral
256.
Zurück zum Zitat Kunou S, Shimada K, Takai M, Sakamoto A, Aoki T, Hikita T, et al. Exosomes secreted from cancer-associated fibroblasts elicit anti-pyrimidine drug resistance through modulation of its transporter in malignant lymphoma. Oncogene. 2021;40(23):3989–4003.PubMedPubMedCentral Kunou S, Shimada K, Takai M, Sakamoto A, Aoki T, Hikita T, et al. Exosomes secreted from cancer-associated fibroblasts elicit anti-pyrimidine drug resistance through modulation of its transporter in malignant lymphoma. Oncogene. 2021;40(23):3989–4003.PubMedPubMedCentral
257.
Zurück zum Zitat Zhang S, Zhang Y, Qu J, Che X, Fan Y, Hou K, et al. Exosomes promote cetuximab resistance via the PTEN/Akt pathway in colon cancer cells. Braz J Med Biol Res. 2017;51(1):e6472.PubMedPubMedCentral Zhang S, Zhang Y, Qu J, Che X, Fan Y, Hou K, et al. Exosomes promote cetuximab resistance via the PTEN/Akt pathway in colon cancer cells. Braz J Med Biol Res. 2017;51(1):e6472.PubMedPubMedCentral
258.
Zurück zum Zitat Pan Y, Lin Y, Mi C. Cisplatin-resistant osteosarcoma cell-derived exosomes confer cisplatin resistance to recipient cells in an exosomal circ_103801-dependent manner. Cell Biol Int. 2021;45(4):858–68.PubMed Pan Y, Lin Y, Mi C. Cisplatin-resistant osteosarcoma cell-derived exosomes confer cisplatin resistance to recipient cells in an exosomal circ_103801-dependent manner. Cell Biol Int. 2021;45(4):858–68.PubMed
259.
Zurück zum Zitat Chang WH, Nguyen TT, Hsu CH, Bryant KL, Kim HJ, Ying H, et al. KRAS-dependent cancer cells promote survival by producing exosomes enriched in Survivin. Cancer Lett. 2021;517:66–77.PubMed Chang WH, Nguyen TT, Hsu CH, Bryant KL, Kim HJ, Ying H, et al. KRAS-dependent cancer cells promote survival by producing exosomes enriched in Survivin. Cancer Lett. 2021;517:66–77.PubMed
260.
Zurück zum Zitat Ning K, Wang T, Sun X, Zhang P, Chen Y, Jin J, et al. UCH-L1-containing exosomes mediate chemotherapeutic resistance transfer in breast cancer. J Surg Oncol. 2017;115(8):932–40.PubMed Ning K, Wang T, Sun X, Zhang P, Chen Y, Jin J, et al. UCH-L1-containing exosomes mediate chemotherapeutic resistance transfer in breast cancer. J Surg Oncol. 2017;115(8):932–40.PubMed
261.
Zurück zum Zitat Yang SJ, Wang DD, Li J, Xu HZ, Shen HY, Chen X, et al. Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene. 2017;623:5–14.PubMed Yang SJ, Wang DD, Li J, Xu HZ, Shen HY, Chen X, et al. Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene. 2017;623:5–14.PubMed
262.
Zurück zum Zitat Wang X, Xu C, Hua Y, Sun L, Cheng K, Jia Z, et al. Exosomes play an important role in the process of psoralen reverse multidrug resistance of breast cancer. J Exp Clin Cancer Res CR. 2016;35(1):186.PubMed Wang X, Xu C, Hua Y, Sun L, Cheng K, Jia Z, et al. Exosomes play an important role in the process of psoralen reverse multidrug resistance of breast cancer. J Exp Clin Cancer Res CR. 2016;35(1):186.PubMed
263.
Zurück zum Zitat He C, Jaffar Ali D, Li Y, Zhu Y, Sun B, Xiao Z. Engineering of HN3 increases the tumor targeting specificity of exosomes and upgrade the anti-tumor effect of sorafenib on HuH-7 cells. PeerJ. 2020;8:e9524.PubMedPubMedCentral He C, Jaffar Ali D, Li Y, Zhu Y, Sun B, Xiao Z. Engineering of HN3 increases the tumor targeting specificity of exosomes and upgrade the anti-tumor effect of sorafenib on HuH-7 cells. PeerJ. 2020;8:e9524.PubMedPubMedCentral
264.
Zurück zum Zitat Wang J, Li D, Zhuang Y, Fu J, Li X, Shi Q, et al. Exosomes derived from bone marrow stromal cells decrease the sensitivity of leukemic cells to etoposide. Oncol Lett. 2017;14(3):3082–8.PubMedPubMedCentral Wang J, Li D, Zhuang Y, Fu J, Li X, Shi Q, et al. Exosomes derived from bone marrow stromal cells decrease the sensitivity of leukemic cells to etoposide. Oncol Lett. 2017;14(3):3082–8.PubMedPubMedCentral
265.
Zurück zum Zitat Liu Y, Song B, Wei Y, Chen F, Chi Y, Fan H, et al. Exosomes from mesenchymal stromal cells enhance imatinib-induced apoptosis in human leukemia cells via activation of caspase signaling pathway. Cytotherapy. 2018;20(2):181–8.PubMed Liu Y, Song B, Wei Y, Chen F, Chi Y, Fan H, et al. Exosomes from mesenchymal stromal cells enhance imatinib-induced apoptosis in human leukemia cells via activation of caspase signaling pathway. Cytotherapy. 2018;20(2):181–8.PubMed
266.
Zurück zum Zitat Dong Y, Lin Y, Gao X, Zhao Y, Wan Z, Wang H, et al. Targeted blocking of miR328 lysosomal degradation with alkalized exosomes sensitizes the chronic leukemia cells to imatinib. Appl Microbiol Biotechnol. 2019;103(23–24):9569–82.PubMed Dong Y, Lin Y, Gao X, Zhao Y, Wan Z, Wang H, et al. Targeted blocking of miR328 lysosomal degradation with alkalized exosomes sensitizes the chronic leukemia cells to imatinib. Appl Microbiol Biotechnol. 2019;103(23–24):9569–82.PubMed
267.
Zurück zum Zitat Zheng Y, Liu L, Wang Y, Xiao S, Mai R, Zhu Z, et al. Glioblastoma stem cell (GSC)-derived PD-L1-containing exosomes activates AMPK/ULK1 pathway mediated autophagy to increase temozolomide-resistance in glioblastoma. Cell Biosci. 2021;11(1):63.PubMedPubMedCentral Zheng Y, Liu L, Wang Y, Xiao S, Mai R, Zhu Z, et al. Glioblastoma stem cell (GSC)-derived PD-L1-containing exosomes activates AMPK/ULK1 pathway mediated autophagy to increase temozolomide-resistance in glioblastoma. Cell Biosci. 2021;11(1):63.PubMedPubMedCentral
268.
Zurück zum Zitat Chuang HY, Su YK, Liu HW, Chen CH, Chiu SC, Cho DY, et al. Preclinical evidence of STAT3 inhibitor pacritinib overcoming temozolomide resistance via downregulating miR-21-enriched exosomes from M2 glioblastoma-associated macrophages. J Clin Med. 2019;8(7):959.PubMedCentral Chuang HY, Su YK, Liu HW, Chen CH, Chiu SC, Cho DY, et al. Preclinical evidence of STAT3 inhibitor pacritinib overcoming temozolomide resistance via downregulating miR-21-enriched exosomes from M2 glioblastoma-associated macrophages. J Clin Med. 2019;8(7):959.PubMedCentral
269.
Zurück zum Zitat Yuwen DL, Sheng BB, Liu J, Wenyu W, Shu YQ. MiR-146a-5p level in serum exosomes predicts therapeutic effect of cisplatin in non-small cell lung cancer. Eur Rev Med Pharmacol Sci. 2017;21(11):2650–8.PubMed Yuwen DL, Sheng BB, Liu J, Wenyu W, Shu YQ. MiR-146a-5p level in serum exosomes predicts therapeutic effect of cisplatin in non-small cell lung cancer. Eur Rev Med Pharmacol Sci. 2017;21(11):2650–8.PubMed
270.
Zurück zum Zitat Sayyed AA, Gondaliya P, Mali M, Pawar A, Bhat P, Khairnar A, et al. MiR-155 inhibitor-laden exosomes reverse resistance to Cisplatin in a 3D tumor spheroid and xenograft model of oral cancer. Mol Pharm. 2021;18(8):3010–25.PubMed Sayyed AA, Gondaliya P, Mali M, Pawar A, Bhat P, Khairnar A, et al. MiR-155 inhibitor-laden exosomes reverse resistance to Cisplatin in a 3D tumor spheroid and xenograft model of oral cancer. Mol Pharm. 2021;18(8):3010–25.PubMed
271.
Zurück zum Zitat Semaan L, Zeng Q, Lu Y, Zhang Y, Zreik MM, Chamseddine MB, et al. MicroRNA-214 enriched exosomes from human cerebral endothelial cells (hCEC) sensitize hepatocellular carcinoma to anti-cancer drugs. Oncotarget. 2021;12(3):185–98.PubMedPubMedCentral Semaan L, Zeng Q, Lu Y, Zhang Y, Zreik MM, Chamseddine MB, et al. MicroRNA-214 enriched exosomes from human cerebral endothelial cells (hCEC) sensitize hepatocellular carcinoma to anti-cancer drugs. Oncotarget. 2021;12(3):185–98.PubMedPubMedCentral
272.
Zurück zum Zitat Zhang K, Shao CX, Zhu JD, Lv XL, Tu CY, Jiang C, et al. Exosomes function as nanoparticles to transfer miR-199a-3p to reverse chemoresistance to cisplatin in hepatocellular carcinoma. Bioscience reports. 2020;40(7):BSR20194026.PubMedPubMedCentral Zhang K, Shao CX, Zhu JD, Lv XL, Tu CY, Jiang C, et al. Exosomes function as nanoparticles to transfer miR-199a-3p to reverse chemoresistance to cisplatin in hepatocellular carcinoma. Bioscience reports. 2020;40(7):BSR20194026.PubMedPubMedCentral
273.
Zurück zum Zitat Kato T, Mizutani K, Kawakami K, Fujita Y, Ehara H, Ito M. CD44v8-10 mRNA contained in serum exosomes as a diagnostic marker for docetaxel resistance in prostate cancer patients. Heliyon. 2020;6(7):e04138.PubMedPubMedCentral Kato T, Mizutani K, Kawakami K, Fujita Y, Ehara H, Ito M. CD44v8-10 mRNA contained in serum exosomes as a diagnostic marker for docetaxel resistance in prostate cancer patients. Heliyon. 2020;6(7):e04138.PubMedPubMedCentral
274.
Zurück zum Zitat Kabakov AE, Yakimova AO. Hypoxia-induced cancer cell responses driving radioresistance of hypoxic tumors: approaches to targeting and radiosensitizing. Cancers. 2021;13(5):1102.PubMedPubMedCentral Kabakov AE, Yakimova AO. Hypoxia-induced cancer cell responses driving radioresistance of hypoxic tumors: approaches to targeting and radiosensitizing. Cancers. 2021;13(5):1102.PubMedPubMedCentral
275.
Zurück zum Zitat Graham K, Unger E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. Int J Nanomed. 2018;13:6049–58. Graham K, Unger E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. Int J Nanomed. 2018;13:6049–58.
276.
Zurück zum Zitat Forster JC, Marcu LG, Bezak E. Approaches to combat hypoxia in cancer therapy and the potential for in silico models in their evaluation. Phys Med PM Int J Devot Appl Phys Med Biol Off J Ital Assoc Biomed Phys (AIFB). 2019;64:145–56. Forster JC, Marcu LG, Bezak E. Approaches to combat hypoxia in cancer therapy and the potential for in silico models in their evaluation. Phys Med PM Int J Devot Appl Phys Med Biol Off J Ital Assoc Biomed Phys (AIFB). 2019;64:145–56.
277.
Zurück zum Zitat Liu L, Zhang Z, Zhou L, Hu L, Yin C, Qing D, et al. Cancer associated fibroblasts-derived exosomes contribute to radioresistance through promoting colorectal cancer stem cells phenotype. Exp Cell Res. 2020;391(2):111956.PubMed Liu L, Zhang Z, Zhou L, Hu L, Yin C, Qing D, et al. Cancer associated fibroblasts-derived exosomes contribute to radioresistance through promoting colorectal cancer stem cells phenotype. Exp Cell Res. 2020;391(2):111956.PubMed
278.
Zurück zum Zitat Wan FZ, Chen KH, Sun YC, Chen XC, Liang RB, Chen L, et al. Exosomes overexpressing miR-34c inhibit malignant behavior and reverse the radioresistance of nasopharyngeal carcinoma. J Transl Med. 2020;18(1):12.PubMedPubMedCentral Wan FZ, Chen KH, Sun YC, Chen XC, Liang RB, Chen L, et al. Exosomes overexpressing miR-34c inhibit malignant behavior and reverse the radioresistance of nasopharyngeal carcinoma. J Transl Med. 2020;18(1):12.PubMedPubMedCentral
279.
Zurück zum Zitat Tang Z, He J, Zou J, Yu S, Sun X, Qin L. Cisplatin-resistant HepG2 cell-derived exosomes transfer cisplatin resistance to cisplatin-sensitive cells in HCC. PeerJ. 2021;9:e11200.PubMedPubMedCentral Tang Z, He J, Zou J, Yu S, Sun X, Qin L. Cisplatin-resistant HepG2 cell-derived exosomes transfer cisplatin resistance to cisplatin-sensitive cells in HCC. PeerJ. 2021;9:e11200.PubMedPubMedCentral
280.
Zurück zum Zitat Min QH, Wang XZ, Zhang J, Chen QG, Li SQ, Liu XQ, et al. Exosomes derived from imatinib-resistant chronic myeloid leukemia cells mediate a horizontal transfer of drug-resistant trait by delivering miR-365. Exp Cell Res. 2018;362(2):386–93.PubMed Min QH, Wang XZ, Zhang J, Chen QG, Li SQ, Liu XQ, et al. Exosomes derived from imatinib-resistant chronic myeloid leukemia cells mediate a horizontal transfer of drug-resistant trait by delivering miR-365. Exp Cell Res. 2018;362(2):386–93.PubMed
281.
Zurück zum Zitat Li G, Lin H, Tian R, Zhao P, Huang Y, Pang X, et al. VEGFR-2 inhibitor apatinib hinders endothelial cells progression triggered by irradiated gastric cancer cells-derived exosomes. J Cancer. 2018;9(21):4049–57.PubMedPubMedCentral Li G, Lin H, Tian R, Zhao P, Huang Y, Pang X, et al. VEGFR-2 inhibitor apatinib hinders endothelial cells progression triggered by irradiated gastric cancer cells-derived exosomes. J Cancer. 2018;9(21):4049–57.PubMedPubMedCentral
282.
Zurück zum Zitat Ni C, Fang QQ, Chen WZ, Jiang JX, Jiang Z, Ye J, et al. Breast cancer-derived exosomes transmit lncRNA SNHG16 to induce CD73+γδ1 Treg cells. Signal Transduct Target Ther. 2020;5(1):41.PubMedPubMedCentral Ni C, Fang QQ, Chen WZ, Jiang JX, Jiang Z, Ye J, et al. Breast cancer-derived exosomes transmit lncRNA SNHG16 to induce CD73+γδ1 Treg cells. Signal Transduct Target Ther. 2020;5(1):41.PubMedPubMedCentral
283.
Zurück zum Zitat Ashrafizadeh M, Zarrabi A, Hushmandi K, Zarrin V, Moghadam ER, Zabolian A, et al. PD-1/PD-L1 axis regulation in cancer therapy: the role of long non-coding RNAs and microRNAs. Life Sci. 2020;256:117899.PubMed Ashrafizadeh M, Zarrabi A, Hushmandi K, Zarrin V, Moghadam ER, Zabolian A, et al. PD-1/PD-L1 axis regulation in cancer therapy: the role of long non-coding RNAs and microRNAs. Life Sci. 2020;256:117899.PubMed
284.
Zurück zum Zitat Dou D, Ren X, Han M, Xu X, Ge X, Gu Y, et al. Cancer-associated fibroblasts-derived exosomes suppress immune cell function in breast cancer via the miR-92/PD-L1 pathway. Front Immunol. 2020;11:2026.PubMedPubMedCentral Dou D, Ren X, Han M, Xu X, Ge X, Gu Y, et al. Cancer-associated fibroblasts-derived exosomes suppress immune cell function in breast cancer via the miR-92/PD-L1 pathway. Front Immunol. 2020;11:2026.PubMedPubMedCentral
285.
Zurück zum Zitat Mirzaei S, Zarrabi A, Hashemi F, Zabolian A, Saleki H, Ranjbar A, et al. Regulation of nuclear factor-kappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: inhibiting or promoting carcinogenesis? Cancer Lett. 2021;509:63–80.PubMed Mirzaei S, Zarrabi A, Hashemi F, Zabolian A, Saleki H, Ranjbar A, et al. Regulation of nuclear factor-kappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: inhibiting or promoting carcinogenesis? Cancer Lett. 2021;509:63–80.PubMed
286.
Zurück zum Zitat Neurath MF. IL-36 in chronic inflammation and cancer. Cytokine Growth Factor Rev. 2020;55:70–9.PubMed Neurath MF. IL-36 in chronic inflammation and cancer. Cytokine Growth Factor Rev. 2020;55:70–9.PubMed
287.
Zurück zum Zitat Hirano T. IL-6 in inflammation, autoimmunity and cancer. Int Immunol. 2021;33(3):127–48.PubMed Hirano T. IL-6 in inflammation, autoimmunity and cancer. Int Immunol. 2021;33(3):127–48.PubMed
288.
Zurück zum Zitat Fishbein A, Hammock BD, Serhan CN, Panigrahy D. Carcinogenesis: failure of resolution of inflammation? Pharmacol Ther. 2021;218:107670.PubMed Fishbein A, Hammock BD, Serhan CN, Panigrahy D. Carcinogenesis: failure of resolution of inflammation? Pharmacol Ther. 2021;218:107670.PubMed
289.
Zurück zum Zitat Guo J, Duan Z, Zhang C, Wang W, He H, Liu Y, et al. Mouse 4T1 breast cancer cell-derived exosomes induce proinflammatory cytokine production in macrophages via miR-183. J Immunol (Baltimore, Md). 2020;205(10):2916–25. Guo J, Duan Z, Zhang C, Wang W, He H, Liu Y, et al. Mouse 4T1 breast cancer cell-derived exosomes induce proinflammatory cytokine production in macrophages via miR-183. J Immunol (Baltimore, Md). 2020;205(10):2916–25.
290.
Zurück zum Zitat Chatterjee S, Chatterjee A, Jana S, Dey S, Roy H, Das MK, et al. Transforming growth factor beta orchestrates PD-L1 enrichment in tumor-derived exosomes and mediates CD8 T-cell dysfunction regulating early phosphorylation of TCR signalome in breast cancer. Carcinogenesis. 2021;42(1):38–47.PubMed Chatterjee S, Chatterjee A, Jana S, Dey S, Roy H, Das MK, et al. Transforming growth factor beta orchestrates PD-L1 enrichment in tumor-derived exosomes and mediates CD8 T-cell dysfunction regulating early phosphorylation of TCR signalome in breast cancer. Carcinogenesis. 2021;42(1):38–47.PubMed
291.
Zurück zum Zitat Shao Q, Deng L, Liu H, Liu Z, Chen J, Jiang F, et al. Involvement of MM cell-derived exosomes in T lymphocytes immune responses. Oncol Lett. 2020;20(4):31.PubMedPubMedCentral Shao Q, Deng L, Liu H, Liu Z, Chen J, Jiang F, et al. Involvement of MM cell-derived exosomes in T lymphocytes immune responses. Oncol Lett. 2020;20(4):31.PubMedPubMedCentral
292.
Zurück zum Zitat Hinata M, Kunita A, Abe H, Morishita Y, Sakuma K, Yamashita H, et al. Exosomes of epstein-barr virus-associated gastric carcinoma suppress dendritic cell maturation. Microorganisms. 2020;8(11):1776.PubMedCentral Hinata M, Kunita A, Abe H, Morishita Y, Sakuma K, Yamashita H, et al. Exosomes of epstein-barr virus-associated gastric carcinoma suppress dendritic cell maturation. Microorganisms. 2020;8(11):1776.PubMedCentral
293.
Zurück zum Zitat Burassakarn A, Srisathaporn S, Pientong C, Wongjampa W, Vatanasapt P, Patarapadungkit N, et al. Exosomes-carrying Epstein-Barr virus-encoded small RNA-1 induces indoleamine 2, 3-dioxygenase expression in tumor-infiltrating macrophages of oral squamous-cell carcinomas and suppresses T-cell activity by activating RIG-I/IL-6/TNF-α pathway. Oral Oncol. 2021;117:105279.PubMed Burassakarn A, Srisathaporn S, Pientong C, Wongjampa W, Vatanasapt P, Patarapadungkit N, et al. Exosomes-carrying Epstein-Barr virus-encoded small RNA-1 induces indoleamine 2, 3-dioxygenase expression in tumor-infiltrating macrophages of oral squamous-cell carcinomas and suppresses T-cell activity by activating RIG-I/IL-6/TNF-α pathway. Oral Oncol. 2021;117:105279.PubMed
294.
Zurück zum Zitat Pakravan N, Abbasi A, Hassan ZM. Immunotherapy using oxygenated water and tumor-derived exosomes potentiates antitumor immune response and attenuates malignancy tendency in mice model of breast cancer. Oxid Med Cell Longev. 2021;2021:5529484.PubMedPubMedCentral Pakravan N, Abbasi A, Hassan ZM. Immunotherapy using oxygenated water and tumor-derived exosomes potentiates antitumor immune response and attenuates malignancy tendency in mice model of breast cancer. Oxid Med Cell Longev. 2021;2021:5529484.PubMedPubMedCentral
295.
Zurück zum Zitat Othman N, Jamal R, Abu N. Cancer-derived exosomes as effectors of key inflammation-related players. Front Immunol. 2019;10:2103.PubMedPubMedCentral Othman N, Jamal R, Abu N. Cancer-derived exosomes as effectors of key inflammation-related players. Front Immunol. 2019;10:2103.PubMedPubMedCentral
296.
Zurück zum Zitat Rishabh K, Khadilkar S, Kumar A, Kalra I, Kumar AP, Kunnumakkara AB. MicroRNAs as modulators of oral tumorigenesis-a focused review. Int J Mol Sci. 2021;22(5):2561.PubMedPubMedCentral Rishabh K, Khadilkar S, Kumar A, Kalra I, Kumar AP, Kunnumakkara AB. MicroRNAs as modulators of oral tumorigenesis-a focused review. Int J Mol Sci. 2021;22(5):2561.PubMedPubMedCentral
297.
Zurück zum Zitat Tan SC, Lim PY, Fang J, Mokhtar MFM, Hanif EAM, Jamal R. Association between MIR499A rs3746444 polymorphism and breast cancer susceptibility: a meta-analysis. Sci Rep. 2020;10(1):3508.PubMedPubMedCentral Tan SC, Lim PY, Fang J, Mokhtar MFM, Hanif EAM, Jamal R. Association between MIR499A rs3746444 polymorphism and breast cancer susceptibility: a meta-analysis. Sci Rep. 2020;10(1):3508.PubMedPubMedCentral
298.
Zurück zum Zitat Zhao M, Chang J, Liu R, Liu Y, Qi J, Wang Y, et al. miR-495 and miR-5688 are down-regulated in non-small cell lung cancer under hypoxia to maintain interleukin-11 expression. Cancer Commun (London, England). 2020;40(9):435–52. Zhao M, Chang J, Liu R, Liu Y, Qi J, Wang Y, et al. miR-495 and miR-5688 are down-regulated in non-small cell lung cancer under hypoxia to maintain interleukin-11 expression. Cancer Commun (London, England). 2020;40(9):435–52.
299.
Zurück zum Zitat Seol HS, Akiyama Y, Lee SE, Shimada S, Jang SJ. Loss of miR-100 and miR-125b results in cancer stem cell properties through IGF2 upregulation in hepatocellular carcinoma. Sci Rep. 2020;10(1):21412.PubMedPubMedCentral Seol HS, Akiyama Y, Lee SE, Shimada S, Jang SJ. Loss of miR-100 and miR-125b results in cancer stem cell properties through IGF2 upregulation in hepatocellular carcinoma. Sci Rep. 2020;10(1):21412.PubMedPubMedCentral
302.
Zurück zum Zitat Guo S, Chen J, Chen F, Zeng Q, Liu WL, Zhang G. Exosomes derived from Fusobacterium nucleatum-infected colorectal cancer cells facilitate tumour metastasis by selectively carrying miR-1246/92b-3p/27a-3p and CXCL16. Gut. 2020;70(8):1507–19. Guo S, Chen J, Chen F, Zeng Q, Liu WL, Zhang G. Exosomes derived from Fusobacterium nucleatum-infected colorectal cancer cells facilitate tumour metastasis by selectively carrying miR-1246/92b-3p/27a-3p and CXCL16. Gut. 2020;70(8):1507–19.
303.
Zurück zum Zitat Wang XH, Jiang ZH, Yang HM, Zhang Y, Xu LH. Hypoxia-induced FOXO4/LDHA axis modulates gastric cancer cell glycolysis and progression. Clin Transl Med. 2021;11(1):e279.PubMedPubMedCentral Wang XH, Jiang ZH, Yang HM, Zhang Y, Xu LH. Hypoxia-induced FOXO4/LDHA axis modulates gastric cancer cell glycolysis and progression. Clin Transl Med. 2021;11(1):e279.PubMedPubMedCentral
304.
Zurück zum Zitat Zhou X, Ma W, Li X, Xu J. Glaucocalyxin a prevents hypoxia-induced epithelial-mesenchymal transition in human gastric cancer cells through the PI3K/Akt signaling pathway. J Recept Signal Transduct Res. 2020;42(2):1–8. Zhou X, Ma W, Li X, Xu J. Glaucocalyxin a prevents hypoxia-induced epithelial-mesenchymal transition in human gastric cancer cells through the PI3K/Akt signaling pathway. J Recept Signal Transduct Res. 2020;42(2):1–8.
305.
Zurück zum Zitat Xia X, Wang S, Ni B, Xing S, Cao H, Zhang Z, et al. Hypoxic gastric cancer-derived exosomes promote progression and metastasis via MiR-301a-3p/PHD3/HIF-1α positive feedback loop. Oncogene. 2020;39(39):6231–44.PubMed Xia X, Wang S, Ni B, Xing S, Cao H, Zhang Z, et al. Hypoxic gastric cancer-derived exosomes promote progression and metastasis via MiR-301a-3p/PHD3/HIF-1α positive feedback loop. Oncogene. 2020;39(39):6231–44.PubMed
306.
Zurück zum Zitat Mohan CD, Srinivasa V, Rangappa S, Mervin L, Mohan S, Paricharak S, et al. Trisubstituted-imidazoles induce apoptosis in human breast cancer cells by targeting the oncogenic PI3K/Akt/mTOR signaling pathway. PLoS One. 2016;11(4):e0153155.PubMedPubMedCentral Mohan CD, Srinivasa V, Rangappa S, Mervin L, Mohan S, Paricharak S, et al. Trisubstituted-imidazoles induce apoptosis in human breast cancer cells by targeting the oncogenic PI3K/Akt/mTOR signaling pathway. PLoS One. 2016;11(4):e0153155.PubMedPubMedCentral
307.
Zurück zum Zitat Kim C, Lee JH, Ko JH, Chinnathambi A, Alharbi SA, Shair OHM, et al. Formononetin regulates multiple oncogenic signaling cascades and enhances sensitivity to bortezomib in a multiple myeloma mouse model. Biomolecules. 2019;9(7):262.PubMedCentral Kim C, Lee JH, Ko JH, Chinnathambi A, Alharbi SA, Shair OHM, et al. Formononetin regulates multiple oncogenic signaling cascades and enhances sensitivity to bortezomib in a multiple myeloma mouse model. Biomolecules. 2019;9(7):262.PubMedCentral
308.
Zurück zum Zitat Lee JH, Chinnathambi A, Alharbi SA, Shair OHM, Sethi G, Ahn KS. Farnesol abrogates epithelial to mesenchymal transition process through regulating Akt/mTOR pathway. Pharmacol Res. 2019;150:104504.PubMed Lee JH, Chinnathambi A, Alharbi SA, Shair OHM, Sethi G, Ahn KS. Farnesol abrogates epithelial to mesenchymal transition process through regulating Akt/mTOR pathway. Pharmacol Res. 2019;150:104504.PubMed
309.
Zurück zum Zitat Abadi AJ, Zarrabi A, Gholami MH, Mirzaei S, Hashemi F, Zabolian A, et al. Small in size, but large in action: microRNAs as potential modulators of PTEN in breast and lung cancers. Biomolecules. 2021;11(2):304.PubMedPubMedCentral Abadi AJ, Zarrabi A, Gholami MH, Mirzaei S, Hashemi F, Zabolian A, et al. Small in size, but large in action: microRNAs as potential modulators of PTEN in breast and lung cancers. Biomolecules. 2021;11(2):304.PubMedPubMedCentral
310.
Zurück zum Zitat Ashrafizadeh M, Najafi M, Ang HL, Moghadam ER, Mahabady MK, Zabolian A, et al. PTEN, a barrier for proliferation and metastasis of gastric cancer cells: from molecular pathways to targeting and regulation. Biomedicines. 2020;8(8):264.PubMedCentral Ashrafizadeh M, Najafi M, Ang HL, Moghadam ER, Mahabady MK, Zabolian A, et al. PTEN, a barrier for proliferation and metastasis of gastric cancer cells: from molecular pathways to targeting and regulation. Biomedicines. 2020;8(8):264.PubMedCentral
311.
Zurück zum Zitat Ashrafizadeh M, Zarrabi A, Samarghandian S, Najafi M. PTEN: What we know of the function and regulation of this onco-suppressor factor in bladder cancer? Eur J Pharmacol. 2020;881:173226.PubMed Ashrafizadeh M, Zarrabi A, Samarghandian S, Najafi M. PTEN: What we know of the function and regulation of this onco-suppressor factor in bladder cancer? Eur J Pharmacol. 2020;881:173226.PubMed
312.
Zurück zum Zitat Wang Y, Lin C. Exosomes miR-22-3p derived from mesenchymal stem cells suppress colorectal cancer cell proliferation and invasion by regulating RAP2B and PI3K/AKT pathway. J Oncol. 2021;2021:3874478.PubMedPubMedCentral Wang Y, Lin C. Exosomes miR-22-3p derived from mesenchymal stem cells suppress colorectal cancer cell proliferation and invasion by regulating RAP2B and PI3K/AKT pathway. J Oncol. 2021;2021:3874478.PubMedPubMedCentral
313.
Zurück zum Zitat Zheng P, Luo Q, Wang W, Li J, Wang T, Wang P, et al. Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional Apolipoprotein E. Cell Death Dis. 2018;9(4):434.PubMedPubMedCentral Zheng P, Luo Q, Wang W, Li J, Wang T, Wang P, et al. Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional Apolipoprotein E. Cell Death Dis. 2018;9(4):434.PubMedPubMedCentral
314.
Zurück zum Zitat Dai G, Yao X, Zhang Y, Gu J, Geng Y, Xue F, et al. Colorectal cancer cell-derived exosomes containing miR-10b regulate fibroblast cells via the PI3K/Akt pathway. Bull Cancer. 2018;105(4):336–49.PubMed Dai G, Yao X, Zhang Y, Gu J, Geng Y, Xue F, et al. Colorectal cancer cell-derived exosomes containing miR-10b regulate fibroblast cells via the PI3K/Akt pathway. Bull Cancer. 2018;105(4):336–49.PubMed
315.
Zurück zum Zitat Nie H, Xie X, Zhang D, Zhou Y, Li B, Li F, et al. Use of lung-specific exosomes for miRNA-126 delivery in non-small cell lung cancer. Nanoscale. 2020;12(2):877–87.PubMed Nie H, Xie X, Zhang D, Zhou Y, Li B, Li F, et al. Use of lung-specific exosomes for miRNA-126 delivery in non-small cell lung cancer. Nanoscale. 2020;12(2):877–87.PubMed
316.
Zurück zum Zitat Hu X, Mu Y, Liu J, Mu X, Gao F, Chen L, et al. Exosomes derived from hypoxic colorectal cancer cells transfer miR-410-3p to regulate tumor progression. J Cancer. 2020;11(16):4724–35.PubMedPubMedCentral Hu X, Mu Y, Liu J, Mu X, Gao F, Chen L, et al. Exosomes derived from hypoxic colorectal cancer cells transfer miR-410-3p to regulate tumor progression. J Cancer. 2020;11(16):4724–35.PubMedPubMedCentral
317.
Zurück zum Zitat Liu MX, Liao J, Xie M, Gao ZK, Wang XH, Zhang Y, et al. miR-93-5p transferred by exosomes promotes the proliferation of esophageal cancer cells via intercellular communication by targeting PTEN. Biomed Environ Sci BES. 2018;31(3):171–85.PubMed Liu MX, Liao J, Xie M, Gao ZK, Wang XH, Zhang Y, et al. miR-93-5p transferred by exosomes promotes the proliferation of esophageal cancer cells via intercellular communication by targeting PTEN. Biomed Environ Sci BES. 2018;31(3):171–85.PubMed
319.
Zurück zum Zitat Ong MS, Cai W, Yuan Y, Leong HC, Tan TZ, Mohammad A, et al. ’Lnc’-ing Wnt in female reproductive cancers: therapeutic potential of long non-coding RNAs in Wnt signalling. Br J Pharmacol. 2017;174(24):4684–700.PubMedPubMedCentral Ong MS, Cai W, Yuan Y, Leong HC, Tan TZ, Mohammad A, et al. ’Lnc’-ing Wnt in female reproductive cancers: therapeutic potential of long non-coding RNAs in Wnt signalling. Br J Pharmacol. 2017;174(24):4684–700.PubMedPubMedCentral
320.
Zurück zum Zitat Bhuvanalakshmi G, Gamit N, Patil M, Arfuso F, Sethi G, Dharmarajan A, et al. Stemness, pluripotentiality, and wnt antagonism: sFRP4, a wnt antagonist mediates pluripotency and stemness in glioblastoma. Cancers. 2018;11(1):25.PubMedCentral Bhuvanalakshmi G, Gamit N, Patil M, Arfuso F, Sethi G, Dharmarajan A, et al. Stemness, pluripotentiality, and wnt antagonism: sFRP4, a wnt antagonist mediates pluripotency and stemness in glioblastoma. Cancers. 2018;11(1):25.PubMedCentral
321.
Zurück zum Zitat Wong CC, Xu J, Bian X, Wu JL, Kang W, Qian Y, et al. In colorectal cancer cells with mutant KRAS, SLC25A22-mediated glutaminolysis reduces DNA demethylation to increase WNT signaling, stemness, and drug resistance. Gastroenterology. 2020;159(6):2163-80.e6.PubMed Wong CC, Xu J, Bian X, Wu JL, Kang W, Qian Y, et al. In colorectal cancer cells with mutant KRAS, SLC25A22-mediated glutaminolysis reduces DNA demethylation to increase WNT signaling, stemness, and drug resistance. Gastroenterology. 2020;159(6):2163-80.e6.PubMed
322.
Zurück zum Zitat Menck K, Heinrichs S, Baden C, Bleckmann A. The WNT/ROR pathway in cancer: from signaling to therapeutic intervention. Cells. 2021;10(1):142.PubMedPubMedCentral Menck K, Heinrichs S, Baden C, Bleckmann A. The WNT/ROR pathway in cancer: from signaling to therapeutic intervention. Cells. 2021;10(1):142.PubMedPubMedCentral
323.
Zurück zum Zitat Wei Y, Wei L, Li J, Ma Z, Zhang Q, Han Z, et al. SLCO4A1-AS1 promotes cell growth and induces resistance in lung adenocarcinoma by modulating miR-4701-5p/NFE2L1 axis to activate WNT pathway. Cancer Med. 2020;9(19):7205–17.PubMedPubMedCentral Wei Y, Wei L, Li J, Ma Z, Zhang Q, Han Z, et al. SLCO4A1-AS1 promotes cell growth and induces resistance in lung adenocarcinoma by modulating miR-4701-5p/NFE2L1 axis to activate WNT pathway. Cancer Med. 2020;9(19):7205–17.PubMedPubMedCentral
324.
Zurück zum Zitat Castagnoli L, Tagliabue E, Pupa SM. Inhibition of the wnt signalling pathway: an avenue to control breast cancer aggressiveness. Int J Mol Sci. 2020;21(23):9069.PubMedCentral Castagnoli L, Tagliabue E, Pupa SM. Inhibition of the wnt signalling pathway: an avenue to control breast cancer aggressiveness. Int J Mol Sci. 2020;21(23):9069.PubMedCentral
325.
Zurück zum Zitat Jiang T, Gao W, Lin S, Chen H, Du B, Liu Q, et al. FNDC1 promotes the invasiveness of gastric cancer via Wnt/β-catenin signaling pathway and correlates with peritoneal metastasis and prognosis. Front Oncol. 2020;10:590492.PubMedPubMedCentral Jiang T, Gao W, Lin S, Chen H, Du B, Liu Q, et al. FNDC1 promotes the invasiveness of gastric cancer via Wnt/β-catenin signaling pathway and correlates with peritoneal metastasis and prognosis. Front Oncol. 2020;10:590492.PubMedPubMedCentral
326.
Zurück zum Zitat Harada T, Yamamoto H, Kishida S, Kishida M, Awada C, Takao T, et al. Wnt5b-associated exosomes promote cancer cell migration and proliferation. Cancer Sci. 2017;108(1):42–52.PubMed Harada T, Yamamoto H, Kishida S, Kishida M, Awada C, Takao T, et al. Wnt5b-associated exosomes promote cancer cell migration and proliferation. Cancer Sci. 2017;108(1):42–52.PubMed
328.
Zurück zum Zitat Yang SS, Ma S, Dou H, Liu F, Zhang SY, Jiang C, et al. Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment. Exp Cell Res. 2020;391(2):111983.PubMed Yang SS, Ma S, Dou H, Liu F, Zhang SY, Jiang C, et al. Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment. Exp Cell Res. 2020;391(2):111983.PubMed
329.
Zurück zum Zitat Alharbi M, Lai A, Guanzon D, Palma C, Zuñiga F, Perrin L, et al. Ovarian cancer-derived exosomes promote tumour metastasis in vivo: an effect modulated by the invasiveness capacity of their originating cells. Clin Sci (London, England). 2019;133(13):1401–19. Alharbi M, Lai A, Guanzon D, Palma C, Zuñiga F, Perrin L, et al. Ovarian cancer-derived exosomes promote tumour metastasis in vivo: an effect modulated by the invasiveness capacity of their originating cells. Clin Sci (London, England). 2019;133(13):1401–19.
330.
Zurück zum Zitat Yuan X, Qian N, Ling S, Li Y, Sun W, Li J, et al. Breast cancer exosomes contribute to pre-metastatic niche formation and promote bone metastasis of tumor cells. Theranostics. 2021;11(3):1429–45.PubMedPubMedCentral Yuan X, Qian N, Ling S, Li Y, Sun W, Li J, et al. Breast cancer exosomes contribute to pre-metastatic niche formation and promote bone metastasis of tumor cells. Theranostics. 2021;11(3):1429–45.PubMedPubMedCentral
331.
Zurück zum Zitat Zuo L, Tao H, Xu H, Li C, Qiao G, Guo M, et al. Exosomes-coated miR-34a displays potent antitumor activity in pancreatic cancer both in vitro and in vivo. Drug Des Dev Ther. 2020;14:3495–507. Zuo L, Tao H, Xu H, Li C, Qiao G, Guo M, et al. Exosomes-coated miR-34a displays potent antitumor activity in pancreatic cancer both in vitro and in vivo. Drug Des Dev Ther. 2020;14:3495–507.
332.
Zurück zum Zitat Yang F, Yan Y, Yang Y, Hong X, Wang M, Yang Z, et al. MiR-210 in exosomes derived from CAFs promotes non-small cell lung cancer migration and invasion through PTEN/PI3K/AKT pathway. Cell Signal. 2020;73:109675.PubMed Yang F, Yan Y, Yang Y, Hong X, Wang M, Yang Z, et al. MiR-210 in exosomes derived from CAFs promotes non-small cell lung cancer migration and invasion through PTEN/PI3K/AKT pathway. Cell Signal. 2020;73:109675.PubMed
333.
Zurück zum Zitat Yan L, Li Q, Sun K, Jiang F. MiR-4644 is upregulated in plasma exosomes of bladder cancer patients and promotes bladder cancer progression by targeting UBIAD1. Am J Transl Res. 2020;12(10):6277–89.PubMedPubMedCentral Yan L, Li Q, Sun K, Jiang F. MiR-4644 is upregulated in plasma exosomes of bladder cancer patients and promotes bladder cancer progression by targeting UBIAD1. Am J Transl Res. 2020;12(10):6277–89.PubMedPubMedCentral
334.
Zurück zum Zitat Chen QL, Xie CF, Feng KL, Cui DY, Sun SL, Zhang JC, et al. microRNAs carried by exosomes promote epithelial-mesenchymal transition and metastasis of liver cancer cells. Am J Transl Res. 2020;12(10):6811–26.PubMedPubMedCentral Chen QL, Xie CF, Feng KL, Cui DY, Sun SL, Zhang JC, et al. microRNAs carried by exosomes promote epithelial-mesenchymal transition and metastasis of liver cancer cells. Am J Transl Res. 2020;12(10):6811–26.PubMedPubMedCentral
335.
Zurück zum Zitat Moradi-Chaleshtori M, Bandehpour M, Soudi S, Mohammadi-Yeganeh S, Hashemi SM. In vitro and in vivo evaluation of anti-tumoral effect of M1 phenotype induction in macrophages by miR-130 and miR-33 containing exosomes. Cancer Immunol Immunother CII. 2021;70(5):1323–39.PubMed Moradi-Chaleshtori M, Bandehpour M, Soudi S, Mohammadi-Yeganeh S, Hashemi SM. In vitro and in vivo evaluation of anti-tumoral effect of M1 phenotype induction in macrophages by miR-130 and miR-33 containing exosomes. Cancer Immunol Immunother CII. 2021;70(5):1323–39.PubMed
336.
Zurück zum Zitat Yu L, Sui B, Fan W, Lei L, Zhou L, Yang L, et al. Exosomes derived from osteogenic tumor activate osteoclast differentiation and concurrently inhibit osteogenesis by transferring COL1A1-targeting miRNA-92a-1-5p. J Extracell Vesicles. 2021;10(3):e12056.PubMedPubMedCentral Yu L, Sui B, Fan W, Lei L, Zhou L, Yang L, et al. Exosomes derived from osteogenic tumor activate osteoclast differentiation and concurrently inhibit osteogenesis by transferring COL1A1-targeting miRNA-92a-1-5p. J Extracell Vesicles. 2021;10(3):e12056.PubMedPubMedCentral
337.
Zurück zum Zitat Fang K, Chen X, Qiu F, Xu J, Xiong H, Zhang Z. Serum-derived exosomes-mediated circular RNA ARHGAP10 modulates the progression of non-small-cell lung cancer through the miR-638/FAM83F axis. Cancer Biother Radiopharm. 2020;37:96–110.PubMed Fang K, Chen X, Qiu F, Xu J, Xiong H, Zhang Z. Serum-derived exosomes-mediated circular RNA ARHGAP10 modulates the progression of non-small-cell lung cancer through the miR-638/FAM83F axis. Cancer Biother Radiopharm. 2020;37:96–110.PubMed
338.
Zurück zum Zitat Sheng Y, Han C, Yang Y, Wang J, Gu Y, Li W, et al. Correlation between LncRNA-LINC00659 and clinical prognosis in gastric cancer and study on its biological mechanism. J Cell Mol Med. 2020;24(24):14467–80.PubMedPubMedCentral Sheng Y, Han C, Yang Y, Wang J, Gu Y, Li W, et al. Correlation between LncRNA-LINC00659 and clinical prognosis in gastric cancer and study on its biological mechanism. J Cell Mol Med. 2020;24(24):14467–80.PubMedPubMedCentral
339.
Zurück zum Zitat Fan Y, Sheng W, Meng Y, Cao Y, Li R. LncRNA PTENP1 inhibits cervical cancer progression by suppressing miR-106b. Artif Cells Nanomed Biotechnol. 2020;48(1):393–407.PubMed Fan Y, Sheng W, Meng Y, Cao Y, Li R. LncRNA PTENP1 inhibits cervical cancer progression by suppressing miR-106b. Artif Cells Nanomed Biotechnol. 2020;48(1):393–407.PubMed
340.
Zurück zum Zitat Cui Y, Pu R, Ye J, Huang H, Liao D, Yang Y, et al. LncRNA FAM230B promotes gastric cancer growth and metastasis by regulating the miR-27a-5p/TOP2A axis. Dig Dis Sci. 2021;66(8):2637–50.PubMed Cui Y, Pu R, Ye J, Huang H, Liao D, Yang Y, et al. LncRNA FAM230B promotes gastric cancer growth and metastasis by regulating the miR-27a-5p/TOP2A axis. Dig Dis Sci. 2021;66(8):2637–50.PubMed
341.
Zurück zum Zitat Xu L, Huan L, Guo T, Wu Y, Liu Y, Wang Q, et al. LncRNA SNHG11 facilitates tumor metastasis by interacting with and stabilizing HIF-1α. Oncogene. 2020;39(46):7005–18.PubMedPubMedCentral Xu L, Huan L, Guo T, Wu Y, Liu Y, Wang Q, et al. LncRNA SNHG11 facilitates tumor metastasis by interacting with and stabilizing HIF-1α. Oncogene. 2020;39(46):7005–18.PubMedPubMedCentral
342.
Zurück zum Zitat Ma Z, Wang YY, Xin HW, Wang L, Arfuso F, Dharmarajan A, et al. The expanding roles of long non-coding RNAs in the regulation of cancer stem cells. Int J Biochem Cell Biol. 2019;108:17–20.PubMed Ma Z, Wang YY, Xin HW, Wang L, Arfuso F, Dharmarajan A, et al. The expanding roles of long non-coding RNAs in the regulation of cancer stem cells. Int J Biochem Cell Biol. 2019;108:17–20.PubMed
343.
Zurück zum Zitat Najafi S, Tan SC, Raee P, Rahmati Y, Asemani Y, Lee EHC, et al. Gene regulation by antisense transcription: a focus on neurological and cancer diseases. Biomed Pharmacother. 2022;145:112265.PubMed Najafi S, Tan SC, Raee P, Rahmati Y, Asemani Y, Lee EHC, et al. Gene regulation by antisense transcription: a focus on neurological and cancer diseases. Biomed Pharmacother. 2022;145:112265.PubMed
345.
Zurück zum Zitat Pandya G, Kirtonia A, Sethi G, Pandey AK, Garg M. The implication of long non-coding RNAs in the diagnosis, pathogenesis and drug resistance of pancreatic ductal adenocarcinoma and their possible therapeutic potential. Biochim Biophys Acta. 2020;1874(2):188423. Pandya G, Kirtonia A, Sethi G, Pandey AK, Garg M. The implication of long non-coding RNAs in the diagnosis, pathogenesis and drug resistance of pancreatic ductal adenocarcinoma and their possible therapeutic potential. Biochim Biophys Acta. 2020;1874(2):188423.
346.
Zurück zum Zitat Chen X, Tang FR, Arfuso F, Cai WQ, Ma Z, Yang J, et al. The emerging role of long non-coding RNAs in the metastasis of hepatocellular carcinoma. Biomolecules. 2019;10(1):66.PubMedCentral Chen X, Tang FR, Arfuso F, Cai WQ, Ma Z, Yang J, et al. The emerging role of long non-coding RNAs in the metastasis of hepatocellular carcinoma. Biomolecules. 2019;10(1):66.PubMedCentral
347.
Zurück zum Zitat Mishra S, Verma SS, Rai V, Awasthee N, Chava S, Hui KM, et al. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell Mol Life Sci CMLS. 2019;76(10):1947–66.PubMed Mishra S, Verma SS, Rai V, Awasthee N, Chava S, Hui KM, et al. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell Mol Life Sci CMLS. 2019;76(10):1947–66.PubMed
348.
Zurück zum Zitat Shi Q, Li Y, Li S, Jin L, Lai H, Wu Y, et al. LncRNA DILA1 inhibits Cyclin D1 degradation and contributes to tamoxifen resistance in breast cancer. Nat Commun. 2020;11(1):5513.PubMedPubMedCentral Shi Q, Li Y, Li S, Jin L, Lai H, Wu Y, et al. LncRNA DILA1 inhibits Cyclin D1 degradation and contributes to tamoxifen resistance in breast cancer. Nat Commun. 2020;11(1):5513.PubMedPubMedCentral
350.
Zurück zum Zitat Chang KC, Diermeier SD, Yu AT, Brine LD, Russo S, Bhatia S, et al. MaTAR25 lncRNA regulates the Tensin1 gene to impact breast cancer progression. Nat Commun. 2020;11(1):6438.PubMedPubMedCentral Chang KC, Diermeier SD, Yu AT, Brine LD, Russo S, Bhatia S, et al. MaTAR25 lncRNA regulates the Tensin1 gene to impact breast cancer progression. Nat Commun. 2020;11(1):6438.PubMedPubMedCentral
351.
Zurück zum Zitat Shermane Lim YW, Xiang X, Garg M, Le MTN, Li-Ann Wong A, Wang L, et al. The double-edged sword of H19 lncRNA: insights into cancer therapy. Cancer Lett. 2021;500:253–62.PubMed Shermane Lim YW, Xiang X, Garg M, Le MTN, Li-Ann Wong A, Wang L, et al. The double-edged sword of H19 lncRNA: insights into cancer therapy. Cancer Lett. 2021;500:253–62.PubMed
352.
Zurück zum Zitat Lei Y, Guo W, Chen B, Chen L, Gong J, Li W. Tumor-released lncRNA H19 promotes gefitinib resistance via packaging into exosomes in non-small cell lung cancer. Oncol Rep. 2018;40(6):3438–46.PubMedPubMedCentral Lei Y, Guo W, Chen B, Chen L, Gong J, Li W. Tumor-released lncRNA H19 promotes gefitinib resistance via packaging into exosomes in non-small cell lung cancer. Oncol Rep. 2018;40(6):3438–46.PubMedPubMedCentral
353.
Zurück zum Zitat Hardin H, Helein H, Meyer K, Robertson S, Zhang R, Zhong W, et al. Thyroid cancer stem-like cell exosomes: regulation of EMT via transfer of lncRNAs. Lab Invest J Tech Methods Pathol. 2018;98(9):1133–42. Hardin H, Helein H, Meyer K, Robertson S, Zhang R, Zhong W, et al. Thyroid cancer stem-like cell exosomes: regulation of EMT via transfer of lncRNAs. Lab Invest J Tech Methods Pathol. 2018;98(9):1133–42.
354.
Zurück zum Zitat Zhang H, Wang J, Ren T, Huang Y, Yu Y, Chen C, et al. LncRNA CASC15 is upregulated in osteosarcoma plasma exosomes and CASC15 knockdown inhibits osteosarcoma progression by regulating miR-338-3p/RAB14 axis. Onco Targets Ther. 2020;13:12055–66.PubMedPubMedCentral Zhang H, Wang J, Ren T, Huang Y, Yu Y, Chen C, et al. LncRNA CASC15 is upregulated in osteosarcoma plasma exosomes and CASC15 knockdown inhibits osteosarcoma progression by regulating miR-338-3p/RAB14 axis. Onco Targets Ther. 2020;13:12055–66.PubMedPubMedCentral
355.
Zurück zum Zitat Ashrafizadeh M, Zarabi A, Hushmandi K, Moghadam ER, Hashemi F, Daneshi S, et al. C-Myc signaling pathway in treatment and prevention of brain tumors. Curr Cancer Drug Targets. 2021;21(1):2–20.PubMed Ashrafizadeh M, Zarabi A, Hushmandi K, Moghadam ER, Hashemi F, Daneshi S, et al. C-Myc signaling pathway in treatment and prevention of brain tumors. Curr Cancer Drug Targets. 2021;21(1):2–20.PubMed
356.
Zurück zum Zitat Lu Y, Chen L, Li L, Cao Y. Exosomes derived from brain metastatic breast cancer cells destroy the blood-brain barrier by carrying lncRNA GS1-600G8.5. BioMed Res Int. 2020;2020:7461727.PubMedPubMedCentral Lu Y, Chen L, Li L, Cao Y. Exosomes derived from brain metastatic breast cancer cells destroy the blood-brain barrier by carrying lncRNA GS1-600G8.5. BioMed Res Int. 2020;2020:7461727.PubMedPubMedCentral
357.
Zurück zum Zitat Yin Z, Zhou Y, Ma T, Chen S, Shi N, Zou Y, et al. Down-regulated lncRNA SBF2-AS1 in M2 macrophage-derived exosomes elevates miR-122-5p to restrict XIAP, thereby limiting pancreatic cancer development. J Cell Mol Med. 2020;24(9):5028–38.PubMedPubMedCentral Yin Z, Zhou Y, Ma T, Chen S, Shi N, Zou Y, et al. Down-regulated lncRNA SBF2-AS1 in M2 macrophage-derived exosomes elevates miR-122-5p to restrict XIAP, thereby limiting pancreatic cancer development. J Cell Mol Med. 2020;24(9):5028–38.PubMedPubMedCentral
358.
Zurück zum Zitat Xu CG, Yang MF, Ren YQ, Wu CH, Wang LQ. Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells. Eur Rev Med Pharmacol Sci. 2016;20(20):4362–8.PubMed Xu CG, Yang MF, Ren YQ, Wu CH, Wang LQ. Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells. Eur Rev Med Pharmacol Sci. 2016;20(20):4362–8.PubMed
359.
Zurück zum Zitat Cui X, Xiao D, Cui Y, Wang X. Exosomes-derived long non-coding RNA HOTAIR reduces laryngeal cancer radiosensitivity by regulating microRNA-454-3p/E2F2 Axis. Onco Targets Ther. 2019;12:10827–39.PubMedPubMedCentral Cui X, Xiao D, Cui Y, Wang X. Exosomes-derived long non-coding RNA HOTAIR reduces laryngeal cancer radiosensitivity by regulating microRNA-454-3p/E2F2 Axis. Onco Targets Ther. 2019;12:10827–39.PubMedPubMedCentral
360.
Zurück zum Zitat Yang YN, Zhang R, Du JW, Yuan HH, Li YJ, Wei XL, et al. Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell Int. 2018;18:164.PubMedPubMedCentral Yang YN, Zhang R, Du JW, Yuan HH, Li YJ, Wei XL, et al. Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell Int. 2018;18:164.PubMedPubMedCentral
361.
Zurück zum Zitat Barbagallo C, Brex D, Caponnetto A, Cirnigliaro M, Scalia M, Magnano A, et al. LncRNA UCA1, upregulated in CRC biopsies and downregulated in serum exosomes, controls mRNA expression by RNA-RNA interactions. Mol Thera Nucleic Acids. 2018;12:229–41. Barbagallo C, Brex D, Caponnetto A, Cirnigliaro M, Scalia M, Magnano A, et al. LncRNA UCA1, upregulated in CRC biopsies and downregulated in serum exosomes, controls mRNA expression by RNA-RNA interactions. Mol Thera Nucleic Acids. 2018;12:229–41.
362.
Zurück zum Zitat Wu Q, Wu X, Ying X, Zhu Q, Wang X, Jiang L, et al. Suppression of endothelial cell migration by tumor associated macrophage-derived exosomes is reversed by epithelial ovarian cancer exosomal lncRNA. Cancer Cell Int. 2017;17:62.PubMedPubMedCentral Wu Q, Wu X, Ying X, Zhu Q, Wang X, Jiang L, et al. Suppression of endothelial cell migration by tumor associated macrophage-derived exosomes is reversed by epithelial ovarian cancer exosomal lncRNA. Cancer Cell Int. 2017;17:62.PubMedPubMedCentral
363.
Zurück zum Zitat Xian J, Zeng Y, Chen S, Lu L, Liu L, Chen J, et al. Discovery of a novel linc01125 isoform in serum exosomes as a promising biomarker for NSCLC diagnosis and survival assessment. Carcinogenesis. 2021;42(6):831–41.PubMed Xian J, Zeng Y, Chen S, Lu L, Liu L, Chen J, et al. Discovery of a novel linc01125 isoform in serum exosomes as a promising biomarker for NSCLC diagnosis and survival assessment. Carcinogenesis. 2021;42(6):831–41.PubMed
364.
Zurück zum Zitat Holdt LM, Kohlmaier A, Teupser D. Molecular roles and function of circular RNAs in eukaryotic cells. Cell Mol Life Sci CMLS. 2018;75(6):1071–98.PubMed Holdt LM, Kohlmaier A, Teupser D. Molecular roles and function of circular RNAs in eukaryotic cells. Cell Mol Life Sci CMLS. 2018;75(6):1071–98.PubMed
365.
Zurück zum Zitat Chen LL. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 2016;17(4):205–11.PubMed Chen LL. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 2016;17(4):205–11.PubMed
366.
Zurück zum Zitat Haque S, Harries LW. Circular RNAs (circRNAs) in health and disease. Genes. 2017;8(12):353.PubMedCentral Haque S, Harries LW. Circular RNAs (circRNAs) in health and disease. Genes. 2017;8(12):353.PubMedCentral
367.
Zurück zum Zitat Meng S, Zhou H, Feng Z, Xu Z, Tang Y, Li P, et al. CircRNA: functions and properties of a novel potential biomarker for cancer. Mol Cancer. 2017;16(1):94.PubMedPubMedCentral Meng S, Zhou H, Feng Z, Xu Z, Tang Y, Li P, et al. CircRNA: functions and properties of a novel potential biomarker for cancer. Mol Cancer. 2017;16(1):94.PubMedPubMedCentral
368.
Zurück zum Zitat Hao Q, Zhang Z. hsa_circRNA_000166 facilitated cell growth and limited apoptosis through targeting miR-326/LASP1 axis in colorectal cancer. Gastroenterol Res Pract. 2020;2020:8834359.PubMedPubMedCentral Hao Q, Zhang Z. hsa_circRNA_000166 facilitated cell growth and limited apoptosis through targeting miR-326/LASP1 axis in colorectal cancer. Gastroenterol Res Pract. 2020;2020:8834359.PubMedPubMedCentral
369.
Zurück zum Zitat Zhou ZF, Wei Z, Yao JC, Liu SY, Wang F, Wang Z, et al. CircRNA_102179 promotes the proliferation, migration and invasion in non-small cell lung cancer cells by regulating miR-330-5p/HMGB3 axis. Pathol Res Pract. 2020;216(11):153144.PubMed Zhou ZF, Wei Z, Yao JC, Liu SY, Wang F, Wang Z, et al. CircRNA_102179 promotes the proliferation, migration and invasion in non-small cell lung cancer cells by regulating miR-330-5p/HMGB3 axis. Pathol Res Pract. 2020;216(11):153144.PubMed
370.
Zurück zum Zitat Zhang X, Yang H, Jia Y, Xu Z, Zhang L, Sun M, et al. circRNA_0005529 facilitates growth and metastasis of gastric cancer via regulating miR-527/Sp1 axis. BMC Mol Cell Biol. 2021;22(1):6.PubMedPubMedCentral Zhang X, Yang H, Jia Y, Xu Z, Zhang L, Sun M, et al. circRNA_0005529 facilitates growth and metastasis of gastric cancer via regulating miR-527/Sp1 axis. BMC Mol Cell Biol. 2021;22(1):6.PubMedPubMedCentral
371.
Zurück zum Zitat Li J, Li Z, Jiang P, Peng M, Zhang X, Chen K, et al. Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res CR. 2018;37(1):177.PubMed Li J, Li Z, Jiang P, Peng M, Zhang X, Chen K, et al. Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res CR. 2018;37(1):177.PubMed
372.
Zurück zum Zitat Ashrafizadeh M, Ahmadi Z, Farkhondeh T, Samarghandian S. Resveratrol targeting the Wnt signaling pathway: a focus on therapeutic activities. J Cell Physiol. 2020;235(5):4135–45.PubMed Ashrafizadeh M, Ahmadi Z, Farkhondeh T, Samarghandian S. Resveratrol targeting the Wnt signaling pathway: a focus on therapeutic activities. J Cell Physiol. 2020;235(5):4135–45.PubMed
373.
Zurück zum Zitat Zhao H, Chen S, Fu Q. Exosomes from CD133(+) cells carrying circ-ABCC1 mediate cell stemness and metastasis in colorectal cancer. J Cell Biochem. 2020;121(5–6):3286–97.PubMed Zhao H, Chen S, Fu Q. Exosomes from CD133(+) cells carrying circ-ABCC1 mediate cell stemness and metastasis in colorectal cancer. J Cell Biochem. 2020;121(5–6):3286–97.PubMed
374.
Zurück zum Zitat Ma J, Qi G, Li L. A novel serum exosomes-based biomarker hsa_circ_0002130 facilitates osimertinib-resistance in non-small cell lung cancer by sponging miR-498. Onco Targets Ther. 2020;13:5293–307.PubMedPubMedCentral Ma J, Qi G, Li L. A novel serum exosomes-based biomarker hsa_circ_0002130 facilitates osimertinib-resistance in non-small cell lung cancer by sponging miR-498. Onco Targets Ther. 2020;13:5293–307.PubMedPubMedCentral
376.
Zurück zum Zitat Xian J, Su W, Liu L, Rao B, Lin M, Feng Y, et al. Identification of three circular RNA cargoes in serum exosomes as diagnostic biomarkers of non-small-cell lung cancer in the chinese population. J Mol Diagn JMD. 2020;22(8):1096–108.PubMed Xian J, Su W, Liu L, Rao B, Lin M, Feng Y, et al. Identification of three circular RNA cargoes in serum exosomes as diagnostic biomarkers of non-small-cell lung cancer in the chinese population. J Mol Diagn JMD. 2020;22(8):1096–108.PubMed
377.
Zurück zum Zitat Chen F, Huang C, Wu Q, Jiang L, Chen S, Chen L. Circular RNAs expression profiles in plasma exosomes from early-stage lung adenocarcinoma and the potential biomarkers. J Cell Biochem. 2020;121(3):2525–33.PubMed Chen F, Huang C, Wu Q, Jiang L, Chen S, Chen L. Circular RNAs expression profiles in plasma exosomes from early-stage lung adenocarcinoma and the potential biomarkers. J Cell Biochem. 2020;121(3):2525–33.PubMed
378.
Zurück zum Zitat Wang J, Zhang Q, Zhou S, Xu H, Wang D, Feng J, et al. Circular RNA expression in exosomes derived from breast cancer cells and patients. Epigenomics. 2019;11(4):411–21.PubMed Wang J, Zhang Q, Zhou S, Xu H, Wang D, Feng J, et al. Circular RNA expression in exosomes derived from breast cancer cells and patients. Epigenomics. 2019;11(4):411–21.PubMed
379.
Zurück zum Zitat Dou Y, Cha DJ, Franklin JL, Higginbotham JN, Jeppesen DK, Weaver AM, et al. Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes. Sci Rep. 2016;6:37982.PubMedPubMedCentral Dou Y, Cha DJ, Franklin JL, Higginbotham JN, Jeppesen DK, Weaver AM, et al. Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes. Sci Rep. 2016;6:37982.PubMedPubMedCentral
380.
Zurück zum Zitat Guo S, Hu C, Zhai X, Sun D. Circular RNA 0006602 in plasma exosomes: a new potential diagnostic biomarker for hepatocellular carcinoma. Am J Transl Res. 2021;13(6):6001–15.PubMedPubMedCentral Guo S, Hu C, Zhai X, Sun D. Circular RNA 0006602 in plasma exosomes: a new potential diagnostic biomarker for hepatocellular carcinoma. Am J Transl Res. 2021;13(6):6001–15.PubMedPubMedCentral
381.
Zurück zum Zitat Ouyang Y, Tang Y, Fu L, Peng S, Wu W, Tan D, et al. Exosomes secreted by chronic hepatitis B patients with PNALT and liver inflammation grade ≥ A2 promoted the progression of liver cancer by transferring miR-25-3p to inhibit the co-expression of TCF21 and HHIP. Cell Prolif. 2020;53(7):e12833.PubMedPubMedCentral Ouyang Y, Tang Y, Fu L, Peng S, Wu W, Tan D, et al. Exosomes secreted by chronic hepatitis B patients with PNALT and liver inflammation grade ≥ A2 promoted the progression of liver cancer by transferring miR-25-3p to inhibit the co-expression of TCF21 and HHIP. Cell Prolif. 2020;53(7):e12833.PubMedPubMedCentral
382.
Zurück zum Zitat Li L, Li C, Wang S, Wang Z, Jiang J, Wang W, et al. Exosomes derived from hypoxic oral squamous cell carcinoma cells deliver miR-21 to normoxic cells to elicit a prometastatic phenotype. Can Res. 2016;76(7):1770–80. Li L, Li C, Wang S, Wang Z, Jiang J, Wang W, et al. Exosomes derived from hypoxic oral squamous cell carcinoma cells deliver miR-21 to normoxic cells to elicit a prometastatic phenotype. Can Res. 2016;76(7):1770–80.
383.
Zurück zum Zitat Park EJ, Jung HJ, Choi HJ, Jang HJ, Park HJ, Nejsum LN, et al. Exosomes co-expressing AQP5-targeting miRNAs and IL-4 receptor-binding peptide inhibit the migration of human breast cancer cells. FASEB J Off Publ Fed Am Soc Exp Biol. 2020;34(2):3379–98. Park EJ, Jung HJ, Choi HJ, Jang HJ, Park HJ, Nejsum LN, et al. Exosomes co-expressing AQP5-targeting miRNAs and IL-4 receptor-binding peptide inhibit the migration of human breast cancer cells. FASEB J Off Publ Fed Am Soc Exp Biol. 2020;34(2):3379–98.
384.
Zurück zum Zitat Yang Y, Li J, Geng Y. Exosomes derived from chronic lymphocytic leukaemia cells transfer miR-146a to induce the transition of mesenchymal stromal cells into cancer-associated fibroblasts. J Biochem. 2020;168(5):491–8.PubMed Yang Y, Li J, Geng Y. Exosomes derived from chronic lymphocytic leukaemia cells transfer miR-146a to induce the transition of mesenchymal stromal cells into cancer-associated fibroblasts. J Biochem. 2020;168(5):491–8.PubMed
385.
Zurück zum Zitat Wang C, Wang Y, Chang X, Ba X, Hu N, Liu Q, et al. Melanoma-derived exosomes endow fibroblasts with an invasive potential via miR-21 target signaling pathway. Cancer Manag Res. 2020;12:12965–74.PubMedPubMedCentral Wang C, Wang Y, Chang X, Ba X, Hu N, Liu Q, et al. Melanoma-derived exosomes endow fibroblasts with an invasive potential via miR-21 target signaling pathway. Cancer Manag Res. 2020;12:12965–74.PubMedPubMedCentral
386.
Zurück zum Zitat Wang Y, Wang P, Zhao L, Chen X, Lin Z, Zhang L, et al. miR-224-5p carried by human umbilical cord mesenchymal stem cells-derived exosomes regulates autophagy in breast cancer cells via HOXA5. Front Cell Dev Biol. 2021;9:679185.PubMedPubMedCentral Wang Y, Wang P, Zhao L, Chen X, Lin Z, Zhang L, et al. miR-224-5p carried by human umbilical cord mesenchymal stem cells-derived exosomes regulates autophagy in breast cancer cells via HOXA5. Front Cell Dev Biol. 2021;9:679185.PubMedPubMedCentral
387.
Zurück zum Zitat Zhou Y, Zhu Y, Dong X, Cao G, Li Y, Fan Y, et al. Exosomes derived from pancreatic cancer cells induce osteoclast differentiation through the miR125a-5p/TNFRSF1B pathway. Onco Targets Ther. 2021;14:2727–39.PubMedPubMedCentral Zhou Y, Zhu Y, Dong X, Cao G, Li Y, Fan Y, et al. Exosomes derived from pancreatic cancer cells induce osteoclast differentiation through the miR125a-5p/TNFRSF1B pathway. Onco Targets Ther. 2021;14:2727–39.PubMedPubMedCentral
388.
Zurück zum Zitat Rezaei R, Baghaei K, Hashemi SM, Zali MR, Ghanbarian H, Amani D. Tumor-derived exosomes enriched by miRNA-124 promote anti-tumor immune response in CT-26 tumor-bearing mice. Front Med. 2021;8:619939. Rezaei R, Baghaei K, Hashemi SM, Zali MR, Ghanbarian H, Amani D. Tumor-derived exosomes enriched by miRNA-124 promote anti-tumor immune response in CT-26 tumor-bearing mice. Front Med. 2021;8:619939.
389.
Zurück zum Zitat Zhang N, Li L, Luo J, Tan J, Hu W, Li Z, et al. Inhibiting microRNA-424 in bone marrow mesenchymal stem cells-derived exosomes suppresses tumor growth in colorectal cancer by upregulating TGFBR3. Arch Biochem Biophys. 2021;709:108965.PubMed Zhang N, Li L, Luo J, Tan J, Hu W, Li Z, et al. Inhibiting microRNA-424 in bone marrow mesenchymal stem cells-derived exosomes suppresses tumor growth in colorectal cancer by upregulating TGFBR3. Arch Biochem Biophys. 2021;709:108965.PubMed
390.
Zurück zum Zitat Vakhshiteh F, Rahmani S, Ostad SN, Madjd Z, Dinarvand R, Atyabi F. Exosomes derived from miR-34a-overexpressing mesenchymal stem cells inhibit in vitro tumor growth: a new approach for drug delivery. Life Sci. 2021;266:118871.PubMed Vakhshiteh F, Rahmani S, Ostad SN, Madjd Z, Dinarvand R, Atyabi F. Exosomes derived from miR-34a-overexpressing mesenchymal stem cells inhibit in vitro tumor growth: a new approach for drug delivery. Life Sci. 2021;266:118871.PubMed
391.
Zurück zum Zitat Sheykhhasan M, Kalhor N, Sheikholeslami A, Dolati M, Amini E, Fazaeli H. Exosomes of mesenchymal stem cells as a proper vehicle for transfecting miR-145 into the breast cancer cell line and its effect on metastasis. Biomed Res Int. 2021;2021:5516078.PubMedPubMedCentral Sheykhhasan M, Kalhor N, Sheikholeslami A, Dolati M, Amini E, Fazaeli H. Exosomes of mesenchymal stem cells as a proper vehicle for transfecting miR-145 into the breast cancer cell line and its effect on metastasis. Biomed Res Int. 2021;2021:5516078.PubMedPubMedCentral
392.
Zurück zum Zitat Du L, Tao X, Shen X. Human umbilical cord mesenchymal stem cell-derived exosomes inhibit migration and invasion of breast cancer cells via miR-21-5p/ZNF367 pathway. Breast Cancer (Tokyo, Japan). 2021;28(4):829–37. Du L, Tao X, Shen X. Human umbilical cord mesenchymal stem cell-derived exosomes inhibit migration and invasion of breast cancer cells via miR-21-5p/ZNF367 pathway. Breast Cancer (Tokyo, Japan). 2021;28(4):829–37.
393.
Zurück zum Zitat Yue S, Ye X, Zhou T, Gan D, Qian H, Fang W, et al. PGRN(-/-) TAMs-derived exosomes inhibit breast cancer cell invasion and migration and its mechanism exploration. Life Sci. 2021;264:118687.PubMed Yue S, Ye X, Zhou T, Gan D, Qian H, Fang W, et al. PGRN(-/-) TAMs-derived exosomes inhibit breast cancer cell invasion and migration and its mechanism exploration. Life Sci. 2021;264:118687.PubMed
394.
Zurück zum Zitat Liu Y, Yang Y, Du J, Lin D, Li F. MiR-3613-3p from carcinoma-associated fibroblasts exosomes promoted breast cancer cell proliferation and metastasis by regulating SOCS2 expression. IUBMB Life. 2020;72(8):1705–14.PubMed Liu Y, Yang Y, Du J, Lin D, Li F. MiR-3613-3p from carcinoma-associated fibroblasts exosomes promoted breast cancer cell proliferation and metastasis by regulating SOCS2 expression. IUBMB Life. 2020;72(8):1705–14.PubMed
395.
Zurück zum Zitat Wang B, Zhang Y, Ye M, Wu J, Ma L, Chen H. Cisplatin-resistant MDA-MB-231 cell-derived exosomes increase the resistance of recipient cells in an exosomal miR-423-5p-dependent manner. Curr Drug Metab. 2019;20(10):804–14.PubMed Wang B, Zhang Y, Ye M, Wu J, Ma L, Chen H. Cisplatin-resistant MDA-MB-231 cell-derived exosomes increase the resistance of recipient cells in an exosomal miR-423-5p-dependent manner. Curr Drug Metab. 2019;20(10):804–14.PubMed
397.
Zurück zum Zitat Xu Y, Liu N, Wei Y, Zhou D, Lin R, Wang X, et al. Anticancer effects of miR-124 delivered by BM-MSC derived exosomes on cell proliferation, epithelial mesenchymal transition, and chemotherapy sensitivity of pancreatic cancer cells. Aging. 2020;12(19):19660–76.PubMedPubMedCentral Xu Y, Liu N, Wei Y, Zhou D, Lin R, Wang X, et al. Anticancer effects of miR-124 delivered by BM-MSC derived exosomes on cell proliferation, epithelial mesenchymal transition, and chemotherapy sensitivity of pancreatic cancer cells. Aging. 2020;12(19):19660–76.PubMedPubMedCentral
398.
Zurück zum Zitat Lan J, Sun L, Xu F, Liu L, Hu F, Song D, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Can Res. 2019;79(1):146–58. Lan J, Sun L, Xu F, Liu L, Hu F, Song D, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Can Res. 2019;79(1):146–58.
399.
Zurück zum Zitat Huang W, Yan Y, Liu Y, Lin M, Ma J, Zhang W, et al. Exosomes with low miR-34c-3p expression promote invasion and migration of non-small cell lung cancer by upregulating integrin α2β1. Signal Transduct Target Ther. 2020;5(1):39.PubMedPubMedCentral Huang W, Yan Y, Liu Y, Lin M, Ma J, Zhang W, et al. Exosomes with low miR-34c-3p expression promote invasion and migration of non-small cell lung cancer by upregulating integrin α2β1. Signal Transduct Target Ther. 2020;5(1):39.PubMedPubMedCentral
400.
Zurück zum Zitat Chen R, Qian Z, Xu X, Zhang C, Niu Y, Wang Z, et al. Exosomes-transmitted miR-7 reverses gefitinib resistance by targeting YAP in non-small-cell lung cancer. Pharmacol Res. 2021;165:105442.PubMed Chen R, Qian Z, Xu X, Zhang C, Niu Y, Wang Z, et al. Exosomes-transmitted miR-7 reverses gefitinib resistance by targeting YAP in non-small-cell lung cancer. Pharmacol Res. 2021;165:105442.PubMed
401.
Zurück zum Zitat Deng Z, Rong Y, Teng Y, Zhuang X, Samykutty A, Mu J, et al. Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis. Oncogene. 2017;36(5):639–51.PubMed Deng Z, Rong Y, Teng Y, Zhuang X, Samykutty A, Mu J, et al. Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis. Oncogene. 2017;36(5):639–51.PubMed
402.
Zurück zum Zitat Lou G, Song X, Yang F, Wu S, Wang J, Chen Z, et al. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol. 2015;8:122.PubMedPubMedCentral Lou G, Song X, Yang F, Wu S, Wang J, Chen Z, et al. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol. 2015;8:122.PubMedPubMedCentral
403.
Zurück zum Zitat Li J, Yu J, Zhang H, Wang B, Guo H, Bai J, et al. Exosomes-derived MiR-302b suppresses lung cancer cell proliferation and migration via TGFβRII inhibition. Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol. 2016;38(5):1715–26. Li J, Yu J, Zhang H, Wang B, Guo H, Bai J, et al. Exosomes-derived MiR-302b suppresses lung cancer cell proliferation and migration via TGFβRII inhibition. Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol. 2016;38(5):1715–26.
404.
Zurück zum Zitat Zhang X, Li F, Tang Y, Ren Q, Xiao B, Wan Y, et al. miR-21a in exosomes from Lewis lung carcinoma cells accelerates tumor growth through targeting PDCD4 to enhance expansion of myeloid-derived suppressor cells. Oncogene. 2020;39(40):6354–69.PubMed Zhang X, Li F, Tang Y, Ren Q, Xiao B, Wan Y, et al. miR-21a in exosomes from Lewis lung carcinoma cells accelerates tumor growth through targeting PDCD4 to enhance expansion of myeloid-derived suppressor cells. Oncogene. 2020;39(40):6354–69.PubMed
405.
Zurück zum Zitat He Z, Li W, Zheng T, Liu D, Zhao S. Human umbilical cord mesenchymal stem cells-derived exosomes deliver microRNA-375 to downregulate ENAH and thus retard esophageal squamous cell carcinoma progression. J Exp Clin Cancer Res CR. 2020;39(1):140.PubMed He Z, Li W, Zheng T, Liu D, Zhao S. Human umbilical cord mesenchymal stem cells-derived exosomes deliver microRNA-375 to downregulate ENAH and thus retard esophageal squamous cell carcinoma progression. J Exp Clin Cancer Res CR. 2020;39(1):140.PubMed
406.
Zurück zum Zitat Lee JC, Zhao JT, Gundara J, Serpell J, Bach LA, Sidhu S. Papillary thyroid cancer-derived exosomes contain miRNA-146b and miRNA-222. J Surg Res. 2015;196(1):39–48.PubMed Lee JC, Zhao JT, Gundara J, Serpell J, Bach LA, Sidhu S. Papillary thyroid cancer-derived exosomes contain miRNA-146b and miRNA-222. J Surg Res. 2015;196(1):39–48.PubMed
407.
Zurück zum Zitat Liu Y, Fu W, Cao X, Li S, Xiong T, Zhang X, et al. Delivery of miR-224-5p by exosomes from cancer-associated fibroblasts potentiates progression of clear cell renal cell carcinoma. Comput Math Methods Med. 2021;2021:5517747.PubMedPubMedCentral Liu Y, Fu W, Cao X, Li S, Xiong T, Zhang X, et al. Delivery of miR-224-5p by exosomes from cancer-associated fibroblasts potentiates progression of clear cell renal cell carcinoma. Comput Math Methods Med. 2021;2021:5517747.PubMedPubMedCentral
408.
Zurück zum Zitat Chang L, Gao H, Wang L, Wang N, Zhang S, Zhou X, et al. Exosomes derived from miR-1228 overexpressing bone marrow-mesenchymal stem cells promote growth of gastric cancer cells. Aging. 2021;13(8):11808–21.PubMedPubMedCentral Chang L, Gao H, Wang L, Wang N, Zhang S, Zhou X, et al. Exosomes derived from miR-1228 overexpressing bone marrow-mesenchymal stem cells promote growth of gastric cancer cells. Aging. 2021;13(8):11808–21.PubMedPubMedCentral
409.
Zurück zum Zitat Pan L, Liang W, Fu M, Huang ZH, Li X, Zhang W, et al. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J Cancer Res Clin Oncol. 2017;143(6):991–1004.PubMed Pan L, Liang W, Fu M, Huang ZH, Li X, Zhang W, et al. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J Cancer Res Clin Oncol. 2017;143(6):991–1004.PubMed
410.
Zurück zum Zitat Xian D, Niu L, Zeng J, Wang L. LncRNA KCNQ1OT1 secreted by tumor cell-derived exosomes mediates immune escape in colorectal cancer by regulating PD-L1 ubiquitination via MiR-30a-5p/USP22. Front Cell Dev Biol. 2021;9:653808.PubMedPubMedCentral Xian D, Niu L, Zeng J, Wang L. LncRNA KCNQ1OT1 secreted by tumor cell-derived exosomes mediates immune escape in colorectal cancer by regulating PD-L1 ubiquitination via MiR-30a-5p/USP22. Front Cell Dev Biol. 2021;9:653808.PubMedPubMedCentral
411.
Zurück zum Zitat Wang YL, Liu LC, Hung Y, Chen CJ, Lin YZ, Wu WR, et al. Long non-coding RNA HOTAIR in circulatory exosomes is correlated with ErbB2/HER2 positivity in breast cancer. Breast (Edinburgh, Scotland). 2019;46:64–9. Wang YL, Liu LC, Hung Y, Chen CJ, Lin YZ, Wu WR, et al. Long non-coding RNA HOTAIR in circulatory exosomes is correlated with ErbB2/HER2 positivity in breast cancer. Breast (Edinburgh, Scotland). 2019;46:64–9.
412.
Zurück zum Zitat Yang H, Qu H, Huang H, Mu Z, Mao M, Xie Q, et al. Exosomes-mediated transfer of long noncoding RNA LINC01133 represses bladder cancer progression via regulating the Wnt signaling pathway. Cell Biol Int. 2021;45(7):1510–22.PubMed Yang H, Qu H, Huang H, Mu Z, Mao M, Xie Q, et al. Exosomes-mediated transfer of long noncoding RNA LINC01133 represses bladder cancer progression via regulating the Wnt signaling pathway. Cell Biol Int. 2021;45(7):1510–22.PubMed
413.
Zurück zum Zitat Liu H, Shen M, Zhao D, Ru D, Duan Y, Ding C, et al. The effect of triptolide-loaded exosomes on the proliferation and apoptosis of human ovarian cancer SKOV3 cells. Biomed Res Int. 2019;2019:2595801.PubMedPubMedCentral Liu H, Shen M, Zhao D, Ru D, Duan Y, Ding C, et al. The effect of triptolide-loaded exosomes on the proliferation and apoptosis of human ovarian cancer SKOV3 cells. Biomed Res Int. 2019;2019:2595801.PubMedPubMedCentral
414.
Zurück zum Zitat Kim MS, Haney MJ, Zhao Y, Yuan D, Deygen I, Klyachko NL, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomed Nanotechnol Biol Med. 2018;14(1):195–204. Kim MS, Haney MJ, Zhao Y, Yuan D, Deygen I, Klyachko NL, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomed Nanotechnol Biol Med. 2018;14(1):195–204.
415.
Zurück zum Zitat Qambrani A, Rehman FU, Tanziela T, Shaikh S, Semcheddine F, Du T, et al. Biocompatible exosomes nanodrug cargo for cancer cell bioimaging and drug delivery. Biomed Mater (Bristol, England). 2021;16(2):025026. Qambrani A, Rehman FU, Tanziela T, Shaikh S, Semcheddine F, Du T, et al. Biocompatible exosomes nanodrug cargo for cancer cell bioimaging and drug delivery. Biomed Mater (Bristol, England). 2021;16(2):025026.
416.
Zurück zum Zitat Dhayapulay A, Kanapathipillai M. Exosomes based geldanamycin delivery to cancer cells with increased therapeutic efficacy. J Biomed Nanotechnol. 2019;15(11):2202–8.PubMed Dhayapulay A, Kanapathipillai M. Exosomes based geldanamycin delivery to cancer cells with increased therapeutic efficacy. J Biomed Nanotechnol. 2019;15(11):2202–8.PubMed
417.
Zurück zum Zitat Wang P, Wang H, Huang Q, Peng C, Yao L, Chen H, et al. Exosomes from M1-polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation. Theranostics. 2019;9(6):1714–27.PubMedPubMedCentral Wang P, Wang H, Huang Q, Peng C, Yao L, Chen H, et al. Exosomes from M1-polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation. Theranostics. 2019;9(6):1714–27.PubMedPubMedCentral
418.
Zurück zum Zitat Thakur A, Sidu RK, Zou H, Alam MK, Yang M, Lee Y. Inhibition of glioma cells’ proliferation by doxorubicin-loaded exosomes via microfluidics. Int J Nanomed. 2020;15:8331–43. Thakur A, Sidu RK, Zou H, Alam MK, Yang M, Lee Y. Inhibition of glioma cells’ proliferation by doxorubicin-loaded exosomes via microfluidics. Int J Nanomed. 2020;15:8331–43.
419.
Zurück zum Zitat Zhang X, Liu L, Tang M, Li H, Guo X, Yang X. The effects of umbilical cord-derived macrophage exosomes loaded with cisplatin on the growth and drug resistance of ovarian cancer cells. Drug Dev Ind Pharm. 2020;46(7):1150–62.PubMed Zhang X, Liu L, Tang M, Li H, Guo X, Yang X. The effects of umbilical cord-derived macrophage exosomes loaded with cisplatin on the growth and drug resistance of ovarian cancer cells. Drug Dev Ind Pharm. 2020;46(7):1150–62.PubMed
420.
Zurück zum Zitat Li Y, Gao Y, Gong C, Wang Z, Xia Q, Gu F, et al. A33 antibody-functionalized exosomes for targeted delivery of doxorubicin against colorectal cancer. Nanomed Nanotechnol Biol Med. 2018;14(7):1973–85. Li Y, Gao Y, Gong C, Wang Z, Xia Q, Gu F, et al. A33 antibody-functionalized exosomes for targeted delivery of doxorubicin against colorectal cancer. Nanomed Nanotechnol Biol Med. 2018;14(7):1973–85.
422.
Zurück zum Zitat Lu ZR, Laney VE, Hall R, Ayat N. Environment-responsive lipid/siRNA nanoparticles for cancer therapy. Adv Healthc Mater. 2021;10(5):2001294. Lu ZR, Laney VE, Hall R, Ayat N. Environment-responsive lipid/siRNA nanoparticles for cancer therapy. Adv Healthc Mater. 2021;10(5):2001294.
423.
Zurück zum Zitat Zhang MM, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. The growth of siRNA-based therapeutics: updated clinical studies. Biochem Pharmacol. 2021;189:114432.PubMed Zhang MM, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. The growth of siRNA-based therapeutics: updated clinical studies. Biochem Pharmacol. 2021;189:114432.PubMed
424.
Zurück zum Zitat Mirzaei S, Gholami MH, Hashemi F, Zabolian A, Hushmandi K, Rahmanian V, et al. Employing siRNA tool and its delivery platforms in suppressing cisplatin resistance: approaching to a new era of cancer chemotherapy. Life Sci. 2021;277:119430.PubMed Mirzaei S, Gholami MH, Hashemi F, Zabolian A, Hushmandi K, Rahmanian V, et al. Employing siRNA tool and its delivery platforms in suppressing cisplatin resistance: approaching to a new era of cancer chemotherapy. Life Sci. 2021;277:119430.PubMed
425.
Zurück zum Zitat Ashrafizade M, Delfi M, Hashemi F, Zabolian A, Saleki H, Bagherian M, et al. Biomedical application of chitosan-based nanoscale delivery systems: Potential usefulness in siRNA delivery for cancer therapy. Carbohydr Polym. 2021;260:117809. Ashrafizade M, Delfi M, Hashemi F, Zabolian A, Saleki H, Bagherian M, et al. Biomedical application of chitosan-based nanoscale delivery systems: Potential usefulness in siRNA delivery for cancer therapy. Carbohydr Polym. 2021;260:117809.
426.
Zurück zum Zitat Najafi S, Tan SC, Aghamiri S, Raee P, Ebrahimi Z, Jahromi ZK, et al. Therapeutic potentials of CRISPR-Cas genome editing technology in human viral infections. Biomed Pharmacother. 2022;148:112743.PubMedPubMedCentral Najafi S, Tan SC, Aghamiri S, Raee P, Ebrahimi Z, Jahromi ZK, et al. Therapeutic potentials of CRISPR-Cas genome editing technology in human viral infections. Biomed Pharmacother. 2022;148:112743.PubMedPubMedCentral
427.
Zurück zum Zitat Khalaf K, Janowicz K, Dyszkiewicz-Konwińska M, Hutchings G, Dompe C, Moncrieff L, et al. CRISPR/Cas9 in cancer immunotherapy: animal models and human clinical trials. Genes. 2020;11(8):921.PubMedCentral Khalaf K, Janowicz K, Dyszkiewicz-Konwińska M, Hutchings G, Dompe C, Moncrieff L, et al. CRISPR/Cas9 in cancer immunotherapy: animal models and human clinical trials. Genes. 2020;11(8):921.PubMedCentral
428.
Zurück zum Zitat Hazafa A, Mumtaz M, Farooq MF, Bilal S, Chaudhry SN, Firdous M, et al. CRISPR/Cas9: a powerful genome editing technique for the treatment of cancer cells with present challenges and future directions. Life Sci. 2020;263:118525.PubMedPubMedCentral Hazafa A, Mumtaz M, Farooq MF, Bilal S, Chaudhry SN, Firdous M, et al. CRISPR/Cas9: a powerful genome editing technique for the treatment of cancer cells with present challenges and future directions. Life Sci. 2020;263:118525.PubMedPubMedCentral
429.
Zurück zum Zitat Martinez-Lage M, Torres-Ruiz R, Puig-Serra P, Moreno-Gaona P, Martin MC, Moya FJ, et al. In vivo CRISPR/Cas9 targeting of fusion oncogenes for selective elimination of cancer cells. Nat Commun. 2020;11(1):5060.PubMedPubMedCentral Martinez-Lage M, Torres-Ruiz R, Puig-Serra P, Moreno-Gaona P, Martin MC, Moya FJ, et al. In vivo CRISPR/Cas9 targeting of fusion oncogenes for selective elimination of cancer cells. Nat Commun. 2020;11(1):5060.PubMedPubMedCentral
430.
Zurück zum Zitat Mohammadinejad R, Sassan H, Pardakhty A, Hashemabadi M, Ashrafizadeh M, Dehshahri A, et al. ZEB1 and ZEB2 gene editing mediated by CRISPR/Cas9 in A549 cell line. Bratislava Med J. 2020;121(1):31–6. Mohammadinejad R, Sassan H, Pardakhty A, Hashemabadi M, Ashrafizadeh M, Dehshahri A, et al. ZEB1 and ZEB2 gene editing mediated by CRISPR/Cas9 in A549 cell line. Bratislava Med J. 2020;121(1):31–6.
432.
Zurück zum Zitat Tao H, Xu H, Zuo L, Li C, Qiao G, Guo M, et al. Exosomes-coated bcl-2 siRNA inhibits the growth of digestive system tumors both in vitro and in vivo. Int J Biol Macromol. 2020;161:470–80.PubMed Tao H, Xu H, Zuo L, Li C, Qiao G, Guo M, et al. Exosomes-coated bcl-2 siRNA inhibits the growth of digestive system tumors both in vitro and in vivo. Int J Biol Macromol. 2020;161:470–80.PubMed
433.
Zurück zum Zitat Koh SA, Kim MK, Lee KH, Kim SW, Kim J-R. RhoGDI2 is associated with HGF-mediated tumor invasion through VEGF in stomach cancer. Clin Exp Metastasis. 2014;31(7):805–15.PubMed Koh SA, Kim MK, Lee KH, Kim SW, Kim J-R. RhoGDI2 is associated with HGF-mediated tumor invasion through VEGF in stomach cancer. Clin Exp Metastasis. 2014;31(7):805–15.PubMed
434.
Zurück zum Zitat Yang X, Zhang XF, Lu X, Jia HL, Liang L, Dong QZ, et al. MicroRNA-26a suppresses angiogenesis in human hepatocellular carcinoma by targeting hepatocyte growth factor-cMet pathway. Hepatology. 2014;59(5):1874–85.PubMed Yang X, Zhang XF, Lu X, Jia HL, Liang L, Dong QZ, et al. MicroRNA-26a suppresses angiogenesis in human hepatocellular carcinoma by targeting hepatocyte growth factor-cMet pathway. Hepatology. 2014;59(5):1874–85.PubMed
435.
Zurück zum Zitat Zhai Y, Wu W, Xi X, Yu R. Adipose-derived stem cells promote proliferation and invasion in cervical cancer by targeting the HGF/c-MET pathway. Cancer Manag Research. 2020;12:11823–32. Zhai Y, Wu W, Xi X, Yu R. Adipose-derived stem cells promote proliferation and invasion in cervical cancer by targeting the HGF/c-MET pathway. Cancer Manag Research. 2020;12:11823–32.
436.
Zurück zum Zitat Xu J, Liu S, Yang X, Cao S, Zhou Y. Paracrine HGF promotes EMT and mediates the effects of PSC on chemoresistance by activating c-Met/PI3K/Akt signaling in pancreatic cancer in vitro. Life Sci. 2020;263:118523.PubMed Xu J, Liu S, Yang X, Cao S, Zhou Y. Paracrine HGF promotes EMT and mediates the effects of PSC on chemoresistance by activating c-Met/PI3K/Akt signaling in pancreatic cancer in vitro. Life Sci. 2020;263:118523.PubMed
437.
Zurück zum Zitat Zhang H, Wang Y, Bai M, Wang J, Zhu K, Liu R, et al. Exosomes serve as nanoparticles to suppress tumor growth and angiogenesis in gastric cancer by delivering hepatocyte growth factor siRNA. Cancer Sci. 2018;109(3):629–41.PubMedPubMedCentral Zhang H, Wang Y, Bai M, Wang J, Zhu K, Liu R, et al. Exosomes serve as nanoparticles to suppress tumor growth and angiogenesis in gastric cancer by delivering hepatocyte growth factor siRNA. Cancer Sci. 2018;109(3):629–41.PubMedPubMedCentral
438.
Zurück zum Zitat Zhang Z, Cheng L, Zhang Q, Kong Y, He D, Li K, et al. Co-targeting Plk1 and DNMT3a in advanced prostate cancer. Adv Sci (Weinheim, Baden-Wurttemberg, Germany). 2021;8(13):e2101458. Zhang Z, Cheng L, Zhang Q, Kong Y, He D, Li K, et al. Co-targeting Plk1 and DNMT3a in advanced prostate cancer. Adv Sci (Weinheim, Baden-Wurttemberg, Germany). 2021;8(13):e2101458.
439.
Zurück zum Zitat Montaudon E, Nikitorowicz-Buniak J, Sourd L, Morisset L, El Botty R, Huguet L, et al. PLK1 inhibition exhibits strong anti-tumoral activity in CCND1-driven breast cancer metastases with acquired palbociclib resistance. Nat Commun. 2020;11(1):4053.PubMedPubMedCentral Montaudon E, Nikitorowicz-Buniak J, Sourd L, Morisset L, El Botty R, Huguet L, et al. PLK1 inhibition exhibits strong anti-tumoral activity in CCND1-driven breast cancer metastases with acquired palbociclib resistance. Nat Commun. 2020;11(1):4053.PubMedPubMedCentral
440.
Zurück zum Zitat Greco KA, Franzen CA, Foreman KE, Flanigan RC, Kuo PC, Gupta GN. PLK-1 silencing in bladder cancer by siRNA delivered With exosomes. Urology. 2016;91(241):e1-7. Greco KA, Franzen CA, Foreman KE, Flanigan RC, Kuo PC, Gupta GN. PLK-1 silencing in bladder cancer by siRNA delivered With exosomes. Urology. 2016;91(241):e1-7.
441.
Zurück zum Zitat Limoni SK, Moghadam MF, Moazzeni SM, Gomari H, Salimi F. Engineered exosomes for targeted transfer of siRNA to HER2 positive breast cancer cells. Appl Biochem Biotechnol. 2019;187(1):352–64.PubMed Limoni SK, Moghadam MF, Moazzeni SM, Gomari H, Salimi F. Engineered exosomes for targeted transfer of siRNA to HER2 positive breast cancer cells. Appl Biochem Biotechnol. 2019;187(1):352–64.PubMed
442.
Zurück zum Zitat Teesalu T, Sugahara KN, Kotamraju VR, Ruoslahti E. C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetration. Proc Natl Acad Sci USA. 2009;106(38):16157–62.PubMedPubMedCentral Teesalu T, Sugahara KN, Kotamraju VR, Ruoslahti E. C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetration. Proc Natl Acad Sci USA. 2009;106(38):16157–62.PubMedPubMedCentral
443.
Zurück zum Zitat Kawakami T, Tokunaga T, Hatanaka H, Kijima H, Yamazaki H, Abe Y, et al. Neuropilin 1 and neuropilin 2 co-expression is significantly correlated with increased vascularity and poor prognosis in nonsmall cell lung carcinoma. Cancer. 2002;95(10):2196–201.PubMed Kawakami T, Tokunaga T, Hatanaka H, Kijima H, Yamazaki H, Abe Y, et al. Neuropilin 1 and neuropilin 2 co-expression is significantly correlated with increased vascularity and poor prognosis in nonsmall cell lung carcinoma. Cancer. 2002;95(10):2196–201.PubMed
444.
Zurück zum Zitat Roth L, Agemy L, Kotamraju VR, Braun G, Teesalu T, Sugahara KN, et al. Transtumoral targeting enabled by a novel neuropilin-binding peptide. Oncogene. 2012;31(33):3754–63.PubMed Roth L, Agemy L, Kotamraju VR, Braun G, Teesalu T, Sugahara KN, et al. Transtumoral targeting enabled by a novel neuropilin-binding peptide. Oncogene. 2012;31(33):3754–63.PubMed
445.
Zurück zum Zitat Bai J, Duan J, Liu R, Du Y, Luo Q, Cui Y, et al. Engineered targeting tLyp-1 exosomes as gene therapy vectors for efficient delivery of siRNA into lung cancer cells. Asian J Pharm Sci. 2020;15(4):461–71.PubMed Bai J, Duan J, Liu R, Du Y, Luo Q, Cui Y, et al. Engineered targeting tLyp-1 exosomes as gene therapy vectors for efficient delivery of siRNA into lung cancer cells. Asian J Pharm Sci. 2020;15(4):461–71.PubMed
448.
Zurück zum Zitat Li H, Yang C, Shi Y, Zhao L. Exosomes derived from siRNA against GRP78 modified bone-marrow-derived mesenchymal stem cells suppress Sorafenib resistance in hepatocellular carcinoma. J Nanobiotechnol. 2018;16(1):103. Li H, Yang C, Shi Y, Zhao L. Exosomes derived from siRNA against GRP78 modified bone-marrow-derived mesenchymal stem cells suppress Sorafenib resistance in hepatocellular carcinoma. J Nanobiotechnol. 2018;16(1):103.
449.
Zurück zum Zitat Kim SM, Yang Y, Oh SJ, Hong Y, Seo M, Jang M. Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting. J Controll Release Off J Controll Release Soc. 2017;266:8–16. Kim SM, Yang Y, Oh SJ, Hong Y, Seo M, Jang M. Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting. J Controll Release Off J Controll Release Soc. 2017;266:8–16.
450.
Zurück zum Zitat Gulei D, Berindan-Neagoe I. Activation of necroptosis by engineered self tumor-derived exosomes loaded with CRISPR/Cas9. Mol Ther Nucleic Acids. 2019;17:448–51.PubMedPubMedCentral Gulei D, Berindan-Neagoe I. Activation of necroptosis by engineered self tumor-derived exosomes loaded with CRISPR/Cas9. Mol Ther Nucleic Acids. 2019;17:448–51.PubMedPubMedCentral
451.
Zurück zum Zitat Gabrusiewicz K, Li X, Wei J, Hashimoto Y, Marisetty AL, Ott M, et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology. 2018;7(4):e1412909.PubMedPubMedCentral Gabrusiewicz K, Li X, Wei J, Hashimoto Y, Marisetty AL, Ott M, et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology. 2018;7(4):e1412909.PubMedPubMedCentral
452.
Zurück zum Zitat Qian M, Wang S, Guo X, Wang J, Zhang Z, Qiu W, et al. Hypoxic glioma-derived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-κB pathways. Oncogene. 2020;39(2):428–42.PubMed Qian M, Wang S, Guo X, Wang J, Zhang Z, Qiu W, et al. Hypoxic glioma-derived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-κB pathways. Oncogene. 2020;39(2):428–42.PubMed
453.
Zurück zum Zitat Wang Z, Yuan Y, Ji X, Xiao X, Li Z, Yi X, et al. The Hippo-TAZ axis mediates vascular endothelial growth factor C in glioblastoma-derived exosomes to promote angiogenesis. Cancer Lett. 2021;513:1–13.PubMed Wang Z, Yuan Y, Ji X, Xiao X, Li Z, Yi X, et al. The Hippo-TAZ axis mediates vascular endothelial growth factor C in glioblastoma-derived exosomes to promote angiogenesis. Cancer Lett. 2021;513:1–13.PubMed
454.
Zurück zum Zitat Naryzhny S, Volnitskiy A, Kopylov A, Zorina E, Kamyshinsky R, Bairamukov V, et al. Proteome of glioblastoma-derived exosomes as a source of biomarkers. Biomedicines. 2020;8(7):216.PubMedCentral Naryzhny S, Volnitskiy A, Kopylov A, Zorina E, Kamyshinsky R, Bairamukov V, et al. Proteome of glioblastoma-derived exosomes as a source of biomarkers. Biomedicines. 2020;8(7):216.PubMedCentral
455.
Zurück zum Zitat Azambuja JH, Ludwig N, Yerneni S, Rao A, Braganhol E, Whiteside TL. Molecular profiles and immunomodulatory activities of glioblastoma-derived exosomes. Neuro-Oncol Adv. 2020;2(1):vdaa 56. Azambuja JH, Ludwig N, Yerneni S, Rao A, Braganhol E, Whiteside TL. Molecular profiles and immunomodulatory activities of glioblastoma-derived exosomes. Neuro-Oncol Adv. 2020;2(1):vdaa 56.
456.
Zurück zum Zitat Sun Z, Wang L, Zhou Y, Dong L, Ma W, Lv L, et al. Glioblastoma stem cell-derived exosomes enhance stemness and tumorigenicity of glioma cells by transferring Notch1 protein. Cell Mol Neurobiol. 2020;40(5):767–84.PubMed Sun Z, Wang L, Zhou Y, Dong L, Ma W, Lv L, et al. Glioblastoma stem cell-derived exosomes enhance stemness and tumorigenicity of glioma cells by transferring Notch1 protein. Cell Mol Neurobiol. 2020;40(5):767–84.PubMed
457.
Zurück zum Zitat Li J, Liao T, Liu H, Yuan H, Ouyang T, Wang J, et al. Hypoxic glioma stem cell-derived exosomes containing linc01060 promote progression of glioma by regulating the MZF1/c-Myc/HIF1α axis. Can Res. 2021;81(1):114–28. Li J, Liao T, Liu H, Yuan H, Ouyang T, Wang J, et al. Hypoxic glioma stem cell-derived exosomes containing linc01060 promote progression of glioma by regulating the MZF1/c-Myc/HIF1α axis. Can Res. 2021;81(1):114–28.
458.
Zurück zum Zitat Guo X, Qiu W, Liu Q, Qian M, Wang S, Zhang Z, et al. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten pathways. Oncogene. 2018;37(31):4239–59.PubMed Guo X, Qiu W, Liu Q, Qian M, Wang S, Zhang Z, et al. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten pathways. Oncogene. 2018;37(31):4239–59.PubMed
459.
Zurück zum Zitat Qian M, Chen Z, Guo X, Wang S, Zhang Z, Qiu W, et al. Exosomes derived from hypoxic glioma deliver miR-1246 and miR-10b-5p to normoxic glioma cells to promote migration and invasion. Lab Investig J Tech Methods Pathol. 2021;101(5):612–24. Qian M, Chen Z, Guo X, Wang S, Zhang Z, Qiu W, et al. Exosomes derived from hypoxic glioma deliver miR-1246 and miR-10b-5p to normoxic glioma cells to promote migration and invasion. Lab Investig J Tech Methods Pathol. 2021;101(5):612–24.
460.
Zurück zum Zitat Lang HL, Hu GW, Zhang B, Kuang W, Chen Y, Wu L, et al. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol Rep. 2017;38(2):785–98.PubMedPubMedCentral Lang HL, Hu GW, Zhang B, Kuang W, Chen Y, Wu L, et al. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol Rep. 2017;38(2):785–98.PubMedPubMedCentral
461.
Zurück zum Zitat Sun X, Ma X, Wang J, Zhao Y, Wang Y, Bihl JC, et al. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget. 2017;8(22):36137–48.PubMedPubMedCentral Sun X, Ma X, Wang J, Zhao Y, Wang Y, Bihl JC, et al. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget. 2017;8(22):36137–48.PubMedPubMedCentral
462.
Zurück zum Zitat Guo X, Qiu W, Wang J, Liu Q, Qian M, Wang S, et al. Glioma exosomes mediate the expansion and function of myeloid-derived suppressor cells through microRNA-29a/Hbp1 and microRNA-92a/Prkar1a pathways. Int J Cancer. 2019;144(12):3111–26.PubMed Guo X, Qiu W, Wang J, Liu Q, Qian M, Wang S, et al. Glioma exosomes mediate the expansion and function of myeloid-derived suppressor cells through microRNA-29a/Hbp1 and microRNA-92a/Prkar1a pathways. Int J Cancer. 2019;144(12):3111–26.PubMed
463.
Zurück zum Zitat Domenis R, Cesselli D, Toffoletto B, Bourkoula E, Caponnetto F, Manini I, et al. Systemic T cells immunosuppression of glioma stem cell-derived exosomes is mediated by monocytic myeloid-derived suppressor cells. PLoS One. 2017;12(1):e0169932.PubMedPubMedCentral Domenis R, Cesselli D, Toffoletto B, Bourkoula E, Caponnetto F, Manini I, et al. Systemic T cells immunosuppression of glioma stem cell-derived exosomes is mediated by monocytic myeloid-derived suppressor cells. PLoS One. 2017;12(1):e0169932.PubMedPubMedCentral
464.
Zurück zum Zitat Figueroa J, Phillips LM, Shahar T, Hossain A, Gumin J, Kim H, et al. Exosomes from glioma-associated mesenchymal stem cells increase the tumorigenicity of glioma stem-like cells via transfer of miR-1587. Can Res. 2017;77(21):5808–19. Figueroa J, Phillips LM, Shahar T, Hossain A, Gumin J, Kim H, et al. Exosomes from glioma-associated mesenchymal stem cells increase the tumorigenicity of glioma stem-like cells via transfer of miR-1587. Can Res. 2017;77(21):5808–19.
465.
Zurück zum Zitat Wen SW, Sceneay J, Lima LG, Wong CS, Becker M, Krumeich S, et al. The biodistribution and immune suppressive effects of breast cancer-derived exosomes. Can Res. 2016;76(23):6816–27. Wen SW, Sceneay J, Lima LG, Wong CS, Becker M, Krumeich S, et al. The biodistribution and immune suppressive effects of breast cancer-derived exosomes. Can Res. 2016;76(23):6816–27.
466.
Zurück zum Zitat Wu Q, Sun S, Li Z, Yang Q, Li B, Zhu S, et al. Breast cancer-released exosomes trigger cancer-associated cachexia to promote tumor progression. Adipocyte. 2019;8(1):31–45.PubMed Wu Q, Sun S, Li Z, Yang Q, Li B, Zhu S, et al. Breast cancer-released exosomes trigger cancer-associated cachexia to promote tumor progression. Adipocyte. 2019;8(1):31–45.PubMed
467.
Zurück zum Zitat Inubushi S, Kawaguchi H, Mizumoto S, Kunihisa T, Baba M, Kitayama Y, et al. Oncogenic miRNAs identified in tear exosomes from metastatic breast cancer patients. Anticancer Res. 2020;40(6):3091–6.PubMed Inubushi S, Kawaguchi H, Mizumoto S, Kunihisa T, Baba M, Kitayama Y, et al. Oncogenic miRNAs identified in tear exosomes from metastatic breast cancer patients. Anticancer Res. 2020;40(6):3091–6.PubMed
468.
Zurück zum Zitat Wen SW, Lima LG, Lobb RJ, Norris EL, Hastie ML, Krumeich S, et al. Breast cancer-derived exosomes reflect the cell-of-origin phenotype. Proteomics. 2019;19(8):e1800180.PubMed Wen SW, Lima LG, Lobb RJ, Norris EL, Hastie ML, Krumeich S, et al. Breast cancer-derived exosomes reflect the cell-of-origin phenotype. Proteomics. 2019;19(8):e1800180.PubMed
469.
Zurück zum Zitat Wang X, Cheng K, Zhang G, Jia Z, Yu Y, Guo J, et al. Enrichment of CD44 in exosomes from breast cancer cells treated with doxorubicin promotes chemoresistance. Front Oncol. 2020;10:960.PubMedPubMedCentral Wang X, Cheng K, Zhang G, Jia Z, Yu Y, Guo J, et al. Enrichment of CD44 in exosomes from breast cancer cells treated with doxorubicin promotes chemoresistance. Front Oncol. 2020;10:960.PubMedPubMedCentral
470.
Zurück zum Zitat Li K, Liu T, Chen J, Ni H, Li W. Survivin in breast cancer-derived exosomes activates fibroblasts by up-regulating SOD1, whose feedback promotes cancer proliferation and metastasis. J Biol Chem. 2020;295(40):13737–52.PubMedPubMedCentral Li K, Liu T, Chen J, Ni H, Li W. Survivin in breast cancer-derived exosomes activates fibroblasts by up-regulating SOD1, whose feedback promotes cancer proliferation and metastasis. J Biol Chem. 2020;295(40):13737–52.PubMedPubMedCentral
471.
Zurück zum Zitat Piao YJ, Kim HS, Hwang EH, Woo J, Zhang M, Moon WK. Breast cancer cell-derived exosomes and macrophage polarization are associated with lymph node metastasis. Oncotarget. 2018;9(7):7398–410.PubMed Piao YJ, Kim HS, Hwang EH, Woo J, Zhang M, Moon WK. Breast cancer cell-derived exosomes and macrophage polarization are associated with lymph node metastasis. Oncotarget. 2018;9(7):7398–410.PubMed
472.
Zurück zum Zitat Sen K, Sheppe AEF, Singh I, Hui WW, Edelmann MJ, Rinaldi C. Exosomes released by breast cancer cells under mild hyperthermic stress possess immunogenic potential and modulate polarization in vitro in macrophages. Int J Hyperth Off J Eur Soc Hyperth Oncol North Am Hyperth Gr. 2020;37(1):696–710. Sen K, Sheppe AEF, Singh I, Hui WW, Edelmann MJ, Rinaldi C. Exosomes released by breast cancer cells under mild hyperthermic stress possess immunogenic potential and modulate polarization in vitro in macrophages. Int J Hyperth Off J Eur Soc Hyperth Oncol North Am Hyperth Gr. 2020;37(1):696–710.
473.
Zurück zum Zitat Rahman MA, Barger JF, Lovat F, Gao M, Otterson GA, Nana-Sinkam P. Lung cancer exosomes as drivers of epithelial mesenchymal transition. Oncotarget. 2016;7(34):54852–66.PubMedPubMedCentral Rahman MA, Barger JF, Lovat F, Gao M, Otterson GA, Nana-Sinkam P. Lung cancer exosomes as drivers of epithelial mesenchymal transition. Oncotarget. 2016;7(34):54852–66.PubMedPubMedCentral
474.
Zurück zum Zitat Wei F, Ma C, Zhou T, Dong X, Luo Q, Geng L, et al. Exosomes derived from gemcitabine-resistant cells transfer malignant phenotypic traits via delivery of miRNA-222-3p. Mol Cancer. 2017;16(1):132.PubMedPubMedCentral Wei F, Ma C, Zhou T, Dong X, Luo Q, Geng L, et al. Exosomes derived from gemcitabine-resistant cells transfer malignant phenotypic traits via delivery of miRNA-222-3p. Mol Cancer. 2017;16(1):132.PubMedPubMedCentral
475.
Zurück zum Zitat Xu H, Jiao X, Wu Y, Li S, Cao L, Dong L. Exosomes derived from PM2.5-treated lung cancer cells promote the growth of lung cancer via the Wnt3a/β-catenin pathway. Oncol Rep. 2019;41(2):1180–8.PubMed Xu H, Jiao X, Wu Y, Li S, Cao L, Dong L. Exosomes derived from PM2.5-treated lung cancer cells promote the growth of lung cancer via the Wnt3a/β-catenin pathway. Oncol Rep. 2019;41(2):1180–8.PubMed
476.
Zurück zum Zitat Wu H, Zeng C, Ye Y, Liu J, Mu Z, Xie Y, et al. Exosomes from irradiated nonsmall cell lung cancer cells reduced sensitivity of recipient cells to anaplastic lymphoma kinase inhibitors. Mol Pharm. 2018;15(5):1892–900.PubMed Wu H, Zeng C, Ye Y, Liu J, Mu Z, Xie Y, et al. Exosomes from irradiated nonsmall cell lung cancer cells reduced sensitivity of recipient cells to anaplastic lymphoma kinase inhibitors. Mol Pharm. 2018;15(5):1892–900.PubMed
477.
Zurück zum Zitat Pan D, Chen J, Feng C, Wu W, Wang Y, Tong J, et al. Preferential localization of MUC1 glycoprotein in exosomes secreted by non-small cell lung carcinoma cells. Int J Mol Sci. 2019;20(2):323.PubMedCentral Pan D, Chen J, Feng C, Wu W, Wang Y, Tong J, et al. Preferential localization of MUC1 glycoprotein in exosomes secreted by non-small cell lung carcinoma cells. Int J Mol Sci. 2019;20(2):323.PubMedCentral
478.
Zurück zum Zitat Deng G, Qu J, Zhang Y, Che X, Cheng Y, Fan Y, et al. Gastric cancer-derived exosomes promote peritoneal metastasis by destroying the mesothelial barrier. FEBS Lett. 2017;591(14):2167–79.PubMed Deng G, Qu J, Zhang Y, Che X, Cheng Y, Fan Y, et al. Gastric cancer-derived exosomes promote peritoneal metastasis by destroying the mesothelial barrier. FEBS Lett. 2017;591(14):2167–79.PubMed
479.
Zurück zum Zitat Wu L, Zhang X, Zhang B, Shi H, Yuan X, Sun Y, et al. Exosomes derived from gastric cancer cells activate NF-κB pathway in macrophages to promote cancer progression. Tumour Biol J Int Soc Oncodev Biol Med. 2016;37(9):12169–80. Wu L, Zhang X, Zhang B, Shi H, Yuan X, Sun Y, et al. Exosomes derived from gastric cancer cells activate NF-κB pathway in macrophages to promote cancer progression. Tumour Biol J Int Soc Oncodev Biol Med. 2016;37(9):12169–80.
480.
Zurück zum Zitat Shen Y, Xue C, Li X, Ba L, Gu J, Sun Z, et al. Effects of gastric cancer cell-derived exosomes on the immune regulation of mesenchymal stem cells by the NF-kB signaling pathway. Stem Cells Dev. 2019;28(7):464–76.PubMed Shen Y, Xue C, Li X, Ba L, Gu J, Sun Z, et al. Effects of gastric cancer cell-derived exosomes on the immune regulation of mesenchymal stem cells by the NF-kB signaling pathway. Stem Cells Dev. 2019;28(7):464–76.PubMed
481.
Zurück zum Zitat Ning X, Zhang H, Wang C, Song X. Exosomes released by gastric cancer cells induce transition of pericytes into cancer-associated fibroblasts. Med Sci Monit Int Med J Exp Clin Res. 2018;24:2350–9. Ning X, Zhang H, Wang C, Song X. Exosomes released by gastric cancer cells induce transition of pericytes into cancer-associated fibroblasts. Med Sci Monit Int Med J Exp Clin Res. 2018;24:2350–9.
482.
Zurück zum Zitat Gao Z, Song C, Li G, Lin H, Lian X, Zhang N, et al. Pyrotinib treatment on HER2-positive gastric cancer cells promotes the released exosomes to enhance endothelial cell progression, which can be counteracted by apatinib. Onco Targets Ther. 2019;12:2777–87.PubMedPubMedCentral Gao Z, Song C, Li G, Lin H, Lian X, Zhang N, et al. Pyrotinib treatment on HER2-positive gastric cancer cells promotes the released exosomes to enhance endothelial cell progression, which can be counteracted by apatinib. Onco Targets Ther. 2019;12:2777–87.PubMedPubMedCentral
483.
Zurück zum Zitat Chen L, Guo P, He Y, Chen Z, Chen L, Luo Y, et al. HCC-derived exosomes elicit HCC progression and recurrence by epithelial-mesenchymal transition through MAPK/ERK signalling pathway. Cell Death Dis. 2018;9(5):513.PubMedPubMedCentral Chen L, Guo P, He Y, Chen Z, Chen L, Luo Y, et al. HCC-derived exosomes elicit HCC progression and recurrence by epithelial-mesenchymal transition through MAPK/ERK signalling pathway. Cell Death Dis. 2018;9(5):513.PubMedPubMedCentral
484.
Zurück zum Zitat Ge Y, Mu W, Ba Q, Li J, Jiang Y, Xia Q, et al. Hepatocellular carcinoma-derived exosomes in organotropic metastasis, recurrence and early diagnosis application. Cancer Lett. 2020;477:41–8.PubMed Ge Y, Mu W, Ba Q, Li J, Jiang Y, Xia Q, et al. Hepatocellular carcinoma-derived exosomes in organotropic metastasis, recurrence and early diagnosis application. Cancer Lett. 2020;477:41–8.PubMed
485.
Zurück zum Zitat He X, Yu J, Xiong L, Liu Y, Fan L, Li Y, et al. Exosomes derived from liver cancer cells reprogram biological behaviors of LO2 cells by transferring Linc-ROR. Gene. 2019;719:144044.PubMed He X, Yu J, Xiong L, Liu Y, Fan L, Li Y, et al. Exosomes derived from liver cancer cells reprogram biological behaviors of LO2 cells by transferring Linc-ROR. Gene. 2019;719:144044.PubMed
486.
Zurück zum Zitat Kimura H, Yamamoto H, Harada T, Fumoto K, Osugi Y, Sada R, et al. CKAP4, a DKK1 receptor, is a biomarker in exosomes derived from pancreatic cancer and a molecular target for therapy. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25(6):1936–47. Kimura H, Yamamoto H, Harada T, Fumoto K, Osugi Y, Sada R, et al. CKAP4, a DKK1 receptor, is a biomarker in exosomes derived from pancreatic cancer and a molecular target for therapy. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25(6):1936–47.
487.
Zurück zum Zitat Linton SS, Abraham T, Liao J, Clawson GA, Butler PJ, Fox T, et al. Tumor-promoting effects of pancreatic cancer cell exosomes on THP-1-derived macrophages. PLoS One. 2018;13(11):e0206759.PubMedPubMedCentral Linton SS, Abraham T, Liao J, Clawson GA, Butler PJ, Fox T, et al. Tumor-promoting effects of pancreatic cancer cell exosomes on THP-1-derived macrophages. PLoS One. 2018;13(11):e0206759.PubMedPubMedCentral
488.
Zurück zum Zitat Tang P, Tao L, Yuan C, Zhang L, Xiu D. Serum derived exosomes from pancreatic cancer patients promoted metastasis: an iTRAQ-based proteomic analysis. Onco Targets Ther. 2019;12:9329–39.PubMedPubMedCentral Tang P, Tao L, Yuan C, Zhang L, Xiu D. Serum derived exosomes from pancreatic cancer patients promoted metastasis: an iTRAQ-based proteomic analysis. Onco Targets Ther. 2019;12:9329–39.PubMedPubMedCentral
489.
Zurück zum Zitat Zhang YF, Zhou YZ, Zhang B, Huang SF, Li PP, He XM, et al. Pancreatic cancer-derived exosomes promoted pancreatic stellate cells recruitment by pancreatic cancer. J Cancer. 2019;10(18):4397–407.PubMedPubMedCentral Zhang YF, Zhou YZ, Zhang B, Huang SF, Li PP, He XM, et al. Pancreatic cancer-derived exosomes promoted pancreatic stellate cells recruitment by pancreatic cancer. J Cancer. 2019;10(18):4397–407.PubMedPubMedCentral
491.
Zurück zum Zitat Masamune A, Yoshida N, Hamada S, Takikawa T, Nabeshima T, Shimosegawa T. Exosomes derived from pancreatic cancer cells induce activation and profibrogenic activities in pancreatic stellate cells. Biochem Biophys Res Commun. 2018;495(1):71–7.PubMed Masamune A, Yoshida N, Hamada S, Takikawa T, Nabeshima T, Shimosegawa T. Exosomes derived from pancreatic cancer cells induce activation and profibrogenic activities in pancreatic stellate cells. Biochem Biophys Res Commun. 2018;495(1):71–7.PubMed
492.
Zurück zum Zitat Zhang Y, Huang S, Li P, Chen Q, Li Y, Zhou Y, et al. Pancreatic cancer-derived exosomes suppress the production of GIP and GLP-1 from STC-1 cells in vitro by down-regulating the PCSK1/3. Cancer Lett. 2018;431:190–200.PubMed Zhang Y, Huang S, Li P, Chen Q, Li Y, Zhou Y, et al. Pancreatic cancer-derived exosomes suppress the production of GIP and GLP-1 from STC-1 cells in vitro by down-regulating the PCSK1/3. Cancer Lett. 2018;431:190–200.PubMed
493.
Zurück zum Zitat Yu Z, Zhao S, Ren L, Wang L, Chen Z, Hoffman RM, et al. Pancreatic cancer-derived exosomes promote tumor metastasis and liver pre-metastatic niche formation. Oncotarget. 2017;8(38):63461–83.PubMedPubMedCentral Yu Z, Zhao S, Ren L, Wang L, Chen Z, Hoffman RM, et al. Pancreatic cancer-derived exosomes promote tumor metastasis and liver pre-metastatic niche formation. Oncotarget. 2017;8(38):63461–83.PubMedPubMedCentral
494.
Zurück zum Zitat Jiao YJ, Jin DD, Jiang F, Liu JX, Qu LS, Ni WK, et al. Characterization and proteomic profiling of pancreatic cancer-derived serum exosomes. J Cell Biochem. 2019;120(1):988–99.PubMed Jiao YJ, Jin DD, Jiang F, Liu JX, Qu LS, Ni WK, et al. Characterization and proteomic profiling of pancreatic cancer-derived serum exosomes. J Cell Biochem. 2019;120(1):988–99.PubMed
495.
Zurück zum Zitat Cheng L, Zhang K, Qing Y, Li D, Cui M, Jin P, et al. Proteomic and lipidomic analysis of exosomes derived from ovarian cancer cells and ovarian surface epithelial cells. J Ovarian Res. 2020;13(1):9.PubMedPubMedCentral Cheng L, Zhang K, Qing Y, Li D, Cui M, Jin P, et al. Proteomic and lipidomic analysis of exosomes derived from ovarian cancer cells and ovarian surface epithelial cells. J Ovarian Res. 2020;13(1):9.PubMedPubMedCentral
496.
Zurück zum Zitat Lee AH, Ghosh D, Quach N, Schroeder D, Dawson MR. Ovarian cancer exosomes trigger differential biophysical response in tumor-derived fibroblasts. Sci Rep. 2020;10(1):8686.PubMedPubMedCentral Lee AH, Ghosh D, Quach N, Schroeder D, Dawson MR. Ovarian cancer exosomes trigger differential biophysical response in tumor-derived fibroblasts. Sci Rep. 2020;10(1):8686.PubMedPubMedCentral
497.
Zurück zum Zitat Yang C, Kim HS, Song G, Lim W. The potential role of exosomes derived from ovarian cancer cells for diagnostic and therapeutic approaches. J Cell Physiol. 2019;234(12):21493–503.PubMed Yang C, Kim HS, Song G, Lim W. The potential role of exosomes derived from ovarian cancer cells for diagnostic and therapeutic approaches. J Cell Physiol. 2019;234(12):21493–503.PubMed
498.
Zurück zum Zitat Chen X, Zhou J, Li X, Wang X, Lin Y, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018;435:80–91.PubMed Chen X, Zhou J, Li X, Wang X, Lin Y, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018;435:80–91.PubMed
499.
Zurück zum Zitat Chen X, Ying X, Wang X, Wu X, Zhu Q, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer deliver microRNA-940 to induce macrophage M2 polarization. Oncol Rep. 2017;38(1):522–8.PubMed Chen X, Ying X, Wang X, Wu X, Zhu Q, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer deliver microRNA-940 to induce macrophage M2 polarization. Oncol Rep. 2017;38(1):522–8.PubMed
500.
Zurück zum Zitat Ghorbanian M, Babashah S, Ataei F. The effects of ovarian cancer cell-derived exosomes on vascular endothelial growth factor expression in endothelial cells. EXCLI J. 2019;18:899–907.PubMedPubMedCentral Ghorbanian M, Babashah S, Ataei F. The effects of ovarian cancer cell-derived exosomes on vascular endothelial growth factor expression in endothelial cells. EXCLI J. 2019;18:899–907.PubMedPubMedCentral
501.
Zurück zum Zitat Bhat A, Yadav J, Thakur K, Aggarwal N, Tripathi T, Chhokar A, et al. Exosomes from cervical cancer cells facilitate pro-angiogenic endothelial reconditioning through transfer of Hedgehog-GLI signaling components. Cancer Cell Int. 2021;21(1):319.PubMedPubMedCentral Bhat A, Yadav J, Thakur K, Aggarwal N, Tripathi T, Chhokar A, et al. Exosomes from cervical cancer cells facilitate pro-angiogenic endothelial reconditioning through transfer of Hedgehog-GLI signaling components. Cancer Cell Int. 2021;21(1):319.PubMedPubMedCentral
502.
Zurück zum Zitat Ren G, Wang Y, Yuan S, Wang B. Dendritic cells loaded with HeLa-derived exosomes simulate an antitumor immune response. Oncol Lett. 2018;15(5):6636–40.PubMedPubMedCentral Ren G, Wang Y, Yuan S, Wang B. Dendritic cells loaded with HeLa-derived exosomes simulate an antitumor immune response. Oncol Lett. 2018;15(5):6636–40.PubMedPubMedCentral
503.
Zurück zum Zitat Salimu J, Webber J, Gurney M, Al-Taei S, Clayton A, Tabi Z. Dominant immunosuppression of dendritic cell function by prostate-cancer-derived exosomes. J Extracell Vesicles. 2017;6(1):1368823.PubMedPubMedCentral Salimu J, Webber J, Gurney M, Al-Taei S, Clayton A, Tabi Z. Dominant immunosuppression of dendritic cell function by prostate-cancer-derived exosomes. J Extracell Vesicles. 2017;6(1):1368823.PubMedPubMedCentral
504.
Zurück zum Zitat Esfandyari S, Elkafas H, Chugh RM, Park HS, Navarro A, Al-Hendy A. Exosomes as biomarkers for female reproductive diseases diagnosis and therapy. Int J Mol Sci. 2021;22(4):2165.PubMedPubMedCentral Esfandyari S, Elkafas H, Chugh RM, Park HS, Navarro A, Al-Hendy A. Exosomes as biomarkers for female reproductive diseases diagnosis and therapy. Int J Mol Sci. 2021;22(4):2165.PubMedPubMedCentral
505.
Zurück zum Zitat Bestard-Escalas J, Reigada R, Reyes J, de la Torre P, Liebisch G, Barceló-Coblijn G. Fatty acid unsaturation degree of plasma exosomes in colorectal cancer patients: a promising biomarker. Int J Mol Sci. 2021;22(10):5060.PubMedPubMedCentral Bestard-Escalas J, Reigada R, Reyes J, de la Torre P, Liebisch G, Barceló-Coblijn G. Fatty acid unsaturation degree of plasma exosomes in colorectal cancer patients: a promising biomarker. Int J Mol Sci. 2021;22(10):5060.PubMedPubMedCentral
506.
Zurück zum Zitat He R, Wang Z, Shi W, Yu L, Xia H, Huang Z, et al. Exosomes in hepatocellular carcinoma microenvironment and their potential clinical application value. Biomed Pharmacother. 2021;138:111529.PubMed He R, Wang Z, Shi W, Yu L, Xia H, Huang Z, et al. Exosomes in hepatocellular carcinoma microenvironment and their potential clinical application value. Biomed Pharmacother. 2021;138:111529.PubMed
507.
Zurück zum Zitat Damanti CC, Gaffo E, Lovisa F, Garbin A, Di Battista P, Gallingani I, et al. MiR-26a-5p as a reference to normalize microRNA qRT-PCR levels in plasma exosomes of pediatric hematological malignancies. Cells. 2021;10(1):101.PubMedCentral Damanti CC, Gaffo E, Lovisa F, Garbin A, Di Battista P, Gallingani I, et al. MiR-26a-5p as a reference to normalize microRNA qRT-PCR levels in plasma exosomes of pediatric hematological malignancies. Cells. 2021;10(1):101.PubMedCentral
508.
Zurück zum Zitat Fu C, Zhang Q, Wang A, Yang S, Jiang Y, Bai L, et al. EWI-2 controls nucleocytoplasmic shuttling of EGFR signaling molecules and miRNA sorting in exosomes to inhibit prostate cancer cell metastasis. Mol Oncol. 2021;15(5):1543–65.PubMedPubMedCentral Fu C, Zhang Q, Wang A, Yang S, Jiang Y, Bai L, et al. EWI-2 controls nucleocytoplasmic shuttling of EGFR signaling molecules and miRNA sorting in exosomes to inhibit prostate cancer cell metastasis. Mol Oncol. 2021;15(5):1543–65.PubMedPubMedCentral
509.
Zurück zum Zitat Cao B, Wang P, Gu L, Liu J. Use of four genes in exosomes as biomarkers for the identification of lung adenocarcinoma and lung squamous cell carcinoma. Oncol Lett. 2021;21(4):249.PubMedPubMedCentral Cao B, Wang P, Gu L, Liu J. Use of four genes in exosomes as biomarkers for the identification of lung adenocarcinoma and lung squamous cell carcinoma. Oncol Lett. 2021;21(4):249.PubMedPubMedCentral
511.
Zurück zum Zitat Ganig N, Baenke F, Thepkaysone ML, Lin K, Rao VS, Wong FC, et al. Proteomic analyses of fibroblast- and serum-derived exosomes identify QSOX1 as a marker for non-invasive detection of colorectal cancer. Cancers. 2021;13(6):1351.PubMedPubMedCentral Ganig N, Baenke F, Thepkaysone ML, Lin K, Rao VS, Wong FC, et al. Proteomic analyses of fibroblast- and serum-derived exosomes identify QSOX1 as a marker for non-invasive detection of colorectal cancer. Cancers. 2021;13(6):1351.PubMedPubMedCentral
512.
Zurück zum Zitat Sinha D, Roy S, Saha P, Chatterjee N, Bishayee AJC. Trends in research on exosomes in cancer progression and anticancer therapy. Cancers. 2021;13(2):326.PubMedPubMedCentral Sinha D, Roy S, Saha P, Chatterjee N, Bishayee AJC. Trends in research on exosomes in cancer progression and anticancer therapy. Cancers. 2021;13(2):326.PubMedPubMedCentral
Metadaten
Titel
Emerging role of exosomes in cancer progression and tumor microenvironment remodeling
verfasst von
Mahshid Deldar Abad Paskeh
Maliheh Entezari
Sepideh Mirzaei
Amirhossein Zabolian
Hossein Saleki
Mohamad Javad Naghdi
Sina Sabet
Mohammad Amin Khoshbakht
Mehrdad Hashemi
Kiavash Hushmandi
Gautam Sethi
Ali Zarrabi
Alan Prem Kumar
Shing Cheng Tan
Marios Papadakis
Athanasios Alexiou
Md Asiful Islam
Ebrahim Mostafavi
Milad Ashrafizadeh
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2022
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-022-01305-4

Weitere Artikel der Ausgabe 1/2022

Journal of Hematology & Oncology 1/2022 Zur Ausgabe

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Das sind die führenden Symptome junger Darmkrebspatienten

Darmkrebserkrankungen in jüngeren Jahren sind ein zunehmendes Problem, das häufig längere Zeit übersehen wird, gerade weil die Patienten noch nicht alt sind. Welche Anzeichen Ärzte stutzig machen sollten, hat eine Metaanalyse herausgearbeitet.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.