Skip to main content

REVIEW article

Front. Oncol., 17 March 2022
Sec. Molecular and Cellular Oncology
This article is part of the Research Topic Reviews in Molecular and Cellular Oncology View all 47 articles

The Potential Role of Exosomes in the Treatment of Brain Tumors, Recent Updates and Advances

Zoufang Huang&#x;Zoufang Huang1†Shayan Keramat&#x;Shayan Keramat2†Mehrdad IzadiradMehrdad Izadirad3Zhe-Sheng Chen*Zhe-Sheng Chen4*Mohammad Soukhtanloo,*Mohammad Soukhtanloo5,6*
  • 1Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
  • 2Department of Hematology and Blood Bank, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
  • 3Department of Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
  • 4Department of Pharmaceutical Sciences, St John’s University, New York, NY, United States
  • 5Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
  • 6Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran

Exosomes are small endosomal derived membrane extracellular vesicles that contain cell-specific cargos such as lipid, protein, DNA, RNA, miRNA, long non-coding RNA, and some other cell components that are released into surrounding body fluids upon the fusion of multivesicular bodies (MVB) and the plasma membrane. Exosomes are a one-of-a-kind cell-to-cell communication mechanism that might pave the way for target therapy. The use of exosomes as a therapeutic potential in a variety of cancers has been and is still being investigated. One of the most important of these has been the use of exosomes in brain tumors therapy. Exosome contents play a crucial role in brain tumor progression by providing a favorable niche for tumor cell proliferation. Also, exosomes that are secreted from tumor cells, lead to the protection of tumor cells and their proliferation in the tumor environment by reducing the inflammatory response and suppression of the immune system. Although some treatment protocols such as surgery, chemotherapy, and radiotherapy are common in brain tumors, they do not result in complete remission in the treatment of some malignant and metastatic brain tumors. Identifying, targeting, and blocking exosomes involved in the progression of brain tumors could be a promising way to reduce brain tumor progression. On the other way, brain tumor therapy with effective therapeutic components such as siRNAs, mRNAs, proteins, could be developed. Finally, our research suggested that exosomes of nanoscale sizes might be a useful tool for crossing the blood-brain barrier and delivering effective content. However, further research is needed to fully comprehend the potential involvement of the exosome in brain tumor therapy protocols.

Introduction

Exosomes are small endosomal derived membrane microvesicles that contain cell-specific cargos such as lipid, protein, DNA, RNA, miRNA, long non-coding RNA, and some other cell components that are released into surrounding body fluids upon the fusion of multivesicular bodies (MVB) and the plasma membrane (1).

In the 1980s, researchers discovered the presence of some structure like a tiny bubble in the extracellular space. Initially, considered as cellular waste resulting from cell damage and has no significant impact on neighboring cells (2). Actually, with the advancement of technology and further studies, they found that it seems the cells in the neighborhood of each other released these bubble-like bodies that are now known as exosomes, to transfer messages between themselves. Gradually, it became clear that these messages were targeting cells farther away from their surroundings (3). Therefore, it is assumed that exosomes represent a novel mode of cell-to-cell communication, and this may show an important role of exosomes in many cellular interactions such as signal transduction and activation of some signaling pathways in recipient cells (4). The role of the vesicles is defined by exosomes composition include of death or survival or proliferation and differentiation, sharing of immune responses, immune modulators, antigen presentation, and some biological response (5).

Thus, exosomes could have a potential role in insights into target therapy. It has been indicated that the composition of exosomes can affect pharmacokinetic properties (6). Exosomes have the potential to aid in the prognosis and diagnosis of illnesses such as cancer, chronic inflammation, cardiovascular diseases, infections, and autoimmune, in addition to its therapeutic potential (7).

Because exosomes are released from many different cell types, including dendritic cells, macrophages, B cells, T cells, epithelial cells, platelets, mast cells, adipocytes, and fibroblasts, it is revealed that nervous system cells, including Schwann cells, astrocytes, and neurons also have a potential role to release exosomes (8, 9). Moreover, it has been demonstrated that exosomes are involved in the function of the nervous system, including the regulation of synaptic communication and nerve regeneration (10). Therefore, in recent times, exosomes in the nervous system are considered as a new bridge for intercellular communication that in addition to participation in normal neuronal physiology, also has an important role in the pathogenic event such as neurodegenerative disease as a pathogen transmitter or as therapeutic potential (11).

The use of exosomes as a therapeutic potential in many cancers has been and is still being investigated. One of the most important of these has been the use of exosomes in the treatment of brain tumors (12, 13).

Up to now, various treatments for brain tumors have been proposed, including surgery, radiotherapy, and chemotherapy. On the other hand, scientists have raised several concerns regarding many side effects and challenges in these types of treatments (14).

Maybe one of the most important challenges is the sensitivity of the brain tissue and the need for high precision in surgery, and on the other hand, the existence of a blood-brain barrier (BBB) to prevent drug treatments from entering the brain environment (15). However, due to the size of the exosome (30–150 nm), it is capable of easily crossing this barrier and pursuing therapeutic aims.

The relevance of these vesicles in brain malignancies, treatments, and pathophysiology will be discussed in this review (16).

Exosome Biogenesis

The endosomal system has an important role in the biogenesis of exosomes. Early endosomes mature into late endosomes and exosomes formed by inward budding of the multivesicular body (MVB) membrane. Intraluminal vesicles (ILVs) are formed by the invagination of late endosomal membranes within large MVBs (17). It has been demonstrated that the endosomal sorting complex required for transport (ESCRT) machinery is important in this process. ESCRT includes four different protein complexes numbered ESCRT‐0, ESCRT‐I, ESCRT‐II, and ESCRT‐III, inside of some associated proteins such as AAA ATPase Vps4, TSG101, and ALIX (18, 19). Each of these protein components has been shown to have specific functions. ESCRT‐0 has an important role in the internalization of ubiquitinated proteins and also, sequestration of these proteins to particular domains of the endosomal membrane. Subsequently, ESCRT-I and ESCRT-II promote the budding process, which will then combine with ESCRT-III, and the final stage of membrane invagination and separation with the total complex of ESCRT is completed. Finally, these processes lead to multivesicular body (MVB) formation (19) (Figure 1).

FIGURE 1
www.frontiersin.org

Figure 1 Early endosomes mature into late endosomes and exosomes are formed by inward budding of the multivesicular body (MVB) membrane. Intraluminal vesicles (ILVs) are formed by the Invagination of late endosomal membranes within large MVBs. It has been demonstrated that the endosomal sorting complex required for transport (ESCRT) machinery is important in this process.

Also, ALIX as an exosomal protein participates in the budding process and the corporation with syndecan has an important role in exosomal cargo selection. In addition, it has been shown that ALIX has an important role in changing the protein composition and also, secretion of exosomes (20). Also, it is demonstrated that ESCRT inhibition can lead to inhibiting the secretion of exosomes, also, in addition to exosome biogenesis ESCRT has another key role in cells’ biological function especially cytokinesis (18, 21). On the other, TSG101, ALIX, VPS4 proteins have been implicated that have a role in promoting the budding of exosome-like vesicles in T lymphocytes (22).

One of the most recent findings in the exosome biogenesis suggested an alternative pathway so-called “ESCRT-independent mechanisms”. It is demonstrated that this mechanism depends on raft-based microdomains and cargo loading involves lipids, sphingosine, hydrophobic modifications, and associated protein such as tetraspanins on rafted membranes (23). The presence of ceramide and sphingolipids molecules generates from ceramide by ceramidase and sphingosine kinase have an important role in the budding process of ILVs formation (24). Also, tetraspanins, which are proteins that organize membrane microdomains which are so-called tetraspanin‐enriched microdomains (TEMs), induce the ILVs formation in ESCRT‐independent mechanism by CD63 (a member of the tetraspanin family) function (23).

Phospholipase D2 (PLD2) is an important enzyme in the lipid modification process, has been shown to play an important role in exosome biogenesis as well as ILV formation. Phospholipase D2, by hydrolysis of phosphatidylcholine to phosphatidic acid (PA), induces a negative membrane curvature, which in turn leads to membrane invagination (25).

Another enzyme, called diacylglycerol kinase α (DGKα), that has a role in phosphate group insertion to the diacylglycerol (DAG), is also involved in the production of phosphatidic acid, which is participated in the release of exosomes (26).

Although, the biogenesis of exosomes has been introduced in two major pathways as ESCRT-dependent or ESCRT-independent mechanisms with different types of exosomes and different types of machinery such as ESCRT machinery, lipids, tetraspanins, PLD2, DGK, and other proteins (2026). But in fact, exosome biogenesis can be the consequence of these two mechanisms Collaboration together. However, although there are many studies related to exosome biogenesis, it still seems that this issue needs further investigation because there are many unknown Pathways.

Applications of Exosomes in Cancer Diagnosis and Treatment

The atmosphere surrounding the research field of exosomes in cancer has dramatically changed in recent years but the use of exosomes in cancer diagnosis or therapy was not impressive (27). However, studies are raising in this field and conducted research on exosomes have shown that exosomes might serve as a new instrument in cancer diagnosis and treatment.

The findings have been revealed that tumor-derived exosomes (TDEs) include components that are distinctive biomarkers and might be used to diagnose tumors. For example, it has been investigated that GPC1+-circulating exosomes increment is a diagnosis factor in patients with pancreatic ductal carcinoma (PDAC) and colorectal cancer (CRC) (28). Investigations in this field regarding lung cancer reveal that circulating exosomes carrying CD151, CD171, and tetraspanin 8 can be detected in lung cancer patients (29). These exosomes could serve as a distinguishing marker to discriminate lung cancer patients from non-lung cancer patients (29).

Besides plasma and serum, other sources such as urine, saliva, cerebrospinal fluid (CSF), and milk can be used for exosome isolation. Studies show that these sources are easy to use for purifying exosomes that have diagnostic applications (30). For example, urine-derived exosomes containing ITGA3 and ITGB1 have been shown to promote metastasis in lung cancer patients when compared to early stage patients (31).. As a result, the use of these exosomes may be effective in predicting metastasis in lung cancer patients (32).. Additionally, it has been shown that saliva-derived vesicles such as exosomes in head and neck carcinoma (HNC) displayed biomarkers like CD44 and CD95L (33). Since, saliva is a more accessible sample than another source, finding these saliva-derived exosomes may be the easiest and fastest way to HNC detection (33).

Interestingly, exosomes can transfer regulator elements such as RNA, DNA, or proteins that affect treatment response assessment. For example, it has been revealed that exosomes could establish chemoresistance in cancer cells, like HER-2 displaying exosomes could neutralize HER-2 antibody (Trastuzumab) and lead to chemoresistance in breast cancer (34). Recently, there has been an increasing interest in exosome-derived lncRNAs as factors in the development of tumor chemoresistance (35). Evidence suggests that lncRNAs transferred by exosomes may contribute to chemoresistance in other cells. For example, Han M et al. demonstrated that exosomes, such as EVs, contain the lncRNA, AFAP1-AS1, which causes trastuzumab resistance in breast cancer patients (36).

There are interesting pieces of evidence regarding exosome applicants in cancer therapy. It seems that the extracellular vesicles exosome can be a promising designable platform for transferring specific content such as drugs, proteins, and regulator RNAs due to their endogenous origin, stability, biocompatibility, and other unique features (37, 38). O’Brien and colleagues showed that exosomes filled by miR-134 could reduce cellular migration and invasion, and enhanced sensitivity to anti-Hsp90 drugs (39). Exosomes also can be enriched by chemotherapeutic drugs and target cancer cells. Bellavia D and colleagues have posited that imatinib or BCR-ABL siRNA-loaded exosomes will be able to target CML cells and unload their cargo to target cells. Forasmuch as IL-3R expression increases on CML cells, they have used this feature for CML cells targeting via producing IL3-Lamp2b expressing exosomes. They reported that these exosomes successfully target cancer cells and imatinib is delivered. As a result, cancer cell proliferation is reduced (40).

Exosomes in Brain Tumors

It has been found that exosomes play a key role in brain tumors that can be effective in tumor growth and progression (41). Exosomes secreted from tumor cells in the brain can create a small communication between a malignant cell and surrounding cells in different ways, such as the release of proteins, mRNAs, or other cellular components involved in cell malignancy (10, 41).

Currently, treatment of brain cancers has been a major challenge compared to cancers in other organs due to the presence of BBB and the complexity of the central nervous system (CNS) microenvironment (42). The therapeutic role of exosomes in various brain cancers such as glioblastoma, neuroblastoma, medulloblastoma, astrocytoma, gliosarcoma, and oligodendroglioma can be effective (15, 43). Although this effect can have different aspects, as it can be effective in the early diagnosis of brain cancer, inhibition of the spread of exosomes containing cancer cell information to other healthy cells, or invitro biogenesis of exosomes and its use to transfer the effective factors for treatment to the microenvironment of the brain (43).

To investigate the effect of exosomes in the treatment of brain cancers, the role of exosomes in the development of various brain cancers and their progression must first be studied in more detail.

Role of Exosome in Primary Brain Tumor

Glioblastoma

Glioblastoma (GBM) is the most aggressive one among tumors of glial origin that rapidly grow and spread into nearby brain tissue and forms from cells called astrocytes that support nerve cells. The survival of patients has been reported usually between 10–15 months (44). Currently, complete remission of GMB is impossible, and actually, treatments slow the progression of cancer and reduce symptoms (45).

Exosomes have an important role in the progression and development of GBM. This communication is between cancer stem cells, tumor cells, microglia, and parenchymal cells, and especially immune cells (46). Although it is similar to a hypothesis, the exosome traces in the glioma progression were detected by purifying the exosome in culture media containing murine glioma. Also, these findings indicated that glioma exosomes contained a variety of proteins involved in tumor progression and surface markers (47). The most important of them are heat shock proteins (HSPs) such as HSPB5, HSP 60, 70, and (48). This finding suggests that the presence of inflammatory cytokines, including interleukin (IL)-1, IL-6, and TNF-α in interaction with HSPs, increases its release, which may contribute to tumor cell immortality and disease progression (49).

In total, it has been shown that exosomes in solid tumors can help tumor progression by suppressing the immune system (49). Also, in GBM, exosomes secreted by tumor cells modulate the immune system reaction by inducing the presence of M2 macrophages around the tumor, thus helping the tumor to progress and escape from the immune system (50). In addition, Annexin A2 that plays a significant role in invasion, metastasis, angiogenesis, and proliferation is one of the important markers that has been found on the surface of exosomes in GBM (51). Despite Annexin A2, CD44 is also present on the surface of exosomes in GBM that plays as a receptor for hyaluronic acid and it is implicated that CD44 is a marker for cell motility, tumor growth, angiogenesis, and cancer stem cell (52, 53). It should be noted, one of the most important challenges in the treatment of GBM is drug resistance (54). Interestingly, It was found that the proteins involved in resistance to treatment that has been named the DNA-dependent protein kinase catalytic subunit (PRKDC), which is involved in the repair of double-strand DNA against radiotherapy, can be transported through exosomes, it is assumed that the inhibition of which can play an important role in the treatment of GBM (51).

Also, miRNA expression was detected in CSF and plasma of GBM patients. These exosomes were containing different miRNA with different activities and roles (55). Regardless of the role and activity of miRNA in GBM patients, their detection can be used as an effective diagnostic marker. In this regard, miR-221 and miR-21 have been evaluated as biomarkers in CSF of GBM patients (56, 57). As well, GBM patients which have received antitumor vaccines, miR-320 and miR-574-3p have been detected in the plasma and have a diagnostic role as biomarkers (43).

Neuroblastoma

Neuroblastoma (NB), as its name implies, is a malignancy involving nerve cells in the immature stages. NB is a malignant and very progressive cancer that is often detected in children under 5 years of age and with higher rates in infants or fetuses (58). It’s important to pay attention to this disease because it is the second most common malignancy in children and also one of the most common malignant cancers of the nervous system (59). NB usually originates in the adrenal glands but progresses rapidly throughout the body, such as the bones, abdomen, neck, chest, and even under the skin (58). Although the etiology of this cancer is not known exactly, the main cause of this disease is considered to be genetic mutations that can be acquired and sporadic or even in rare cases inherited from parents (60). According to the staging set by the International Neuroblastoma Staging System (INSS), it has been shown that in advanced stages, the NB deviates from the localized form and can progress rapidly to other tissues. This rapid progression similar to a metastatic process can be effectively correlated with the activity of exosomes (61).

Despite the GBM, there are few exosome studies in the field of NB, and the role of exosomes in the progression of NB remains unknown. The studies that have been done in this field so far are often in the field of treatment resistance and providing a suitable environment for tumor development (62). MYCN-amplified (a proto-oncogene that is associated with poor prognosis in NB) NB cells have been shown to play an important role in tumor progression and development by secreting some exosomal miRNAs and result in changing the tumor environment in favor of tumor growth (63).

Also, it has been demonstrated that exosomes with miR-155 which transferred from monocytes/macrophages to NB cells, and miRNA-21 which has been secreted from NB cells in around of tumor environment have a key role in resistance to chemotherapy, a process that occurs through miR-21/TLR8-NF-κB/exosomal miR-155/TERF1 signaling pathway (64, 65). Therefore, it can be said that inhibition of these exosomes maybe help the patients with NB to be cured and lead to a favorable prognosis in NB patients (62).

One of the most recent findings regarding exosomes in NB is the study of exosomal hsa-miR199a-3p (66). It has been shown that upregulation of exosomal hsa-miR199a-3p can be associated with high-risk and poor prognosis of NB. Although hsa-miR199a-3p is generally found in other malignancies and has opposite effects in different cancers, in NB has a progressive effect and poor prognosis (66). The mechanism of the hsa-miR199a-3p function is such that it reduces the expression of an enzyme called NEDD4. This enzyme plays a very important role in suppressing tumor activity by interacting with Myc via ubiquitination and degradation of Myc protein and catalyzing PTEN mono-ubiquitination and regulating PTEN nuclear translocation (67). As a result, inhibition of exosomal hsa-miR199a-3p can play an effective role in improving NB. Also, detecting the increase in the level of exosomal hsa-miR199a-3p can be a biomarker to detect NB in ​​the early stages or to determine the prognosis (66, 67).

Recently, it has been demonstrated that a high level of exosomal miR-375 correlates with BM metastasis in NB patients (68). Therefore, exosomal miR-375 may be an important novel biomarker in detecting BM metastatic progression, and also may represent a novel potential target for NB patients with BM metastasis (68, 69).

Role of Exosomes in Metastasis to the Brain

To date, although many advances have been made in the treatment of various cancers, from chemotherapy to radiotherapy, immunotherapy, and surgical skills, the focus of these therapies has been on anti-tumor or anti-cancer activities (70). The missing puzzle of therapeutic protocols, on the other hand, maybe therapy against anti-metastatic activities. In recent years, many studies have been conducted on the role of exosomes in tumor metastasis (71).

Metastatic brain tumors are one of the most common brain tumors that can be secondary to a variety of tumors in different tissues of the body. Therefore, these tumors are also called secondary tumors (72). In addition, metastatic brain tumors are important because they grow rapidly and have more destructive effects on brain tissue than primary tumors. On the other hand, multiple metastatic tumors may involve the brain at the same time (73). Regarding the risk of metastatic brain tumors, it can be said that most malignancies and systemic tumors have a risk of metastasis to the brain, but what is more common is that tumors of the breast, lung, renal, and colon increase the risk of metastasis to the brain (74, 75). It has been shown that more than half of people with metastatic brain tumors have a history of non-small cell lung cancer (76, 77). Also, this risk is about 30% for patients with a history of breast cancer (74). Exosomal microRNAs have a crucial role in the development of malignancies, as previously stated. As a result, more research into these topics might be worthwhile (78). For example, High expression of miR-451a and miR-4257 have been shown to be closely related to non-small cell lung cancer tumor progression and poor prognosis (79). miR-21 is also associated with the recurrence and progression of lung cancer (79, 80). Actually, high levels of exosomal miR-23a that has angiogenesis activity, are found in non-small cell lung cancer patients which can also be effective in metastasis to the brain (81, 82).. It has been demonstrated that cancer cells in breast cancers secrete a large amount of exosomal miR-122 (83). By affecting normal cells in pre-metastatic sites, miR-122 stops glucose uptake during this process, thus providing the energy needed for the unbridled proliferation of tumor cells and the development of metastasis (84). Therefore, its inhibition can play an essential role in preventing the metastasis and progression of tumor cells (84, 85). The cells identified as receptors for miR-122 were fibroblasts, brain astrocytes, and neurons, indicating the importance of exosomal miR-122 role in metastasis to the brain (85, 86).

Also, miR-105 is secreted from Breast cancer cells and affects endothelial cells. This effect can be such that it leads to the destruction of endothelial barriers, which enhances the metastasis process (77). It has also been shown that the destruction of endothelial barriers leads to the destruction of the BBB, indicating the effective role of exosomal miR-105 in metastasis to the brain (87).

One of the most important microRNAs that recently have been identified and discussed in the study of metastasis between breast cancer cells and other tissues, especially the brains, is miR-181c (88). The direct effect of miR-181c on brain metastasis is due to the destruction of the BBB (81). The mechanism of degradation by miR-181c is by promoting the pathway of cofilin protein activation and finally by destroying actin filaments by it (81) (Figure 2).

FIGURE 2
www.frontiersin.org

Figure 2 (A) miR-181c that secreted from metastatic breast cancer cells could transfer by exosomes to epithelial cells and suppresses the expression of PDPK1. (B) Then, it results in activated cofilin which disassembles actin filaments and makes the BBB permeable. Following, cancer cells could cross the BBB.

In total, if we want to investigate other roles of exosomal contents (such as cytokines, enzymes, chemokines, etc.) in metastasis to the brain, we could consider their effective role in changing normal fibroblasts to tumor-associated fibroblasts and creating a suitable niche for the development of tumor cells (82, 89). Also, reducing the level of inflammation and escaping from the immune system, protects tumor cells and their proliferation in the tumor environment (90).

Further researches could yield novel insights into the molecular mechanisms of metastasis and the development of advanced therapeutic strategies to prevent the formation of brain metastasis.

Conclusion

Brain tumors are important because, in addition to physical complications, they also have cognitive complications (91). To date, the treatment of brain tumors in medicine has been associated with many challenges. Surgery is still considered the most important step in treating brain tumors, but brain surgery is one of the most difficult types of surgery (92). In addition, the long recovery period after brain surgery and its complications should not be underestimated (92). Chemotherapy and radiotherapy are also prescribed for malignant tumors in the next step. Although Chemotherapy and radiotherapy have gradually become more effective with advances in medical science, it still does not bring complete remission in the treatment of some malignant and metastatic brain tumors. In addition, complications of Chemotherapy are also undeniable (93, 94).

In molecular studies of the pathogenesis of brain tumors, researchers focus on genetics and epigenetics, while cellular communications with each other and their surroundings play a very important role in tumor progression (95). Intercellular communications are formed by membrane extracellular vesicles, exosomes typically play a critical role in the transmission of biological processes between tumor cells and other cells and tissues (4, 96).

As mentioned, exosomes play an important role in the progression of brain tumors, and one of the most important roles of exosomes in brain tumors is in the metastasis of tumors to the brain, for example, by transmitting the contents, or in other words, the signal for tumor growth and progression from tumor cells in other tissues to the brain (97, 98). The use of exosomes in the treatment of brain tumors could be in two ways: 1) Identifying, targeting, and inhibiting exosomes involved in the progression of brain tumors. 2) Treatment of brain tumors by exosomes carrying effective contents such as mRNAs, proteins, and lipids. Although not routinely used today and treatments such as whole-brain radiation therapy, surgery, stereotactic radiosurgery, chemotherapy, anticonvulsant drugs, and steroids are the main guidelines for treating patients with brain tumors (99).

The most prominent feature of exosomes, their size, can be used to indicate the applicability of exosomes for the treatment of brain tumors. The nanometer size of exosomes could be a good tool for crossing the BBB and delivering effective contents. However, further studies are needed to properly understand the potential role of the exosome in treatment protocols. Also, the exosome has been shown to be a safe way to increase the stability of its contents. Therefore, in addition to the ability of the exosome to cross the BBB, the use of the exosome as a suitable coating in protecting the useful contents for the treatment of brain tumors and their delivery can be effective (100).

Author Contributions

ZH and SK prepared the backbone of the manuscript. ZH, SK, and MI wrote the original draft of the manuscript. Z-SC and MS refined the manuscript. Z-SC critically revised the manuscript. ZH and Z-SC supported the project. All authors approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Zhang JX, Tang WS. Output Feedback H∞ Control for Uncertain Piecewise Linear Systems. J Adv Res Dyn Control Syst (2008) 2008:14. doi: 10.1007/s10883-007-9030-8

CrossRef Full Text | Google Scholar

2. Doyle LM, Wang MZ. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells (2019) 8:727. doi: 10.3390/cells8070727

CrossRef Full Text | Google Scholar

3. Zhang H-G, Grizzle WE. Exosomes: A Novel Pathway of Local and Distant Intercellular Communication That Facilitates the Growth and Metastasis of Neoplastic Lesions. Am J Pathol (2014) 184(1):28–41. doi: 10.1016/j.ajpath.2013.09.027

CrossRef Full Text | Google Scholar

4. Corrado C, Raimondo S, Chiesi A, Ciccia F, De Leo G, Alessandro R. Exosomes as Intercellular Signaling Organelles Involved in Health and Disease: Basic Science and Clinical Applications. Int J Mol Sci (2013) 14(3):5338–66. doi: 10.3390/ijms14035338

CrossRef Full Text | Google Scholar

5. Li Q, Wang H, Peng H, Huyan T, Cacalano NA. Exosomes: Versatile Nano Mediators of Immune Regulation. Cancers (Basel) (2019) 11(10):1557. doi: 10.3390/cancers11101557

CrossRef Full Text | Google Scholar

6. Kim DH, Kothandan VK, Kim HW, Kim KS, Kim JY, Cho HJ, et al. Noninvasive Assessment of Exosome Pharmacokinetics In Vivo: A Review. Pharmaceutics (2019) 11(12):649. doi: 10.3390/pharmaceutics11120649

CrossRef Full Text | Google Scholar

7. Kim H, Jang H, Cho H, Choi J, Hwang KY, Choi Y, et al. Recent Advances in Exosome-Based Drug Delivery for Cancer Therapy. Cancers (Basel) (2021) 13(17):4435. doi: 10.3390/cancers13174435

CrossRef Full Text | Google Scholar

8. Golchin A, Hosseinzadeh S, Ardeshirylajimi A. The Exosomes Released From Different Cell Types and Their Effects in Wound Healing. J Cell Biochem (2018) 119(7):5043–52. doi: 10.1002/jcb.26706

CrossRef Full Text | Google Scholar

9. Budnik V, Ruiz-Canada C, Wendler F. Extracell Vesicles Round Off Communication in the Nervous System. Nat Rev Neurosci (2016) 17(3):160–72. doi: 10.1038/nrn.2015.29

CrossRef Full Text | Google Scholar

10. Liu W, Bai X, Zhang A, Huang J, Xu S, Zhang J. Role of Exosomes in Central Nervous System Diseases. Front Mol Neurosci (2019) 12:240. doi: 10.3389/fnmol.2019.00240

CrossRef Full Text | Google Scholar

11. Pascual M, Ibáñez F, Guerri C. Exosomes as Mediators of Neuron-Glia Communication in Neuroinflammation. Neural Regen Res 15(5):796–801. doi: 10.4103/1673-5374.268893

CrossRef Full Text | Google Scholar

12. Li Z, Yang H, Ye L, Quan R, Chen M. Role of Exosomal miRNAs in Brain Metastasis Affected by Radiotherapy. Transl Neurosci (2021) 12(1):127–37. doi: 10.1515/tnsci-2020-0163

CrossRef Full Text | Google Scholar

13. Mirzaei H, Sahebkar A, Jaafari MR, Goodarzi M, Mirzaei HR. Diagnostic and Therapeutic Potential of Exosomes in Cancer: The Beginning of a New Tale? J Cell Physiol (2017) 232(12):3251–60. doi: 10.1002/jcp.25739

CrossRef Full Text | Google Scholar

14. Graham CA, Cloughesy TF. Brain Tumor Treatment: Chemotherapy and Other New Developments. Semin Oncol Nurs (2004) 29(4):260–72. doi: 10.1016/S0749-2081(04)00090-7

CrossRef Full Text | Google Scholar

15. Bhowmik A, Khan R, Ghosh MK. Blood Brain Barrier: A Challenge for Effectual Therapy of Brain Tumors. BioMed Res Int (2015) 2015:320941. doi: 10.1155/2015/320941

CrossRef Full Text | Google Scholar

16. Elliott RO, He M. Unlocking the Power of Exosomes for Crossing Biological Barriers in Drug Delivery. Pharmaceutics (2021) 13(1):122. doi: 10.3390/pharmaceutics13010122

CrossRef Full Text | Google Scholar

17. Hessvik NP, Llorente A. Current Knowledge on Exosome Biogenesis and Release. Cell Mol Life Sci (2018) 75(2):193–208. doi: 10.1007/s00018-017-2595-9

CrossRef Full Text | Google Scholar

18. Vietri M, Radulovic M, Stenmark H. The Many Functions of ESCRTs. Nat Rev Mol Cell Biol (2020) 21(1):25–42. doi: 10.1038/s41580-019-0177-4

CrossRef Full Text | Google Scholar

19. Juan T, Fürthauer M. Biogenesis and Function of ESCRT-Dependent Extracellular Vesicles. Semin Cell Dev Biol (2018) 74:66–77. doi: 10.1016/j.semcdb.2017.08.022

CrossRef Full Text | Google Scholar

20. Fares J, Kashyap R, Zimmermann P. Syntenin: Key Player in Cancer Exosome Biogenesis and Uptake? Cell Adh Migr (2017) 11(2):124–6. doi: 10.1080/19336918.2016.1225632

CrossRef Full Text | Google Scholar

21. Catalano M, O’Driscoll L. Inhibiting Extracellular Vesicles Formation and Release: A Review of EV Inhibitors. J Extracell Vesicles (2020) 9(1):1703244. doi: 10.1080/20013078.2019.1703244

CrossRef Full Text | Google Scholar

22. Ventimiglia LN, Alonso MA. Biogenesis and Function of T Cell-Derived Exosomes. Front Cell Dev Biol (2016) 4:84. doi: 10.3389/fcell.2016.00084

CrossRef Full Text | Google Scholar

23. Kapustin AN, Chatrou MLL, Drozdov I, Zheng Y, Davidson SM, Soong D, et al. Vascular Smooth Muscle Cell Calcification is Mediated by Regulated Exosome Secretion. Circ Res (2015) 116(8):1312–23. doi: 10.1161/CIRCRESAHA.116.305012

CrossRef Full Text | Google Scholar

24. Zhou K, Blom T. Trafficking and Functions of Bioactive Sphingolipids: Lessons From Cells and Model Membranes. Lipid Insights (2015) 8:LPI–S31615. doi: 10.4137/LPI.S31615

CrossRef Full Text | Google Scholar

25. Egea-Jimenez AL, Zimmermann P. Thematic Review Series: Exosomes and Microvesicles: Lipids as Key Components of Their Biogenesis and Functions: Phospholipase D and Phosphatidic Acid in the Biogenesis and Cargo Loading of Extracellular Vesicles. J Lipid Res (2018) 59(9):1554. doi: 10.1194/jlr.R083964

CrossRef Full Text | Google Scholar

26. Bertin G, Averbeck D. Cadmium: Cellular Effects, Modifications of Biomolecules, Modulation of DNA Repair and Genotoxic Consequences (a Review). Biochimie (2006) 88(11):1549–59. doi: 10.1016/j.biochi.2006.10.001

CrossRef Full Text | Google Scholar

27. Zhang H-G, Grizzle WE. Exosomes and Cancer: A Newly Described Pathway of Immune Suppression. Clin Cancer Res (2011) 17(5):959–64. doi: 10.1158/1078-0432.CCR-10-1489

CrossRef Full Text | Google Scholar

28. Frampton AE, Prado MM, López-Jiménez E, Fajardo-Puerta AB, Jawad ZAR, Lawton P, et al. Glypican-1 is Enriched in Circulating-Exosomes in Pancreatic Cancer and Correlates With Tumor Burden. Oncotarget (2018) 9(27):19006–13. doi: 10.18632/oncotarget.24873

CrossRef Full Text | Google Scholar

29. Sandfeld-Paulsen B, Jakobsen KR, Bæk R, Folkersen BH, Rasmussen TR, Meldgaard P, et al. Exosomal Proteins as Diagnostic Biomarkers in Lung Cancer. J Thorac Oncol (2016) 11(10):1701–10. doi: 10.1016/j.jtho.2016.05.034

CrossRef Full Text | Google Scholar

30. Soares Martins T, Catita J, Martins Rosa I, AB da Cruz e Silva O, Henriques AG. Exosome Isolation From Distinct Biofluids Using Precipitation and Column-Based Approaches. PloS One (2018) 13(6):e0198820. doi: 10.1371/journal.pone.0198820

CrossRef Full Text | Google Scholar

31. Zhou B, Xu K, Zheng X, Chen T, Wang J, Song Y, et al. Application of Exosomes as Liquid Biopsy in Clinical Diagnosis. Signal Transduct Target Ther (2020) 5(1):1–14. doi: 10.1038/s41392-020-00258-9

CrossRef Full Text | Google Scholar

32. Wang N, Song X, Liu L, Niu L, Wang X, Song X, et al. Circulating Exosomes Contain Protein Biomarkers of Metastatic non-Small-Cell Lung Cancer. Cancer Sci (2018) 109(5):1701–9. doi: 10.1111/cas.13581

CrossRef Full Text | Google Scholar

33. Principe S, Hui AB, Bruce J, Sinha A, Liu F, Kislinger T. Tumor-Derived Exosomes and Microvesicles in Head and Neck Cancer: Implications for Tumor Biology and Biomarker Discovery. Proteomics (2013) 13(10–11):1608–23. doi: 10.1002/pmic.201200533

CrossRef Full Text | Google Scholar

34. Dong X, Bai X, Ni J, Zhang H, Duan W, Graham P, et al. Exosomes and Breast Cancer Drug Resistance. Cell Death Dis (2020) 11(11):1–14. doi: 10.1038/s41419-020-03189-z

CrossRef Full Text | Google Scholar

35. Izadirad M, Jafari L, James AR, Unfried JP, Wu Z-X, Chen Z-S. Long Noncoding RNAs Have Pivotal Roles in Chemoresistance of Acute Myeloid Leukemia. Drug Discov Today (2021) 26(7):1735–43. doi: 10.1016/j.drudis.2021.03.017

CrossRef Full Text | Google Scholar

36. Han M, Gu Y, Lu P, Li J, Cao H, Li X, et al. Exosome-Mediated lncRNA AFAP1-AS1 Promotes Trastuzumab Resistance Through Binding With AUF1 and Activating ERBB2 Translation. Mol Cancer (2020) 19(1):1–18. doi: 10.1186/s12943-020-1145-5

CrossRef Full Text | Google Scholar

37. Johnsen KB, Gudbergsson JM, Skov MN, Pilgaard L, Moos T, Duroux M. A Comprehensive Overview of Exosomes as Drug Delivery Vehicles—Endogenous Nanocarriers for Targeted Cancer Therapy. Biochim Biophys Acta (BBA)-Reviews Cancer (2014) 1846(1):75–87. doi: 10.1016/j.bbcan.2014.04.005

CrossRef Full Text | Google Scholar

38. He C, Zheng S, Luo Y, Wang B. Exosome Theranostics: Biology and Translational Medicine. Theranostics (2018) 8(1):237. doi: 10.7150/thno.21945

CrossRef Full Text | Google Scholar

39. O’Brien K, Lowry MC, Corcoran C, Martinez VG, Daly M, Rani S, et al. miR-134 in Extracellular Vesicles Reduces Triple-Negative Breast Cancer Aggression and Increases Drug Sensitivity. Oncotarget (2015) 6(32):32774. doi: 10.18632/oncotarget.5192

CrossRef Full Text | Google Scholar

40. Bellavia D, Raimondo S, Calabrese G, Forte S, Cristaldi M, Memeo L, et al. T H E R a N O s T I C s Interleukin 3- Receptor Targeted Exosomes Inhibit In Vitro and In Vivo Chronic Myelogenous Leukemia Cell Growth. Theranostics (2017) 7(5):1333–45. doi: 10.7150/thno.17092

CrossRef Full Text | Google Scholar

41. Osaki M, Okada F. Exosomes and Their Role in Cancer Progression. Yonago Acta Med (2019) 62(2):182–90. doi: 10.33160/yam.2019.06.002

CrossRef Full Text | Google Scholar

42. Wang Z, Sun H, Sebastian Yakisich J. Overcoming the Blood-Brain Barrier for Chemotherapy: Limitations, Challenges and Rising Problems. Anti-Cancer Agents Med Chem (Formerly Curr Med Chem Agents) (2014) 14(8):1085–93. doi: 10.2174/18715206113139990029

CrossRef Full Text | Google Scholar

43. Ciregia F, Urbani A, Palmisano G. Extracellular Vesicles in Brain Tumors and Neurodegenerative Diseases. Front Mol Neurosci (2017) 10:276. doi: 10.3389/fnmol.2017.00276

CrossRef Full Text | Google Scholar

44. Zaman M, Fatima N, Zaman A, Sajid M, Zaman U, Zaman S. Diagnostic Challenges in Prostate Cancer and 68Ga-PSMA PET Imaging: A Game Changer? Asian Pac J Cancer Prev APJCP (2017) 18(10):2625–8. doi: 10.22034/APJCP.2017.18.10.2625

CrossRef Full Text | Google Scholar

45. Burster T, Traut R, Yermekkyzy Z, Mayer K, Westhoff M-A, Bischof J, et al. Critical View of Novel Treatment Strategies for Glioblastoma: Failure and Success of Resistance Mechanisms by Glioblastoma Cells. Front Cell Dev Biol (2021) 2290. doi: 10.3389/fcell.2021.695325

CrossRef Full Text | Google Scholar

46. Chen Y, Jin Y, Wu N. Role of Tumor-Derived Extracellular Vesicles in Glioblastoma. Cells (2021) 10(3):512. doi: 10.3390/cells10030512

CrossRef Full Text | Google Scholar

47. Bălașa A, Șerban G, Chinezu R, Hurghiș C, Tămaș F, Manu D. The Involvement of Exosomes in Glioblastoma Development, Diagnosis, Prognosis, and Treatment. Brain Sci (2020) 10(8):553. doi: 10.3390/brainsci10080553

CrossRef Full Text | Google Scholar

48. Graziano F, Rappa F, Gammazza M, Logozzi M, Fais S, Maugeri R, et al. Exosomal Chaperones and miRNAs in Gliomagenesis: State-Of-Art and Theranostics Perspectives. Int J Mol Sci (2018) 19(9):2626. doi: 10.3390/ijms19092626

CrossRef Full Text | Google Scholar

49. Whiteside TL. The Effect of Tumor-Derived Exosomes on Immune Regulation and Cancer Immunotherapy. Futur Oncol (2017) 13(28):2583–92. doi: 10.2217/fon-2017-0343

CrossRef Full Text | Google Scholar

50. Gabrusiewicz K, Li X, Wei J, Hashimoto Y, Marisetty AL, Ott M. Glioblastoma Stem Cell-Derived Exosomes Induce M2 Macrophages and PDL1 Expression on Human Monocytes. Oncoimmunology (2018) 7(4):e1412909. doi: 10.1080/2162402X.2017.1412909

CrossRef Full Text | Google Scholar

51. 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. doi: 10.3390/biomedicines8070216

CrossRef Full Text | Google Scholar

52. Szatanek R, Baj-Krzyworzeka M. CD44 and Tumor-Derived Extracellular Vesicles (TEVs). Possible Gateway to Cancer Metastasis. Int J Mol Sci (2021) 22(3):1463. doi: 10.3390/ijms22031463

CrossRef Full Text | Google Scholar

53. Chen C, Zhao S, Karnad A, Freeman JW. The Biology and Role of CD44 in Cancer Progression: Therapeutic Implications. J Hematol Oncol (2018) 11(1):1–23. doi: 10.1186/s13045-018-0605-5

CrossRef Full Text | Google Scholar

54. Shi J, Zhang Y, Yao B, Sun P, Hao Y, Piao H, et al. Role of Exosomes in the Progression, Diagnosis, and Treatment of Gliomas. Med Sci Monit Int Med J Exp Clin Res (2020) 26:e924023–1. doi: 10.12659/MSM.924023

CrossRef Full Text | Google Scholar

55. Xia X, Wang Y, Huang Y, Zhang H, Lu H, Zheng JC. Exosomal miRNAs in Central Nervous System Diseases: Biomarkers, Pathological Mediators, Protective Factors and Therapeutic Agents. Prog Neurobiol (2019) 183:101694. doi: 10.1016/j.pneurobio.2019.101694

CrossRef Full Text | Google Scholar

56. Areeb Z, Stylli SS, Koldej R, Ritchie DS, Siegal T, Morokoff AP, et al. MicroRNA as Potential Biomarkers in Glioblastoma. J Neurooncol (2015) 125(2):237–48. doi: 10.1007/s11060-015-1912-0

CrossRef Full Text | Google Scholar

57. Jiang G, Mu J, Liu X, Peng X, Zhong F, Yuan W, et al. Prognostic Value of miR-21 in Gliomas: Comprehensive Study Based on Meta-Analysis and TCGA Dataset Validation. Sci Rep (2020) 10(1):1–10. doi: 10.1038/s41598-020-61155-3

CrossRef Full Text | Google Scholar

58. Colon NC, Chung DH. Neuroblastoma. Adv Pediatr (2011) 58(1):297–311. doi: 10.1016/j.yapd.2011.03.011

CrossRef Full Text | Google Scholar

59. Maris JM. Recent Advances in Neuroblastoma. N Engl J Med (2010) 362(23):2202–11. doi: 10.1056/NEJMra0804577

CrossRef Full Text | Google Scholar

60. Cao Y, Jin Y, Yu J, Wang J, Yan J, Zhao Q. Research Progress of Neuroblastoma Related Gene Variations. Oncotarget (2017) 8(11):18444. doi: 10.18632/oncotarget.14408

CrossRef Full Text | Google Scholar

61. Sokol E, Desai AV. The Evolution of Risk Classification for Neuroblastoma. Children (2019) 6(2):27. doi: 10.3390/children6020027

CrossRef Full Text | Google Scholar

62. Richard H, Pokhrel A, Chava S, Pathania A, Katta SS, Challagundla KB. Exosomes: Novel Players of Therapy Resistance in Neuroblastoma. Adv Exp Med Biol (2020) 1277:75–85. doi: 10.1007/978-3-030-50224-9_5

CrossRef Full Text | Google Scholar

63. Haug BH, Hald ØH, Utnes P, Roth SA, Løkke C, Flaegstad T, et al. Exosome-Like Extracellular Vesicles From MYCN-Amplified Neuroblastoma Cells Contain Oncogenic miRNAs. Anticancer Res (2015) 35(5):2521–30.

Google Scholar

64. Hu W, Liu C, Bi Z-Y, Zhou Q, Zhang H, Li L-L, et al. Comprehensive Landscape of Extracellular Vesicle-Derived RNAs in Cancer Initiation, Progression, Metastasis and Cancer Immunology. Mol Cancer (2020) 19:1–23. doi: 10.1186/s12943-020-01199-1

CrossRef Full Text | Google Scholar

65. Challagundla KB, Wise PM, Neviani P, Chava H, Murtadha M, Xu T, et al. Exosome-Mediated Transfer of microRNAs Within the Tumor Microenvironment and Neuroblastoma Resistance to Chemotherapy. J Natl Cancer Inst (2015) 107(7):djv135. doi: 10.1093/jnci/djv135

CrossRef Full Text | Google Scholar

66. Ma J. Exosomal Hsa-Mir199a-3p Promotes Proliferation and Migration in Neuroblastoma. Front Oncol (2019) 9:459. doi: 10.3389/fonc.2019.00459

CrossRef Full Text | Google Scholar

67. Huang X, Chen J, Cao W, Yang L, Chen Q, He J, et al. The Many Substrates and Functions of NEDD4-1. Cell Death Dis (2019) 10(12):1–12. doi: 10.1038/s41419-019-2142-8

CrossRef Full Text | Google Scholar

68. Colletti M, Tomao L, Galardi A, Paolini A, Di Paolo V, De Stefanis C, et al. Neuroblastoma-Secreted Exosomes Carrying miR-375 Promote Osteogenic Differentiation of Bone-Marrow Mesenchymal Stromal Cells. J Extracell Vesicles (2020) 9(1):1774144. doi: 10.1080/20013078.2020.1774144

CrossRef Full Text | Google Scholar

69. Ding F, Liu J, Zhang X. microRNA-375 Released From Extracellular Vesicles of Bone Marrow Mesenchymal Stem Cells Exerts Anti-Oncogenic Effects Against Cervical Cancer. Stem Cell Res Ther (2020) 11(1):1–16. doi: 10.1186/s13287-020-01908-z

CrossRef Full Text | Google Scholar

70. Wang Y, Deng W, Li N, Neri S, Sharma A, Jiang W, et al. Combining Immunotherapy and Radiotherapy for Cancer Treatment: Current Challenges and Future Directions. Front Pharmacol (2018) 9:185. doi: 10.3389/fphar.2018.00185

CrossRef Full Text | Google Scholar

71. Steinbichler TB, Dudás J, Riechelmann H, Skvortsova II. The Role of Exosomes in Cancer Metastasis. Semin Cancer Biol (2017) 44:170–81. doi: 10.1016/j.semcancer.2017.02.006

CrossRef Full Text | Google Scholar

72. Boire A, Brastianos PK, Garzia L, Valiente M. Brain Metastasis. Nat Rev Cancer (2020) 20(1):4–11. doi: 10.1038/s41568-019-0220-y

CrossRef Full Text | Google Scholar

73. Mendez JS, DeAngelis LM. Metastatic Complications of Cancer Involving the Central and Peripheral Nervous Systems. Neurol Clin (2018) 36(3):579–98. doi: 10.1016/j.ncl.2018.04.011

CrossRef Full Text | Google Scholar

74. Saunus JM, Momeny M, Simpson PT, Lakhani SR, Da Silva L. Molecular Aspects of Breast Cancer Metastasis to the Brain. Genet Res Int (2011) 2011:219189. doi: 10.4061/2011/219189

CrossRef Full Text | Google Scholar

75. Mongan JP, Fadul CE, Cole BF, Zaki BI, Suriawinata AA, Ripple GH, et al. Brain Metastases From Colorectal Cancer: Risk Factors, Incidence, and the Possible Role of Chemokines. Clin Colorectal Cancer (2009) 8(2):100–5. doi: 10.3816/CCC.2009.n.016

CrossRef Full Text | Google Scholar

76. Jiang C, Chen X, Alattar M, Wei J, Liu H. MicroRNAs in Tumorigenesis, Metastasis, Diagnosis and Prognosis of Gastric Cancer. Cancer Gene Ther (2015) 22(6):291–301. doi: 10.1038/cgt.2015.19

CrossRef Full Text | Google Scholar

77. Li H-Y, Liang J-L, Kuo Y-L, Lee H-H, Calkins MJ, Chang H-T, et al. miR-105/93-3p Promotes Chemoresistance and Circulating miR-105/93-3p Acts as a Diagnostic Biomarker for Triple Negative Breast Cancer. Breast Cancer Res (2017) 19(1):1–14. doi: 10.1186/s13058-017-0918-2

CrossRef Full Text | Google Scholar

78. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, et al. Cancer-Secreted miR-105 Destroys Vascular Endothelial Barriers to Promote Metastasis. Cancer Cell (2014) 25(4):501–15. doi: 10.1016/j.ccr.2014.03.007

CrossRef Full Text | Google Scholar

79. Liu S, Zhan Y, Luo J, Feng J, Lu J, Zheng H, et al. Roles of Exosomes in the Carcinogenesis and Clinical Therapy of non-Small Cell Lung Cancer. BioMed Pharmacother (2019) 111:338–46. doi: 10.1016/j.biopha.2018.12.088

CrossRef Full Text | Google Scholar

80. Bai H, Wu S. miR-451: A Novel Biomarker and Potential Therapeutic Target for Cancer. Onco Targets Ther (2019) 12:11069. doi: 10.2147/OTT.S230963

CrossRef Full Text | Google Scholar

81. Tominaga N, Kosaka N, Ono M, Katsuda T, Yoshioka Y, Tamura K, et al. Brain Metastatic Cancer Cells Release microRNA-181c-Containing Extracellular Vesicles Capable of Destructing Blood–Brain Barrier. Nat Commun (2015) 6(1):1–12. doi: 10.1038/ncomms7716

CrossRef Full Text | Google Scholar

82. Ping Q, Yan R, Cheng X, Wang W, Zhong Y, Hou Z, et al. Cancer-Associated Fibroblasts: Overview, Progress, Challenges, and Directions. Cancer Gene Ther (2021) 28:984–99. doi: 10.1038/s41417-021-00318-4

CrossRef Full Text | Google Scholar

83. Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-Cancer-Secreted miR-122 Reprograms Glucose Metabolism in Premetastatic Niche to Promote Metastasis. Nat Cell Biol (2015) 17(2):183–94. doi: 10.1038/ncb3094

CrossRef Full Text | Google Scholar

84. Zhang Y, Huang H, Zhang Y, Liao N. Combined Detection of Serum MiR-221-3p and MiR-122-5p Expression in Diagnosis and Prognosis of Gastric Cancer. J Gastric Cancer (2019) 19(3):315–28. doi: 10.5230/jgc.2019.19.e28

CrossRef Full Text | Google Scholar

85. Morad G, Moses MA. Brainwashed by Extracellular Vesicles: The Role of Extracellular Vesicles in Primary and Metastatic Brain Tumour Microenvironment. J Extracell Vesicles (2019) 8(1):1627164. doi: 10.1080/20013078.2019.1627164

CrossRef Full Text | Google Scholar

86. Malone K, Tsirka SE. Breast-To-Brain Metastasis: A Focus on the Pre-Metastatic Niche. J Cancer Metastasis Treat (2021) 7:40. doi: 10.20517/2394-4722.2021.37

CrossRef Full Text | Google Scholar

87. Sereno M, Videira M, Wilhelm I, Krizbai IA, Brito MA. miRNAs in Health and Disease: A Focus on the Breast Cancer Metastatic Cascade Towards the Brain. Cells (2020) 9(8):1790. doi: 10.3390/cells9081790

CrossRef Full Text | Google Scholar

88. Song H, Zhang X, Chen R, Miao J, Wang L, Cui L, et al. Cortical Neuron-Derived Exosomal MicroRNA-181c-3p Inhibits Neuroinflammation by Downregulating CXCL1 in Astrocytes of a Rat Model With Ischemic Brain Injury. Neuroimmunomodulation (2019) 26(5):217–33. doi: 10.1159/000502694

CrossRef Full Text | Google Scholar

89. Wang J, Guan X, Zhang Y, Ge S, Zhang L, Li H, et al. Exosomal miR-27a Derived From Gastric Cancer Cells Regulates the Transformation of Fibroblasts Into Cancer-Associated Fibroblasts. Cell Physiol Biochem (2018) 49(3):869–83. doi: 10.1159/000493218

CrossRef Full Text | Google Scholar

90. Tan S, Xia L, Yi P, Han Y, Tang L, Pan Q, et al. Exosomal miRNAs in Tumor Microenvironment. J Exp Clin Cancer Res (2020) 39(1):1–15. doi: 10.1186/s13046-020-01570-6

CrossRef Full Text | Google Scholar

91. Coomans MB, van der Linden SD, Gehring K, Taphoorn MJB. Treatment of Cognitive Deficits in Brain Tumour Patients: Current Status and Future Directions. Curr Opin Oncol (2019) 31(6):540. doi: 10.1097/CCO.0000000000000581

CrossRef Full Text | Google Scholar

92. D’Andrea G, Palombi L, Minniti G, Pesce A, Marchetti P. Brain Metastases: Surgical Treatment and Overall Survival. World Neurosurg (2017) 97:169–77. doi: 10.1016/j.wneu.2016.09.054

CrossRef Full Text | Google Scholar

93. Béhin A, Delattre J-Y. Complications of Radiation Therapy on the Brain and Spinal Cord. In: Seminars in Neurology. Copyright© 2004. 333 Seventh Avenue, New: Thieme Medical Publishers, Inc. (2004). p. 405–17. doi: 10.1055/s-2004-861535

CrossRef Full Text | Google Scholar

94. Allen JC. Complications of Chemotherapy in Patients With Brain and Spinal Cord Tumors. Pediatr Neurosurg (1991) 17(4):218–24. doi: 10.1159/000120601

CrossRef Full Text | Google Scholar

95. Campos-Sanchez E, Martínez-Cano J, del Pino Molina L, López-Granados E, Cobaleda C. Epigenetic Deregulation in Human Primary Immunodeficiencies. Trends Immunol (2019) 40(1):49–65. doi: 10.1016/j.it.2018.11.005

CrossRef Full Text | Google Scholar

96. Gurunathan S, Kang M-H, Jeyaraj M, Qasim M, Kim J-H. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells (2019) 8(4):307. doi: 10.3390/cells8040307

CrossRef Full Text | Google Scholar

97. Solé C, Lawrie CH. MicroRNAs and Metastasis. Cancers (Basel) (2020) 12(1):96. doi: 10.3390/cancers12010096

CrossRef Full Text | Google Scholar

98. Dilsiz N. Role of Exosomes and Exosomal microRNAs in Cancer. Futur Sci OA (2020) 6(4):FSO465. doi: 10.2144/fsoa-2019-0116

CrossRef Full Text | Google Scholar

99. Ammirati M, Nahed BV, Andrews D, Chen CC, Olson JJ. Congress of Neurological Surgeons Systematic Review and Evidence-Based Guidelines on Treatment Options for Adults With Multiple Metastatic Brain Tumors. Neurosurgery (2019) 84(3):E180–2. doi: 10.1093/neuros/nyy548

CrossRef Full Text | Google Scholar

100. Akuma P, Okagu OD, Udenigwe CC. Naturally Occurring Exosome Vesicles as Potential Delivery Vehicle for Bioactive Compounds. Front Sustain Food Syst (2019) 3:23. doi: 10.3389/fsufs.2019.00023

CrossRef Full Text | Google Scholar

Keywords: exosomes, extracellular vesicles, brain tumor, tumor environment, blood-brain barrier, treatment

Citation: Huang Z, Keramat S, Izadirad M, Chen Z-S and Soukhtanloo M (2022) The Potential Role of Exosomes in the Treatment of Brain Tumors, Recent Updates and Advances. Front. Oncol. 12:869929. doi: 10.3389/fonc.2022.869929

Received: 05 February 2022; Accepted: 21 February 2022;
Published: 17 March 2022.

Edited by:

Fei Chen, Wayne State University, United States

Reviewed by:

Chitra Thakur, Stony Brook University, United States
Zhuoyue Bi, Wayne State University, United States

Copyright © 2022 Huang, Keramat, Izadirad, Chen and Soukhtanloo. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zhe-Sheng Chen, chenz@stjohns.edu; Mohammad Soukhtanloo, soukhtanlooM@mums.ac.ir

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.