Skip to main content

Effects of exosomes on pre-metastatic niche formation in tumors

Abstract

A pre-metastatic niche is a microenvironment prepared for the colonization of circulating tumor cells in specific organs. Exosomes are extracellular vesicles with a variety of biological functions. Exosomes play an irreplaceable role in the development of pre-metastatic niches, and mainly function as communication medium. In this review, we analyzed the effects of exosomes on pre-metastatic niches from various perspectives, including inflammation, immune response, angiogenesis, organotropism, matrix remodeling and biomarker expression. In particular, exosomes express programmed death ligand 1 (PD-L1) and cause the immune escape of tumor cells. The immunomodulatory effects of exosomes and their potential in liquid diagnosis have drawn our attention. The potential value of exosomes and pre-metastatic niches will be realized in the field of immunity therapy.

Background

Tumor metastasis usually indicates a poor prognosis and is the leading cause of cancer-related death [1]. Thus, meaningful research is being done on how to prevent tumor metastasis. In recent years, the concept of a pre-metastatic niche has provided new ideas to prevent tumor metastasis. The pre-metastatic niche refers to the microenvironment, which is well prepared for tumor cells to colonize in and disseminate to distant organ sites. Lyden firstly proposed the idea of the pre-metastatic niche [2]. The significance of the pre-metastatic niche has received increasing attention in recent years [3, 4]. Cao summarized the characteristics and four stages of the pre-metastatic niche. The key components of the pre-metastatic niche include tumor-derived secreted factors (TDSFs), extracellular vesicles (EVs), bone marrow-derived cells (BMDCs), suppressive immune cells and host stromal cells [5].

Exosomes are extracellular vesicles with a diameter of 30–150 nm [6,7,8]. Exosomes were found in blood, urine, saliva and other bodily fluids [9]. As a key player in intercellular communication, exosomes transmit information through their cargo levels, including proteins, lipids, DNA, messenger RNA and microRNAs [10,11,12,13,14,15]. Exosome-mediated intercellular communication mainly occurs in the following three ways: First, the exosome membrane protein can bind to the target cell membrane protein, thereby activating the signal pathway in the target cell. Second, in the extracellular matrix, a protease cleaves the exosome membrane protein, which then binds to receptors on the cell membrane to activate the signaling pathway within the cell. Third, the exosome membrane can directly fuse with the target cell membrane, causing nonselective release of the proteins, mRNA and microRNA of the exosomes [16, 17]. Recently, reports have shown that the formation of a pre-metastatic niche depends on tumor-derived exosomes [2, 12, 18,19,20]. The function of exosomes depends on the type of cells from which they are derived [21, 22]. Studies have shown that tumor-derived exosomes are involved in the exchange of genetic information between tumor cells and basal cells, resulting in the production of a large number of new blood vessels, which promotes tumor growth and invasion [23, 24].

This article summarizes the role of exosomes in the pre-metastasis niche, to identify new means of cancer immunotherapy. Screening for biomarkers contained within exosomes can provide diagnostic and prognostic value.

Exosomes promote the upregulation of inflammatory molecules in the pre-metastatic niche

Chronic inflammation is a driving force for tumor development and metastasis. Thus, the local inflammatory microenvironment is one of the basic factors for the formation of a pre-metastatic niche.

Hoshino reported the regulatory effect of integrins (ITGs) on the proinflammatory factor S100. They identified differentially expressed genes (DEGs) by RNA sequencing in Kupffer cells (KCs) treated with different cell-derived exosomes. The results showed that S100A8 and S100P were upregulated more than four-fold after treatment with BxPC-3-LiT exosomes compared to exosomes produced by BxPC-3-LiT ITGβ5KD (BxPC-3-LiT cells with knocked down ITGβ5 expression). After treatment of WI-38 fibroblasts with 4175-LuT exosomes, S100A4, S100A6, S100A10, S100A11, S100A13 and S100A16 were upregulated five-fold compared to the 4175-LuT ITGβ4KD (4175-LuT cells with knocked down ITGβ4 expression) exosomes. Thus, exosome integrins could upregulate the expression of proinflammatory S100 molecules in the distant tissue microenvironment. How do tumor-derived exosomes upregulate the expression of proinflammatory S100? The activation of Src, and its subsequent phosphorylation, may be the main pathway [25].

The local inflammatory microenvironment can induce tumor cells to produce tumor-derived secreted factors (TDSFs), such as vascular endothelial growth factor (VEGF), tumor necrosis factor alpha (TNF-α), transforming growth factor-β (TGF-β) and interleukin (IL). These TDSFs in turn affect myeloid cells through paracrine means to stimulate their migration to future pre-metastatic niche formation sites [26]. Under the stimulation of TDSFs, host stromal cells in the pre-metastatic niche may upregulate the expression of inflammatory factors. BMDCs or immune cells are recruited to the pre-metastatic niche and accelerate the secretion of inflammatory factors. In addition, tumor-derived exosomes carry inflammatory factors into the bloodstream, which in turn reach the pre-metastatic niche. Through the above possible ways, an inflammatory microenvironment favorable for tumors is finally formed in the pre-metastatic niche.

Exosomes induce immune suppression or immune surveillance in the pre-metastatic niche

Immune escape induced by exosome-derived PD-L1

In recent years, programmed death receptor 1 (PD-1) and programmed death ligand 1 (PD-L1) have attracted much attention. PD-1 is mainly expressed on macrophages, activated T cells and B cells, while PD-L1 is highly expressed in tumor tissues, tumor-associated antigen-presenting cells (APCs) and stromal cells [27,28,29,30]. Under normal circumstances, T cells can recognize and attack tumor cells. However, when PD-1 binds to PD-L1, it provides an inhibitory signal, induces T cell apoptosis and inhibits T cell activation and proliferation. Therefore, blocking the PD-1/PD-L1 pathway can enhance the killing effect of T cells and improve the immune response [31, 32].

Currently, immunotherapy with PD-1/PD-L1 inhibitors has moved from the laboratory to clinical applications. Anti-PD-1/PD-L1 antibodies have fewer adverse effects than conventional radiotherapy and chemotherapy. More importantly, the patient’s remission period was longer after immunotherapy. Anti-PD-1 antibodies have shown reliable effects in clinical treatment [33]. However, patients treated with PD-1/PD-L1 inhibitors have significant individual differences [34, 35]. Studies have shown that patients with high expression of PD-L1 have better clinical efficacy with PD-1/PD-L1 blockers [36]. Researchers demonstrated that histone deacetylase (HDAC) inhibitors combined with anti-PD-1 immunotherapy enhanced the therapeutic efficiency in a melanoma mouse model [37]. Therefore, the direction of future research is how to improve the remission rate, expand the indications for anti-PD-1/PD-L1 antibodies, and reduce adverse reactions.

Recently, scientists have confirmed that cancer cells release exosomes carrying PD-L1, which spread directly from the tumor tissue to all parts of the body and comprehensively crack down on and suppress the human immune system. PD-L1 in exosomes has the same structure as it does on the surface of tumor cells and can also bind to PD-1 on T cells. Cell experiments showed that exosomes incorporated with PD-L1 can inhibit the proliferation of CD8+ T cells (Fig. 1). In mouse experiments, exosomes carrying PD-L1 promoted tumor growth and reduced the number of T cells in the spleen and lymph nodes [38]. Therefore, when the exosomes carrying PD-L1 reach the pre-metastatic microenvironment, the immune system is inhibited here, and the development of the pre-metastatic niche is promoted.

Fig. 1
figure 1

Exosome PD-L1 has a similar function to tumor PD-L1. Exosome PD-L1 can also bind to PD-1 on T cells, induce T cell apoptosis, and inhibit T cell activation and proliferation

Inhibitory immune cell recruitment

Tumor exosomes can cause recruitment of suppressive immune cells. After exosomes upregulate the expression of proinflammatory factors, the local inflammatory microenvironment induces tumor cells to produce chemokines and cytokines. These factors act synergistically with the exosomes produced by tumor cells to recruit tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), regulatory T (Treg) cells and myeloid-derived suppressor cells (MDSCs) to distant secondary sites [5]. These immune cells may inhibit antitumor immune responses [39, 40]. Hematopoietic stem and progenitor cells can also differentiate into MDSCs, thereby promoting immunosuppression within the pre-metastatic niche [41].

Impaired function of T cells, NK cells and antigen-presenting cells

Exosomes contain many substances that can impair immune cell function, thereby forming an immunosuppressive premetastatic microenvironment. Prolyl hydroxylase-mediated oxygen sensing can inhibit CD4 + and CD8 + T cell functions [42]. Cancer exosomes can cause NK cell dysfunction, inhibit antigen-presenting cells, block T cell activation, and enhance T cell apoptosis to block adaptive immune responses [43, 44]. In addition, four known immunomodulatory proteins have been identified in melanoma EVs, the combination of which reduces DC maturation in vitro [45].

Molecular regulation

In addition to immunosuppression at the cellular level, some molecules are also involved in the immunomodulation of the pre-metastatic niche. During the process of MDSCs promoting immunosuppression in the pre-metastatic niche of liver cancer, TNF receptor-2 plays a supporting role [46]. In the pre-metastatic niche, ARG1 and reactive oxygen species (ROS) can inhibit antitumor T cells [47]. In addition to T cells, B cells are also inhibited. Through the TGF-βR1/TGF-βR2 signaling pathway, cancer-induced regulatory B (Breg) cells further potentiate the immunosuppression and metastasis function of MDSCs. Under the regulation of Breg cells, MDSCs produce more ROS and NO to inhibit CD8+ T cells [48].

Immune surveillance of the heterogeneity of tumor-derived exosomes

It is well-known that tumor cells are heterogeneous, which can cause differences in tumor growth rate, tumor invasion, metastasis, drug sensitivity, and prognosis. Similarly, the latest research suggests that exosomes are also heterogeneous.

Unlike the above conclusions, “nonmetastatic” exosomes have the opposite effect of exosomes produced by metastatic tumor cells. Nonmetastatic exosomes can inhibit tumor cell metastasis by increasing the number of lung-patrolling monocytes, recruiting NK cells, and inducing macrophage differentiation and phagocytosis [49]. Moreover, the level of exosome PD-L1 in metastatic melanoma cells was significantly higher than that in primary melanoma cell exosomes [38]. This experiment confirmed the heterogeneity of exosomes derived from different cells.

To sum up, exosomes affect the immune response in pre-metastatic niches through multiple mechanisms. Exosomes are heterogeneous. In the pre-metastatic niche, the effects of exosomes on immunity were divided into positive and negative aspects. On the one hand, as positive effects (promoting immune surveillance), non-metastatic exosomes increase lung mononuclear cells, recruit NK cells and induce macrophage phagocytosis. On the other hand, as negative effects (causing immunosuppression), exosomes not only mediate immune escape through PD-L1, but also promote the recruitment of suppressive immune cells impairing immune cell function. In addition, some immunosuppressive molecules, such as TNF receptor-2, ARG1, ROS and NO, also inhibit the immune function of the pre-metastatic niche.

Exosomes increase angiogenesis and vascular permeability in the pre-metastatic niche

The microenvironment prior to metastasis can increase angiogenesis and vascular permeability to facilitate metastasis. Studies have reported that exosomes released from hypoxic tumors are more likely to cause angiogenesis and vascular leakage [50, 51].

Mesenchymal-derived microvesicles activate endothelial cells via Akt phosphorylation and promote the formation of a metastatic vascular microenvironment [52]. Human kidney cancer stem cell-derived CD105-positive microvesicles can also stimulate angiogenesis, thereby promoting the formation of the pre-metastatic niche [53]. In addition, the exosome surface expresses soluble E-cadherin, which promotes tumor angiogenesis [54]. Furthermore, exosomes can express carbonic anhydrase 9 to promote angiogenesis [55]. In the pre-metastasis niche, new blood vessels not only transport TDSFs and EVs but also help bring circulating tumor cells to secondary sites. Then, exosomes begin to increase vascular permeability.

Peinado found that exosomes from the highly malignant B16-F10 enhanced lung endothelial permeability in mice compared to exosomes from nonmetastatic cell lines [19]. Melanoma-derived exosomes also induce vascular leakage and receptor tyrosine kinase Met to reprogram bone marrow progenitor cells in the niche [19]. It was observed by exosome tracing that 1833-BoT and 4175-LuT exosomes promoted pulmonary vascular leakage 24 h after injection [25]. MiR-105 secreted by metastatic breast cancer cells may disrupt the vascular endothelial barrier in the early metastatic niche, thereby increasing the vascular permeability of distant organs and promoting metastasis [56]. In addition, breast cancer cells can express matrix metalloproteinase (MMP) and cyclooxygenase. These enzymes promote vascular remodeling and increase vascular permeability to accelerate tumor cell metastasis [57].

A recent study showed that exosome secreted by colorectal cancer (CRC) cells was rich in miR-25-3p, which promoted angiogenesis and disrupted the tight junctions of vascular endothelial cells by targeting KLF2 and KLF4 [58]. In addition, the miR-25-3p of CRC cell-derived exosomes significantly induced vascular leakage and CRC metastasis in the liver and lung of mice [58]. Tumor-derived exosomes are considered to be the major drivers of the pre-metastatic niche. The above studies demonstrated that exosomes were involved in angiogenesis and increased vascular permeability in order to facilitate the formation of the pre-metastatic niche. Before the tumor is transferred, a microenvironment suitable for tumor metastasis has been created for tumor metastasis. This is another evidence that tumor-derived exosomes promote tumor metastasis.

Tumor exosomes determine organotropism metastasis

If the tumor cells are regarded as seeds and the pre-metastasis niche is regarded as soil, then the exosomes are similar to fertilizers, which can make the barren land fertile and facilitate the colonization of tumor cells [59,60,61,62,63]. This hypothesis helps us understand exosome-mediated organ-specific metastasis.

Hoshino found that tumor-derived exosomes were important factors in organ-specific metastasis. Tumor-derived exosomes prepare a favorable microenvironment at future metastatic sites, thereby mediating nonrandom transfer patterns. First, future metastatic sites uptake exosomes. Second, exosomes redirect metastatic distribution. Third, ITGs on the surface of exosomes determine organotropism. Quantitative mass spectrometry and western blot analysis as well as qualitative mass spectrometry showed that ITGα6 was present in lung-directed exosomes, while ITGβ5 was present in liver-directed exosomes. These results indicate that the different expression of exosome ITGs is the basis of organotropism. Fourth, different cells absorb tropic exosomes. Lung-tropic 4175 exosomes mainly colocalized with S100A4-positive fibroblasts in the lung. By contrast, pancreatic cancer exosomes derived from BxPC-3-LiT cells mainly fused with Kupffer cells (Fig. 2). These data demonstrate that specific tissue-resident stromal cells differentially uptake tumor exosomes in metastatic target organs. Finally, exosome tropism requires ITGβ4 and ITGβ5. After knocking down ITGβ4 expression in 4175-LuT cells (4175β4KD), a more than threefold reduction in labeled ITGβ4KD exosomes in the lung was observed. Conversely, ITGβ4 overexpression in 1833-BoT exosomes was sufficient to increase exosome uptake in the lung. Similarly, ITGβ5 knockdown in BxPC-3-LiT exosomes decreased liver uptake by sevenfold compared with uptake by control BxPC-3-LiT exosomes. The above results demonstrate that exosome ITGs are responsible for organ-specific metastasis [25].

Fig. 2
figure 2

Different cells uptake tropic exosomes. Lung-tropic 4175 exosomes mainly colocalized with S100A4-positive fibroblasts in the lung; pancreatic cancer exosomes derived from BxPC-3-LiT cells mainly fused with Kupffer cells

The above evidence proves that tumor cells prepare the receiving organs by releasing exosomes. Cancer cells spread from the origin to distant organs through blood circulation. The process of dissemination is not random, and cancer cells will preferentially look for specific organs under the guidance of exosomes and build nests there. This kind of destination-seeking behavior involves the interaction of “seeds” and “soil”. Seeds can fertilize the “soil” through exosomes before they reach the soil, consequently preparing for tumor metastasis.

Stroma cell activation and remodeling of the extracellular matrix (ECM) in pre-metastatic niches

The matrix environment of the pre-metastatic niche is mainly composed of fibroblasts, endothelial cells and extracellular matrix (ECM). Fibroblasts not only produce inflammatory and growth factors but also express fibronectin (FN) and matrix metalloproteinase (MMP) [64]. Moreover, endothelial cells secrete proinflammatory factors S100A8 and S100A9 [65]. A prominent part of the pre-metastasis matrix of liver cancer is hepatic stellate cells, which can promote ECM deposition and upregulate the expression of VEGF, IL-1a and TGF-β [66, 67] . Inflammatory factors produced by stromal cells may contribute to the formation of local inflammatory microenvironments. Growth factors secreted by stromal cells may promote angiogenesis in the pre-metastatic niche. In addition, hypoxia is also an important factor of ECM. Hypoxia can induce the expression of HIF, VEGF and G-CSF, thereby promoting the formation of a pre-metastatic niche [68, 69].

Melanoma-derived exosomes can reprogram human adult dermal fibroblasts (HADF) to cause extracellular acidification [70]. Multiple types of BMDCs promote matrix remodeling in the pre-metastatic niche, and upregulation of fibronectin (FN) [2, 71]. Costa-Silva showed that exosome macrophage migration inhibitory factor (MIF) induces the release of TGF-β from Kupffer cells, which in turn promotes the production of FN in hepatic stellate cells (HSTCs). FN deposits subsequently promote the stagnation of bone marrow-derived macrophages and neutrophils in the liver, completing the formation of the pre-metastatic niche [72]. Exosomes expressing ITGα6β4 and ITGα6β1 co-localize with laminin, and exosomes expressing ITGαvβ5 colocalize with fibronectin [25]. Therefore, we hypothesize that specific exosome integrins may interact with extracellular matrices, and that deposited laminin and fibronectin may help increase the adhesion of extracellular matrices to facilitate colonization of circulating tumor cells.

Roles of coding or non-coding RNA carried out by exosomes in the pre-metastatic niche

Tumor-derived exosomes have been reported to contain DNA fragments that provide exosome origin information. Once released into the peripheral cycle, the information contained in DNA fragments may direct the metastatic behavior of circulating tumor cells [73, 74]. Mutant genes were found in serum exosomes from patients with pancreatic cancer. For example, p53 and KRAS mutations have been verified [75]. In addition, DNA fragments with the ability to amplify the c-Myc gene were found in medulloblastoma cell-derived exosomes [76].

In addition, exosome proteins have been identified to be associated with the cell cycle, cell signaling process, cytoskeleton formation, apoptosis, oxidative stress, focal adhesions formation, and cell mobility [77]. Exosome integrins play an important role in the pre-metastatic niche, which has been described in detail above. Studies have shown that osteopontin derived from tumor microbubbles contributes to the colonization of tumor cells and BMDC mobilization [78].

Furthermore, microRNAs associated with metastasis in exosomes have also been reported [79]. Tumor-secreted microRNAs bind to TLR8 in human immune cells, triggering tumor metastasis [80]. Brain astrocyte-derived exosomes mediate PTEN targeting of miRNA-19a, thereby promoting PTEN silencing in tumor cells. Decreased PTEN upregulates the expression of chemokine CCL2, which recruits bone marrow cells in the brain [81]. Exosomes derived from prostate cancer contain differentially expressed genes (DEGs), such as miR-21-5p and miR-139-5p. These microRNAs derived from exosomes cooperate to adjust the pre-metastatic niche [82]. Studies have reported that lncRNAs have important functions for pre-metastatic niches [83]. Multiple studies have shown that lncRNAs can be used as biomarkers for tumor diagnosis [84,85,86] . Circular RNA (circRNA) is a closed loop structure, and its expression is more stable and less prone to degradation. Circular RNA has spatiotemporal specificity and is highly conserved among species. Recently, the role of circRNA in exosomes has been reported. In the future, the function of circRNA in the pre-metastatic niche may be gradually excavated.

From the above studies, we conclude that both coding and non-coding RNA derived from exosomes promote the formation of pre-metastatic niches. At the present, the induction of pre-metastatic niche formation by exosome miRNAs has attracted wide attention. In the future, exosome-derived lncRNA and circRNA will also show their prominence.

Molecules carried by exosomes may be tumor biomarkers

In recent years, research on finding biomarkers for detecting diseases has grown exponentially. Some studies have used exosomes containing non-coding RNA as potential biomarkers for detecting different malignancies. MiRNAs isolated from tumor-derived exosomes are more stable and therefore considered to be more reliable biomarkers. In addition, exosome-derived lncRNA and circRNA are also fascinating potential biomarkers.

Biomarkers for tumor diagnosis

The levels of miR-21, miR-150, miR-223, and miR-1229 in serum exosomes of colorectal cancer (CRC) patients were significantly higher compared to those of the control group. Similarly, in vitro experiments revealed that these exosome miRNAs were expressed at higher levels in CRC cell lines than in nontumor cell lines [87]. In gastric cancer (GC), plasma LINC00152 levels in patients with gastric cancer were significantly higher than in healthy controls [88]. The above findings suggest that molecules carried by exosomes can be potential biomarkers for the early detection of disease.

Biomarkers for metastasis predication

Scientists have found that exosomes in the blood of melanoma patients, especially those with metastatic melanoma, have significantly higher PD-L1 levels than those of healthy individuals [38]. Hoshino found more highly-expressed ITGβ4 in exosomes from patients with lung metastasis compared with those from in situ cancer or liver metastasis patients. The exosomes isolated before lung metastasis of breast cancer patients express the highest level of ITGβ4. In addition, levels of ITGαv in the exosomes of patients with liver metastases were elevated compared with those of patients with orthotopic tumors or lung metastases [25]. In gastric cancer patients, serum exosomes ZFAS1 are highly expressed. High levels of ZFAS1 were significantly associated with lymphatic metastasis and TNM staging [89]. Moreover, low levels of miR-217 in serum exosomes are associated with lymph node metastasis or distant metastasis [90].

Biomarkers for the evaluation of prognosis

For patients with prostate cancer, upregulation of miR-375 and miR-1290 may indicate a lower overall survival rate [91]. It has been reported that elevated serum exosome CRNDE-h is closely related to poor prognosis in patients with CRC [92]. In addition, there is a significant difference in the expression of miR-17-92 clusters in CRC patients with poor prognosis compared with healthy patients [93]. PD-1 levels are associated with the prognosis of classical Hodgkin’s lymphoma. Inhibiting the expression of PD-1 can improve the prognoses of patients [94]. High expression of PD-L1 in gastric cancer tissues may be associated with poor prognosis [95]. In patients with triple-negative breast cancer, PD-L1 is highly expressed in the tumor tissues [96].

In short, exosome-derived PDL-1 and noncoding RNA affect tumor biological functions such as invasion, metastasis and proliferation. Therefore, these molecules have the potential to affect prognosis.

Biomarkers for the evaluation of treatment effectiveness

Recently, in 44 melanoma patients treated with Keytruda, scientists found that among the 21 patients who responded to treatment, the level of exosome PD-L1 in the blood before treatment was significantly lower than the level of exosome PD-L1 of the other 23 patients who did not respond to treatment. In terms of prognosis, patients with low levels of PD-L1 in blood exosomes had a better prognosis. In the future, monitoring PD-L1 or other biomarkers on circulating exosomes may be an effective way to predict and observe therapeutic effects [38].

Traditional tissue biopsy is invasive and cannot be used for the early diagnosis of tumors. For years, researchers have been looking for non-invasive cancer detection tools. Currently, with the in-depth study of plasma exosomes, liquid biopsy has been considered for the early diagnosis of tumors [97]. Exosomes secreted by tumor cells carry different types of contents, and these molecules have multiple key biological activities. [98]. Exosomes can be abundantly released from cancer cells and can be broadly distributed in body fluids. Exosomes are very stable and are capable of protecting proteins and nucleic acids with its lipid bilayer. In short, exosomes possess obvious advantages in terms of quantity, stability, and accessibility.

Previous studies have shown the advantages of exosomes in predicting metastasis, prognosis and therapeutic effects on tumors. However, the credibility of a single molecule is limited, and some researchers suggest using a combination of the most reliable biomarkers to improve the sensitivity and specificity of detecting tumors [99].

Exosome-mediated potential anti-tumor therapy

Exosome-mediated chemotherapy resistance

Currently, chemotherapy is still one of the main methods of cancer treatment. However, tumor cell resistance to chemotherapy makes treatment difficult. Therefore, understanding the mechanisms of chemical resistance and identifying key molecules are the basis for solving the current dilemma.

Some studies have linked exosomes to chemotherapy resistance by the action of cargo miRNAs on recipient cells. It has been observed that miRNAs are tumor suppressors, leading to uncontrolled cell growth and resistance to anti-tumor drugs [100]. Furthermore, the chemical resistance of 5-FU can result in the enhanced invasion and migration of tumor cells. This phenomenon is related to the loss of miR-200 [101]. P73 is an isoform of p53. Both in vivo and in vitro experiments have shown that ΔNP73 can increase the proliferation of CRC cells and confer resistance to oxaliplatin by exosome transfer [102].

The pre-metastatic niche provides new ideas for targeted therapy

Targeted therapy is the treatment of established carcinogenic sites at the cellular and molecular levels. When a targeted drug enters the body, it specifically binds to the oncogenic site, causing the tumor cells to die without affecting normal tissue cells, so molecular targeted therapy agents have also called “bio-missiles”. With the development of molecular biology, molecular targeted therapy has gradually become a new means of cancer treatment in recent years.

Although targeted therapy has been recognized by the medical community for its unique advantages, the effects on cancer treatment are limited. There are significant individual differences in targeted therapeutic effects. Drugs that are useful to some individuals may not be effective for other patients. Therefore, finding a powerful target in the early process of tumor development is the key to breaking through the bottleneck of targeted therapy.

Tumor-derived exosomes transmit information between cells to promote the formation of pre-metastatic niches. Thus, blocking the delivery of exosome cargos to recipient cells may be a powerful strategy for preventing tumor metastasis. There are many targets that could be used to inhibit the formation of pre-metastatic niches (for instance, blocking the production of proinflammatory factors, inhibiting the recruitment of BMDCs, preventing angiogenesis and vascular penetration, destroying the local matrix, and reactivating the anti-tumor immune response). These targets could develop potential ways to prevent and control cancer metastasis in the future.

Conclusions

In summary, exosomes play multiple roles in the development of pre-metastatic niches. Exosomes can not only promote the upregulation of inflammatory molecules, induce angiogenesis and infiltration, and regulate immune function in both directions but also determine organotropism metastasis and cause extracellular matrix remodeling [103] (Fig. 3).

Fig. 3
figure 3

Effects of exosomes on tumor pre-metastatic niche formation. Exosomes promote the upregulation of inflammatory molecules; exosomes induce immune suppression; exosome increase angiogenesis and vascular permeability; exosome determine organotropism metastasis; Exosome promote matrix remodeling

Many studies have confirmed that exosomes can promote the formation of pre-metastatic niches. However, the functions, mechanisms, and contribution rates of exosomes remain to be further explored. The problems that need to be solved urgently include 1. What is the difference in effect between tumor-derived exosomes and immune cell- or stromal cell-derived exosomes on the pre-metastatic niche? 2. The composition of exosomes is complex, so what role do these components play? Is there an interaction between the different components? 3. After the primary tumor is surgically removed, does the effect of the exosomes on the pre-metastatic niche terminate? 4. What is the reliability and validity of using exosomes as biomarkers to judge tumor metastasis? 5. How can the theoretical basis of exosomes and pre-metastatic niches be applied to clinical treatment?

To conclude, exosomes play an essential role in the pre-metastatic niche, and further exploration of the function of exosomes in the pre-metastatic niche will help to determine treatments in the early stage of cancer. In addition, circulating exosomes, as biomarkers, have great potential in liquid diagnostics. As exosomes have the biological role of transporting cargo, exosomes may be used as a carrier for targeted drugs to accurately destroy tumor cells [104]. In the future, tumors are expected to become as chronic as diseases such as diabetes. Just as people with diabetes monitor blood glucose with a blood glucose meter, tumor patients will be able to track tumor progression through exosome biomarkers.

Abbreviations

APCs:

Antigen presenting cells

BMDCs:

Bone marrow-derived cells

CRC:

Colorectal cancer

DC:

Dendritic cell

ECM:

Extracellular matrix

EVs:

Extracellular vesicle

FN:

Fibronectin

GC:

Gastric cancer

HADF:

Human adult dermal fibroblasts

HSTCs:

Hepatic stellate cells

KCs:

Kupffer cells

LncRNA:

Long noncoding RNA

MIF:

Macrophage migration inhibitory factor

NK:

Natural killer cells

PD-1:

Programmed death receptor 1

PD-L1:

Programmed death ligand 1

TAMs:

Tumor-associated macrophage

TANs:

Tumor-associated neutrophil

TDSFs:

Tumor-derived secreted factors

VEGF:

Vascular endothelial growth factor

References

  1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. https://doi.org/10.1016/j.cell.2011.02.013.

    Article  CAS  PubMed  Google Scholar 

  2. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820–7. https://doi.org/10.1038/nature04186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chin AR, Wang SE. Cancer tills the Premetastatic field: mechanistic basis and clinical implications. Clin Cancer Res. 2016;22(15):3725–33. https://doi.org/10.1158/1078-0432.CCR-16-0028.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sleeman JP. The metastatic niche and stromal progression. Cancer Metastasis Rev. 2012;31(3–4):429–40. https://doi.org/10.1007/s10555-012-9373-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Liu Y, Cao XT. Characteristics and significance of the pre-metastatic niche. Cancer Cell. 2016;30(5):668–81. https://doi.org/10.1016/j.ccell.2016.09.011.

    Article  CAS  PubMed  Google Scholar 

  6. Arscott WT, Camphausen KA. Exosome characterization from ascitic fluid holds promise for identifying markers of colorectal cancer. Biomark Med. 2011;5(6):821–2.

    Article  Google Scholar 

  7. Record M, Carayon K, Poirot M, Silvente-Poirot S. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochimica Et Biophysica Acta-Molecular and Cell Biology of Lipids. 2014;1841(1):108–20. https://doi.org/10.1016/j.bbalip.2013.10.004.

    Article  CAS  Google Scholar 

  8. EL Andaloussi S, Maeger I, Breakefield XO, Wood MJA. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):348–58. https://doi.org/10.1038/nrd3978.

    Article  CAS  Google Scholar 

  9. Boukouris S, Mathivanan S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteomics Clin Appl. 2015;9(3–4):358–67. https://doi.org/10.1002/prca.201400114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Choi DS, Kim DK, Kim YK, Gho YS. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 2013;13(10–11):1554–71. https://doi.org/10.1002/pmic.201200329.

    Article  CAS  PubMed  Google Scholar 

  11. Martins VR, Dias MS, Hainaut P. Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr Opin Oncol. 2013;25(1):66–75. https://doi.org/10.1097/CCO.0b013e32835b7c81.

    Article  CAS  PubMed  Google Scholar 

  12. Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol. 2011;21(2):139–46. https://doi.org/10.1016/j.semcancer.2011.01.002.

    Article  CAS  PubMed  Google Scholar 

  13. Tetta C, Ghigo E, Silengo L, Deregibus MC, Camussi G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine. 2013;44(1):11–9. https://doi.org/10.1007/s12020-012-9839-0.

    Article  CAS  PubMed  Google Scholar 

  14. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–U672. https://doi.org/10.1038/ncb1596.

    Article  CAS  PubMed  Google Scholar 

  15. Zoller M. Pancreatic cancer diagnosis by free and exosomal miRNA. World J Gastrointest Pathophysiol. 2013;4(4):74–90. https://doi.org/10.4291/wjgp.v4.i4.74.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important in intercellular communication. J Proteome. 2010;73(10):1907–20. https://doi.org/10.1016/j.jprot.2010.06.006.

    Article  CAS  Google Scholar 

  17. Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20(9):1487–95. https://doi.org/10.1038/sj.leu.2404296.

    Article  CAS  PubMed  Google Scholar 

  18. Sceneay J, Smyth MJ, Moller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32(3–4):449–64. https://doi.org/10.1007/s10555-013-9420-1.

    Article  CAS  PubMed  Google Scholar 

  19. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18(6):883-+. https://doi.org/10.1038/nm.2753.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hood JL, Roman SS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71(11):3792–801. https://doi.org/10.1158/0008-5472.CAN-10-4455.

    Article  CAS  PubMed  Google Scholar 

  21. Zhang W, Yang JX, Cao DY, You Y, Shen K, Peng P. Regulation of exosomes released from normal ovarian epithelial cells and ovarian cancer cells. Tumor Biol. 2016;37(12):15763–71. https://doi.org/10.1007/s13277-016-5394-2.

    Article  CAS  Google Scholar 

  22. 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Mears R, Craven RA, Hanrahan S, Totty N, Upton C, Young SL, et al. Proteomic analysis of melanoma-derived exosomes by two-dimensional polyacrylamide gel electrophoresis and mass spectrometry. Proteomics. 2004;4(12):4019–31. https://doi.org/10.1002/pmic.200400876.

    Article  CAS  PubMed  Google Scholar 

  24. Vaksman O, Trope C, Davidson B, Reich R. Exosome-derived miRNAs and ovarian carcinoma progression. Carcinogenesis. 2014;35(9):2113–20. https://doi.org/10.1093/carcin/bgu130.

    Article  CAS  PubMed  Google Scholar 

  25. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Mark MT, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329-+. https://doi.org/10.1038/nature15756.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Deng J, Liu Y, Lee H, Herrmann A, Zhang W, Zhang C, et al. S1PR1-STAT3 signaling is crucial for myeloid cell colonization at future metastatic sites. Cancer Cell. 2012;21(5):642–54. https://doi.org/10.1016/j.ccr.2012.03.039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Boussiotis VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med. 2016;375(18):1767–78. https://doi.org/10.1056/NEJMra1514296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800. https://doi.org/10.1038/nm730.

    Article  CAS  PubMed  Google Scholar 

  29. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015;125(9):3384–91. https://doi.org/10.1172/JCI80011.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Gordon SR, Aute RLM, Dulken BW, Hutter G, George BM, Ccracken MNM, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545(7655):495-+. https://doi.org/10.1038/nature22396.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Meng X, Huang Z, Teng F, Xing L, Yu J. Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy. Cancer Treat Rev. 2015;41(10):868–76. https://doi.org/10.1016/j.ctrv.2015.11.001.

    Article  CAS  PubMed  Google Scholar 

  32. Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4(127):127ra137. https://doi.org/10.1126/scitranslmed.3003689.

    Article  CAS  Google Scholar 

  33. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275–87. https://doi.org/10.1038/nrc.2016.36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, et al. Association of Pembrolizumab with Tumor Response and Survival among Patients with Advanced Melanoma. JAMA. 2016;315(15):1600–9. https://doi.org/10.1001/jama.2016.4059.

    Article  CAS  PubMed  Google Scholar 

  35. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375(9):819–29. https://doi.org/10.1056/NEJMoa1604958.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gangadhar TC, Salama AK. Clinical applications of PD-1-based therapy: a focus on pembrolizumab (MK-3475) in the management of melanoma and other tumor types. Onco Targets Ther. 2015;8:929–37. https://doi.org/10.2147/OTT.S53164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Woods DM, Sodre AL, Villagra A, Sarnaik A, Sotomayor EM, Weber J. HDAC inhibition upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade. Cancer Immunol Res. 2015;3(12):1375–85. https://doi.org/10.1158/2326-6066.CIR-15-0077-T.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560(7718):382-+. https://doi.org/10.1038/s41586-018-0392-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16(8):717–27. https://doi.org/10.1038/ncb3015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu Y, Cao X. Immunosuppressive cells in tumor immune escape and metastasis. J Mol Med-Jmm. 2016;94(5):509–22. https://doi.org/10.1007/s00109-015-1376-x.

    Article  CAS  Google Scholar 

  41. Giles AJ, Reid CM, Evans JD, Murgai M, Vicioso Y, Highfill SL, et al. Activation of hematopoietic stem/progenitor cells promotes immunosuppression within the pre-metastatic niche. Cancer Res. 2016;76(6):1335–47. https://doi.org/10.1158/0008-5472.CAN-15-0204.

    Article  CAS  PubMed  Google Scholar 

  42. Clever D, Roychoudhuri R, Constantinides MG, Askenase MH, Sukumar M, Klebanoff CA, et al. Oxygen Sensing by T Cells Establishes an Immunologically Tolerant Metastatic Niche. Cell. 2016;166(5):1117-+. https://doi.org/10.1016/j.cell.2016.07.032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ludwig S, Floros T, Theodoraki MN, Hong CS, Jackson EK, Lang S, et al. Suppression of lymphocyte functions by plasma exosomes correlates with disease activity in patients with head and neck cancer. Clin Cancer Res. 2017;23(16):4843–54. https://doi.org/10.1158/1078-0432.CCR-16-2819.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Czernek L, Duchler M. Functions of Cancer-derived extracellular vesicles in immunosuppression. Arch Immunol Ther Exp. 2017;65(4):311–23. https://doi.org/10.1007/s00005-016-0453-3.

    Article  CAS  Google Scholar 

  45. Maus RLG, Jakub JW, Nevala WK, Christensen TA, Noble-Orcutt K, Sachs Z, et al. Human Melanoma-Derived Extracellular Vesicles Regulate Dendritic Cell Maturation. Front Immunol. 2017; 8. doi: Artn 35810.3389/Fimmu.2017.00358.

  46. Ham B, Wang N, D'Costa Z, Fernandez MC, Bourdeau F, Auguste P, et al. TNF Receptor-2 facilitates an immunosuppressive microenvironment in the liver to promote the colonization and growth of hepatic metastases. Cancer Res. 2015;75(24):5235–47. https://doi.org/10.1158/0008-5472.CAN-14-3173.

    Article  CAS  PubMed  Google Scholar 

  47. Tacke RS, Lee HC, Goh C, Courtney J, Polyak SJ, Rosen HR, et al. Myeloid suppressor cells induced by hepatitis C virus suppress T-cell responses through the production of reactive oxygen species. Hepatology. 2012;55(2):343–53. https://doi.org/10.1002/hep.24700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bodogai M, Moritoh K, Lee-Chang C, Hollander CM, Sherman-Baust CA, Wersto RP, et al. Immunosuppressive and prometastatic functions of myeloid-derived suppressive cells rely upon education from tumor-associated B cells. Cancer Res. 2015;75(17):3456–65. https://doi.org/10.1158/0008-5472.CAN-14-3077.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Plebanek MP, Angeloni NL, Vinokour E, Li J, Henkin A, Martinez-Marin D, et al. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun. 2017;8 doi: Artn 131910.1038/S41467–017–01433-3.

  50. Kucharzewska P, Christianson HC, Welch JE, Svensson KJ, Fredlund E, Ringner M, et al. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci U S A. 2013;110(18):7312–7. https://doi.org/10.1073/pnas.1220998110.

    Article  PubMed  PubMed Central  Google Scholar 

  51. King HW, Michael MZ, Gleadle JM. Hypoxic enhancement of exosome release by breast cancer cells. Bmc Cancer. 2012;12 doi: Artn 42110.1186/1471–2407–12-421.

  52. Pasquier J, Thawadi HA, Ghiabi P, Abu-Kaoud N, Maleki M, Guerrouahen BS, et al. Microparticles mediated cross-talk between tumoral and endothelial cells promote the constitution of a pro-metastatic vascular niche through Arf6 up regulation. Cancer Microenvironment. 2014;7(1–2):41–59. https://doi.org/10.1007/s12307-013-0142-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung Premetastatic niche. Cancer Res. 2011;71(15):5346–56. https://doi.org/10.1158/0008-5472.can-11-0241.

    Article  CAS  PubMed  Google Scholar 

  54. Tang MKS, Yue PYK, Ip PP, Huang RL, Lai HC, Cheung ANY, et al. Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nat Commun. 2018;9(1):2270. https://doi.org/10.1038/s41467-018-04695-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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. https://doi.org/10.1016/j.bbrc.2017.08.107.

    Article  CAS  PubMed  Google Scholar 

  56. Zhou WY, Fong MY, Min YF, 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. https://doi.org/10.1016/j.ccr.2014.03.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY, Nadal C, et al. Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature. 2007;446(7137):765–U762. https://doi.org/10.1038/nature05760.

    Article  CAS  PubMed  Google Scholar 

  58. Zeng Z, Li Y, Pan Y, Lan X, Song F, Sun J, et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun. 2018;9(1):5395. https://doi.org/10.1038/s41467-018-07810-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Muller A, Homey B, Soto H, Ge NF, Catron D, Buchanan ME, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410(6824):50–6. https://doi.org/10.1038/35065016.

    Article  CAS  PubMed  Google Scholar 

  60. Lu X, Kang YB. Organotropism of breast cancer metastasis. J Mammary Gland Biol Neoplasia. 2007;12(2–3):153–62. https://doi.org/10.1007/s10911-007-9047-3.

    Article  PubMed  Google Scholar 

  61. Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu WP, Giri DD, et al. Genes that mediate breast cancer metastasis to lung. Nature. 2005;436(7050):518–24. https://doi.org/10.1038/nature03799.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bos PD, Zhang XHF, Nadal C, Shu WP, Gomis RR, Nguyen DX, et al. Genes that mediate breast cancer metastasis to the brain. Nature. 2009;459(7249):1005–U1137. https://doi.org/10.1038/nature08021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Minn AJ, Kang YB, Serganova I, Gupta GP, Giri DD, Doubrovin M, et al. Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors. J Clin Investig. 2005;115(1):44–55. https://doi.org/10.1172/Jc1200522320.

    Article  CAS  PubMed  Google Scholar 

  64. Yamamura Y, Asai N, Enomoto A, Kato T, Mii S, Kondo Y, et al. Akt-Girdin signaling in cancer-associated fibroblasts contributes to tumor progression. Cancer Res. 2015;75(5):813–23. https://doi.org/10.1158/0008-5472.CAN-14-1317.

    Article  CAS  PubMed  Google Scholar 

  65. Hiratsuka S, Watanabe A, Sakurai Y, Akashi-Takamura S, Ishibashi S, Miyake K, et al. The S100A8-serum amyloid A3-TLR4 paracrine cascade establishes a pre-metastatic phase. Nat Cell Biol. 2008;10(11):1349–55. https://doi.org/10.1038/ncb1794.

    Article  CAS  PubMed  Google Scholar 

  66. Eveno C, Hainaud P, Rampanou A, Bonnin P, Bakhouche S, Dupuy E, et al. Proof of prometastatic niche induction by hepatic stellate cells. J Surg Res. 2015;194(2):496–504. https://doi.org/10.1016/j.jss.2014.11.005.

    Article  PubMed  Google Scholar 

  67. Mikuriya Y, Tashiro H, Kuroda S, Nambu J, Kobayashi T, Amano H, et al. Fatty liver creates a pro-metastatic microenvironment for hepatocellular carcinoma through activation of hepatic stellate cells. Int J Cancer. 2015;136(4):E3–E13. https://doi.org/10.1002/ijc.29096.

    Article  CAS  PubMed  Google Scholar 

  68. Chafe SC, Lou Y, Sceneay J, Vallejo M, Hamilton MJ, McDonald PC, et al. Carbonic anhydrase IX promotes myeloid-derived suppressor cell mobilization and establishment of a metastatic niche by stimulating G-CSF production. Cancer Res. 2015;75(6):996–1008. https://doi.org/10.1158/0008-5472.CAN-14-3000.

    Article  CAS  PubMed  Google Scholar 

  69. Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, et al. Hypoxia-induced Lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the Premetastatic niche. Cancer Cell. 2009;15(1):35–44. https://doi.org/10.1016/j.ccr.2008.11.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. La Shu S, Yang Y, Allen CL, Maguire O, Minderman H, Sen A, et al. Metabolic reprogramming of stromal fibroblasts by melanoma exosome microRNA favours a pre-metastatic microenvironment. Sci Rep. 2018;8. https://doi.org/10.1038/s41598-018-31323-7.

  71. Hiratsuka S, Watanabe A, Aburatani H, Maru Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat Cell Biol. 2006;8(12):1369–U1331. https://doi.org/10.1038/ncb1507.

    Article  CAS  PubMed  Google Scholar 

  72. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang HY, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816-+. https://doi.org/10.1038/ncb3169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kahlert C, Kalluri R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med-Jmm. 2013;91(4):431–7. https://doi.org/10.1007/s00109-013-1020-6.

    Article  CAS  Google Scholar 

  74. Williams C, Rodriguez-Barrueco R, Silva JM, Zhang W, Hearn S, Elemento O, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24(6):766–9. https://doi.org/10.1038/cr.2014.44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kahlert C, Melo SA, Protopopov A, Tang JB, Seth S, Koch M, et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 2014;289(7):3869–75. https://doi.org/10.1074/jbc.C113.532267.

  76. Balaj L, Lessard R, Dai LX, Cho YJ, Pomeroy SL, Breakefield XO, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2 doi: Artn 18010.1038/Ncomms1180.

  77. Kourembanas S. Exosomes: Vehicles of Intercellular Signaling, Biomarkers, and Vectors of Cell Therapy. Annu Rev Physiol. 2015;77:13–27. https://doi.org/10.1146/annurev-physiol-021014-071641.

    Article  CAS  PubMed  Google Scholar 

  78. Fremder E, Munster M, Aharon A, Miller V, Gingis-Velitski S, Voloshin T, et al. Tumor-derived microparticles induce bone marrow-derived cell mobilization and tumor homing: a process regulated by osteopontin. Int J Cancer. 2014;135(2):270–81. https://doi.org/10.1002/ijc.28678.

    Article  CAS  PubMed  Google Scholar 

  79. Bigagli E, Luceri C, Guasti D, Cinci L. Exosomes secreted from human colon cancer cells influence the adhesion of neighboring metastatic cells: role of microRNA-210. Cancer Biol Ther. 2016;17(10):1062–9. https://doi.org/10.1080/15384047.2016.1219815.

    Article  CAS  PubMed Central  Google Scholar 

  80. Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, et al. MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci U S A. 2012;109(31):E2110–6. https://doi.org/10.1073/pnas.1209414109.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Zhang L, Zhan SY, Yao J, Lowery FJ, Zhang QL, Huang WC, et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature. 2015;527(7576):100–4. https://doi.org/10.1038/nature15376.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Sanchez CA, Andahur EI, Valenzuela R, Castellon EA, Fulla JA, Ramos CG, et al. Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget. 2016;7(4):3993–4008. https://doi.org/10.18632/oncotarget.6540.

    Article  PubMed  Google Scholar 

  83. Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, et al. Exosome-transmitted lncARSR promotes Sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 2016;29(5):653–68. https://doi.org/10.1016/j.ccell.2016.03.004.

    Article  CAS  PubMed  Google Scholar 

  84. Chen M, Xu R, Ji H, Greening DW, Rai A, Izumikawa K, et al. Transcriptome and long noncoding RNA sequencing of three extracellular vesicle subtypes released from the human colon cancer LIM1863 cell line. Sci Rep. 2016;6:38397. https://doi.org/10.1038/srep38397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Zhang J, Liu SC, Luo XH, Tao GX, Guan M, Yuan H, et al. Exosomal long noncoding RNAs are differentially expressed in the Cervicovaginal lavage samples of cervical cancer patients. J Clin Lab Anal. 2016;30(6):1116–21. https://doi.org/10.1002/jcla.21990.

    Article  CAS  PubMed  Google Scholar 

  86. Dong L, Lin WR, Qi P, Xu MD, Wu XB, Ni SJ, et al. Circulating long RNAs in serum extracellular vesicles: their characterization and potential application as biomarkers for diagnosis of colorectal Cancer. Cancer Epidemiol Biomark Prev. 2016;25(7):1158–66. https://doi.org/10.1158/1055-9965.EPI-16-0006.

    Article  CAS  Google Scholar 

  87. Ogata-Kawata H, Izumiya M, Kurioka D, Honma Y, Yamada Y, Furuta K, et al. Circulating Exosomal microRNAs as Biomarkers of Colon Cancer. Plos One. 2014;9(4) doi: ARTN e9292110.1371/journal.pone.0092921.

  88. Li Q, Shao YF, Zhang XJ, Zheng T, Miao M, Qin LJ, et al. Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumor Biol. 2015;36(3):2007–12. https://doi.org/10.1007/s13277-014-2807-y.

    Article  CAS  Google Scholar 

  89. 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. https://doi.org/10.1007/s00432-017-2361-2.

    Article  CAS  PubMed  Google Scholar 

  90. Yu B, Du Q, Li H, Liu HY, Ye X, Zhu B, et al. Diagnostic potential of serum exosomal colorectal neoplasia differentially expressed long non-coding RNA (CRNDE-p) and microRNA-217 expression in colorectal carcinoma. Oncotarget. 2017;8(48):83745–53. https://doi.org/10.18632/oncotarget.19407.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Huang XY, Yuan TZ, Liang MH, Du MJ, Xia S, Dittmar R, et al. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur Urol. 2015;67(1):33–41. https://doi.org/10.1016/j.eururo.2014.07.035.

    Article  CAS  PubMed  Google Scholar 

  92. Liu T, Zhang X, Gao SY, Jing FM, Yang YM, Du LT, et al. Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer. Oncotarget. 2016;7(51):85551–63. https://doi.org/10.18632/oncotarget.13465.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Matsumura T, Sugimachi K, Iinuma H, Takahashi Y, Kurashige J, Sawada G, et al. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br J Cancer. 2015;113(2):275–81. https://doi.org/10.1038/bjc.2015.201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Koh YW, Jeon YK, Yoon DH, Suh C, Huh J. Programmed death 1 expression in the peritumoral microenvironment is associated with a poorer prognosis in classical Hodgkin lymphoma. Tumour Biol. 2016;37(6):7507–14. https://doi.org/10.1007/s13277-015-4622-5.

    Article  CAS  PubMed  Google Scholar 

  95. Wu CP, Zhu YB, Jiang JT, Zhao JM, Zhang XG, Xu N. Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochem. 2006;108(1):19–24. https://doi.org/10.1016/j.acthis.2006.01.003.

    Article  PubMed  Google Scholar 

  96. Pan BJ, Xu C, Ping GQ, Song GX, Zhang WM, Wang C, et al. Correlation analysis of PD-L1 expression and prognosis in triple-negative breast cancers. Zhonghua Bing Li Xue Za Zhi. 2017;46(12):822–6. https://doi.org/10.3760/cma.j.issn.0529-5807.2017.12.002.

    Article  CAS  PubMed  Google Scholar 

  97. Gulei D, Petrut B, Tigu AB, Onaciu A, Fischer-Fodor E, Atanasov AG, et al. Exosomes at a glance - common nominators for cancer hallmarks and novel diagnosis tools. Crit Rev Biochem Mol Biol. 2018;53(5):564–77. https://doi.org/10.1080/10409238.2018.1508276.

    Article  CAS  PubMed  Google Scholar 

  98. Bu H, He D, He X, Wang K. Exosomes: isolation, analysis, and applications in cancer detection and therapy. Chembiochem. 2018. https://doi.org/10.1002/cbic.201800470.

  99. Ruiz-Lopez L, Blancas I, Garrido JM, Mut-Salud N, Moya-Jodar M, Osuna A, et al. The role of exosomes on colorectal cancer: a review. J Gastroenterol Hepatol. 2018;33(4):792–9. https://doi.org/10.1111/jgh.14049.

    Article  PubMed  Google Scholar 

  100. Akao Y, Khoo F, Kumazaki M, Shinohara H, Miki K, Yamada N. Extracellular disposal of tumor-suppressor miRs-145 and-34a via microvesicles and 5-FU resistance of human colon cancer cells. Int J Mol Sci. 2014;15(1):1392–401. https://doi.org/10.3390/ijms15011392.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Senfter D, Holzner S, Kalipciyan M, Staribacher A, Walzl A, Huttary N, et al. Loss of miR-200 family in 5-fluorouracil resistant colon cancer drives lymphendothelial invasiveness in vitro. Hum Mol Genet. 2015;24(13):3689–98. https://doi.org/10.1093/hmg/ddv113.

    Article  CAS  PubMed  Google Scholar 

  102. Soldevilla B, Rodriguez M, San Millan C, Garcia V, Fernandez-Perianez R, Gil-Calderon B, et al. Tumor-derived exosomes are enriched in Delta Np73, which promotes oncogenic potential in acceptor cells and correlates with patient survival. Hum Mol Genet. 2014;23(2):467–78. https://doi.org/10.1093/hmg/ddt437.

    Article  CAS  PubMed  Google Scholar 

  103. 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;6:6716. https://doi.org/10.1038/ncomms7716.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Samanta S, Rajasingh S, Drosos N, Zhou Z, Dawn B, Rajasingh J. Exosomes: new molecular targets of diseases. Acta Pharmacol Sin. 2018;39(4):501–13. https://doi.org/10.1038/aps.2017.162.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was supported by The National Natural Science Foundation of China (81560385, 81671542), The Medical Scientific and Technological Research Project of Henan Province (201702027), Youth Innovation Fund Project of The First Affiliated Hospital of Zhengzhou University (YNQN2017035), The China Postdoctoral Science Foundation (2017 M610462), The National Natural Science Foundation of Henan Province (182300410342) and The Health Commission Technology Talents Overseas Training Project of Henan Province (2018140).

Availability of data and materials

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

ZQS, ZYJ and WTY provided direction and guidance throughout the preparation of this manuscript. YXG, XJ wrote and edited the manuscript. JBL, DDF and WWW reviewed and made significant revisions to the manuscript. QBZ, CC and HJW collected and prepared the related papers. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Weitang Yuan, Zhenyu Ji or Zhenqiang Sun.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, Y., Ji, X., Liu, J. et al. Effects of exosomes on pre-metastatic niche formation in tumors. Mol Cancer 18, 39 (2019). https://doi.org/10.1186/s12943-019-0995-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12943-019-0995-1

Keywords