Open Access

Key role of exosomes derived from M2 macrophages in maintaining cancer cell stemness (Review)

  • Authors:
    • Weiqiong Zhang
    • Ruiping Zhou
    • Xin Liu
    • Lin You
    • Chang Chen
    • Xiaoling Ye
    • Jie Liu
    • Youde Liang
  • View Affiliations

  • Published online on: September 14, 2023     https://doi.org/10.3892/ijo.2023.5574
  • Article Number: 126
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer stem cells (CSCs) constitute a specific subset of cells found within tumors that are responsible for initiating, advancing and resisting traditional cancer treatments. M2 macrophages, also known as alternatively activated macrophages, contribute to the development and progression of cancer through their involvement in promoting angiogenesis, suppressing the immune system, supporting tumor growth and facilitating metastasis. Exosomes, tiny vesicles released by cells, play a crucial role in intercellular communications and have been shown to be associated with cancer development and progression by influencing the immune response; thus, they may serve as markers for diagnosis and prognosis. Currently, investigating the impact of exosomes derived from M2 macrophages on the maintenance of CSCs is a crucial area of research with the aim of developing novel therapeutic strategies to target this process and improve outcomes for individuals with cancer. Understanding the biological functions of exosomes derived from M2 macrophages and their involvement in cancer may lead to the formulation of novel diagnostic tools and treatments for this disease. By targeting M2 macrophages and the exosomes they secrete, promising prospects emerge for cancer treatment, given their substantial contribution to cancer development and progression. Further research is required to fully grasp the intricate interactions between CSCs, M2 macrophages and exosomes in cancer, and to identify fresh targets for cancer therapy. The present review explores the pivotal roles played by exosomes derived from M2 cells in maintaining the stem‑like properties of cancer cells.

1. Introduction

Cancer stem cells (CSCs) constitute a distinct set of cells found within a tumor that share similar properties to normal stem cells (1). These cells are important for cancer development and progression, as well as in making tumors resistant to chemotherapy and radiation therapy (2). CSCs exhibit self-renewal potential and can differentiate into various cell types found within the tumor, allowing them to continually regenerate the tumor and form more aggressive tumors, even after initial treatment has been administered. They have been identified in various types of cancers, including glioma, breast cancer, lung carcinoma, and leukemia (3-5). Due to their resistance to conventional cancer treatments, they are considered to be one of the contributing factors for tumor recurrence following treatment (3-5).

The CSC niche, similar to the adult stem cell niche, is a component of the tumor microenvironment (TME) that regulates stem cell activities through interactions between cells and secreted molecules (6). The TME consists of a variety of components, such as cytokine networks, immune cells, perivascular cells, fibroblasts, extracellular matrix (ECM) components, and endothelial cells (7). Tumor-associated macrophages (TAMs) are the most prevalent immune cells in the TME and can be recruited by CSCs to participate in TME formation, which can aid in CSC survival. M2 macrophages, which are alternatively activated macrophages, have a significant influence on cancer development and progression (8), unlike M1 macrophages, which are responsible for the immune response against infections and inflammation (8).

Exosomes are small extracellular vesicles that range from 40 to 160 nm in diameter and originate from endosomes (9). It has been shown that nearly all cells secrete exosomes, which can be found in various biofluids and cell culture media (10). Exosomes represent a novel mechanism for intercellular communication between donor and recipient cells, and individuals with cancer have been found to possess increased quantities of exosomes compared with healthy individuals, highlighting the potential role of exosome-mediated cellular and communication in cancer. This communication promotes tumor formation, angiogenesis, metastasis, progression, immune evasion, and drug resistance (11). Due to their favorable biocompatibility properties and capacity for customization to target specific cells, exosomes hold promise as carriers for therapeutic payloads like microRNAs (miRNAs/miRNAs), small interfering RNA (siRNAs), and small-molecule drugs. This potential creates possibilities for transforming conventional cancer treatment approaches (12). Nevertheless, the quest for suitable exosomal molecules and donor cells to establish an effective exosomal drug delivery system remains a substantial challenge.

Extensive research has been performed to understand the complex system of communication facilitated by exosomes within the TME (9). Within the TME, macrophages play a pivotal role in intercellular communication through the release of exosomes. Macrophages are broadly categorized into two types based on their activation status: Classically activated M1 macrophages and alternatively activated M2 macrophages, which are influenced by a range of stimuli (13). TAMs exhibit a mixed M1/M2 phenotype, and macrophage-derived exosomes may vary based on their parental cell properties. For example, exosomes derived from M2 macrophages may contain higher levels of specific miRNAs compared to those derived from M1 macrophages, thereby impacting cancer progression and drug resistance (14). Hence, understanding the specific exosomes secreted by distinct macrophage phenotypes may offer therapeutic opportunities. Macrophage-derived exosomes constitute a substantial proportion of blood-borne exosomes and these may be used as potential biomarkers for diagnosing cancer through minimally invasive liquid biopsies (15). Moreover, exosomes released by macrophages can trigger immune responses that restrain cancer progression, highlighting their possible application in anti-tumor treatments (16,17). Currently, the role of M2-derived exosomes in maintaining cancer cell stemness is an active area of investigation for developing novel therapeutic approaches to target this process and improve outcomes in cancer patients.

2. CSCs in tumor progression

CSCs are a subpopulation of tumor cells that exhibit self-renewal and differentiation abilities, similar to those of normal stem cells (5,18). CSCs are hypothesized to be responsible for initiating and maintaining tumor growth, as well as conferring resistance to chemotherapy and radiation therapy. These versatile cancer cells have the capacity to differentiate into various cell types found in tumors, which enables them to drive primary tumor growth and contribute to the development of new tumors (19). Various surface markers, such as CD34+/CD38−, are used to identify CSCs in a wide range of cancers (20). By way of their pluripotency, CSCs play a pivotal role in tumorigenesis, cellular proliferation, and metastasis. Furthermore, CSCs can self-renew, making them functionally immortal. Although only a small percentage of cancer cells exhibit stemness properties, they can differentiate into a range of cancer cell types that constitute the majority of tumor cells (18). CSCs are often more tumorigenic than non-stem cancer cells. Although chemotherapy and radiotherapy can effectively kill a significant portion of the tumor mass, CSCs are typically resistant to these treatments, making it difficult to achieve significant clinical improvement (2,4,21). CSCs also have the capacity to generate a wide range of cell types within a tumor, resulting in heterogeneous progeny (22). This diversity of phenotypes stems from the inherent plasticity of CSCs, allowing them to transition between different cell states or phenotypic states. These transitions can occur spontaneously or in response to signals from the tumor microenvironment, such as changes in oxygen levels, nutrient availability, or interactions with other cells and signaling molecules (22). The phenotypic variations exhibited by CSCs are extensive and can vary depending on the tumor type and context (22). There are several common phenotypic states observed in CSCs: i) Stem-like state: CSCs maintain stem cell-like properties, characterized by their ability to self-renew and differentiate into multiple cell lineages. These cells often show increased expression of stem cell markers and signaling pathways associated with stemness (23). ii) Differentiated state: CSCs can undergo partial or complete differentiation into various cell types present in the tumor, resembling the non-CSC population. This differentiation can lead to the formation of bulk tumor cells with limited self-renewal potential (24). iii) Hybrid state: CSCs can exhibit a hybrid phenotype that combines both stem-like and differentiated characteristics. These cells possess certain stem cell properties while also displaying markers or features associated with more differentiated cells. The hybrid state may confer increased resistance to therapies and enhanced metastatic potential (24). iv) Epithelial-to-mesenchymal transition (EMT): CSCs can undergo a process known as EMT, which is associated with increased invasiveness and metastatic potential. During EMT, CSCs lose epithelial characteristics and acquire mesenchymal traits, including enhanced motility, resistance to apoptosis, and ECM remodeling abilities (25). v) Metabolic plasticity: CSCs can adapt their metabolic profile to utilize different energy sources and survive in a range of different microenvironments. They can switch between glycolysis and oxidative phosphorylation (a process known as the Warburg effect), which provides them with a survival advantage under nutrient-deprived conditions (26). Understanding and targeting the cellular plasticity of CSCs is crucial for developing effective therapeutic strategies against cancer. The ability of CSCs to transition between different phenotypic states enables them to evade therapies, contribute to tumor heterogeneity, and drive tumor relapse and metastasis (25). Overall, the characteristics of CSCs offer a valuable avenue for comprehending cancer development and the potential treatment of various cancer types.

3. The role of M2 macrophages in tumor progression

Within the scientific community, an ongoing debate persists regarding the specific mechanism underlying the origin of macrophages. However, it is widely recognized that macrophages can be classified into two distinct lineages: Bone marrow-derived macrophages and tissue-resident macrophages (27,28). Tissue-resident macrophages develop during embryonic development and are self-sustaining within their specific area, while bone marrow-derived macrophages arise from monocytes differentiated by bone marrow progenitors (29). These monocytes migrate from the bloodstream to tissues during normal and inflammatory conditions and are activated by various factors. These macrophage populations have distinct distributions within the TME, with tissue-resident macrophages spreading to neighboring tumor cells early on, promoting EMT and increasing invasion (30). Furthermore, tissue-resident macrophages raise regulatory T-cell numbers to help tumor cells escape from the immune system (31). Hence, tissue-resident macrophages may present a promising target for treating tumors. There are various macrophage subtypes that can be characterized with specific markers.

Macrophages are a heterogeneous population of immune cells with a range of phenotypes and functions that actively regulate tumor progression. Among these, the M1 and M2 macrophage subtypes hold significant roles in tumor regulation. M1 macrophages exhibit a pro-inflammatory phenotype when exposed to Type 1 T helper cytokines, such as IFN-γ, and TNF-α. They secrete anti-tumor pro-inflammatory cytokines such as IL-8, TNF-α, IL-1β, and IFN-γ (32,33). Conversely, M2 macrophages are primarily activated by Type 1 T helper (Th2) cytokines, including IL-13 and IL-4, resulting in anti-inflammatory properties and tumorigenesis. M2 macrophages can be further classified into distinct subsets based on specific stimuli and markers. The M2a subset, characterized by CD206 and CD68, contributes to fibrosis, allergic responses, and parasite elimination. The M2b subset, identified by CD86 receptors, plays a vital role in immune responses (34,35). The M2c subset, distinguished by the expression of CD163 receptors, is induced by IL-10, TGF-β, or glucocorticoids and serves a critical function in anti-inflammatory processes (36,37). Finally, the M2d subset, associated with tumor progression, exhibits increased secretion of vascular endothelial growth factor (VEGF) and IL-10, along with reduced expression of TNF-α and IL-12 (38,39). Nonetheless, the precise mechanism underlying the programming of M2d macrophages remains a subject of controversy. Fig. 1 provides a depiction of the expression of markers associated with different subtypes of M2 macrophages.

Macrophages in the TME may have divergent effects on cancer progression based on their polarization status. Initially, macrophages may exhibit a pro-inflammatory response and inhibit tumor growth; however, the evidence supporting this remains limited (40). As a tumor expands, Th2 cells guide macrophages toward a pro-tumor phenotype, which promotes tumor development (41). M2 macrophages have been shown to regulate multiple aspects of tumorigenesis, such as angiogenesis, metastasis, and chemo-resistance. M2 macrophages can promote tumor cell intravasation and extravasation by secreting VEGF and epidermal growth factor (42,43). They also modulate tumor metastasis by regulating EMT and promoting ECM degradation. The majority of TME-associated macrophages tend to be M2, which creates an immunosuppressive microenvironment (44). An overview of the interactions between CSCs and M2 macrophages is shown in Fig. 2. Therefore, gaining insights into the characteristics of M2 macrophages can enhance our understanding of cancer states and enable the development of new approaches for inhibiting or eradicating cancer.

4. Characteristics and composition of macrophage-derived exosomes

The process by which exosomes are formed from macrophages follows a similar pattern to that observed in other cells. It is initiated by the inward budding of the cellular membrane, leading to the formation of endosomes (9). These endosomes then generate intraluminal vesicles (ILVs) within the cytoplasm, gradually transforming into multivesicular bodies (MVBs) (9). Throughout this process, the sorting of exosomal cargo can be influenced by various external factors. For example, in response to IL-4 stimuli, macrophage-derived exosomes selectively incorporate miRNAs (45). Moreover, macrophages activate the peroxisome proliferator-activated receptor γ pathway and transfer phosphatase and tensin homolog (PTEN) into exosomes when exposed to the microenvironment of apoptotic lung cancer cells undergoing irradiation (46). Additionally, the activation of the P2X7 signaling pathway triggered by extracellular ATP enables macrophages to transfer IL-1β and other proteins into exosomes, leading to an elevation in intracellular calcium levels (Fig. 3) (47,48).

Typically, exosomes from the inward budding of the cellular membrane can create endosomes, which develop ILVs that eventually mature into MVBs through cargo sorting and external factors. In the typical scenario, MVBs are predominantly degraded by lysosomes, and only a small fraction of them are released as exosomes through exocytosis facilitated by Rab proteins and small GTPases (49). However, when lysosomes in macrophages are defective, increased secretion of exosomes is observed (50), indicating the substantial reliance on macrophage-derived exosomes on lysosomal function. Conversely, autophagy has a diminishing effect on exosomal secretion since autophagosomes can merge with MVBs to form amphisomes, which can then be degraded by lysosomes (51). Conversely, reducing the expression of lysosome-associated membrane protein-2 and lysosome-associated membrane protein-1 hinders the fusion of amphisomes with lysosomes, resulting in an increased release of exosomes. Notably, not only endogenous factors but also exogenous stimuli, such as cellular stress impact the secretion of macrophage-derived exosomes (52). For example, macrophages release a higher number of exosomes upon stimulation by lipopolysaccharide (LPS), which upregulates the expression of Rab27b and Rab27a. However, this effect can be counteracted by IL-25 (53). In the tumor microenvironment, hypoxia can also enhance the release of macrophage-derived exosomes compared to physiological conditions (54). These exosomes become independent components of the tumor microenvironment and can affect other cells through various mechanisms. For instance, For example, LPS/IFN-γ-induced macrophage-derived exosomes can bind to extracellular endoplasmic reticulum aminopeptidase 1, which promotes macrophage phagocytosis and nitric oxide synthesis (55). Macrophage-derived exosome surface markers are specific to cell and/or tissue types and can determine the types of recipient cells (56). Different proteins, such as RAB27A and syntaxin 3, are used by macrophages from various tissues to regulate exosomal biogenesis and docking with recipient cells (57). Overall, these findings suggest that macrophage-derived exosomes play a role in cell-to-cell communication and have a wide range of effects depending on their contents and surface markers.

5. M2 macrophage-derived exosomes in cancer cell stemness

M2 macrophage-derived exosomes transfer various biomolecules, including growth factors, cytokines, miRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) (Table I) have been shown to promote self-renewal and survival of CSCs (58). They can also modulate the signaling pathways and molecular processes that regulate CSCs, thereby maintaining and expanding the cancer stem cell population (58).

Table I

Role of M2 macrophage-derived exosomal non-coding RNAs in regulating cancer progression.

Table I

Role of M2 macrophage-derived exosomal non-coding RNAs in regulating cancer progression.

Types of non-coding RNAsName of non-coding RNAsRelated target/signaling pathwayCancer type(Refs.)
miRNAmiR-155-5pZC3H12BColon cancer(65)
miR-155-5pBRG1Colon cancer(66)
miR-186-5pDLC1Colon cancer(152)
miR-21-5pBRG1Colon cancer(152)
miR-221-3pCDKN1BEpithelial ovarian cancer(89)
miR-487aTIA1Gastric cancer(84)
miR-588CYLDGastric cancer(85)
miR-27b-3pMLL4/PRDM1Glioblastoma(87)
miR-15aPI3K/AKT/mTOR signalingGlioma(93)
miR-92aPI3K/AKT/mTOR signalingGlioma(93)
miR-660-5pKLF3Hepatocellular carcinoma(88)
miR-222-3pPDLIM2/PFKLLaryngeal squamous cell carcinoma(153)
miR-1911-5pCELF2/ZBTB4 signalingLung cancer(77)
miR-942FOXO1Lung cancer(81)
miR-3917GRK6Lung cancer(78)
miR-501-3pWD repeat domain 82Lung cancer(80)
miR-155-3pWD repeat domain 82 Medulloblastoma(154)
miR-31-5pLATS2/Hippo signalingOral squamous cell carcinoma(91)
miR-221-3p SOCS3/JAK2/STAT3Osteosarcoma(90)
miR-193b-3pTRIM62Pancreatic cancer(70)
miR-21-5pKLF3Pancreatic cancer(76)
miR-155-5pE2F2Pancreatic cancer(68)
miR-221-5pE2F2Pancreatic cancer(68)
miR-365BTG2/FAK/AKTPancreatic cancer(71)
miR-501-3p TGFBR3-mediated
TGF-β signaling
Pancreatic cancer(73)
miR-21-5pPTEN/Akt signalingRenal cell carcinoma(155)
lncRNAlncMMPAGlycolysis pathwayHepatocellular carcinoma(106)
H19ULK1Bladder cancer(108)
lncRNA CRNDEPTENGastric cancer(115)
AGAP2-AS1 miR-296/Notch2signalingLung cancer(112)
AFAP1-AS1miR-26a/ATF2 signalingEsophageal cancer(113)
circRNACirc_0008253Not examinedGastric cancer(121)
Circ_0020256miR-432-5p/E2F3 signaling Cholangiocarcinoma(122)
Circ_0001610 miR-139-5p/cyclin
B1 signaling
Endometrial cancer(123)
Circ_TNFRSF21mMiR-3619-5p/ROCK signalingCutaneous squamous cell carcinoma(124)

[i] lncRNA, long non-coding RNA; ATF2, activating transcription factor 2; BRG1, BRM/SWI2-related gene 1; CDKN1B, cyclin-dependent kinase inhibitor 1B; CELF2/ZBTB4, CUGBP Elav-like family member 2/zinc finger and BTB domain containing 4; CYLD, cylindromatosis; DCL1, deleted in liver cancer 1; FOXO1, forkhead box O1; KLF3, Krüppel-like factor 3; LATS2, large tumor suppressor kinase 2; MLL4/PRDM1, mixed-lineage leukemia 4/positive regulatory domain I; PDLIM2/PFKL, PDZ and LIM domain 2/phosphofructo-1-kinase isozyme B; PTEN, phosphatase and tensin homolog; SOCS3, suppressor of cytokine signaling 3; TGFBR3, transforming growth factor beta receptor 3; TRIM62, tripartite motif containing 62; ULK1, Unc-51 like autophagy activating kinase 1; ZC3H12B, zinc finger CCCH-type containing 12B.

Additionally, M2 macrophage-derived exosomes have been shown to modify the behavior of immune cells, thereby suppressing the anti-tumor immune response (59). This may further promote the survival and expansion of the cancer stem cell populations and contribute to tumor progression.

miRNA-mediated regulation of cancer cell stemness

Exosomes, which mediate cell-to-cell interactions, possess the ability to exchange genetic molecules, such as miRNAs (60). miRNAs are key to the development, polarization, and metabolic control of macrophages (61-63). To maintain a balanced mRNA environment, exosomes can rapidly eliminate excessive miRNAs in an activation-dependent manner (45). Consequently, exosomes derived from macrophages stimulated with LPS or IL-4 exhibit noticeable enrichment of specific miRNAs (64). Numerous studies have demonstrated that alterations in exosomal miRNAs derived from macrophages can significantly impact important post-transcriptional control functions in neighboring cells, including cancer cells.

To date, a wide range of miRNAs sourced from exosomes of M2 macrophages have been implicated in the regulation of cancer stemness. In human colon cancer, exosomal miR-155-5p derived from M2 macrophages has been shown to augment the proliferation and anti-apoptotic capabilities of SW48 and HT29 cells. It also promotes immune escape by downregulating zinc finger CCCH-type containing 12B and upregulating IL-6, thereby increasing CD3+ T cell proliferation and the proportion of IFN-γ+ T cells (65). Another study revealed that the collaborative action of exosomal miR-155-5p and miR-21-5p from M2 macrophages promotes cell migration and invasion in colon cancer by suppressing BRG1 expression. (66); BRG1 is a crucial regulator in maintaining colorectal cancer stem cells (67). In pancreatic cancer, exosomal miR-155-5p and miR-221-5p derived from M2 macrophages were found to stimulate angiogenesis and pancreatic cancer growth by inhibiting E2F2 expression (68). E2F2 transcriptionally regulates multiple targets involved in various characteristics of CSCs, including proliferation, self-renewal, metastasis, and drug resistance (69). Furthermore, exosomal miR-193b-3p from M2 macrophages was observed to enhance the proliferation, migration, invasion, and glutamine uptake of pancreatic cancer cells by downregulating tripartite motif containing 62 (70). Similarly, exosomal miR-365 from M2 macrophages suppressed the expression of BTG2, activating the FAK/AKT pathway and promoting pancreatic cancer development (71). Inhibiting the FAK/AKT signaling pathway was found to reduce the viability of human breast cancer stem cells (72). Additionally, exosomal miR-501-3p derived from M2 macrophages was identified to hinder the tumor suppressor TGFβ receptor 3 gene, thereby facilitating the progression of pancreatic cancer through the activation of the TGF-β signaling pathway (73). It is important to note that the TGF-β signaling pathway has been demonstrated to enhance pancreatic cancer stemness (74,75). Interestingly, miR-21-5p from extracellular vesicles derived from M2 macrophages was found to promote the differentiation and activity of pancreatic cancer stem cells by suppressing Krüppel-like factor 3 (KLF3) expression (76). In lung cancer, exosomal miR-1911-5p from M2 macrophages facilitated cell migration and invasion in lung adenocarcinoma by downregulating zinc finger and BTB domain containing 4 expression, which is mediated by CUGBP Elav-like family member 2 (77). Additionally, exosomal miR-3917 from M2 macrophages promoted lung cancer progression by inhibiting G protein-coupled receptor kinase 6 (78), a factor involved in maintaining self-renewal of hematopoietic stem cells (79). Furthermore, exosomal miR-501-3p from M2 macrophages represses WD repeat domain 82, contributing to the progression of lung cancer (80). Notably, M2 macrophage-derived exosomal miR-942 suppresses forkhead box protein O1 (FOXO1) expression, promoting the progression of lung adenocarcinoma (81), FOXO1 acts as a key inhibitor of cancer cell stemness in various cancer types (82,83). In gastric cancer, exosomal miR-487a from M2 macrophages advances disease progression by suppressing TIA1 expression (84). Moreover, exosomal miR-588 from M2 macrophages contributes to cisplatin resistance in gastric cancer cells by partially inhibiting cylindromatosis expression (85), which regulates the proliferation of esophageal cancer stem-like cells through the cylindromatosis pathway (86).

In glioblastoma, M2 macrophage-derived exosomal miR-27b-3p represses mixed-lineage leukemia 4/positive regulatory domain 1 signaling, resulting in the activation of IL-33 and maintenance of stem-like properties in glioblastoma stem cells (87). Studies on hepatocellular carcinoma indicated that exosomal miR-27b-3p from M2 macrophages enhanced tumor development by suppressing KLF3 (88), whereas inhibition of KLF3 was found to stimulate the differentiation and activity of pancreatic cancer stem cells (76). In epithelial ovarian cancer, exosomal miR-221-3p from M2 macrophages promotes disease progression by suppressing cyclin-dependent kinase inhibitor 1B. Similarly, exosomal miR-221-3p derived from M2 macrophages enhance the growth and metastasis of osteosarcoma by repressing suppressor of cytokine signaling 3, activating JAK2/STAT3 signaling (89), which plays a critical role in maintaining cancer cell stemness (90). In oral squamous cell carcinoma, exosomal miR-31-5p from M2 macrophages hinders the tumor suppressor large tumor suppressor kinase 2 gene, facilitating cancer progression by inhibiting the Hippo signaling pathway (91), which promotes the transition EMT and the maintenance of cancer stem cells (92). Conversely, in glioma, M2 macrophage-derived exosomal miR-15a and miR-92a suppress cyclin D1 and RAP1B, respectively, resulting in the inhibition of cell migration and invasion via the PI3K/AKT/mTOR pathway (93). The PI3K/AKT/mTOR signaling pathway has been shown to regulate cancer stemness (94). Notably, exosomal miR-223 derived from macrophages exhibits divergent effects in different cancer types, despite its involvement in regulating the biological function of cancer stem cells. The expression levels of miR-223 vary across different cancer types, with increased expression in metastatic gastric and ovarian cancers, but decreased expression in hepatocellular and esophageal cancer (95,96). This indicates a contradictory role for miR-223 in cancer. Furthermore, exosomal miR-223 from macrophages can promote drug resistance in epithelial ovarian carcinoma cells by inhibiting PTEN expression (97), while in breast cancer cells, it can induce invasion and metastasis by suppressing Mef2c expression (98). However, conflicting findings suggest that exosomal miR-223 from macrophages inhibits cancer cell proliferation in hepatocellular cancer cells by downregulating stathmin 1 and insulin-like growth factor 1 receptor expression (99). It is possible that miR-223 induces macrophages to adopt either an anti-tumor or pro-tumor phenotype in different pathological conditions, leading to pleiotropic effects in cancer cells, exhibiting both suppressive and promotive roles.

lncRNA-mediated regulation of cancer cell stemness

lncRNAs are RNA molecules that are >200 nucleotides in length and lack protein-coding capacity. Despite their non-coding nature, they play crucial roles in various cellular processes, including regulation of gene expression, epigenetic modifications, chromatin remodeling, and mRNA processing. In recent years, extensive research has highlighted the significance of lncRNAs in cancer development and progression (100,101). Dysregulation of lncRNAs can disrupt normal cellular processes, leading to uncontrolled cell growth, survival, and metastasis, which are all characteristic features of cancer. Previous research has primarily focused on studying the functions and mechanisms of lncRNAs within individual cells, but there has been limited investigation into the role of lncRNAs carried by exosomes secreted by macrophages in facilitating communication between cells. However, recent studies have shown that exosomes released by macrophages can deliver a specific lncRNA called HIF-1α-stabilizing lncRNA to breast cancer cells. This delivery process then influences the glycolysis of cancer cells by interacting with a protein called prolyl hydroxylase domain 2, leading to the stabilization of HIF-1α (102). HIF-1α is known to play a role in the development of cancer stemness under conditions of hypoxia (103). These findings suggest that exosomal lncRNAs derived from macrophages have a positive impact on cancer stemness. Additionally, Yin et al (104) discovered that exosomal lncRNA SET-binding factor 2 antisense RNA 1 derived from M2 macrophages can be transferred to pancreatic cancer cells, promoting cancer progression by suppressing miR-122-5p and increasing the expression of a protein called X-linked inhibitor of apoptosis protein. Notably, miR-122-5p has been associated with cervical cancer stem cell self-renewal and differentiation (105). In hepatocellular carcinoma, exosomes derived from M2 macrophages play a role in promoting malignancy by transferring lncMMPA to tumor cells. This transfer inhibits miR-548 and leads to the upregulation of aldehyde dehydrogenase 1 family member A3 (ALDH1A3) expression (106), whereas inhibition of ALDH1A3 can impede cancer cell stemness (107). In bladder cancer, M2 macrophage-derived lncRNA H19 promotes autophagy in bladder cells by stabilizing Unc-51-like kinase 1 (108). Notably, lncRNA H19 has been extensively studied for its involvement in promoting cancer stemness across various cancer types (109-111). In lung cancer, exosomal lncRNA AGAP2 antisense RNA 1 derived from M2 macrophages enhances radiotherapy immunity by reducing miRNA-296 and increasing NOTCH2 expression. Activation of the Notch2 signaling pathway has been associated with promoting lung cancer stemness (112). In esophageal cancer, exosomal lncRNA AFAP1 antisense RNA 1 from M2 macrophages affects cell migration and metastasis by repressing miR-26a expression, thereby promoting activating transcription factor 2 activity (113). It is worth noting that miR-26a can regulate the activating enhancer binding Protein 2 α/Nanog signaling axis related to glioma cancer stemness (114). In gastric cancer, M2 macrophage-derived lncRNA colorectal neoplasia differentially expressed (CRNDE) contributes to cisplatin resistance (115). Furthermore, lncRNA CRNDE has been implicated in the regulation of biological characteristics of glioma stem cells (116). In summary, these findings shed light on the role of exosomal lncRNAs in mediating interactions between cells within TME.

circRNA-mediated regulation of cancer cell stemness

CircRNAs are a type of non-coding RNA characterized by a closed-loop structure and play crucial roles in gene regulation, development, and disease progression. Emerging studies suggest that circRNAs may participate in modulating CSCs across various cancer types (117,118). Specifically, certain circRNAs have been shown to influence the differentiation and self-renewal of CSCs in breast, glioblastoma, and colorectal cancer (119). Additionally, certain circRNAs have been found to modulate the expression of genes that are important for CSC maintenance and survival (120). One mechanism through which circRNAs exert their influence on CSCs is by interacting with miRNAs. By acting as miRNA sponges, circRNAs can bind to and sequester miRNAs, thereby regulating the expression of miRNA target genes crucial for CSC function. For example, Yu et al (121), revealed that macrophage-derived exosomes regulate gastric cancer cell resistance to oxaliplatin by encapsulating circ_0008253. However, the role of circ_0008253 in CSCs has not yet been investigated. Another study by Chen et al (122) revealed that M2 macrophage-derived exosomal circ_0020256 enhances cholangiocarcinoma progression by targeting miR-432-5p/E2F3 axis. Gu et al (123) identified that M2 macrophage-derived exosomal circ_0001610 reduces endometrial cancer radiosensitivity. Moreover, M2 macrophage-derived exosomal circ_TNFRSF21 facilitates angiogenesis in cutaneous squamous cell carcinoma through the regulation of miR-3619-5p/Rho-associated coiled-coil containing protein serine/threonine kinase signaling (124). Nevertheless, the specific roles of these circRNAs in CSCs remain to be investigated.

M2 macrophage-derived exosomal proteins in cancer stemness

Proteomics has been widely employed to analyze protein expression profiles (125). By utilizing proteomics, it was uncovered that treating macrophages with LPS/IFN-γ led to the upregulation of 24 proteins and the downregulation of eight proteins in macrophage-derived exosomes (55). Additionally, proteomic analysis of the culture media from the co-culture of colorectal cancer and Ana-1 cells identified an enrichment of proteins related to RNA processing, including several subunits of the 20S proteasome and ribosomal proteins, in M2 macrophage-derived exosomes (126). These findings suggest that proteins present in M2 macrophage-derived exosomes may contribute to tumor survival by degrading misfolded or denatured proteins in cancer cells. Nevertheless, to obtain a more accurate representation of the in vivo environment, it is crucial to isolate solid tumor tissue-derived exosomes, which requires suitable models and an adequate number of samples. Due to the sensitivity of macrophages and their exosomes to the surrounding conditions, the contents of macrophage-derived exosomes and macrophage phenotypes can vary under different conditions or in different types of cancer. The activities of proteins originating from macrophage-derived exosomes are primarily determined by the specific recipient cells they interact with. In the context of high-grade serous ovarian carcinoma, the inclusion of GATA binding protein 3 within macrophage-derived exosomes has been observed to facilitate the advancement of tumors (127). In gastric cancer, macrophages are the primary source of apolipoprotein E (ApoE), which is also abundant in their exosomes. When cancer cells internalize ApoE-containing exosomes derived from macrophages, they activate the PI3K/AKT signaling pathway, thereby promoting cancer cell migration (128).

Furthermore, macrophage-derived exosomes can influence the adaptability of cancer cells and contribute to metastasis. Kim et al (46) demonstrated that macrophages can deliver an increased amount of PTEN protein to recipient cells via exosomes when exposed to irradiated apoptotic lung cancer cells, thereby impeding EMT. Conversely, exosomal ADAM domain 15, a protein secreted by M2 macrophages, inhibits cancer cell migration and growth (129). In addition to macromolecular proteins, cytokines also play a critical role in cancer and can be found in M2 macrophage-derived exosomes. For instance, mouse macrophages release various cytokines in exosomes upon LPS stimulation (64). In one study, M2 macrophages cultured with apoptotic breast cancer cells after chemotherapy increased the production of IL-6 in their exosomes, which were then transferred to cancer cells (130). This process promoted cancer cell metastasis and proliferation by enhancing STAT phosphorylation. While cytokines have been extensively investigated in the context of non-exosomal secretion pathways, exploring whether these cytokines exhibit enhanced efficacy through exosomal pathways would be valuable.

Other macrophage-derived exosomal cargos in cancer stemness

Several studies have provided compelling evidence that macrophage-derived exosomes contain a diverse array of components, including miRNAs, lncRNAs, proteins, mRNA, tRNA and ribosomes (131). Recent research has highlighted the significant role of exosomal mitochondrial/nuclear DNA derived from cancer cells in tumor immunity (132). However, the presence of functional endogenous DNA in macrophage-derived exosomes remains uncertain. Nonetheless, artificial dsDNA has been detected in macrophage-derived exosomes in pancreatic cancer (14). Importantly, M2 macrophage-derived exosomes enriched with arginase-1 can stimulate the migration and proliferation of glioblastoma cells (133). Moreover, once macrophage-derived exosomes are released into the extracellular microenvironment, they may serve as primitive particles within the ECM (9). This is supported by the observation that components in macrophage-derived exosomes are capable of synthesizing thromboxane B2, thromboxane, and specific proteins (126). Further investigation is warranted to explore the potential independent functions of macrophage-derived exosomes separate from their parent cells.

6. Therapeutic agents targeting M2 macrophages for cancer treatment in clinical trials

Both preclinical and clinical investigations have underscored the therapeutic potential of targeting two signaling pathways, namely the C-C chemokine receptor type 2 (CCR2)-C-C Motif chemokine ligand 2 (CCL2) axis and the C-C Motif chemokine ligand 12 (CXCL12)-C-X-C Motif chemokine receptor 4 (CXCR4) pathway, to hinder the recruitment and infiltration of TAMs into the TME. These approaches hold promise for patients with solid tumors (134). For example, an anti-CCL2 antibody (carlumab) inhibited macrophage infiltration in mice and is presently being tested in clinical trials (NCT 00992186) for the treatment of solid tumors and metastatic castrate-resistant prostate cancer (135). However, carlumab alone only produces a temporary reduction in serum CCL2 levels without significant antitumor effects. Nevertheless, when combined with conventional chemotherapeutic regimens such as paclitaxel and carboplatin, it enhances the antitumor response. Similarly, clinical trials have revealed that inhibiting the CXCL12-CXCR4 signaling pathway can result in TAM exclusion and effective treatment of solid tumors (136). For example, a CXCR4 antagonist called (Plerixafor) impedes tumor angiogenesis by inhibiting the release of VEGF-A from TAMs and has been employed in the treatment of solid tumors and pediatric cancers (137-139). LY2510924, a CXCR4 antagonist, has also undergone clinical trials (NCT02737072) for the treatment of solid tumors (134).

TAM recruitment and polarization are heavily influenced by colony-stimulating factor-1 receptor (CSF-1R)/colony-stimulating factor-1 (CSF-1) pathway, thus, there have been endeavors to block this signal in TAMs for the treatment of solid tumors (140). Clinical trials have been conducted with Emactuzumab (RG7155) (NCT01494688). This treatment has demonstrated a reduction in CD163+/CSF-1R+ macrophages in diffuse-type giant cell tumors and an increase in CD8+/CD4+ ratio (134). Clinical trials are also underway to explore the combination of Emactuzumab with chemotherapy (NCT02760797) and immunotherapy (NCT02323191) for solid tumor treatment. Clinical trials have utilized CSF-1R-specific inhibitors for the treatment of solid tumors. Both CSF-1R inhibitors and antibodies have exhibited therapeutic improvements in clinical trials. An example of this is the utilization of the CSF-1R inhibitor BLZ945, either alone or in conjunction with anti-programmed cell death protein 1 (PD1) antibody immunotherapy (NCT02829723), which has demonstrated the ability to impede macrophage recruitment and promote a change in macrophage polarization towards phenotypes that are beneficial in combating tumors (134). The efficacy of this combination is presently being assessed in clinical trials as a potential treatment for advanced-stage solid tumors.

Although strategies that aim to eliminate or hinder the recruitment of TAMs can delay tumor progression, they often have systemic toxicities as they target all macrophages and can be quickly compensated by TAMs. Moreover, discontinuation of CCR2/CCL2 inhibitors can lead to accelerated metastasis in breast cancer due to the sudden release of monocytes that were previously trapped in the bone marrow (141). To overcome these limitations, alternative approaches have garnered interest, such as re-educating macrophages to adopt an anti-tumor phenotype. One such approach involves using inhibitors that block receptor signals on macrophages responsible for modulating phagocytosis. Tumor cells frequently overexpress a signaling molecule called CD47, which acts as a 'do not eat me' signal and suppresses macrophage phagocytic capacity by interacting with signal regulatory protein α (SIRPα). Anti-CD47 antibodies can disrupt the CD47-SIRPα axis, restoring the ability of macrophages to engulf tumors (126). Several conventional anti-CD47 antibodies have shown success in preclinical and clinical trials. For example, the anti-CD47 antibody Hu5F9-G4 inhibits the interaction between CD47 and SIRPα, promoting macrophage-mediated phagocytosis and the elimination of cancer cells; this antibody has been tested in clinical trials (NCT02953509 and NCT02216409) for the treatment of solid tumors and various hematological malignancies (142). Additionally, polypeptides or recombinant proteins derived from SIRPα, such as engineered high-affinity SIRPα proteins, can act as decoys by binding to CD47 and disrupting CD47-SIRPα signaling. Studies have demonstrated that the recombinant protein TTI-621, consisting of the N-terminal domain of SIRPα fused to human IgG1, suppresses tumor growth by enhancing macrophage-mediated phagocytosis of solid tumor cells (143). Currently, TTI-621 is undergoing clinical investigation for the treatment of solid tumors (NCT02663518 and NCT02890368). However, CD47 is expressed on both healthy and tumor cells. Therefore, targeting CD47 will inevitably lead to the elimination of healthy cells that express CD47, including red blood cells and thrombocytes. The unintended consequences of this approach, such as thrombocythemia and anemia, result in a reduced maximum tolerated dose during clinical trials. Consequently, these off-target effects limit the impact on the tumor (144).

CD40, a member of the TNF receptor superfamily, is expressed on various antigen-presenting cells and certain tumor cells. Activation of TAMs by agonistic anti-CD40 antibodies has been shown to stimulate the secretion of pro-inflammatory cytokines such as nitric oxide and TNF-α, activating effector T cells and restoring tumor immune surveillance. Selicrelumab in combination with immunotherapy, such as atezolizumab, has been tested in clinical trials (NCT02304393) for the treatment of solid tumors and has been demonstrated to significantly enhance macrophage phagocytic activity (134).

Toll-like receptor (TLR) activation is essential for stimulating the innate immune response of macrophages, driving them toward the M1 phenotype associated with anti-tumor activity. The activation of multiple TLR signals enhances macrophage phagocytic activity and promotes anti-tumor responses. For example, TLR4 and TLR5 agonists have been observed to polarize a greater number of CD206+ M2 TAMs towards the CD86+ M1 phenotype, effectively suppressing tumor growth without significant toxicity (145). Clinical trials have evaluated the TLR9 agonist, IMO-2125, for the treatment of refractory solid tumors and metastatic melanoma (NCT04126876 and NCT03052205), resulting in macrophage polarization towards an anti-tumor phenotype and subsequent tumor regression (134). However, TLR agonists can upregulate the expression of programmed death-ligand 1 (PD-L1) in macrophages, limiting the anti-tumor response. To overcome this limitation, the combination of IMO-2125 with immunotherapy, such as ipilimumab, has been explored to enhance the effectiveness of cancer treatment. Additionally, SD101, another TLR9 agonist, is currently undergoing clinical trials (NCT03007732) in combination with PD-1 blockade to augment therapeutic efficacy (134). Table II provides a summary of ongoing clinical trials investigating therapeutic agents targeting M2 macrophages for cancer treatment.

Table II

Therapeutic agents targeting M2 macrophages for cancer treatment in clinical trials.

Table II

Therapeutic agents targeting M2 macrophages for cancer treatment in clinical trials.

Type of compoundCompoundsTumor typeClinical trialClinical phase
CCL2 inhibitorCarlumabSolid tumorsNCT01204996I
CD40 agonistSelicrelumabSolid tumorsNCT02304393I
SEA-CD40Solid tumorsNCT02376699I
CD47-SIRPa inhibitorHu5F9-G4Advanced solid malignanciesNCT02216409I
CSF-1R antagonistsSNFX-6352Advanced solid tumorsNCT03238027I
CSF-1R antibodyEmactuzumabAdvanced solid tumorsNCT01494688I
FPA008Advanced solid tumorsNCT02526017I
CSF-1R inhibitorPLX3397Advanced solid tumorsNCT01596751II
ARRY-382Advanced solid tumorsNCT02880371II
PexidartinibAdvanced solid tumorsNCT02734433I
BLZ945Advanced solid tumorsNCT02829723I
JNJ-40346527Prostate cancerNCT03177460I
IMC-CS4Advanced solid tumorsNCT01346358I
PXL7486Advanced solid tumorsNCT01804530I
AMG 820Solid tumorsNCT01444404I
CXCR4 antagonistPlerixaforSolid tumorsNCT01225419II
CXCR4 antagonist peptideLY2510924Solid tumorsNCT02737072I
IL-1R antagonistAnakinraAdvanced solid tumorsNCT01624766I
PI3Kg inhibitorIPI-549Advanced solid tumorsNCT02637531Ib
SIRPa-IgG1 FcTTI-621Solid tumorsNCT02663518I
TLR4 agonistGSK1795091Advanced solid tumorsNCT03447314I
TLR7/8 agonistTelratolimod (MEDI9197)Solid tumorsNCT02556463I
TLR9 agonistIMO-2125Refractory solid tumors, metastatic melanomaNCT03052205I
SD101Solid tumorsNCT03007732II
Vasculature-modulating agent Ang2/VEGFVanucizumabAdvanced/metastatic solid tumorsNCT02665416I
Vitamin-D-binding protein, macrophage-activating factorEF-022 (Efranat)Solid tumorsNCT02052492I

[i] Ang2, angiopoietin-2; CCL12, C-C motif chemokine ligand 2; CSF-1R, colony stimulating factor 1 receptor; CXCR4, C-X-C motif chemokine receptor 4; IL-1R, interleukin-1 receptor; SIRPa, signal regulatory protein alpha; TLR4, Toll Like receptor 4; TLR7/8, Toll like receptor 7/8; TLR9, Toll like receptor 9; VEGF, vascular endothelial growth factor.

7. Possible therapeutic approaches involving exosomes derived from macrophages in cancer treatment

The utilization of macrophage-derived exosomes in cancer therapy is potentially significant. These exosomes have the ability to deliver targeted drugs and nanomaterials to specific recipient cells through cargo transfer. They exhibit biocompatibility and can facilitate drug transport across natural barriers, including the blood-brain barrier, enabling the delivery of brain-derived neurotrophic factors for the treatment of central nervous system diseases and brain tumor therapy while minimizing toxicity (146). Furthermore, macrophage-derived exosomes have the potential to mitigate adverse reactions in drug therapy. Studies have demonstrated that cisplatin-loaded exosomes derived from M1 macrophages enhance the anti-cancer effects of a drug by suppressing cancer cell proliferation, inducing apoptosis, and improving drug sensitivity (147). Additionally, macrophages, particularly M1 macrophages, can transport paclitaxel and overcome drug resistance mechanisms, resulting in potent anti-tumor effects (148).

Exploring the biogenesis and composition of macrophage-derived exosomes holds promise for advancing anti-tumor therapies. Inhibiting exosome formation and secretion may offer the potential for anti-tumor treatment. Exosomes released by macrophages can induce cancer cells to express PD-L1 proteins, which play a crucial role in tumor immune escape. However, the use of GW4869, an inhibitor of exosomal secretion, can counteract this induction (149,150). Inhibiting exosome biogenesis in vivo may have implications for normal cellular functions, potentially leading to adverse effects on other intracellular transport processes. Therefore, it is crucial to exercise caution and conduct thorough investigations when developing cancer therapeutic strategies targeting exosome biogenesis. In terms of composition, ovatodiolide, a macrocyclic bioactive compound, has demonstrated its ability to reduce the abundance of M2 macrophage-derived exosomal miR-21, consequently suppressing bladder carcinogenesis (151). This discovery suggests that macrophage-derived exosomal miRNAs hold promising clinical potential for cancer treatment. In conclusion, the development of therapeutic approaches for the complex roles of macrophage-derived exosomes should prioritize optimizing treatment efficacy while minimizing adverse reactions during the design of clinical trials.

8. Conclusions and future perspectives

M2 macrophages and the exosomes they release play a vital role in supporting CSCs and facilitating cancer progression. Through secretion of growth factors, cytokines, and immunosuppressive substances, M2 macrophages contribute to tumor growth and progression. These M2-derived exosomes further promote tumor development by transferring genetic material and influencing the immune response against cancer. The exploration of M2 cell-derived exosomes holds significant implications for the development of novel approaches in cancer treatment. Two potential avenues for therapeutic intervention involve targeting M2 macrophages and their exosomes, as well as utilizing exosome-based therapies. By directing efforts towards M2 macrophages and their exosomes, it becomes possible to limit their support to the tumor and stimulate an immune response against cancer. Additionally, exosomes can serve as a therapeutic tool by delivering therapeutic agents to cancer cells and modulating the immune response. Further research is required to fully comprehend the role of M2-derived exosomes in cancer and to develop effective therapeutic strategies. Specifically, there is a need to obtain deeper insights into the mechanisms by which M2-derived exosomes contribute to the maintenance of cancer stem cells and to develop targeted therapeutic strategies focusing on M2-derived exosomes in cancer due to their critical involvement in disease progression. In conclusion, studying M2 cell-derived exosomes in cancer has the potential to offer a better understanding of the mechanisms driving cancer progression and to guide the development of innovative therapeutic approaches. However, extensive research is essential to gain a comprehensive understanding of the role of M2 cell-derived exosomes in cancer.

Availability of data and materials

Not applicable.

Authors' contributions

YL, WZ and RZ were involved in the conceptualization of the study. XL, LY, CC, JL and XY were involved in the preparation of the figures and tables, and edited the manuscript. WZ, JL and RZ were involved in the curation of the data for inclusion in the review. WZ and YL were involved in the writing and preparation of the original draft of the manuscript. YL was involved in funding acquisition. All authors have read and agreed to the published version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the Open Research Fund Program of Hubei-MOST KLOS & KLOBME (grant no. 202203) and the Science and Technology Project of Yantian District in Shenzhen City, Guangdong Province, China (grant no. YTWS20220206).

References

1 

Clevers H: The cancer stem cell: Premises, promises and challenges. Nat Med. 17:313–319. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Nassar D and Blanpain C: Cancer stem cells: Basic concepts and therapeutic Implications. Annu Rev Pathol. 11:47–76. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Dawood S, Austin L and Cristofanilli M: Cancer stem cells: Implications for cancer therapy. Oncology (Williston Park). 28:1101–1107. 11102014.PubMed/NCBI

4 

Vlashi E and Pajonk F: Cancer stem cells, cancer cell plasticity and radiation therapy. Semin Cancer Biol. 31:28–35. 2015. View Article : Google Scholar

5 

Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S and Kossatz-Boehlert U: Cancer stem cells-origins and biomarkers: Perspectives for targeted personalized therapies. Front Immunol. 11:12802020. View Article : Google Scholar : PubMed/NCBI

6 

Plaks V, Kong N and Werb Z: The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 16:225–238. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Neophytou CM, Panagi M, Stylianopoulos T and Papageorgis P: The role of tumor microenvironment in cancer metastasis: Molecular mechanisms and therapeutic opportunities. Cancers (Basel). 13:20532021. View Article : Google Scholar : PubMed/NCBI

8 

Liu J, Geng X, Hou J and Wu G: New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 21:3892021. View Article : Google Scholar : PubMed/NCBI

9 

Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI

10 

Zhou B, Xu K, Zheng X, Chen T, Wang J, Song Y, Shao Y and Zheng S: Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct Target Ther. 5:1442020. View Article : Google Scholar : PubMed/NCBI

11 

Nicolini A, Ferrari P and Biava PM: Exosomes and cell communication: From tumour-derived exosomes and their role in tumour progression to the use of exosomal cargo for cancer treatment. Cancers (Basel). 13:8222021. View Article : Google Scholar : PubMed/NCBI

12 

Zhang Y, Liu Q, Zhang X, Huang H, Tang S, Chai Y, Xu Z, Li M, Chen X, Liu J and Yang C: Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J Nanobiotechnology. 20:2792022. View Article : Google Scholar : PubMed/NCBI

13 

Long KB, Collier AI and Beatty GL: Macrophages: Key orchestrators of a tumor microenvironment defined by therapeutic resistance. Mol Immunol. 110:3–12. 2019. View Article : Google Scholar

14 

Binenbaum Y, Fridman E, Yaari Z, Milman N, Schroeder A, Ben David G, Shlomi T and Gil Z: Transfer of miRNA in macrophage-derived exosomes induces drug resistance in pancreatic adenocarcinoma. Cancer Res. 78:5287–5299. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Ismail N, Wang Y, Dakhlallah D, Moldovan L, Agarwal K, Batte K, Shah P, Wisler J, Eubank TD, Tridandapani S, et al: Macrophage microvesicles induce macrophage differentiation and miR-223 transfer. Blood. 121:984–995. 2013. View Article : Google Scholar :

16 

Behzadi E, Hosseini HM, Halabian R and Fooladi AAI: Macrophage cell-derived exosomes/staphylococcal enterotoxin B against fibrosarcoma tumor. Microb Pathog. 111:132–138. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Cheng L, Wang Y and Huang L: Exosomes from M1-polarized macrophages potentiate the cancer vaccine by creating a pro-inflammatory microenvironment in the lymph node. Mol Ther. 25:1665–1675. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Batlle E and Clevers H: Cancer stem cells revisited. Nat Med. 23:1124–1134. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Akbar Samadani A, Keymoradzdeh A, Shams S, Soleymanpour A, Elham Norollahi S, Vahidi S, Rashidy-Pour A, Ashraf A, Mirzajani E, Khanaki K, et al: Mechanisms of cancer stem cell therapy. Clin Chim Acta. 510:581–592. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Zhao W, Li Y and Zhang X: Stemness-related markers in cancer. Cancer Transl Med. 3:87–95. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Babaei G, Aziz SG and Jaghi NZZ: EMT, cancer stem cells and autophagy; the three main axes of metastasis. Biomed Pharmacother. 133:1109092021. View Article : Google Scholar

22 

Das PK, Pillai S, Rakib MA, Khanam JA, Gopalan V, Lam AKY and Islam F: Plasticity of cancer stem cell: Origin and role in disease progression and therapy resistance. Stem Cell Rev Rep. 16:397–412. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Huang T, Song X, Xu D, Tiek D, Goenka A, Wu B, Sastry N, Hu B and Cheng SY: Stem cell programs in cancer initiation, progression, and therapy resistance. Theranostics. 10:8721–8743. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Zhu P and Fan Z: Cancer stem cells and tumorigenesis. Biophys Rep. 4:178–188. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Chen W, Dong J, Haiech J, Kilhoffer MC and Zeniou M: Cancer stem cell quiescence and plasticity as major challenges in cancer therapy. Stem Cells Int. 2016:17409362016. View Article : Google Scholar : PubMed/NCBI

26 

Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, Wang X, Dong Z, Chen F and Cui H: Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 5:82020. View Article : Google Scholar : PubMed/NCBI

27 

Shi C and Pamer EG: Monocyte recruitment during infection and inflammation. Nat Rev Immunol. 11:762–774. 2011. View Article : Google Scholar : PubMed/NCBI

28 

van Furth R and Cohn ZA: The origin and kinetics of mononuclear phagocytes. J Exp Med. 128:415–435. 1968. View Article : Google Scholar : PubMed/NCBI

29 

Epelman S, Lavine KJ and Randolph GJ: Origin and functions of tissue macrophages. Immunity. 41:21–35. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Casanova-Acebes M, Dalla E, Leader AM, LeBerichel J, Nikolic J, Morales BM, Brown M, Chang C, Troncoso L, Chen ST, et al: Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature. 595:578–584. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Gratchev A, Schledzewski K, Guillot P and Goerdt S: Alternatively activated antigen-presenting cells: Molecular repertoire, immune regulation, and healing. Skin Pharmacol Appl Skin Physiol. 14:272–279. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Orecchioni M, Ghosheh Y, Pramod AB and Ley K: Macrophage polarization: Different gene signatures in M1(LPS+) vs classically and M2(LPS-) vs alternatively activated macrophages. Front Immunol. 10:10842019. View Article : Google Scholar

33 

Tong Y, Guo YJ, Zhang Q, Bi HX, Kai K and Zhou RY: Combined treatment with dihydrotestosterone and lipopolysaccharide modulates prostate homeostasis by upregulating TNF-α from M1 macrophages and promotes proliferation of prostate stromal cells. Asian J Androl. 24:513–520. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Hu W, Lin J, Lian X, Yu F, Liu W, Wu Y, Fang X, Liang X and Hao W: M2a and M2b macrophages predominate in kidney tissues and M2 subpopulations were associated with the severity of disease of IgAN patients. Clin Immunol. 205:8–15. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Wen Y, Lu X, Ren J, Privratsky JR, Yang B, Rudemiller NP, Zhang J, Griffiths R, Jain MK, Nedospasov SA, et al: KLF4 in macrophages attenuates TNFα-mediated kidney injury and fibrosis. J Am Soc Nephrol. 30:1925–1938. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Loegl J, Hiden U, Nussbaumer E, Schliefsteiner C, Cvitic S, Lang I, Wadsack C, Huppertz B and Desoye G: Hofbauer cells of M2a, M2b and M2c polarization may regulate feto-placental angiogenesis. Reproduction. 152:447–455. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Lurier EB, Dalton D, Dampier W, Raman P, Nassiri S, Ferraro NM, Rajagopalan R, Sarmady M and Spiller KL: Transcriptome analysis of IL-10-stimulated (M2c) macrophages by next-generation sequencing. Immunobiology. 222:847–856. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Wang Q, Ni H, Lan L, Wei X, Xiang R and Wang Y: Fra-1 protooncogene regulates IL-6 expression in macrophages and promotes the generation of M2d macrophages. Cell Res. 20:701–712. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Ferrante CJ, Pinhal-Enfield G, Elson G, Cronstein BN, Hasko G, Outram S and Leibovich SJ: The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL-4Rα) signaling. Inflammation. 36:921–931. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Cassetta L and Pollard JW: Targeting macrophages: Therapeutic approaches in cancer. Nat Rev Drug Discov. 17:887–904. 2018. View Article : Google Scholar : PubMed/NCBI

41 

DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N and Coussens LM: CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 16:91–102. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER, Graf T, Pollard JW, Segall J and Condeelis J: A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res. 64:7022–7029. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Esser S, Lampugnani MG, Corada M, Dejana E and Risau W: Vascular endothelial growth factor induces VE-cadherin tyrosine phosphorylation in endothelial cells. J Cell Sci. 111:1853–1865. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Su S, Liu Q, Chen J, Chen J, Chen F, He C, Huang D, Wu W, Lin L, Huang W, et al: A positive feedback loop between mesenchymal-like cancer cells and macrophages is essential to breast cancer metastasis. Cancer Cell. 25:605–620. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Squadrito ML, Baer C, Burdet F, Maderna C, Gilfillan GD, Lyle R, Ibberson M and De Palma M: Endogenous RNAs modulate microRNA sorting to exosomes and transfer to acceptor cells. Cell Rep. 8:1432–1446. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Kim YB, Ahn YH, Jung JH, Lee YJ, Lee JH and Kang JL: Programming of macrophages by UV-irradiated apoptotic cancer cells inhibits cancer progression and lung metastasis. Cell Mol Immunol. 16:851–867. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Välimäki E, Cypryk W, Virkanen J, Nurmi K, Turunen PM, Eklund KK, Åkerman KE, Nyman TA and Matikainen S: Calpain activity is essential for ATP-driven unconventional vesicle-mediated protein secretion and inflammasome activation in human macrophages. J Immunol. 197:3315–3325. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Qu Y, Franchi L, Nunez G and Dubyak GR: Nonclassical IL-1 beta secretion stimulated by P2X7 receptors is dependent on inflammasome activation and correlated with exosome release in murine macrophages. J Immunol. 179:1913–1925. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Pegtel DM and Gould SJ: Exosomes. Annu Rev Biochem. 88:487–514. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Yang Q, Nanayakkara GK, Drummer C, Sun Y, Johnson C, Cueto R, Fu H, Shao Y, Wang L, Yang WY, et al: Low-intensity ultrasound-induced anti-inflammatory effects are mediated by several new mechanisms including gene induction, immunosuppressor cell promotion, and enhancement of exosome biogenesis and docking. Front Physiol. 8:8182017. View Article : Google Scholar : PubMed/NCBI

51 

Xu J, Camfield R and Gorski SM: The interplay between exosomes and autophagy-partners in crime. J Cell Sci. 131:jcs2152102018. View Article : Google Scholar

52 

Babuta M, Furi I, Bala S, Bukong TN, Lowe P, Catalano D, Calenda C, Kodys K and Szabo G: Dysregulated autophagy and lysosome function are linked to exosome production by Micro-RNA 155 in alcoholic liver disease. Hepatology. 70:2123–2141. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Li ZG, Scott MJ, Brzóska T, Sundd P, Li YH, Billiar TR, Wilson MA, Wang P and Fan J: Lung epithelial cell-derived IL-25 negatively regulates LPS-induced exosome release from macrophages. Mil Med Res. 5:242018.PubMed/NCBI

54 

Brahimi-Horn MC, Chiche J and Pouysségur J: Hypoxia and cancer. J Mol Med (Berl). 85:1301–1307. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Goto Y, Ogawa Y, Tsumoto H, Miura Y, Nakamura TJ, Ogawa K, Akimoto Y, Kawakami H, Endo T, Yanoshita R and Tsujimoto M: Contribution of the exosome-associated form of secreted endoplasmic reticulum aminopeptidase 1 to exosome-mediated macrophage activation. Biochim Biophys Acta Mol Cell Res. 1865:874–888. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Sancho-Albero M, Navascués N, Mendoza G, Sebastián V, Arruebo M, Martín-Duque P and Santamaría J: Exosome origin determines cell targeting and the transfer of therapeutic nanoparticles towards target cells. J Nanobiotechnology. 17:162019. View Article : Google Scholar : PubMed/NCBI

57 

Lai B, Wang J, Fagenson A, Sun Y, Saredy J, Lu Y, Nanayakkara G, Yang WY, Yu D, Shao Y, et al: Twenty novel disease group-specific and 12 new shared macrophage pathways in eight groups of 34 diseases including 24 inflammatory organ diseases and 10 types of tumors. Front Immunol. 10:26122019. View Article : Google Scholar : PubMed/NCBI

58 

Bryl R, Piwocka O, Kawka E, Mozdziak P, Kempisty B and Knopik-Skrocka A: Cancer stem cells-the insight into non-coding RNAs. Cells. 11:36992022. View Article : Google Scholar : PubMed/NCBI

59 

Zheng N, Wang T, Luo Q, Liu Y, Yang J, Zhou Y, Xie G, Ma Y, Yuan X and Shen L: M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance. Oncoimmunology. 12:22109592023. View Article : Google Scholar : PubMed/NCBI

60 

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Roy S: miRNA in macrophage development and function. Antioxid Redox Signal. 25:795–804. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Li H, Jiang T, Li MQ, Zheng XL and Zhao GJ: Transcriptional regulation of macrophages polarization by MicroRNAs. Front Immunol. 9:11752018. View Article : Google Scholar : PubMed/NCBI

63 

Yao Q, Song Z, Wang B and Zhang JA: Emerging roles of microRNAs in the metabolic control of immune cells. Cancer Lett. 433:10–17. 2018. View Article : Google Scholar : PubMed/NCBI

64 

McDonald MK, Tian Y, Qureshi RA, Gormley M, Ertel A, Gao R, Aradillas Lopez E, Alexander GM, Sacan A, Fortina P and Ajit SK: Functional significance of macrophage-derived exosomes in inflammation and pain. Pain. 155:1527–1539. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Ma YS, Wu TM, Ling CC, Yu F, Zhang J, Cao PS, Gu LP, Wang HM, Xu H, Li L, et al: M2 macrophage-derived exosomal microRNA-155-5p promotes the immune escape of colon cancer by downregulating ZC3H12B. Mol Ther Oncolytics. 20:484–498. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Lan J, Sun L, Xu F, Liu L, Hu F, Song D, Hou Z, Wu W, Luo X, Wang J, et al: M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 79:146–158. 2019. View Article : Google Scholar

67 

Yoshikawa T, Fukuda A, Omatsu M, Namikawa M, Sono M, Fukunaga Y, Masuda T, Araki O, Nagao M, Ogawa S, et al: Brg1 is required to maintain colorectal cancer stem cells. J Pathol. 255:257–269. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Yang Y, Guo Z, Chen W, Wang X, Cao M, Han X, Zhang K, Teng B, Cao J, Wu W, et al: M2 macrophage-derived exosomes promote angiogenesis and growth of pancreatic ductal adenocarcinoma by targeting E2F2. Mol Ther. 29:1226–1238. 2021. View Article : Google Scholar :

69 

Xie D, Pei Q, Li J, Wan X and Ye T: Emerging role of E2F family in cancer stem cells. Front Oncol. 11:7231372021. View Article : Google Scholar : PubMed/NCBI

70 

Zhang K, Li YJ, Peng LJ, Gao HF, Liu LM and Chen H: M2 macrophage-derived exosomal miR-193b-3p promotes progression and glutamine uptake of pancreatic cancer by targeting TRIM62. Biol Direct. 18:12023. View Article : Google Scholar : PubMed/NCBI

71 

Li X, Xu H, Yi J, Dong C, Zhang H, Wang Z, Miao L and Zhou W: miR-365 secreted from M2 Macrophage-derived extracellular vesicles promotes pancreatic ductal adenocarcinoma progression through the BTG2/FAK/AKT axis. J Cell Mol Med. 25:4671–4683. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Thakur R, Trivedi R, Rastogi N, Singh M and Mishra DP: Inhibition of STAT3, FAK and Src mediated signaling reduces cancer stem cell load, tumorigenic potential and metastasis in breast cancer. Sci Rep. 5:101942015. View Article : Google Scholar : PubMed/NCBI

73 

Yin Z, Ma T, Huang B, Lin L, Zhou Y, Yan J, Zou Y and Chen S: Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-β signaling pathway. J Exp Clin Cancer Res. 38:3102019. View Article : Google Scholar

74 

Katoh M and Katoh M: WNT signaling and cancer stemness. Essays Biochem. 66:319–331. 2022. View Article : Google Scholar : PubMed/NCBI

75 

Tang Q, Chen J, Di Z, Yuan W, Zhou Z, Liu Z, Han S, Liu Y, Ying G, Shu X and Di M: TM4SF1 promotes EMT and cancer stemness via the Wnt/β-catenin/SOX2 pathway in colorectal cancer. J Exp Clin Cancer Res. 39:2322020. View Article : Google Scholar

76 

Chang J, Li H, Zhu Z, Mei P, Hu W, Xiong X and Tao J: microRNA-21-5p from M2 macrophage-derived extracellular vesicles promotes the differentiation and activity of pancreatic cancer stem cells by mediating KLF3. Cell Biol Toxicol. 38:577–590. 2022. View Article : Google Scholar :

77 

Guan B, Dai X, Zhu Y and Geng Q: M2 macrophage-derived exosomal miR-1911-5p promotes cell migration and invasion in lung adenocarcinoma by down-regulating CELF2-activated ZBTB4 expression. Anticancer Drugs. 34:238–247. 2023. View Article : Google Scholar : PubMed/NCBI

78 

Song S, Zhao Y, Wang X, Tong X, Chen X and Xiong Q: M2 macrophages-derived exosomal miR-3917 promotes the progression of lung cancer via targeting GRK6. Biol Chem. 404:41–57. 2022. View Article : Google Scholar : PubMed/NCBI

79 

Le Q, Yao W, Chen Y, Yan B, Liu C, Yuan M, Zhou Y and Ma L: GRK6 regulates ROS response and maintains hematopoietic stem cell self-renewal. Cell Death Dis. 7:e24782016. View Article : Google Scholar : PubMed/NCBI

80 

Lei J, Chen P, Zhang F, Zhang N, Zhu J, Wang X and Jiang T: M2 macrophages-derived exosomal microRNA-501-3p promotes the progression of lung cancer via targeting WD repeat domain 82. Cancer Cell Int. 21:912021. View Article : Google Scholar : PubMed/NCBI

81 

Wei K, Ma Z, Yang F, Zhao X, Jiang W, Pan C, Li Z, Pan X, He Z, Xu J, et al: M2 macrophage-derived exosomes promote lung adenocarcinoma progression by delivering miR-942. Cancer Lett. 526:205–216. 2022. View Article : Google Scholar

82 

Yu JM, Sun W, Wang ZH, Liang X, Hua F, Li K, Lv XX, Zhang XW, Liu YY, Yu JJ, et al: TRIB3 supports breast cancer stemness by suppressing FOXO1 degradation and enhancing SOX2 transcription. Nat Commun. 10:57202019. View Article : Google Scholar : PubMed/NCBI

83 

Firat E and Niedermann G: FoxO proteins or loss of functional p53 maintain stemness of glioblastoma stem cells and survival after ionizing radiation plus PI3K/mTOR inhibition. Oncotarget. 7:54883–54896. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Yang X, Cai S, Shu Y, Deng X, Zhang Y, He N, Wan L, Chen X, Qu Y and Yu S: Exosomal miR-487a derived from m2 macrophage promotes the progression of gastric cancer. Cell Cycle. 20:434–444. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Cui HY, Rong JS, Chen J, Guo J, Zhu JQ, Ruan M, Zuo RR, Zhang SS, Qi JM and Zhang BH: Exosomal microRNA-588 from M2 polarized macrophages contributes to cisplatin resistance of gastric cancer cells. World J Gastroenterol. 27:6079–6092. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Xu DD, Zhou PJ, Wang Y, Zhang L, Fu WY, Ruan BB, Xu HP, Hu CZ, Tian L, Qin JH, et al: Reciprocal activation between STAT3 and miR-181b regulates the proliferation of esophageal cancer stem-like cells via the CYLD pathway. Mol Cancer. 15:402016. View Article : Google Scholar : PubMed/NCBI

87 

Zhao G, Ding L, Yu H, Wang W, Wang H, Hu Y, Qin L, Deng G, Xie B, Li G and Qi L: M2-like tumor-associated macrophages transmit exosomal miR-27b-3p and maintain glioblastoma stem-like cell properties. Cell Death Discov. 8:3502022. View Article : Google Scholar : PubMed/NCBI

88 

Tian B, Zhou L, Wang J and Yang P: miR-660-5p-loaded M2 macrophages-derived exosomes augment hepatocellular carcinoma development through regulating KLF3. Int Immunopharmacol. 101:1081572021. View Article : Google Scholar : PubMed/NCBI

89 

Li X and Tang M: Exosomes released from M2 macrophages transfer miR-221-3p contributed to EOC progression through targeting CDKN1B. Cancer Med. 9:5976–5988. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Liu W, Long Q, Zhang W, Zeng D, Hu B, Liu S and Chen L: miRNA-221-3p derived from M2-polarized tumor-associated macrophage exosomes aggravates the growth and metastasis of osteosarcoma through SOCS3/JAK2/STAT3 axis. Aging (Albany NY). 13:19760–19775. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Yuan Y, Wang Z, Chen M, Jing Y, Shu W, Xie Z, Li Z, Xu J, He F, Jiao P, et al: Macrophage-derived exosomal miR-31-5p promotes oral squamous cell carcinoma tumourigenesis through the large tumor suppressor 2-mediated hippo signalling pathway. J Biomed Nanotechnol. 17:822–837. 2021. View Article : Google Scholar : PubMed/NCBI

92 

Li Z, Wang Y, Zhu Y, Yuan C, Wang D, Zhang W, Qi B, Qiu J, Song X, Ye J, et al: The Hippo transducer TAZ promotes epithelial to mesenchymal transition and cancer stem cell maintenance in oral cancer. Mol Oncol. 9:1091–1105. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Yao J, Wang Z, Cheng Y, Ma C, Zhong Y, Xiao Y, Gao X and Li Z: M2 macrophage-derived exosomal microRNAs inhibit cell migration and invasion in gliomas through PI3K/AKT/mTOR signaling pathway. J Transl Med. 19:992021. View Article : Google Scholar : PubMed/NCBI

94 

Gao XF, He HQ, Zhu XB, Xie SL and Cao Y: LncRNA SNHG20 promotes tumorigenesis and cancer stemness in glioblastoma via activating PI3K/Akt/mTOR signaling pathway. Neoplasma. 66:532–542. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Gao Y, Lin L, Li T, Yang J and Wei Y: The role of miRNA-223 in cancer: Function, diagnosis and therapy. Gene. 616:1–7. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Haneklaus M, Gerlic M, O'Neill LA and Masters SL: miR-223: Infection, inflammation and cancer. J Intern Med. 274:215–226. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Zhu X, Shen H, Yin X, Yang M, Wei H, Chen Q, Feng F, Liu Y, Xu W and Li Y: Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J Exp Clin Cancer Res. 38:812019. View Article : Google Scholar : PubMed/NCBI

98 

Yang M, Chen J, Su F, Yu B, Su F, Lin L, Liu Y, Huang JD and Song E: Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol Cancer. 10:1172011. View Article : Google Scholar : PubMed/NCBI

99 

Aucher A, Rudnicka D and Davis DM: MicroRNAs transfer from human macrophages to hepato-carcinoma cells and inhibit proliferation. J Immunol. 191:6250–6260. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Peng WX, Koirala P and Mo YY: LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Chen F, Chen J, Yang L, Liu J, Zhang X, Zhang Y, Tu Q, Yin D, Lin D, Wong PP, et al: Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol. 21:498–510. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Zhang Q, Han Z, Zhu Y, Chen J and Li W: Role of hypoxia inducible factor-1 in cancer stem cells (Review). Mol Med Rep. 23:172021.

104 

Yin Z, Zhou Y, Ma T, Chen S, Shi N, Zou Y, Hou B and Zhang C: Down-regulated lncRNA SBF2-AS1 in M2 macrophage-derived exosomes elevates miR-122-5p to restrict XIAP, thereby limiting pancreatic cancer development. J Cell Mol Med. 24:5028–5038. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Gao Z, Wang Q, Ji M, Guo X, Li L and Su X: Exosomal lncRNA UCA1 modulates cervical cancer stem cell self-renewal and differentiation through microRNA-122-5p/SOX2 axis. J Transl Med. 19:2292021. View Article : Google Scholar : PubMed/NCBI

106 

Xu M, Zhou C, Weng J, Chen Z, Zhou Q, Gao J, Shi G, Ke A, Ren N, Sun H and Shen Y: Tumor associated macrophages-derived exosomes facilitate hepatocellular carcinoma malignance by transferring lncMMPA to tumor cells and activating glycolysis pathway. J Exp Clin Cancer Res. 41:2532022. View Article : Google Scholar : PubMed/NCBI

107 

Gelardi ELM, Colombo G, Picarazzi F, Ferraris DM, Mangione A, Petrarolo G, Aronica E, Rizzi M, Mori M, La Motta C and Garavaglia S: A selective competitive inhibitor of aldehyde dehydrogenase 1A3 hinders cancer cell growth, invasiveness and stemness in vitro. Cancers (Basel). 13:3562021. View Article : Google Scholar : PubMed/NCBI

108 

Guo Y, Sun W, Gao W, Li L, Liang Y, Mei Z, Liu B and Wang R: Long noncoding RNA H19 derived from M2 tumor-associated macrophages promotes bladder cell autophagy via stabilizing ULK1. J Oncol. 2022:34654592022. View Article : Google Scholar : PubMed/NCBI

109 

Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, Wang Y, Wang T and Hou Y: Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 8:3932–3948. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Shima H, Kida K, Adachi S, Yamada A, Sugae S, Narui K, Miyagi Y, Nishi M, Ryo A, Murata S, et al: Lnc RNA H19 is associated with poor prognosis in breast cancer patients and promotes cancer stemness. Breast Cancer Res Treat. 170:507–516. 2018. View Article : Google Scholar : PubMed/NCBI

111 

Wang F, Rong L, Zhang Z, Li M, Ma L, Ma Y, Xie X, Tian X and Yang Y: LncRNA H19-derived miR-675-3p promotes epithelial-mesenchymal transition and stemness in human pancreatic cancer cells by targeting the STAT3 pathway. J Cancer. 11:4771–4782. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Zhang F, Sang Y, Chen D, Wu X, Wang X, Yang W and Chen Y: M2 macrophage-derived exosomal long non-coding RNA AGAP2-AS1 enhances radiotherapy immunity in lung cancer by reducing microRNA-296 and elevating NOTCH2. Cell Death Dis. 12:4672021. View Article : Google Scholar : PubMed/NCBI

113 

Mi X, Xu R, Hong S, Xu T, Zhang W and Liu M: M2 macrophage-derived exosomal lncRNA AFAP1-AS1 and MicroRNA-26a affect cell migration and metastasis in esophageal cancer. Mol Ther Nucleic Acids. 22:779–790. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Huang W, Zhong Z, Luo C, Xiao Y, Li L, Zhang X, Yang L, Xiao K, Ning Y, Chen L, et al: The miR-26a/AP-2α/Nanog signaling axis mediates stem cell self-renewal and temozolomide resistance in glioma. Theranostics. 9:5497–5516. 2019. View Article : Google Scholar :

115 

Xin L, Zhou LQ, Liu C, Zeng F, Yuan YW, Zhou Q, Li SH, Wu Y, Wang JL, Wu DZ and Lu H: Transfer of LncRNA CRNDE in TAM-derived exosomes is linked with cisplatin resistance in gastric cancer. EMBO Rep. 22:e521242021. View Article : Google Scholar : PubMed/NCBI

116 

Zheng J, Li XD, Wang P, Liu XB, Xue YX, Hu Y, Li Z, Li ZQ, Wang ZH and Liu YH: CRNDE affects the malignant biological characteristics of human glioma stem cells by negatively regulating miR-186. Oncotarget. 6:25339–25355. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Feng Z, Meng S, Zhou H, Xu Z, Tang Y, Li P, Liu C, Huang Y and Wu M: Functions and potential applications of circular RNAs in cancer stem cells. Front Oncol. 9:5002019. View Article : Google Scholar : PubMed/NCBI

118 

Yu T, Wang Y, Fan Y, Fang N, Wang T, Xu T and Shu Y: CircRNAs in cancer metabolism: A review. J Hematol Oncol. 12:902019. View Article : Google Scholar : PubMed/NCBI

119 

Zhou P, Chen X, Shi K, Qu H and Xia J: The characteristics, tumorigenicities and therapeutics of cancer stem cells based on circRNAs. Pathol Res Pract. 233:1538222022. View Article : Google Scholar : PubMed/NCBI

120 

Zhuang Z, Jia L, Li W and Zheng Y: The emerging roles of circular RNAs in regulating the fate of stem cells. Mol Cell Biochem. 476:231–246. 2021. View Article : Google Scholar

121 

Yu D, Chang Z, Liu X, Chen P, Zhang H and Qin Y: Macrophage-derived exosomes regulate gastric cancer cell oxaliplatin resistance by wrapping circ 0008253. Cell Cycle. 22:705–717. 2023. View Article : Google Scholar

122 

Chen S, Chen Z, Li Z, Li S, Wen Z, Cao L, Chen Y, Xue P, Li H and Zhang D: Tumor-associated macrophages promote cholangiocarcinoma progression via exosomal Circ_0020256. Cell Death Dis. 13:942022. View Article : Google Scholar : PubMed/NCBI

123 

Gu X, Shi Y, Dong M, Jiang L, Yang J and Liu Z: Exosomal transfer of tumor-associated macrophage-derived hsa_ circ_0001610 reduces radiosensitivity in endometrial cancer. Cell Death Dis. 12:8182021. View Article : Google Scholar

124 

Ma J, Huang L, Gao YB, Li MX, Chen LL and Yang L: M2 macrophage facilitated angiogenesis in cutaneous squamous cell carcinoma via circ_TNFRSF21/miR-3619-5p/ROCK axis. Kaohsiung J Med Sci. 38:761–771. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Panis C, Pizzatti L, Souza GF and Abdelhay E: Clinical proteomics in cancer: Where we are. Cancer Let. 382:231–239. 2016. View Article : Google Scholar

126 

Zhu Y, Chen X, Pan Q, Wang Y, Su S, Jiang C, Li Y, Xu N, Wu L, Lou X and Liu S: A comprehensive proteomics analysis reveals a secretory path- and status-dependent signature of exosomes released from tumor-associated macrophages. J Proteome Res. 14:4319–4331. 2015. View Article : Google Scholar : PubMed/NCBI

127 

El-Arabey AA, Denizli M, Kanlikilicer P, Bayraktar R, Ivan C, Rashed M, Kabil N, Ozpolat B, Calin GA, Salama SA, et al: GATA3 as a master regulator for interactions of tumor-associated macrophages with high-grade serous ovarian carcinoma. Cell Signal. 68:1095392020. View Article : Google Scholar : PubMed/NCBI

128 

Zheng P, Luo Q, Wang W, Li J, Wang T, Wang P, Chen L, Zhang P, Chen H, Liu Y, et al: Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional apolipoprotein E. Cell Death Dis. 9:4342018. View Article : Google Scholar : PubMed/NCBI

129 

Lee HD, Koo BH, Kim YH, Jeon OH and Kim DS: Exosome release of ADAM15 and the functional implications of human macrophage-derived ADAM15 exosomes. FASEB J. 26:3084–3095. 2012. View Article : Google Scholar : PubMed/NCBI

130 

Yu X, Zhang Q, Zhang X, Han Q, Li H, Mao Y, Wang X, Guo H, Irwin DM, Niu G and Tan H: Exosomes from macrophages exposed to apoptotic breast cancer cells promote breast cancer proliferation and metastasis. J Cancer. 10:2892–2906. 2019. View Article : Google Scholar : PubMed/NCBI

131 

Wang M, Zhou L, Yu F, Zhang Y, Li P and Wang K: The functional roles of exosomal long non-coding RNAs in cancer. Cell Mol Life Sci. 76:2059–2076. 2019. View Article : Google Scholar : PubMed/NCBI

132 

Sharma A and Johnson A: Exosome DNA: Critical regulator of tumor immunity and a diagnostic biomarker. J Cell Physiol. 235:1921–1932. 2020. View Article : Google Scholar

133 

Azambuja JH, Ludwig N, Yerneni SS, Braganhol E and Whiteside TL: Arginase-1+ exosomes from reprogrammed macrophages promote glioblastoma progression. Int J Mol Sci. 21:39902020. View Article : Google Scholar : PubMed/NCBI

134 

Chen Y, Jin H, Song Y, Huang T, Cao J, Tang Q and Zou Z: Targeting tumor-associated macrophages: A potential treatment for solid tumors. J Cell Physiol. 236:3445–3465. 2021. View Article : Google Scholar

135 

Pienta KJ, Machiels JP, Schrijvers D, Alekseev B, Shkolnik M, Crabb SJ, Li S, Seetharam S, Puchalski TA, Takimoto C, et al: Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 (CCL2), in metastatic castration-resistant prostate cancer. Invest New Drugs. 31:760–768. 2013. View Article : Google Scholar

136 

Moisan F, Francisco EB, Brozovic A, Duran GE, Wang YC, Chaturvedi S, Seetharam S, Snyder LA, Doshi P and Sikic BI: Enhancement of paclitaxel and carboplatin therapies by CCL2 blockade in ovarian cancers. Mol Oncol. 8:1231–1239. 2014. View Article : Google Scholar : PubMed/NCBI

137 

Xu F, Wei Y, Tang Z, Liu B and Dong J: Tumor-associated macrophages in lung cancer: Friend or foe? (Review). Mol Med Rep. 22:4107–4115. 2020.PubMed/NCBI

138 

DiPersio JF, Uy GL, Yasothan U and Kirkpatrick P: Plerixafor. Nat Rev Drug Discov. 8:105–106. 2009. View Article : Google Scholar : PubMed/NCBI

139 

Wang J, Tannous BA, Poznansky MC and Chen H: CXCR4 antagonist AMD3100 (plerixafor): From an impurity to a therapeutic agent. Pharmacol Res. 159:1050102020. View Article : Google Scholar : PubMed/NCBI

140 

Hume DA and MacDonald KP: Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood. 119:1810–1820. 2012. View Article : Google Scholar

141 

Bonapace L, Coissieux MM, Wyckoff J, Mertz KD, Varga Z, Junt T and Bentires-Alj M: Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 515:130–133. 2014. View Article : Google Scholar : PubMed/NCBI

142 

Advani R, Flinn I, Popplewell L, Forero A, Bartlett NL, Ghosh N, Kline J, Roschewski M, LaCasce A, Collins GP, et al: CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin's lymphoma. N Engl J Med. 379:1711–1721. 2018. View Article : Google Scholar : PubMed/NCBI

143 

Petrova PS, Viller NN, Wong M, Pang X, Lin GH, Dodge K, Chai V, Chen H, Lee V, House V, et al: TTI-621 (SIRPαFc): A CD47-blocking innate immune checkpoint inhibitor with broad antitumor activity and minimal erythrocyte binding. Clin Cancer Res. 23:1068–1079. 2017. View Article : Google Scholar

144 

Bouwstra R, van Meerten T and Bremer E: CD47-SIRPα blocking-based immunotherapy: Current and prospective therapeutic strategies. Clin Transl Med. 12:e9432022. View Article : Google Scholar

145 

Zheng JH, Nguyen VH, Jiang SN, Park SH, Tan W, Hong SH, Shin MG, Chung IJ, Hong Y, Bom HS, et al: Two-step enhanced cancer immunotherapy with engineered Salmonella typhimurium secreting heterologous flagellin. Sci Transl Med. 9:eaak95372017. View Article : Google Scholar : PubMed/NCBI

146 

Yuan D, Zhao Y, Banks WA, Bullock KM, Haney M, Batrakova E and Kabanov AV: Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials. 142:1–12. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Li J, Li N and Wang J: M1 macrophage-derived exosome-encapsulated cisplatin can enhance its anti-lung cancer effect. Minerva Med. Apr 8–2020.Epub ahead of print. View Article : Google Scholar

148 

Kim MS, Haney MJ, Zhao Y, Mahajan V, Deygen I, Klyachko NL, Inskoe E, Piroyan A, Sokolsky M, Okolie O, et al: Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine. 12:655–664. 2016. View Article : Google Scholar

149 

Bellmunt À M, López-Puerto L, Lorente J and Closa D: Involvement of extracellular vesicles in the macrophage-tumor cell communication in head and neck squamous cell carcinoma. PLoS One. 14:e02247102019. View Article : Google Scholar : PubMed/NCBI

150 

Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, Schwille P, Brügger B and Simons M: Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 319:1244–1247. 2008. View Article : Google Scholar : PubMed/NCBI

151 

Wu ATH, Srivastava P, Yadav VK, Tzeng DTW, Iamsaard S, Su EC, Hsiao M and Liu MC: Ovatodiolide, isolated from Anisomeles indica, suppresses bladder carcinogenesis through suppression of mTOR/β-catenin/CDK6 and exosomal miR-21 derived from M2 tumor-associated macrophages. Toxicol Appl Pharmacol. 401:1151092020. View Article : Google Scholar

152 

Guo J, Wang X, Guo Q, Zhu S, Li P, Zhang S and Min L: M2 macrophage derived extracellular vesicle-mediated transfer of MiR-186-5p promotes colon cancer progression by targeting DLC1. Int J Biol Sci. 18:1663–1676. 2022. View Article : Google Scholar : PubMed/NCBI

153 

Wang P, Li GY, Zhou L, Jiang HL, Yang Y and Wu HT: Exosomes from M2 macrophages promoted glycolysis in FaDu cells by inhibiting PDLIM2 expression to stabilize PFKL. Neoplasma. 69:1041–1053. 2022. View Article : Google Scholar : PubMed/NCBI

154 

Song L, Luan B, Xu Q, Shi R and Wang X: microRNA-155-3p delivered by M2 macrophages-derived exosomes enhances the progression of medulloblastoma through regulation of WDR82. J Transl Med. 20:132022. View Article : Google Scholar : PubMed/NCBI

155 

Zhang Z, Hu J, Ishihara M, Sharrow AC, Flora K, He Y and Wu L: The miRNA-21-5p payload in exosomes from M2 macrophages drives tumor cell aggression via PTEN/Akt signaling in renal cell carcinoma. Int J Mol Sci. 23:30052022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2023
Volume 63 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang W, Zhou R, Liu X, You L, Chen C, Ye X, Liu J and Liang Y: Key role of exosomes derived from M2 macrophages in maintaining cancer cell stemness (Review). Int J Oncol 63: 126, 2023
APA
Zhang, W., Zhou, R., Liu, X., You, L., Chen, C., Ye, X. ... Liang, Y. (2023). Key role of exosomes derived from M2 macrophages in maintaining cancer cell stemness (Review). International Journal of Oncology, 63, 126. https://doi.org/10.3892/ijo.2023.5574
MLA
Zhang, W., Zhou, R., Liu, X., You, L., Chen, C., Ye, X., Liu, J., Liang, Y."Key role of exosomes derived from M2 macrophages in maintaining cancer cell stemness (Review)". International Journal of Oncology 63.5 (2023): 126.
Chicago
Zhang, W., Zhou, R., Liu, X., You, L., Chen, C., Ye, X., Liu, J., Liang, Y."Key role of exosomes derived from M2 macrophages in maintaining cancer cell stemness (Review)". International Journal of Oncology 63, no. 5 (2023): 126. https://doi.org/10.3892/ijo.2023.5574