Skip to main content

Mutual regulation of PD-L1 immunosuppression between tumor-associated macrophages and tumor cells: a critical role for exosomes

Abstract

Tumor cells primarily employ the PD-1/PD-L1 pathway to thwart the anti-tumor capabilities of T lymphocytes, inducing immunosuppression. This occurs through the direct interaction of PD-L1 with PD-1 on T lymphocyte surfaces. Recent research focusing on the tumor microenvironment has illuminated the pivotal role of immune cells, particularly tumor-associated macrophages (TAMs), in facilitating PD-L1-mediated immunosuppression. Exosomes, characterized by their ability to convey information and be engulfed by cells, significantly contribute to promoting TAM involvement in establishing PD-L1-mediated immunosuppression within the tumor microenvironment. Exosomes, characterized by their ability to convey information and be engulfed by cells, significantly contribute to promoting TAM involvement in establishing PD-L1-mediated immunosuppression within the tumor microenvironment. In addition to receiving signals from tumor-derived exosomes that promote PD-L1 expression, TAMs also exert control over PD-L1 expression in tumor cells through the release of exosomes. This paper aims to summarize the mechanisms by which exosomes participate in this process, identify crucial factors that influence these mechanisms, and explore innovative strategies for inhibiting or reversing the tumor-promoting effects of TAMs by targeting exosomes.

Introduction

Exosomes, which are nanoscale vesicles produced via exocytosis by both normal and tumor cells, play a critical role in mediating the interaction between tumor and immune cells within the tumor microenvironment [1]. They represent a distinct mode of signaling in addition to direct cell-to-cell communication and indirect communication through chemokines or cytokines [2, 3]. Exosomes contribute to the immunosuppressive characteristics observed in almost all lymphocyte and bone marrow cell populations, thereby assisting tumor cells in evading immune surveillance [4].

Tumor-associated macrophages (TAMs) are a type of immune cells present in the tumor microenvironment, primarily located in the tumor stroma where they regulate inflammatory responses. Recent evidence has highlighted that TAMs not only contribute to tumor progression but also play a role in immunosuppression [5, 6].

PD-1/PD-L1 signaling pathway serves as a critical checkpoint for tumor immune evasion [7]. Tumor-infiltrating T cells, influenced by the tumor microenvironment, exhibit high levels of PD-1 expression. However, their ability to kill tumor cells is significantly hindered upon interaction with PD-L1 expressed on the tumor surface [8].

Although immune checkpoint inhibitors have revolutionized cancer treatment, the response rate in patients receiving these therapies has not met expectations. This necessitates further exploration of the underlying mechanisms leading to resistance against immune-based therapies.

TAMs play a crucial role in mediating PD-1/PD-L1 immunosuppression, primarily through three mechanisms: exosome secretion, PD-L1 expression, and cytokine secretion [9]. This perspective highlights that TAMs actively contribute to the immune evasion of malignancies. Exosomes act as passive agents, implicating TAMs in the continuous invasion of tumors, and they are among several factors in the tumor microenvironment that influence the polarization of tumor-associated macrophages. Studying how exosomes promote the development of PD-1/PD-L1 immunosuppression between tumor and immune cells will help develop strategies to improve immunotherapy efficacy and overcome immune resistance. This paper reviews the recent progress in this field and its potential mechanisms. Experimental evidence supports exosomal targeting to enhance immune effectiveness, emphasizing the importance of exosomal targeting as a key strategy to combat immune resistance.

Exosomes regulate the pro-tumor and anti-tumor state of TAMs

In a broader context, TAMs do not represent a specific type of macrophage, but rather a heterogeneous population of macrophages with diverse molecular expressions and functional states infiltrating the tumor microenvironment. They can be roughly categorized into anti-tumor M1 type and pro-tumor M2 type, although TAMs predominantly exhibit an M2 phenotype [10]. M1 macrophages are characterized by the expression of molecules such as CD86 and possess pro-inflammatory functions [11]. On the other hand, M2 macrophages, which have high IL-10 expression and low IL-12 expression, can be further subdivided into different subtypes (M2a, M2b, M2c, M2d) with unique combinations of molecular markers that support the malignant behavior of most tumors [12]. It is worth noting that the polarization state of TAM and the ratio of different subtypes change dynamically with specific time, which is related to the complex stimulus signals in the tumor microenvironment [13]. Additionally, intermediate cells between the two extreme M1 and M2 markers may simultaneously express markers associated with both M1 and M2 phenotypes [14].

Tumor-derived exosomes facilitate macrophage recruitment and have been shown to influence the polarization of M1/M2 type macrophages in various tumor types [15,16,17]. Exosomes play a significant regulatory role in promoting the pro-tumor phenotype of macrophages by carrying nucleic acids, proteins, lipids, and other important regulatory substances [18]. For instance, the incorporation of MiR-3591-3p into glioma exosomes promotes M2-type polarization of macrophages through the JAK2/PI3K/Akt/mTOR and STAT3 pathways, thereby enhancing tumor invasion and migration [19]. Gallbladder cancer cells effectively utilize exosomes to transport leptin to macrophages, thereby influencing M2 macrophage polarization. Inhibiting leptin reverses this process’s pro-tumor growth effect [20]. By controlling macrophage polarization via exosomes, tumors facilitate the development of a pre-metastatic niche. Researchers discovered that the polarization of pulmonary macrophages by Caveolin-1 transported by exosomes could be inextricably linked to pre-metastatic microenvironment creation before breast cancer (BC) cells metastasize to the lung [21]. Tumor cell-produced microRNA (miR)-21 and miR29a exosomes stimulate the production of Toll-like receptor (TLR)-mediated pro-inflammatory proteins in macrophages, which are linked to NF-κB activation and collectively implicated in the pre-tumor inflammatory phase [22]. In addition to promoting malignant tumor growth and distant spread, M2 macrophages polarized by tumor-derived exosomes also contribute to reducing tumor treatment sensitivity. For instance, non-small cell lung cancer induces macrophage M2 polarization by releasing phosphoribosyl pyrophosphate synthetases 2 exosomes, thereby increasing the cisplatin toxicity threshold for tumor cells [23]. The direction of TAMs polarization is also influenced by exosomes produced by different cell types in the tumor microenvironment. Cancer-associated fibroblasts educate monocytes to differentiate into the M2 macrophage phenotype, which supports BC growth. Exosomes produced by this phenotype also exhibit high expression of the highly invasive miR-181a. Furthermore, it has been demonstrated that miR-320a, released by fibroblasts, controls the direction of M2-type polarization of macrophages [24]. The progression of prostate cancer has also been shown to be related to the regulation of the M2-type polarization direction of macrophages by miR-320a secreted by fibroblasts [25].

TAM-derived exosomes promote tumor growth

Exosomes serve as a critical medium for communication between TAMs and tumor cells within the tumor microenvironment. TAM-derived exosomes play a significant role in influencing tumor formation through various mechanisms, highlighting the importance of their components [26, 27]. These exosomes derived from TAMs contribute to the continued malignant growth of tumors by regulating tumor cell stemness, promoting tumor angiogenesis, facilitating tumor metastasis, and inducing treatment resistance [28,29,30,31]. Importantly, TAM-derived exosomes also contribute to the resistance of tumor cells towards targeted drugs, in addition to chemotherapy drugs [32, 33]. Furthermore, TAM-derived exosomes play a crucial role in metabolic reprogramming. For instance, exosomes containing long non-coding RNA derived from TAMs activate glycolysis in hepatocellular carcinoma (HCC) via the miR-548 s/ALDH1A3 pathway, demonstrating that metabolic reprogramming is an essential mechanism through which exosomes promote malignant proliferation [34].

Several factors, including hypoxia [35], epithelial-mesenchymal transition [36], gene mutations [37], and various signaling pathways [38], influence TAMs mediated by exosomes that promote tumor growth. Hypoxia has long been recognized as a critical physiological component in the tumor microenvironment. Tumor-derived exosomes, which are released at higher levels and exhibit changes in heterogeneity under hypoxic conditions, play a role in transferring hypoxic information within the tumor microenvironment. These exosomes possess distinct characteristics in terms of cargo content, cellular recognition, and internalization by target cells [39, 40]. Exosomes facilitate the interaction between TAMs and tumor cells or other immune cells in the tumor microenvironment, thereby promoting tumor growth and invasion.

Exosomes mediate the regulation of PD-L1 expression by TAMs

Yang et al.’s research has provided long-standing evidence that PD-L1 is present simultaneously in tumor cells and the exosomes they produce [41]. PD-L1 exosomes produced by tumor cells enhance the immunosuppressive ability of the tumor microenvironment by directly binding with T cells and indirectly binding with TLRs on myeloid cells [42]. Exosomes carrying PD-L1 compromise the body’s ability to fight tumors by transmitting PD-L1 to other cells in the tumor microenvironment, such as macrophages, actively suppressing T cells’ immunological functions [41]. Phagocytes, including macrophages, have a more professional approach to phagocytosis than non-phagocytic cells and absorb PD-L1 exosomes in the tumor microenvironment to escape immune surveillance [41, 43]. Gastric cancer (GC)-derived exosomes cause macrophages to polarize towards the M2 type and express PD-1, leading to increased production of IL-10 and accelerated tumor growth by preventing T cell activation [44]. Gu and his colleagues found that miR-92a-3p exosomes from GC can also be ingested by pulmonary macrophages, activating the ERK signaling pathway by inhibiting the expression of PTEN in macrophages and promoting the expression of PD-L1 in macrophages through transcriptional regulation [45]. Similarly, exosomes from TAMs also regulate the expression of PD-L1 in tumor cells. M2-type TAMs use exosomes to enhance PD-L1 expression in GC cells [46]. Conversely, M1 macrophages downregulate PD-L1 expression in GC cells by loading miR-16-5p exosomes [47]. Crosstalk between different immune cell types in the tumor microenvironment also plays an important role in promoting the expression of PD-L1 on TAMs. Mesenchymal stem cells produce exosomes carrying TGF-β, C1q, and signaling proteins, which induce overexpression of PD-L1 in undifferentiated monocytes and macrophages (Fig. 1) [48].

Fig. 1
figure 1

Mechanism of exosomes mediated TAM regulation of PD-L1 expression. Exosomes regulate the expression of tumor cells and TAM PD-L1 mainly through two ways: (a) Direct delivery dependent on PD-L1; (b) By acting on PTEN, STAT, TOLL receptor, NF-κB mediated signaling pathways

Mechanism of exosomes mediated TAM regulation of PD-L1 expression

PTEN/PD-L1 axis

Strong evidence has linked the deletion of the PTEN gene to tumor PD-L1 expression in various types of cancer [49,50,51]. Breast cancer (BC) cells increase PD-L1 expression in macrophages to evade the immunological response, cooperating with cancer cells through a PTEN-mediated signaling pathway. Exosomes generated from BC cells are enriched in miR-27a-3p, contributing to this process [52]. Similarly, miR-23a-3p, abundant in exosomes produced from hepatocellular carcinoma (HCC), reduces the expression of PD-L1 in TAMs through PTEN-mediated signaling pathways [53]. When comparing miR-183-5p exosomes produced by intrahepatic cholangiocarcinoma cells with primary human intrahepatic bile duct epithelial cells, higher expression was observed in the HCC. This encourages the activation of PD-L1 in macrophages through the PTEN/AKT/PD-L1 signaling pathway, enhancing tumor immune evasion and reducing patient survival [54]. Surprisingly, TAMs possess higher levels of PD-L1 than colorectal cancer (CRC) cells, and CRC cells stimulate PD-L1 expression in macrophages by controlling the PTEN/AKT pathway via miR-21-5P exosomes (Table 1) [55].

Table 1 Exosomes that regulate PD-L1 immunosuppression

STAT/PD-L1 axis

The Signal Transducer and Activator of Transcription (STAT) family, consisting of numerous members, exerts a significant regulatory influence on tumor proliferation, immune evasion, and metastasis [65]. Loading exosomes with lysyl oxidase-like 4 (LOXL4) produced by liver cancer cells promotes macrophage PD-L1 expression, mediated by interferon and STATs [56]. Microparticles loaded with PD-L1 in triple-negative breast cancer stimulate the TBK1/STAT6 signaling pathway, polarizing internalized macrophages into an immunosuppressive phenotype and promoting the establishment of immunosuppressive features in the tumor microenvironment [66]. Macrophage-derived exosomes promote PD-L1 expression in laryngeal squamous cell carcinoma through the STAT3 transcription pathway, which was further confirmed by the addition of STAT3 inhibitors [57]. Glioblastoma plays a crucial role in inducing the formation of an immunosuppressive phenotype in macrophages through the STAT3-mediated signaling pathway, facilitating dynamic communication between malignant tumor cells [58]. Melatonin-exposed HCC cells reduce PD-L1 expression in macrophages via the STAT3 pathway mediated by exosomes, thereby altering the characteristics of exosomes initially promoting liver tumor growth [59]. Exosomes from CRC carrying miR-200a and miR-21-5p also stimulate the STAT1 pathway within the macrophage, which is crucial in encouraging PD-L1 expression [55].

TOLL/PD-L1 axis

The intricate network of TLR-mediated signaling pathways is a vital component in the complex landscape of cancer development. As a pattern recognition receptor, TLRs serve as the crucial link between innate and adaptive immunity, allowing for effective immune responses against invading pathogens and malignant cells alike [67]. The expression of PD-L1 in M2 macrophages through TLR4-dependent signaling effectively shapes the immunosuppressive phenotype of macrophages, preventing T cell proliferation and providing opportunities for tumor immune escape. These secretory autophagosomes, derived from malignant tumors, are found in the pleural and abdominal fluids of tumors [60]. Exosomes secreted by non-small cell lung cancer have an immunosuppressive effect on macrophages, dependent on PD-L1 activation via TLR2 signaling [62]. Exosomes produced by tumor cells in chronic lymphocytic leukemia activate TLR7 to mediate PD-L1 production in monocytes and release cytokines that stimulate tumor growth [63].

NF-κB/PD-L1 axis

NF-κB has long been recognized as a key player in inflammatory-mediated tumorigenesis [68], and accumulating evidence suggests that activated NF-κB is a key regulator of different functional states of TAMs and can directly mediate TAM PD-L1 expression [69]. Morrissey et al. also emphasized the role of NF-κB-dependent pathways in the action of tumor-derived exosomes on TAMs, which, in addition to Toll-like receptors, mediate enhanced glycolysis and lactate production through independent activation of NOS2 and HIF-1α, thereby promoting M2-type macrophage polarization. Increased lactate-driven de novo synthesis of PD-L1 expression is observed in TAMs, which mediates pre-metastatic niche formation; this effect is unrelated to exosome PD-L1 translocation [62].

Factors affecting exosomes-mediated TAMs regulation of PD-L1-mediated immunosuppression

Cellular stress environment

The ER stress marker was closely and positively related to the expression of M2 macrophages PD-L1 in oral squamous cell carcinoma. In vivo and in vitro experiments revealed that co-culturing with tumor cells subjected to ER stress could promote PD-L1 expression in macrophages [70]. Similarly, ER stress was found to upregulate PD-L1 expression in macrophages in breast cancer (BC) and hepatocellular carcinoma (HCC) tissues through exosomes [52, 53]. Reactive oxygen species (ROS) generated by redox reactions in the tumor immune microenvironment played a significant role in regulating PD-L1 on macrophages. Elevated reactive oxygen species (ROS) downregulated exosomal miR-155-5p expression in tumor cells, impacting PD-L1 expression. Neutralization with N-acetyl-L-cysteine (NAC) restored miR-155-5p levels, reducing macrophage migration and tumor infiltration while enhancing CD8 T cell function [71]. Tumor-derived exosomes were shown to produce lactate, which played a crucial role in metabolic reprogramming and increased macrophage PD-L1 expression through NF-κB signaling [62]. The immunosuppressive effects of adenosine in HCC were specific. Exosomes from CD73-expressing liver cancer cells (circTMEM181 exosomes) induce macrophages to upregulate CD39 expression, leading to adenosine accumulation and changes in the immune microenvironment, forming resistance to anti-PD-1 drugs (Table 2) [75].

Table 2 Factors affecting the regulation of PD-L1 expression exosomes

Key genes and proteins

Golgi apparatus protein 1 was found to transport PD-L1 protein and upregulate PD-L1 expression in macrophages in hepatocellular carcinoma (HCC), thus affecting the response of liver cancer to anti-PD-L1 therapy. This process depended on proteasomes to reduce PD-L1 protein consumption and promote PD-L1 extracellular transport by inhibiting Rab27b. Treatment with proteasome inhibitors reduced PD-L1 expression, providing evidence for the critical role of Golgi protein 1 in promoting the formation of tumor immunosuppressive environments [73]. CMTM6, an essential protein regulating PD-L1 expression, has gained unprecedented attention [76]. Pang et al. reported that CMTM6 was closely associated with macrophage infiltration and PD-L1 expression in oral squamous cell carcinoma. Transfer of CMTM6 to macrophages through exosomes promoted ERK1/2-mediated M2 polarization [77]. The expression of CMTM6 and PD-L1 in colorectal cancer (CRC) with mismatch repair defect was positively correlated with the density of M2 macrophages. The expression of CMTM6 in M2 macrophages was the best biomarker for predicting the effectiveness of PD-1/PD-L1 inhibitors in CRC patients [78].

Others

Sulfasalazine has been found to disrupt the redox balance of tumor cells, leading to increased production of reactive oxygen species and inhibition of tumor progression. In melanoma, sulfasalazine inhibits cystine-glutamate exchange, which promotes the production of two transcription factors, IRF4 and EGR1, that regulate PD-L1 expression. This enhances the secretion of PD-L1 exosomes and induces M2 polarization in macrophages, reducing sensitivity to immune checkpoint blockade (ICB) treatment [72]. Counteracting the effect of LOXL4 exosomes on increasing macrophage PD-L1 expression in hepatocellular cancer can be achieved by using copper or hydrogen peroxide scavengers [56]. In vitro experiments simulating the mechanical strain experienced by tumor cells during growth have shown that it promotes the release of PD-L1 exosomes from breast cancer cells in the tumor microenvironment, which are then internalized by macrophages [74]. Patients infected with hepatitis B virus produce exosomes containing viral DNA and proteins, which upregulate PD-L1 expression in endocytic monocytes/macrophages, contributing to immune suppression [79]. Melatonin, known for its immune regulatory effects, has been utilized in tumor treatment. In hepatocellular carcinoma cells, melatonin therapy reverses the downregulation of macrophage PD-L1 expression through the inhibition of the STAT3-mediated signaling pathway [59].. Researchers studying non-small cell lung cancer patients with varying degrees of obstructive sleep apnea have found that intermittent hypoxia induces hypoxia-inducible factor-1, which enhances macrophage PD-L1 expression induced by exosomes [80].

Discussion

Tumors have the tendency and capability to regulate the expression of PD-L1 in tumor-associated macrophages (TAMs). TAMs undergo a transformation into tumor-promoting phenotypes after being influenced by exosomes released by tumor cells. This transformation then establishes a positive feedback loop through exosomes, leading to increased PD-L1 expression in tumor cells. This article reviews the mechanism of exosomes regulating PD-L1 expression between tumor-associated macrophages (TAM) and tumor cells. It has been observed that PTEN, STAT, TLR and NF-κB have the highest frequency of occurrence in different types of tumor signaling pathways, presenting potential targets for inhibiting TAMs’ regulation of the PD-1/PD-L1 immunosuppressive pathway. The specificity of exosomes is determined by various factors such as cell type, cellular state, and environmental factors including mechanical stress, ER stress, reactive oxygen species, adenosine, lactic acid, among others. Proteins like Rab27, TP53, and CMTM6 play key roles in regulating the formation of exosomes that carry information involved in PD-L1 expression regulation. Factors like melatonin, sulfasalazine, hepatitis B virus, and obstructive sleep apnea also influence the level of PD-L1 expression regulated by TAMs through exosomes to varying degrees. These findings provide valuable research directions for targeting exosome-mediated PD-L1 expression and blocking the immunosuppressive effect of TAMs on tumor PD-L1.

Targeting TAMs to enhance the efficacy of immunotherapy has been clinically validated through two main strategies: TAMs consumption and TAMs reeducation. Due to the unique biological characteristics of exosomes, which are easily customizable and carry substances, targeting or modifying exosomes has emerged as a novel approach to inhibiting the M2 polarization of TAMs, achieving TAMs reeducation, and improving the effectiveness of tumor treatment [81,82,83]. Exosomes can overcome the toxicity of nanoparticles and limitations of phagocytic clearance by targeting specific cargoes to lesions and tissues through cellular phagocytosis and homing mechanisms [84]. CpGODN, a tumor vaccine modified by apoptotic bodies, not only polarizes M1 macrophages to produce more tumor suppressor factors but also enables these polarized macrophages to infect nearby macrophages, leading to a cascade reaction of tumor inhibitory effects [85]. Exosomes play a crucial role in controlling macrophage dynamic polarization in vivo, allowing them to transition from a pro-tumor phenotype to an anti-tumor one [86]. Exosomes are engineered and modified to deliver functional molecules that re-educate TAMs [87,88,89]. Researchers often target microRNA and chemotherapy drugs for exosome payloads, and antisense oligonucleotides targeting key transcription factors and inhibitors targeting enzymes are also being considered [90,91,92,93]. Transfection of genes expressing pigment epithelial-derived factors into parental cells resulted in the ability of derived exosomes to promote the repolarization of macrophages from the M2 phenotype to the M1 phenotype [94]. Ovarian cancer cells overexpressing ETS1 secrete exosomes that are more easily internalized by macrophages, promoting M2 polarization to facilitate tumor cell metastasis to the omentum. Targeting Integrin αVβ5, which plays a crucial role in inhibiting the polarization process, with cilengitide weakens the driving effect of these exosomes [95].

It is noteworthy that engineering modifications of extracellular vesicles derived from tumor-associated macrophages (TAMs) offer several advantages in tumor treatment compared to vesicles derived from other cell types [96, 97]. The investigation into the modification of exosomes derived from tumor-associated macrophages (TAMs) to enhance the effectiveness of drug delivery is experiencing rapid growth, particularly in the context of M1 subtype, showcasing notable efficacy in reversing tumor chemoresistance [98,99,100,101]. For instance, Choo et al. successfully repolarized M2 TAMs into M1 macrophages using exosomes derived from macrophages, thereby enhancing the anti-tumor efficacy of anti-PD-L1 therapy [102]. By carrying anti-PD-L1 nanobodies or binding with anti-CD73 antibodies, exosomes derived from M1 TAMs acquire the ability to reverse tumor immune suppression and increase the sensitivity of tumor cells to radiation therapy [103, 104]. Hybrid cells combining macrophages and tumor cells can produce exosomes that effectively activate T cells to kill tumor cells, combining the immune-stimulating abilities of macrophages with the “homing ability” of circulating tumor cells. When combined with anti-PD-L1 therapy, these hybrid exosomes inhibit tumor progression and provide insights into personalized tumor immunotherapy [105]. Additionally, it is worth mentioning that Artemisia-derived nanovesicles have been found to enhance the effectiveness of PD-L1 immune checkpoint inhibitors, suggesting that cross-species approaches with common biological characteristics hold promise for tumor treatment [106].

The clinical application of PD-L1/PD-1 immune checkpoint inhibitors has transformed the treatment of advanced unresectable tumors, surpassing traditional radiotherapy and chemotherapy. However, immunotherapy still poses challenges, such as low response rates and drug resistance. Therefore, researchers must focus on screening patients who respond effectively to immunotherapy and developing strategies to improve and maintain drug efficacy. In recent years, the discovery of the tumor-promoting effect of immune cells in the tumor microenvironment has provided important clues for understanding tumor progression. To further identify and expand the population that responds to immune therapy, it is necessary to conduct more in-depth research on the mechanism of tumor immune suppression mediated by TAMs, providing more diverse treatment options. TAMs are among the most abundant immune cells in the tumor microenvironment, and their dynamic polarization and phagocytosis suggest that exosomes play a crucial role in TAM-mediated tumor PD-L1/PD-1 immunosuppression. The evolving understanding of exosomes, from metabolic waste transporters to important communication mediators between cells, has deepened research in the field of cancer. It is anticipated that exosomes will significantly contribute to suppressing TAMs and enhancing PD-L1 immune development.

Availability of data and materials

Not applicable.

Abbreviations

PD-1:

Programmed cell death protein 1

PD-L1:

Programmed cell death ligand 1

TAM:

Tumor-associated macrophages

MiR:

MicroRNA

BC:

Breast cancer

TLR:

Toll-like receptor

STAT:

Signal transformer and activator of the transcription

HCC:

Hepatocellular carcinoma

ER:

Endoplasmic reticulum

LOXL4:

Lysyl oxidase-like 4

CRC:

Colorectal cancer

References

  1. Kallinger I, Rubenich DS, Głuszko A, Kulkarni A, Spanier G, Spoerl S, Taxis J, Poeck H, Szczepański MJ, Ettl T, et al. Tumor gene signatures that correlate with release of extracellular vesicles shape the immune landscape in head and neck squamous cell carcinoma. Clin Exp Immunol. 2023;213.

  2. Hwang I. Cell-cell communication via extracellular membrane vesicles and its role in the immune response. Mol Cells. 2013;36:105–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mittelbrunn M, Sánchez-Madrid F. Intercellular communication: diverse structures for exchange of genetic information. Nat Rev Mol Cell Biol. 2012;13:328–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kugeratski FG, Kalluri R. Exosomes as mediators of immune regulation and immunotherapy in cancer. FEBS J. 2021;288:10–35.

    Article  PubMed  Google Scholar 

  5. Fu LQ, Du WL, Cai MH, Yao JY, Zhao YY, Mou XZ. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol. 2020;353:104119.

    Article  CAS  PubMed  Google Scholar 

  6. Nowak M, Klink M. The role of tumor-associated macrophages in the progression and Chemoresistance of ovarian Cancer. Cells/. 2020;9.

  7. Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, Hundal J, Arthur CD, Krebber WJ, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:577–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pu Y, Ji Q. Tumor-associated macrophages regulate PD-1/PD-L1 immunosuppression. Front Immunol. 2022;13:874589.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Boutilier AJ, Elsawa SF. Macrophage polarization states in the tumor microenvironment. Int J Mol Sci. 2021;22.

  11. Stöger JL, Gijbels MJ, van der Velden S, Manca M, van der Loos CM, Biessen EA, Daemen MJ, Lutgens E, de Winther MP. Distribution of macrophage polarization markers in human atherosclerosis. Atherosclerosis. 2012;225:461–8.

    Article  PubMed  Google Scholar 

  12. Zhang Q, Sioud M. Tumor-associated macrophage subsets: shaping polarization and targeting. Int J Mol Sci. 2023;24.

  13. Pan Y, Yu Y, Wang X, Zhang T. Tumor-associated macrophages in tumor immunity. Front Immunol. 2020;11:583084.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Chong BF, Tseng LC, Hosler GA, Teske NM, Zhang S, Karp DR, Olsen NJ, Mohan C. A subset of CD163+ macrophages displays mixed polarizations in discoid lupus skin. Arthritis Res Ther. 2015;17:324.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Frank AC, Ebersberger S, Fink AF, Lampe S, Weigert A, Schmid T, Ebersberger I, Syed SN, Brüne B. Apoptotic tumor cell-derived microRNA-375 uses CD36 to alter the tumor-associated macrophage phenotype. Nat Commun. 2019;10:1135.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Baig MS, Roy A, Rajpoot S, Liu D, Savai R, Banerjee S, Kawada M, Faisal SM, Saluja R, Saqib U, et al. Tumor-derived exosomes in the regulation of macrophage polarization. Inflamm Res. 2020;69:435–51.

    Article  CAS  PubMed  Google Scholar 

  17. Pritchard A, Tousif S, Wang Y, Hough K, Khan S, Strenkowski J, Chacko BK, Darley-Usmar VM, Deshane JS. Lung tumor cell-derived exosomes promote M2 macrophage polarization. Cells. 2020;9.

  18. Chen Q, Li Y, Gao W, Chen L, Xu W, Zhu X. Exosome-mediated crosstalk between tumor and tumor-associated macrophages. Front Mol Biosci. 2021;8:764222.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Li M, Xu H, Qi Y, Pan Z, Li B, Gao Z, Zhao R, Xue H, Li G. Tumor-derived exosomes deliver the tumor suppressor miR-3591-3p to induce M2 macrophage polarization and promote glioma progression. Oncogene. 2022;41:4618–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhao S, Liu Y, He L, Li Y, Lin K, Kang Q, Liu L, Zou H. Gallbladder Cancer cell-derived exosome-mediated transfer of leptin promotes cell invasion and migration by modulating STAT3-mediated M2 macrophage polarization. Anal Cell Pathol (Amst). 2022;2022:9994906.

    PubMed  PubMed Central  Google Scholar 

  21. Wang Y, Li Y, Zhong J, Li M, Zhou Y, Lin Q, Zong S, Luo W, Wang J, Wang K, et al. Tumor-derived Cav-1 promotes pre-metastatic niche formation and lung metastasis in breast cancer. Theranostics. 2023;13:1684–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, Lovat F, Fadda P, Mao C, Nuovo GJ, et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci U S A. 2012;109:E2110–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu G, Luo Y, Hou P. PRPS2 enhances resistance to cisplatin via facilitating exosomes-mediated macrophage M2 polarization in non-small cell lung Cancer. Immunol Investig. 2022;51:1423–36.

    Article  CAS  Google Scholar 

  24. Pakravan K, Mossahebi-Mohammadi M, Ghazimoradi MH, Cho WC, Sadeghizadeh M, Babashah S. Monocytes educated by cancer-associated fibroblasts secrete exosomal miR-181a to activate AKT signaling in breast cancer cells. J Transl Med. 2022;20:559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhao M, Zhuang A, Fang Y. Cancer-associated fibroblast-derived Exosomal miRNA-320a promotes macrophage M2 polarization in vitro by regulating PTEN/PI3Kγ signaling in pancreatic Cancer. J Oncol. 2022;2022:9514697.

    Article  PubMed  PubMed Central  Google Scholar 

  26. 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. 2018;9:434.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Guan H, Peng R, Fang F, Mao L, Chen Z, Yang S, Dai C, Wu H, Wang C, Feng N, et al. Tumor-associated macrophages promote prostate cancer progression via exosome-mediated miR-95 transfer. J Cell Physiol. 2020;235:9729–42.

    Article  CAS  PubMed  Google Scholar 

  28. Li W, Xin X, Li X, Geng J, Sun Y. Exosomes secreted by M2 macrophages promote cancer stemness of hepatocellular carcinoma via the miR-27a-3p/TXNIP pathways. Int Immunopharmacol. 2021;101:107585.

    Article  CAS  PubMed  Google Scholar 

  29. 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. 2021;29:1226–38.

    Article  CAS  PubMed  Google Scholar 

  30. Ludwig N, Yerneni SS, Azambuja JH, Pietrowska M, Widłak P, Hinck CS, Głuszko A, Szczepański MJ, Kärmer T, Kallinger I, et al. TGFβ(+) small extracellular vesicles from head and neck squamous cell carcinoma cells reprogram macrophages towards a pro-angiogenic phenotype. J Extracell Vesicles. 2022;11:e12294.

    Article  PubMed  Google Scholar 

  31. Cheng HY, Hsieh CH, Lin PH, Chen YT, Hsu DS, Tai SK, Chu PY, Yang MH. Snail-regulated exosomal microRNA-21 suppresses NLRP3 inflammasome activity to enhance cisplatin resistance. J Immunother Cancer. 2022;10.

  32. Wan X, Xie B, Sun H, Gu W, Wang C, Deng Q, Zhou S. Exosomes derived from M2 type tumor-associated macrophages promote osimertinib resistance in non-small cell lung cancer through MSTRG.292666.16-miR-6836-5p-MAPK8IP3 axis. Cancer Cell Int. 2022;22:83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yuan S, Chen W, Yang J, Zheng Y, Ye W, Xie H, Dong L, Xie J. Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways. Oncol Lett. 2022;24:356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Xu M, Zhou C, Weng J, Chen Z, Zhou Q, Gao J, Shi G, Ke A, Ren N, Sun H, 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. 2022;41:253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lu Q, Wang X, Zhu J, Fei X, Chen H, Li C. Hypoxic tumor-derived Exosomal Circ0048117 facilitates M2 macrophage polarization acting as miR-140 sponge in esophageal squamous cell carcinoma. Onco Targets Ther. 2020;13:11883–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sun D, Luo T, Dong P, Zhang N, Chen J, Zhang S, Dong L, Janssen HLA, Zhang S. M2-polarized tumor-associated macrophages promote epithelial-mesenchymal transition via activation of the AKT3/PRAS40 signaling pathway in intrahepatic cholangiocarcinoma. J Cell Biochem. 2020;121:2828–38.

    Article  CAS  PubMed  Google Scholar 

  37. Katopodi T, Petanidis S, Domvri K, Zarogoulidis P, Anestakis D, Charalampidis C, Tsavlis D, Bai C, Huang H, Freitag L, et al. Kras-driven intratumoral heterogeneity triggers infiltration of M2 polarized macrophages via the circHIPK3/PTK2 immunosuppressive circuit. Sci Rep. 2021;11:15455.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Xu Z, Chen Y, Ma L, Chen Y, Liu J, Guo Y, Yu T, Zhang L, Zhu L, Shu Y. Role of exosomal non-coding RNAs from tumor cells and tumor-associated macrophages in the tumor microenvironment. Mol Ther. 2022;30:3133–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, Yeoh KW, Kon OL, Tam JP, Sze SK. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 2019;38:5158–73.

    Article  CAS  PubMed  Google Scholar 

  40. He G, Peng X, Wei S, Yang S, Li X, Huang M, Tang S, Jin H, Liu J, Zhang S, et al. Exosomes in the hypoxic TME: from release, uptake and biofunctions to clinical applications. Mol Cancer. 2022;21:19.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Yang Y, Li CW, Chan LC, Wei Y, Hsu JM, Xia W, Cha JH, Hou J, Hsu JL, Sun L, Hung MC. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28:862–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Morrissey SM, Yan J. Exosomal PD-L1: roles in tumor progression and immunotherapy. Trends Cancer. 2020;6:550–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Feng D, Zhao WL, Ye YY, Bai XC, Liu RQ, Chang LF, Zhou Q, Sui SF. Cellular internalization of exosomes occurs through phagocytosis. Traffic. 2010;11:675–87.

    Article  CAS  PubMed  Google Scholar 

  44. Wang F, Li B, Wei Y, Zhao Y, Wang L, Zhang P, Yang J, He W, Chen H, Jiao Z, Li Y. Tumor-derived exosomes induce PD1(+) macrophage population in human gastric cancer that promotes disease progression. Oncogenesis. 2018;7:41.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Gu J, Chu X, Huo Y, Liu C, Chen Q, Hu S, Pei Y, Ding P, Pang S, Wang M. Gastric cancer-derived exosomes facilitate pulmonary metastasis by activating ERK-mediated immunosuppressive macrophage polarization. J Cell Biochem. 2023;124.

  46. Wang Y, Shang K, Zhang N, Zhao J, Cao B. Tumor-associated macrophage-derived exosomes promote the progression of gastric Cancer by regulating the P38MAPK signaling pathway and the immune checkpoint PD-L1. Cancer Biother Radiopharm. 2021.

  47. Li Z, Suo B, Long G, Gao Y, Song J, Zhang M, Feng B, Shang C, Wang D. Exosomal miRNA-16-5p derived from M1 macrophages enhances T cell-dependent immune response by regulating PD-L1 in gastric Cancer. Front Cell Dev Biol. 2020;8:572689.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Biswas S, Mandal G, Roy Chowdhury S, Purohit S, Payne KK, Anadon C, Gupta A, Swanson P, Yu X, Conejo-Garcia JR, Bhattacharyya A. Exosomes produced by mesenchymal stem cells drive differentiation of myeloid cells into immunosuppressive M2-polarized macrophages in breast Cancer. J Immunol. 2019;203:3447–60.

    Article  CAS  PubMed  Google Scholar 

  49. Ugai T, Zhao M, Shimizu T, Akimoto N, Shi S, Takashima Y, Zhong R, Lau MC, Haruki K, Arima K, et al. Association of PIK3CA mutation and PTEN loss with expression of CD274 (PD-L1) in colorectal carcinoma. Oncoimmunology. 2021;10:1956173.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Qiu R, Wang W, Li J, Wang Y. Roles of PTEN inactivation and PD-1/PD-L1 activation in esophageal squamous cell carcinoma. Mol Biol Rep. 2022;49:6633–45.

    Article  CAS  PubMed  Google Scholar 

  51. Palicelli A, Croci S, Bisagni A, Zanetti E, De Biase D, Melli B, Sanguedolce F, Ragazzi M, Zanelli M, Chaux A, et al. What do we have to know about PD-L1 expression in prostate Cancer? A systematic literature review (part 6): correlation of PD-L1 expression with the status of mismatch repair system, BRCA, PTEN, and other genes. Biomedicines. 2022;10.

  52. Yao X, Tu Y, Xu Y, Guo Y, Yao F, Zhang X. Endoplasmic reticulum stress-induced exosomal miR-27a-3p promotes immune escape in breast cancer via regulating PD-L1 expression in macrophages. J Cell Mol Med. 2020;24:9560–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liu J, Fan L, Yu H, Zhang J, He Y, Feng D, Wang F, Li X, Liu Q, Li Y, et al. Endoplasmic reticulum stress causes liver Cancer cells to release Exosomal miR-23a-3p and up-regulate programmed death ligand 1 expression in macrophages. Hepatology. 2019;70:241–58.

    Article  CAS  PubMed  Google Scholar 

  54. Luo C, Xin H, Zhou Z, Hu Z, Sun R, Yao N, Sun Q, Borjigin U, Wu X, Fan J, et al. Tumor-derived exosomes induce immunosuppressive macrophages to foster intrahepatic cholangiocarcinoma progression. Hepatology. 2022;76:982–99.

    Article  CAS  PubMed  Google Scholar 

  55. Yin Y, Liu B, Cao Y, Yao S, Liu Y, Jin G, Qin Y, Chen Y, Cui K, Zhou L, et al. Colorectal Cancer-derived small extracellular vesicles promote tumor immune evasion by upregulating PD-L1 expression in tumor-associated macrophages. Adv Sci (Weinh). 2022;9:2102620.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Tan HY, Wang N, Zhang C, Chan YT, Yuen MF, Feng Y. Lysyl oxidase-like 4 fosters an immunosuppressive microenvironment during Hepatocarcinogenesis. Hepatology. 2021;73:2326–41.

    Article  CAS  PubMed  Google Scholar 

  57. Bellmunt ÀM, López-Puerto L, Lorente J, Closa D. Involvement of extracellular vesicles in the macrophage-tumor cell communication in head and neck squamous cell carcinoma. PLoS One. 2019;14:e0224710.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Gabrusiewicz K, Li X, Wei J, Hashimoto Y, Marisetty AL, Ott M, Wang F, Hawke D, Yu J, Healy LM, et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology. 2018;7:e1412909.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Cheng L, Liu J, Liu Q, Liu Y, Fan L, Wang F, Yu H, Li Y, Bu L, Li X, et al. Exosomes from melatonin treated Hepatocellularcarcinoma cells Alter the Immunosupression status through STAT3 pathway in macrophages. Int J Biol Sci. 2017;13:723–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wen ZF, Liu H, Gao R, Zhou M, Ma J, Zhang Y, Zhao J, Chen Y, Zhang T, Huang F, et al. Tumor cell-released autophagosomes (TRAPs) promote immunosuppression through induction of M2-like macrophages with increased expression of PD-L1. J Immunother Cancer. 2018;6:151.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Du A, Yang Q, Sun X, Zhao Q. Exosomal circRNA-001264 promotes AML immunosuppression through induction of M2-like macrophages and PD-L1 overexpression. Int Immunopharmacol. 2023;124:110868.

    Article  CAS  PubMed  Google Scholar 

  62. Morrissey SM, Zhang F, Ding C, Montoya-Durango DE, Hu X, Yang C, Wang Z, Yuan F, Fox M, Zhang HG, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021;33:2040-2058.e2010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Haderk F, Schulz R, Iskar M, Cid LL, Worst T, Willmund KV, Schulz A, Warnken U, Seiler J, Benner A, et al. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci Immunol. 2017;2.

  64. Liu Y, Yan H, Gu H, Zhang E, He J, Cao W, Qu J, Xu R, Cao L, He D, et al. Myeloma-derived IL-32γ induced PD-L1 expression in macrophages facilitates immune escape via the PFKFB3-JAK1 axis. Oncoimmunology. 2022;11:2057837.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Verhoeven Y, Tilborghs S, Jacobs J, De Waele J, Quatannens D, Deben C, Prenen H, Pauwels P, Trinh XB, Wouters A, et al. The potential and controversy of targeting STAT family members in cancer. Semin Cancer Biol. 2020;60:41–56.

    Article  CAS  PubMed  Google Scholar 

  66. Li C, Qiu S, Jin K, Zheng X, Zhou X, Jin D, Xu B, Jin X. Tumor-derived microparticles promote the progression of triple-negative breast cancer via PD-L1-associated immune suppression. Cancer Lett. 2021;523:43–56.

    Article  CAS  PubMed  Google Scholar 

  67. Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-like receptor signaling and its role in cell-mediated immunity. Front Immunol. 2022;13:812774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ben-Neriah Y, Karin M. Inflammation meets cancer, with NF-κB as the matchmaker. Nat Immunol. 2011;12:715–23.

    Article  CAS  PubMed  Google Scholar 

  69. Antonangeli F, Natalini A, Garassino MC, Sica A, Santoni A, Di Rosa F. Regulation of PD-L1 expression by NF-κB in Cancer. Front Immunol. 2020;11:584626.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yuan Y, Jiao P, Wang Z, Chen M, Du H, Xu L, Xu J, Dai Y, Wu FG, Zhang Y, Wu H. Endoplasmic reticulum stress promotes the release of exosomal PD-L1 from head and neck cancer cells and facilitates M2 macrophage polarization. Cell Commun Signal. 2022;20:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li X, Wang S, Mu W, Barry J, Han A, Carpenter RL, Jiang BH, Peiper SC, Mahoney MG, Aplin AE, et al. Reactive oxygen species reprogram macrophages to suppress antitumor immune response through the exosomal miR-155-5p/PD-L1 pathway. J Exp Clin Cancer Res. 2022;41:41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu N, Zhang J, Yin M, Liu H, Zhang X, Li J, Yan B, Guo Y, Zhou J, Tao J, et al. Inhibition of xCT suppresses the efficacy of anti-PD-1/L1 melanoma treatment through exosomal PD-L1-induced macrophage M2 polarization. Mol Ther. 2021;29:2321–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Chen J, Lin Z, Liu L, Zhang R, Geng Y, Fan M, Zhu W, Lu M, Lu L, Jia H, et al. GOLM1 exacerbates CD8(+) T cell suppression in hepatocellular carcinoma by promoting exosomal PD-L1 transport into tumor-associated macrophages. Signal Transduct Target Ther. 2021;6:397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wang Y, Goliwas KF, Severino PE, Hough KP, Van Vessem D, Wang H, Tousif S, Koomullil RP, Frost AR, Ponnazhagan S, et al. Mechanical strain induces phenotypic changes in breast cancer cells and promotes immunosuppression in the tumor microenvironment. Lab Investig. 2020;100:1503–16.

    Article  CAS  PubMed  Google Scholar 

  75. Lu JC, Zhang PF, Huang XY, Guo XJ, Gao C, Zeng HY, Zheng YM, Wang SW, Cai JB, Sun QM, et al. Amplification of spatially isolated adenosine pathway by tumor-macrophage interaction induces anti-PD1 resistance in hepatocellular carcinoma. J Hematol Oncol. 2021;14:200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Burr ML, Sparbier CE, Chan YC, Williamson JC, Woods K, Beavis PA, Lam EYN, Henderson MA, Bell CC, Stolzenburg S, et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature. 2017;549:101–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Pang X, Wang SS, Zhang M, Jiang J, Fan HY, Wu JS, Wang HF, Liang XH, Tang YL. OSCC cell-secreted exosomal CMTM6 induced M2-like macrophages polarization via ERK1/2 signaling pathway. Cancer Immunol Immunother. 2021;70:1015–29.

    Article  CAS  PubMed  Google Scholar 

  78. Wu X, Lan X, Hu W, Zhang W, Lai X, Xu S, Li J, Qiu W, Wang W, Xiao J, et al. CMTM6 expression in M2 macrophages is a potential predictor of PD-1/PD-L1 inhibitor response in colorectal cancer. Cancer Immunol Immunother. 2021;70:3235–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kakizaki M, Yamamoto Y, Yabuta S, Kurosaki N, Kagawa T, Kotani A. The immunological function of extracellular vesicles in hepatitis B virus-infected hepatocytes. PLoS One. 2018;13:e0205886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Liu Y, Lu M, Chen J, Li S, Deng Y, Yang S, Ou Q, Li J, Gao P, Luo Z, et al. Extracellular vesicles derived from lung cancer cells exposed to intermittent hypoxia upregulate programmed death ligand 1 expression in macrophages. Sleep Breath. 2022;26:893–906.

    Article  PubMed  Google Scholar 

  81. Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Control Release. 2015;219:396–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29:341–5.

    Article  CAS  PubMed  Google Scholar 

  83. Wiklander OP, Nordin JZ, O’Loughlin A, Gustafsson Y, Corso G, Mäger I, Vader P, Lee Y, Sork H, Seow Y, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316.

    Article  PubMed  Google Scholar 

  84. Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics. 2021;11:3183–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Zhao G, Liu H, Wang Z, Yang H, Zhao H, Zhang Y, Ge K, Wang X, Luo L, Zhou X, et al. Exosome transportation-mediated immunosuppression relief through cascade amplification for enhanced apoptotic body vaccination. Acta Biomater. 2022;153:529–39.

    Article  CAS  PubMed  Google Scholar 

  86. Kim H, Park HJ, Chang HW, Back JH, Lee SJ, Park YE, Kim EH, Hong Y, Kwak G, Kwon IC, et al. Exosome-guided direct reprogramming of tumor-associated macrophages from protumorigenic to antitumorigenic to fight cancer. Bioact Mater. 2023;25:527–40.

    CAS  PubMed  Google Scholar 

  87. Zhang L, Lin Y, Li S, Guan X, Jiang X. In situ reprogramming of tumor-associated macrophages with internally and externally engineered exosomes. Angew Chem Int Ed Engl. 2023;62:e202217089.

    Article  CAS  PubMed  Google Scholar 

  88. Li C, Guan N, Liu F. T7 peptide-decorated exosome-based nanocarrier system for delivery of Galectin-9 siRNA to stimulate macrophage repolarization in glioblastoma. J Neuro-Oncol. 2023;162:93–108.

    Article  CAS  Google Scholar 

  89. Nie W, Wu G, Zhang J, Huang LL, Ding J, Jiang A, Zhang Y, Liu Y, Li J, Pu K, Xie HY. Responsive exosome Nano-bioconjugates for synergistic Cancer therapy. Angew Chem Int Ed Engl. 2020;59:2018–22.

    Article  CAS  PubMed  Google Scholar 

  90. Li L, He D, Guo Q, Zhang Z, Ru D, Wang L, Gong K, Liu F, Duan Y, Li H. Exosome-liposome hybrid nanoparticle codelivery of TP and miR497 conspicuously overcomes chemoresistant ovarian cancer. J Nanobiotechnology. 2022;20:50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Gunassekaran GR, Poongkavithai Vadevoo SM, Baek MC, Lee B. M1 macrophage exosomes engineered to foster M1 polarization and target the IL-4 receptor inhibit tumor growth by reprogramming tumor-associated macrophages into M1-like macrophages. Biomaterials. 2021;278:121137.

    Article  CAS  PubMed  Google Scholar 

  92. Kamerkar S, Leng C, Burenkova O, Jang SC, McCoy C, Zhang K, Dooley K, Kasera S, Zi T, Sisó S, et al. Exosome-mediated genetic reprogramming of tumor-associated macrophages by exoASO-STAT6 leads to potent monotherapy antitumor activity. Sci Adv. 2022;8:eabj7002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Azambuja JH, Ludwig N, Yerneni SS, Braganhol E, Whiteside TL. Arginase-1+ exosomes from reprogrammed macrophages promote glioblastoma progression. Int J Mol Sci. 2020;21.

  94. Moradi-Chaleshtori M, Koochaki A, Shojaei S, Paryan M, Safarzadeh M, Hashemi SM, Mohammadi-Yeganeh S. Overexpression of pigment epithelium-derived factor in breast cancer cell-derived exosomes induces M1 polarization in macrophages. Immunol Lett. 2022;248:31–6.

    Article  CAS  PubMed  Google Scholar 

  95. Zhou W, Zhou Y, Chen X, Ning T, Chen H, Guo Q, Zhang Y, Liu P, Zhang Y, Li C, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268:120546.

    Article  CAS  PubMed  Google Scholar 

  96. Huis i’nt Veld RV, Lara P, Jager MJ, Koning RI, Ossendorp F, Cruz LJ. M1-derived extracellular vesicles enhance photodynamic therapy and promote immunological memory in preclinical models of colon cancer. J Nanobiotechnol. 2022;20:252.

    Article  Google Scholar 

  97. Han R, Yu L, Zhao C, Li Y, Ma Y, Zhai Y, Qian Z, Gu Y, Li S. Inhibition of SerpinB9 to enhance granzyme B-based tumor therapy by using a modified biomimetic nanoplatform with a cascade strategy. Biomaterials. 2022;288:121723.

    Article  CAS  PubMed  Google Scholar 

  98. Baek S, Jeon M, Jung HN, Lee W, Hwang JE, Lee JS, Choi Y, Im HJ. M1 macrophage-derived exosome-mimetic Nanovesicles with an enhanced Cancer targeting ability. ACS Appl Bio Mater. 2022;5:2862–9.

    Article  CAS  PubMed  Google Scholar 

  99. Zhao Y, Zheng Y, Zhu Y, Zhang Y, Zhu H, Liu T. M1 macrophage-derived exosomes loaded with gemcitabine and Deferasirox against Chemoresistant pancreatic Cancer. Pharmaceutics. 2021;13.

  100. Zhang X, Liu L, Tang M, Li H, Guo X, Yang X. The effects of umbilical cord-derived macrophage exosomes loaded with cisplatin on the growth and drug resistance of ovarian cancer cells. Drug Dev Ind Pharm. 2020;46:1150–62.

    Article  CAS  PubMed  Google Scholar 

  101. Zhao Y, Zheng Y, Zhu Y, Li H, Zhu H, Liu T. Docetaxel-loaded M1 macrophage-derived exosomes for a safe and efficient chemoimmunotherapy of breast cancer. J Nanobiotechnol. 2022;20:359.

    Article  CAS  Google Scholar 

  102. Choo YW, Kang M, Kim HY, Han J, Kang S, Lee JR, Jeong GJ, Kwon SP, Song SY, Go S, et al. M1 macrophage-derived Nanovesicles potentiate the anticancer efficacy of immune checkpoint inhibitors. ACS Nano. 2018;12:8977–93.

    Article  CAS  PubMed  Google Scholar 

  103. Ma X, Yao M, Gao Y, Yue Y, Li Y, Zhang T, Nie G, Zhao X, Liang X. Functional immune cell-derived exosomes engineered for the trilogy of radiotherapy sensitization. Adv Sci (Weinh). 2022;9:e2106031.

    Article  PubMed  Google Scholar 

  104. Zhou Q, Ding W, Qian Z, Zhu Q, Sun C, Yu Q, Tai Z, Xu K. Immunotherapy strategy targeting programmed cell death ligand 1 and CD73 with macrophage-derived mimetic Nanovesicles to treat bladder Cancer. Mol Pharm. 2021;18:4015–28.

    Article  CAS  PubMed  Google Scholar 

  105. Wang S, Li F, Ye T, Wang J, Lyu C, Qing S, Ding Z, Gao X, Jia R, Yu D, et al. Macrophage-tumor chimeric exosomes accumulate in lymph node and tumor to activate the immune response and the tumor microenvironment. Sci Transl Med. 2021;13:eabb6981.

    Article  CAS  PubMed  Google Scholar 

  106. Liu J, Xiang J, Jin C, Ye L, Wang L, Gao Y, Lv N, Zhang J, You F, Qiao H, Shi L. Medicinal plant-derived mtDNA via nanovesicles induces the cGAS-STING pathway to remold tumor-associated macrophages for tumor regression. J Nanobiotechnol. 2023;21:78.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the public drawing platform Figdraw (www.figdraw.com) for supporting our illustration making.

Funding

This study was supported by the Science and Technology Project for Youth Talent of Changzhou Health Commission (QN201703), Young Talent Development Plan of Changzhou Health Commission (CZQM2020024), Major Science and Technology Project of Changzhou Health Commission (ZD202004, ZD202007), and China Postdoctoral Science Foundation (2020M670064ZX).

Author information

Authors and Affiliations

Authors

Contributions

BW, DC, wrote the original draft of the article and drew the illustration. DM, RC, DL, WZ, CF, and MJ contributed to the conceptualization and revised the draft. All authors participated in the revision of the manuscript. All authors contributed to the article and approved the submitted version.

Corresponding author

Correspondence to Mei Ji.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

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 licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, B., Cheng, D., Ma, D. et al. Mutual regulation of PD-L1 immunosuppression between tumor-associated macrophages and tumor cells: a critical role for exosomes. Cell Commun Signal 22, 21 (2024). https://doi.org/10.1186/s12964-024-01473-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-024-01473-5

Keywords