Open Access

Mechanism of exosomes in the tumor microenvironment in the abscopal effect (Review)

  • Authors:
    • Guicheng Kuang
    • Zirui Wang
    • Chengyu Luo
    • Jingyan Luo
    • Jing Wang
  • View Affiliations

  • Published online on: November 7, 2022     https://doi.org/10.3892/ijo.2022.5450
  • Article Number: 2
  • Copyright: © Kuang 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

Previously, the abscopal effect, which is an antitumor therapeutic effect on untreated tumor locations elsewhere in the body as a result of treatment of the targeted region, was rarely reported, and its mechanism remains unknown. Increasing evidence has shown that the immune system is implicated in the abscopal effect, and that combining immunotherapy and radiation can assist to improve its frequency. Understanding how different types of cells and cell‑derived exosomes cause the abscopal effect in the tumor microenvironment (TME) is crucial to increasing the clinical occurrence of the abscopal effect in the TME.

1. Introduction

Multiple novel radiotherapies have emerged in recent years, including brachytherapy, carbon ion radiotherapy and proton therapy, which can be further subdivided into photothermal therapy and photodynamic therapy (1-3). The abscopal effect describes the shrinkage of unirradiated tumors that occurs concurrently with irradiated tumors in patients with multiple tumors. Since Mole's first proposal of this effect (4), there had only been a few cases reported, until the association with the immune system was demonstrated by previous research that found the abscopal effect was not observed in mice with immunodeficiency (5). With immunotherapy becoming a more effective treatment for tumors (6,7), clinical studies have revealed that a combination of radiotherapy and immunotherapy, such as the immune checkpoint inhibitor anti-cytotoxic T lymphocyte-associated antigen-4 (anti-CTLA-4), anti-programmed death-1 (anti-PD-1) and anti-programmed cell death 1 ligand 1 (anti-PD-L1), produces the abscopal effect in the treatment of lung cancer (8), prostate cancer (9), melanoma (10), breast cancer (11), liver cancer (12), type B3 thymoma (13) and glioblastoma (GBM). As a result, the mechanism of the abscopal effect and how to produce it by combining immunotherapy and radiotherapy has become a hotspot in tumor research, which is also the focus of the present review.

2. Immune mechanism of the abscopal effect

The immunological mechanism of action behind the abscopal effect remains unknown, although various studies have shown that it is dependent on T cells (5,14,15) and macrophages (7). Thus far, it is known that tumor cells are triggered to produce tumor-associated antigens (TAAs) when their DNA is damaged. TAAs are then phagocytosed by antigen-presenting cells (APCs) before being activated by major histocompatibility complex (MHC) molecules on CD8+ T cells. Since CD8+ T cells not only have a direct impact on primary tumors but also reach untreated tumors via the blood and lymph circulation, they attach to tumor cells and destroy them, thus exerting an antitumor effect (16).

Exosomes are secreted by cells for intercellular signal transduction and information exchange. They carry nucleic acid, proteins and lipids to the target cell by acting on its surface or fusing with it (17). Exosomes derived from various cells serve an immunosuppressive or immunoenhancing role (18), and participate in carcinogenesis, proliferation and metastasis (19), thus playing distinct roles in the abscopal effect. Furthermore, there are alterations in exosome secretion following radiation (20,21). The subsequent sections discuss the mechanisms by which distinct cells produce exosomes after radiation to modify the abscopal effect.

3. Role of tumor cells

Tumor cells become more immunogenic after being irradiated because their DNA is damaged, thus causing the production of TAAs. The endoplasmic reticulum, which contains calreticulin and the disulfide isomerase ERp57, migrates to the plasma membrane and delivers an 'eat-me' signal to APCs, enhancing their phagocytosis and abscopal action. On the other hand, due to the formation of cytoplasmic double stranded DNA (dsDNA) induced by radiation, GMP-AMP synthase (cGAS) and dsDNA initiate the formation of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) (22). The increased cGAMP level combines with Stimulator of interferon genes (SINGs) to help regulate the activity of downstream immune stimulating genes, and ultimately promote the maturation and migration of dendritic cells and the activity of CD8+ T cells, thus playing an antitumor role (23,24). Certain exosomes produced by tumor cells can enhance the antitumor effect. They are highly rich in proteins, such as CD40L, which activates the CD40 signaling pathway in dendritic cells (DCs), and induces DCs to mature and produce IL-12, thus promoting anti-tumor immunity (25).

Previous research indicated that tumors with neoepitopes, such as epidermal growth factor receptor (EGFR) vIII in GBM, are more vulnerable to the abscopal effect (7), albeit the mechanism is unknown and requires additional exploration. This tumor type also produces exosomes with high EGFRvIII levels (26).

The radiation-induced damage to DNA, on the other hand, can be repaired and therefore blocked by the DNA exonuclease 3 repair exonuclease 1 inside the tumor, and the degree to which the calreticulin is exposed will be lowered by tumor cell autophagy. Furthermore, CD47 on the plasma membrane can inhibit phagocytosis of tumor cells by the immune system and emit a 'do-not-eat-me' signal (27) to offset the impact of 'eat-me' signals (28). Furthermore, the expression of PD-L1 on the surface of tumor cells increases, and the combination of PD-L1 and PD-1 on the surface of CD8+ T cells acts as an immunological brake, weakening the action of effector T cells (29-31), and therefore limiting the abscopal effect (Fig. 1).

There are immunosuppressive exosomes as well as immunoenhancing exosomes. Some of them produce cell membrane proteins, including PD-L1 and MHC. According to a previous study, PD-L1 immunosuppression on tumor cell-derived exosomes is considerably greater than that on the membrane (32). These PD-L1-rich exosomes perform the same function as PD-1 on the tumor surface, and their immunosuppressive impact is amplified when they express both PD-L1 and MHC molecules (33). Furthermore, certain cancer types produce exosomes that increase PD-1 expression on CD8+ T cells (34) and increase PD-L1 expression on the surface of macrophages (35), shielding tumor cells from CD8+ T cells. Exosomes rich in TGF-β and IL-10 may increase tumor migration and invasion (36), and tumor-derived exosomes can cause tumor immune evasion via the T-cell immunoglobulin and mucin domain 1 (TIM-1) signaling pathway (37). Furthermore, tumor-derived exosomes are high in Fas-L (38), which acts on the surface of T cells to trigger their death, thus markedly reducing antitumor immunity. Meanwhile, these exosomes' surfaces may be rich in integrin β 3 (ITGB3), which can activate focal adhesion kinase and influence intracellular signaling cascades, promoting tumor spread (39). The distinction is that tumor cells from the original location secrete exosomes rich in integrin β-like 1 (ITGBL1) to act on the distant site, thus activating the EVs-ITGBL1-CAFs-TNFAIP3-NF-κB signaling axis (40) to modify the tumor microenvironment (TME) and accelerate tumor metastasis, severely reducing the abscopal effect. A previous study has found that GBM-induced exosomes contain CD274, DNA and other chemicals that influence the transcriptional activator 3 STAT3 signaling pathway and promote macrophage polarization to M2 cells (41). Finally, exosomes from lymphoma cells include the inhibitor of apoptosis protein survivin, which inhibits natural killer (NK) cell surveillance and cytotoxicity (42).

Aside from proteins, nucleic acids in exosomes play an important role in the abscopal action. DNA, for example, can stimulate the STING signaling system, boost antitumor immunity and promote the abscopal effect (43). Furthermore, colorectal cancer-induced long non-coding RNA (lncRNA)-rich exosomes operate on the TME to stimulate the proliferation and differentiation of T helper (Th)17 cells and enhance the antitumor action (44). Meanwhile, microRNAs (miRNAs or miRs) in exosomes can influence IL-10 activation, resulting in an immunosuppressive TME that inhibits the development of the abscopal effect. Among these miRNAs, miR-212-3p has been shown to diminish MHC-2 expression and induce immunological tolerance in DCs, allowing tumors to escape immune surveillance (45). It has been reported that miR-934 (46), miR-301a-3p (47), miR-21-3p, miR-125b-5p and miR-181d-5p (48) can cause macrophages to polarize toward M2 cells, hence promoting tumor spread. These tumor-derived exosomes can act not only on the local TME, but also on the distant tumor site via the circulatory system.

There are numerous strategies for reducing the release of tumor-induced exosomes, which limit the abscopal effect. The first is to target and block exosomes from being released via routes such as endosomal sorting complex required for transport (ESCRT), tumor susceptibility gene 101 protein and other ESCRT proteins or ESCRT auxiliary molecules (49). The second is to influence the exosome acceptance pathway. The third option is to disrupt the functional pathway, such as by utilizing anti-PD-L1 or anti-PD-1 antibodies to prevent exosomes from acting on cells. In addition, radiotherapy can reduce the secretion of exosomes that promote tumor proliferation and metastasis by 25.8% (50). Furthermore, brachytherapy can boost the release of particular exosomes rich in high mobility group to induce macrophage polarization to M1 cells (51), hence considerably enhancing the abscopal effect. A combination of radiotherapy and immunotherapy can restrict the production and activity of immunosuppressive exosomes, and it remains to be explored which combined option should be utilized to do so while also increasing the release of immunoenhancing exosomes.

Upon radiotherapy, tumor cells will release damage-associated molecular patterns (DAMPs) such as ATP, high mobility group box 1 protein (HMGB1), nucleic acid, prostaglandin E2 (PGE2), sphingosine 1-phosphate, IL-6 and granulocyte macrophage colony stimulating factor (GM-CSF) (29,52,53) to exert their different biochemical effects.

4. Roles of tumor-associated molecular patterns

HMGB1 has a two-fold effect

On one hand, HMGB1 binds to receptor for advanced glycation end product (RAGE), a self-receptor, to promote tumor development and immunological tolerance while also inhibiting the abscopal effect. When its oxidation sites are blocked, however, HMGB1 increases immunity and enhances the abscopal effect (54,55).

ATP's role

The production of ATP at the start of radiation can stimulate the activation of DCs and effector T cells, boosting the abscopal effect (12,56-58). However, a previous study found that high ATP release could cause PD-1 overexpression in tumor cells as well as regulatory T cell aggregation. It could also cause the decrease of the entire immune process via APCs that promote immunological tolerance (59), thus substantially reducing the abscopal effect. Meanwhile, radiotherapy-induced dying tumor cells predominantly release ATP, which is converted to adenosine monophosphate (AMP) and adenosine by ecto-5′-nucleotidase expressed on macrophages, thus activating the A2a adenosine receptor on macrophages and suppressing macrophage-mediated antitumor immunity (60). In addition, ATP and adenosine diphosphate (ADP) are cleaved into AMP by CD39, and then CD73 converts AMP to adenosine, thereby inhibiting the stimulation of CD8+ T cells, activating regulatory T cells (Tregs) and promoting the differentiation of M2 macrophages to inhibit antitumor immune responses (61). CD39 and CD73 together play an important role in transforming an ATP-mediated proinflammatory TME into an adenosine-mediated immunosuppressive microenvironment (62). Furthermore, ATP and its metabolites ADP and AMP all have immunosuppressive roles in inhibiting the abscopal effect; thus, sedatives of several target adenosines are being studied in the clinical practice to restore the abscopal effect.

TGF-β

TGF-β affects the generation and activity of a range of immune cells. It regulates acquired immunity by directly stimulating Treg cell proliferation while suppressing the production and function of CD8+ T cells and antigen-presenting DCs. Similarly, TGF-β regulates the innate immune system by decreasing NK cells and controlling the complex activities of macrophages and neutrophils, as well as counteracting the anti-CTLA-4 and anti-PD-1 effects of immunosuppressive medications. As a result, research recently being conducted aimed to diminish TGF-β's impact on immunological drugs and prevent the reduction in acquired immunity by limiting its function, thus improving the abscopal effect (63,64).

To summarize, tumor cells perform a dual role in the abscopal effect. In general, tumors tend to block the abscopal effect for self-protection as the disease advances. As a result, efforts should be made to increase tumor cell immunogenicity and immunostimulation while decreasing their immunosuppressive effect.

Effect of APCs

DCs are the most effective APCs. Their Toll-like receptor (TLR) receives radiotherapy-induced TAAs via a TLR-signaling network (65), thus allowing DCs to identify, phagocytose and process TAAs. After swallowing TAAs, APCs can produce antigen peptides and costimulatory signals that activate T cells and amplify the abscopal effect. This boosting effect is smaller than that obtained directly from APCs (66), but it is sufficient to promote the abscopal effect. Thus, the tumor is driven to create 'danger signals', which activate DCs and promote the T cell response via MHC molecules. As a result, MHC-1 and MHC-2-rich exosomes generated by mature DCs have been used in clinical immunotherapy (67). Furthermore, exosomes generated by DCs during irradiation can strongly activate NK cells to destroy tumor cells (68). In comparison, despite their large number, immunological DC-induced exosomes had a significantly lesser effect than mature DC-induced microvesicles, which was analogous to their role in the abscopal effect (69). Activated DCs significantly enhanced the CD8+ T/Treg ratio in both primary (irradiated) and secondary (unirradiated) tumors, thus amplifying the abscopal effect (70). Furthermore, the insensitivity of DCs to irradiation, in comparison to that of tumor cells, ensures their survival and activity under high-dose irradiation, thus facilitating that their relatively high antigen-presenting effects are retained in the TME when high-dose irradiation is required to ensure therapeutic outcomes (71).

Tumor cell-induced DCs, on the other hand, produce TGF-β, IL-27 and other molecules that inhibit their own activity, activate Tregs and limit antitumor immunity function, thus decreasing the immune response and the abscopal effect (72,73). Immature DCs severely impede the function of effector T cells and suppress the immune response, resulting in the body's failure to generate the abscopal effect (74). One of the most essential features of solid tumors is a hypoxic TME, which substantially inhibits APC activity and reduces the antitumor impact. To reinforce the abscopal effect (75), it is worthwhile examining methods to improve DCs' antitumor immunological action while decreasing their immunosuppressive role.

Role of macrophages

Tumor-associated macrophages (TAMs) phagocytose tumor cells by recognizing their TAAs. TAMs can cause the abscopal effect in two ways: i) By attaching to T cells to achieve the antitumor immunological response or ii) By self-activation to reach the distal tumor location and kill tumor cells. Previous research has demonstrated that anti-PD-L1 can directly activate macrophages to boost the abscopal effect (7), and that it is a promising technique to promote the combination of activated macrophages with targeted radiation to enhance tumor cell damage and, thus, the abscopal effect (76).

TAMs are categorized into several subtypes, the most common of which are M1 and M2. M1 cells primarily contribute to antitumor immunity by directly phagocytosing tumor cells and secreting cytokines such as TNF to macrophages of the M1 phenotype. M1 macrophages can improve antitumor immunity by promoting the activation of effector T cells and the maturation of DCs by producing cytokines such as TNF-β, IL-6 and IL-23 (77). Meanwhile, M1 macrophages produce exosomes that interfere with the NF-κB signaling pathway while activating the caspase 3 signaling pathway, resulting in M1 macrophage polarization (78). M2 macrophages, on the other hand, predominantly suppress the immune system through the production of angiogenesis factors, growth factors and proteases, thus hastening the development of malignant tumors. M2 macrophages release exosomal miR-590-3p, which passes through the target LATS1 and activates YAP/β-catenin to decrease the immunological response (79). They also release integrin αMβ2-rich exosomes, which activate the MMP-9 signaling pathway in receptor tumor cells, as well as apolipoprotein E-rich exosomes, which promote tumor spread and proliferation (80,81). When M2 macrophages produce miRNA-21, it inhibits cell death, increases PI3K/AKT signaling pathway activation by downregulating PTEN (82), and boosts drug resistance in gastric cancer cells as well as tumor proliferation and metastasis, thus decreasing the abscopal effect.

Furthermore, M1 and M2 cells can convert into each other type, resulting in a shift in macrophage activity between antitumor and pro-tumor effects, which has a marked impact on the development of the abscopal effect.

Hypoxia (83) and suppression of NF-κB activity within the TME (84) can both cause M2 polarization in macrophages, resulting in an increase in the M2/M1 cell ratio. M2 macrophages produce a considerable quantity of TGF-β, which suppresses APC activity, and promotes tumorigenesis and development (85). Furthermore, the production of IL-4 by CD4+ T cells promotes M2 macrophage polarization (86), a process triggered by IL-10 and IL-11 (87). Meanwhile, inflammatory cytokines produced by cytotoxic T lymphocytes (CTLs), such as IFN, drive M1 macrophage polarization (88).

The polarization of M1 and M2 cells is mutually hostile and complex, and it is strongly associated with radiation. According to a previous study, when a cumulative quantity of 10 Gy is obtained based on a daily dosage of radiation of 2 Gy/fraction/day, the number of M1 cells increases while the number of M2 cells declines (89). When local low-dose irradiation is administered, M2 cells polarize back to M1 cells, promoting CTL penetration into the TME (90), improving the therapeutic effect. On the other hand, another study found that M2 cells were less sensitive to radiation than M1 cells, so that the ratio of M2/M1 cells increased when the cumulative radiation dosage reached a particular level (91). These seemingly contradicting outcomes are strongly linked to the radiation dose and method. In any case, M2 infiltration and polarization during radiation has a significant impact on patients' prognosis, and diminish their survival rate (92). Therefore, it is worth investigating how to activate M1 cells while suppressing M2 cell activation, as well as how to re-convert M2 cells into M1 cells to minimize the M2/M1 cell ratio.

TAMs steadily increase the expression of PD-1 while exerting antitumor immunological actions. Since PD-1 expression is inversely correlated with macrophage phagocytic and antigen-presenting capabilities (93), tumor escape can be inhibited by reducing PD-1 expression, and anti-PD-1 treatment can boost macrophage antitumor efficacy. TAMs, on the other hand, overexpress indoleamine 2,3-dioxygenase (IDO) (94), which inhibits CTL activity. Furthermore, CD40 on the surface of APCs will be highly expressed, weakening their function; thus, currently, studies are using multi-functional radiotherapy-associated biological materials to inhibit the expression of CD40 by delivering an anti-CD40 antibody to a tumor in situ, which allows the antitumor effects to be maintained (95).

TAMs also cause exosomes containing miR-29a-3p and miR-21-5p to inhibit STAT3, resulting in significant Treg proliferation (96), whereas exosomes containing miR-155 can control the TME, prompting macrophages to polarize into M1 cells (97,98). When TAMs produce lysyl oxidase like 4-rich exosomes, they promote the production of their own PD-L1, resulting in an immunosuppressive phenotype that inhibits the activity of CD8+ T cells (99), significantly decreasing antitumor immunity and the development of the abscopal effect. Furthermore, macrophages can produce GM-CSF-rich exosomes to enhance tumor angiogenesis (100), supply oxygen and nutrients to tumors, and hence provide possibilities for tumor migration and invasion.

Despite the several conditions that can restrict APCs' function, it has been observed that when antigens are available, boosting APCs function alone can induce a sufficiently significant abscopal effect. As a result, targeted activation of APCs contributes to the intensification of the abscopal effect.

Role of T cells

There are various T cell types, and all play crucial roles in the generation of the abscopal effect, with some of them enhancing each other, while others antagonize each other.

CD8+ T cells

CD8+ T cells have a potent antitumor function (101). The release of exosomes by CD8+ T cells is important, as they ingest exosomes originating from tumor cells and immune cells (102) to regulate their own functions and antitumor actions, which enhances the abscopal effect. CTLs recognize tumor cells via their TCR on the cell surface and eliminate them via two traditional mechanisms, namely i) The Fas-based molecular and ii) The perforin-granzyme-based processes (103). In the first mechanism, Fas ligands (FasL) exist on the T cell surface, and the surface of these exosomes is also rich in FasL, which acts on the Fas receptor on the surface of tumor, thus mediating tumor cell apoptosis (104,105). FasL bind to their receptors on tumor cells, activate the caspase protein, and promote the apoptosis of tumor cells through the Fas/FasL pathway. The second mechanism involves the release of perforin through CTLs. In terms of the first mechanism, both CD8+ T and NK cells release exosomes rich in CD56 and perforin (106). Perforin mediates the lysis and destruction of receptor tumor cells, thereby 'punching a hole' in the membrane of the tumor cell, allowing the granzyme to enter the tumor cells, and eventually leading to the rupture and death of tumor cells. Therefore, the degree of infiltration of CD8+ T cells is positively correlated with their antitumor effects.

The killing effect of CTLs can be activated when they are targeted by the auto-specific antigen of certain tumors, such as melanoma-associated antigen 1 (107) in melanoma and EGFRvIII (7) in GBM; therefore, these tumors are more susceptible to the abscopal effect. Furthermore, CD8+ T cells stimulate DCs by releasing exosomes rich in cytoplasmic DNA, and DCs are activated via the cGAS/STING signaling pathway (108). By creating immunological synapses and suppressing apoptosis, DCs stimulate CD8+ T cells, forming a powerful immune impact that can boost the abscopal effect.

During the antitumor process, CD8+ T cells in the TME are gradually fatigued, and the depleted CD8+ T cells produce a high number of lncRNA-rich exosomes. These exosomes impact the production of INF-γ and IL-2 in normal CD8+ T cells (109), and the ratio of CD8+ T/Tregs decreases, thus blocking the abscopal effect. Furthermore, CTLA-4 expression in CD8+ T cells would gradually increase, thus reducing the lethal impact of CTL on malignancies. Furthermore, PD-1 expression on CTLs increases during the antitumor process, thus blocking TCR signals by inactivating CD28 (110). CTLs unite with tumor cells that are highly expressing PD-L1 to further block their tumor-killing impact. Notably, IFN-γ secreted by effector T cells, which reduces the abscopal effect, increases PD-L1 expression on tumor cells. Aside from CTLA-4 and PD-1, it has recently been shown that Lag-3, Tim-3 and TIGIT likewise suppress CTL function while conducting their own (111).

Role of Tregs

Tregs have a high immunosuppressive impact (112), and their primary role is to induce immunological tolerance re-establishment. PD-1 expression on Tregs can enhance Treg self-proliferation and CD8+ T cell death (113), as well as strongly block CTL activity, and this immunosuppressive impact is regulated by IL-2. The level of IL-2 in the body is positively correlated with the immunosuppressive ability of Tregs; that is, when the number of IL-2 declines, the number and activity of Tregs decrease (114), and when the number of IL-2 increases, the number and activity of Tregs increase (114). As a result of their dependence on IL-2, Tregs' function can be impeded. Moreover, Tregs counteract the tumor immunity in a variety of ways (115), including the release of immunosuppressive factors such as IL-10 and TGF-β. The tumor sends a 'recruitment' signal to Tregs, such as C-X-C motif chemokine ligand 12, C-C motif chemokine ligand (CCL)17, CCL22 and CCL28, to increase the number of Tregs (116), resulting in a further decrease in the CD8+ T/Tregs ratio in the TME, thus inhibiting the abscopal effect and enhancing tumor immune tolerance.

The presence of CD73 on the surface of Treg-derived exosomes is required for Tregs to mediate immunosuppressive effects, and its mechanism is similar to that of Th17 cells, which facilitates the conversion of ATP to ADP and AMP (117,118). When the TCR on the surface of Tregs is activated, it increases exosome secretion, inhibits the tumor-killing activity of CTLs and inhibits the proliferation of effector T cells (119). The activated TCR promotes the release of IL-4 and IL-10, which is linked to miR-150-rich exosomes generated by Tregs (120). Treg-secreted miR-146a-5p-rich exosomes can play a role via suppressing STAT1 and interleukin 1 receptor associated kinase 2 (121).

Tregs serve an important function in preventing DC maturation. They produce exosomes and deliver them to DCs for intercellular communication with miRNAs in DCs (122), which significantly reduces antitumor immunity. Let-7d miRNA is encased in exosomes and preferentially acts on Th1 cells, reducing their proliferation and immunity by inhibiting cyclooxygenase 2 (123). Certain Treg-derived exosomes have a 25-100-fold higher IL-35 concentration in the cell surface, and IL-35 acts on target cells to promote the expression of PD-1, Tim3 and Lag3 (124), suppressing antitumor immunity. In summary, Tregs can suppress antitumor immunity and the abscopal effect by secreting various exosomes.

After radiotherapy, CXCR4 antagonist can promote the depletion of Tregs and enhance the antitumor and anti-metastasis therapeutic effect (125,126). Depletion also encourages T cells to mature into effector memory T cells (127), as well as inhibiting Treg proliferation, resulting in a higher CD8+ T cell/Treg ratio. Tregs are less vulnerable to radiation than CTLs, and their increased activity can be maintained even at larger doses (128); therefore, the radiation dose can be utilized to modify the CD8+ T cell/Treg ratio. According to a previous study, hypo-fractionated stereotactic radiation therapy can increase the number of CD8+ T cells while decreasing the number of Tregs (129).

Dual effect of Th17 cells

Th17 cells, a novel assistant cell distinct from Th1 and Th2 cells (130), have a dual effect on tumors, resulting in their 'double-edged sword' function in the occurrence and progression of the abscopal effect. Th17 infiltration was also observed in tumors that showed the abscopal effect, such as prostate, lung and breast cancer, and melanoma (131,132). Th17 cells, on the one hand, can release CCL20, recruit DCs to the tumor, activate CD8+ T cells and release IFN-γ to increase the antitumor response (133,134), thus contributing to the abscopal effect. Th17 cells, on the other hand, have an immunosuppressive effect after entering tumors. They produce IL-17, attract myeloid-derived suppressor cells (MDSCs), and promote their own proliferation and development (135,136). The concentration of VEGF is positively correlated with the concentration of IL-17 (137), and IL-17 can increase tumor angiogenesis, thus enhancing tumor proliferation and metastasis. Meanwhile, the anti-abscopal extracellular nucleotidase CD39 and CD73 on the surface of Th17 cells decompose ATP into AMP to achieve an immunosuppressive effect (117).

The role of Th17 cells in malignancies, on the other hand, is complex, and their processes are unknown. Meanwhile, their effects differ depending on the tumor type. Thus, further research is required. To summarize, different types of T cells play diverse roles in the abscopal effect. Only by further studying and specifying the role and mechanism of distinct T cells, as well as the connections between T cells and other cells, the human body's antitumor immunity will be better understood and maintained.

Role of MDSCs

MDSCs, which are immature myeloid cells produced and secreted by the bone marrow, are recruited to the TME to control the immune response and build an immunological-tolerant TME. Meanwhile, additional MDSCs will be created by the bone marrow, which is triggered by chronic inflammatory signals sent out by the TME (138), thus creating a feedback loop that interferes with tumor therapy. According to a previous study, tumor cells in a model lacking MDSCs were quickly removed by activated antitumor immune cells (139), indicating the critical role of MDSCs in the immunosuppressive process. Exosomes secreted by MDSCs play an important role in suppressing the abscopal effect and promoting tumor immune escape, proliferation and migration. Thanks to their contents (>4,000 types of protein), MDSCs-derived exosomes and MDSCs support each other, strengthen the immunosuppressive effect, and promote tumor proliferation and survival (140).

It has been demonstrated that, even during radiation, MDSCs suppress the abscopal effect, since they arrive at the tumor site 10 days after radiotherapy (139), and decrease antitumor immunity by releasing cytokines such as TGF-β and strongly expressing PD-L1 (141). Proliferation of MDSCs can be aided in a variety of ways. VEGF has been shown to play a significant role in promoting the proliferation of MDSCs (142), and an increase in MDSCs and a decrease in effector T cells have been observed in tumor models with high VEGF expression (143), implying that VEGF inhibits the abscopal effect by promoting MDSCs.

IL-6 is linked to aggressive tumor development and recruitment of MDSCs (144), and this process reduces IL-6-silencing small hairpin RNA, upregulates miR-155 and miR-21, and activates STAT3, thus initiating and enhancing proliferation of MDSCs (145). Furthermore, miR-9, miR-494 and miR-21 in these exosomes govern the cell cycle, boost MDSCs proliferation and expansion, increase MDSCs accumulation in the TME, and enhance MDSCs immunosuppressive capacity, all of which promote tumor progression (146,147). The involvement of nucleic acid in MDSCs exosomes is clearer, since MDSCs-derived exosomes contain a high concentration of nucleic acid. Their miR-146a content, which is 18-fold higher than that of cells, acts on TNF receptor-associated factor 6 and other NF-κB pathway-related receptors, thereby regulating the genesis and apoptosis of cervical cancer cells (148,149).

Moreover, the proliferation of MDSCs is promoted by PGE2, which activates the p38MAPK/ERK signaling pathway to enhance the release of TGF-β (150,151). HMGB1 is a high-content protein in MDSCs-derived exosomes, which can induce the production and accumulation of MDSCs. Other MDSCs-derived exosomes rich in TGF-β1, IL-10 and IL-6 may be ingested by macrophages and T cells, causing a significant increase in Tregs to play a stronger immunosuppressive effect, a reduction in the proliferation of Th cells, weakened CTL cytotoxic activity and a slight increase in the lymphocyte apoptosis rate. Specifically, the content of TGF-β1 within these exosomes is 4.3-fold higher than that within the cell, thus promoting tumor angiogenesis and metastasis (148,152,153). Moreover, it has been shown that MDSC-derived exosomes are associated with the resistance of chemotherapeutic drugs (152).

In summary, MDSCs can inhibit the generation of the abscopal effect via a variety of mechanisms. Furthermore, the greater the accumulation of MDSCs in the TME, the stronger its immunosuppressive ability, the more conducive to tumor growth, ultimately having a negative effect on the overall treatment of the tumor. As a result, they may be utilized as a prognostic marker in patients, and targeting MDSCs can help restore antitumor immunity and boost the abscopal effect.

Role of eosinophils

Eosinophils have been observed to infiltrate numerous tumors, directly interact with tumor cells, govern tumor formation by modifying the TME and contribute to antitumor immunity via a number of pathways. Eosinophils have high levels of IL-1 receptor 1, TLR4 and RAGE expression. They also recruit to the tumor site following radiotherapy-induced DAMPs such as HMGB1 and IL-33 signals (154).

Eosinophils have a two-pronged effect on malignancies. On one hand, they cause CD8+ T cells to penetrate tumors, normalize blood vessels and drive macrophage polarization to M1 cells, resulting in a marked antitumor action (155). Although the underlying mechanism is unknown, eosinophils can significantly boost the abscopal effect. On the other hand, eosinophils play an inhibitory function in immunity (156). When triggered by thymic stromal lymphopoietin, they restrict DC maturation and block tumor apoptosis by secreting a substantial quantity of IL-10, IL-4, IL-5 and IL-13 (157), thus increasing tumor growth. TGF-β (158) is also released by eosinophils to impact the abscopal effect. Furthermore, they produce IDO (159) to enhance TME's immunosuppressive function, boost M2 polarization of macrophages in the TME by releasing IL-13 and IL-4 (160), and decrease the antitumor effect. As a result, inhibiting the tumor-promoting function of eosinophils, so that they improve the antitumor action of macrophages to promote the abscopal effect, is worth investigating.

There are vigorous battles between antitumor cells, such as DCs and NK, CD8+ T, M1 cells and CTLs, and immunosuppressive cells, such as M2 cells, MDSCs and Tregs, during the generation of the abscopal effect after radiotherapy (Fig. 2). Such battles are regulated by cells with dual functions, including Th17 cells and eosinophils (Fig. 3), resulting in complex and mutual influences on these antitumor and immunosuppressive cells: CD8+ T cells and Tregs interact to produce adenosine (72); CD8+ T cells bind to eosinophils; the TME enhances the immunosuppressive effect of MDSCs; and MDSCs and Tregs mutually promote the expansion of each other.

Immunosuppressive cells gradually win the fight to suppress the abscopal effect later in the process. Despite the fact that the current immunotherapy has overcome the disadvantages of antitumor cells, the lack of knowledge of the underlying mechanism, combined with the complexity of the aforementioned battles, hampers the formation of the abscopal effect over time (Fig. 4).

5. Clinical application

Radiotherapy combined with immunotherapy

Radiotherapy is an important cancer treatment that can boost the abscopal effect. Radiotherapy was previously only utilized to treat local malignancies due to technological limitations and its harmful effects on normal tissues. In addition to its cell-killing impact, radiation modulates immunomodulation. Radiotherapy, for example, renders tumor cells more vulnerable to T cell attack. After each 10-25 Gy low-fraction radiation session, the expression of MHC-1 molecules on the surface of human melanoma cells was increased (161), which enhances the presentation of antigens, making it easier for these tumor cells to be destroyed and removed by T lymphocytes. Furthermore, various immune cells respond differently to radiation; for example, a radiation dosage of 0.94 Gy strongly inhibits Treg proliferation (162). Due to the lack of studies on different tumor radiotherapy doses and the susceptibility of different cells to radiotherapy, it is difficult to utilize radiotherapy alone to overcome the inhibitory impact of the TME, which is why the abscopal effect was uncommon in the past.

However, immunotherapy compensates for this rarity. Multiple clinical and pre-clinical studies (Table I) have shown that, compared with the effect of radiotherapy or immunotherapy alone, the combination of radiotherapy and immunotherapy can significantly increase the incidence and intensity of the abscopal effect (7,13,163-166). Commonly used immune checkpoint inhibitors are anti-CTLA-4, anti-PD-1 and anti-PD-L1 antibodies, all of which strengthen the effect of T cells on tumor cells and can be combined with radiotherapy to enhance the incidence rate of the abscopal effect. A recent study showed that the combination of 8 Gy x3F radiotherapy and anti-PD-L1 monoclonal antibody can enhance the abscopal effect, significantly reduce MDSCs and promote CD8+ T cell infiltration (167). Another clinical study indicated that radiotherapy combined with immune adjuvant GM-CSF treatment can trigger the abscopal effect in 30% of patients with cancer (168). Similarly, anti-CD40 antibody could maintain the antitumor effect of APCs (95), and FMS-like tyrosine kinase 3 ligand could recruit and stimulate APCs (169), so both of them can enhance the effect of APCs, and increase the incidence of the abscopal effect when combined with radiotherapy. In addition, when combined with tumor cells, both anti-CD47 and anti-CD73 antibodies can promote APCs to phagocytose tumor cells (170-172), and further exert their antitumor role.

Table I

Compared with radiotherapy or immunotherapy alone, the combination of radiotherapy and immunotherapy can significantly increase the incidence and intensity of the abscopal effect.

Table I

Compared with radiotherapy or immunotherapy alone, the combination of radiotherapy and immunotherapy can significantly increase the incidence and intensity of the abscopal effect.

Tumor typeRadiation doseTypes of immunotherapyCombined treatment sequence
Melanoma30×10CTLA4Immunotherapy before radiotherapy
Colon adenocarcinoma3×8 Gy Anti-PD1/anti-CD137Immunotherapy after radiotherapy
Glioblastoma10 GyAnti-PD-L1Immunotherapy after radiotherapy
NSCLC9 Gy × 3/6 Gy × 5Anti-CTLA-4Immunotherapy after radiotherapy
Adenocarcinoma30 Gy (10 fractions)AtezolizumabImmunotherapy after radiotherapy
Head and neck squamous cell carcinoma4,500 cGyAtezolizumabImmunotherapy before radiotherapy
NSCLC3 fractions of 12 GyAnti-PD-L1Immunotherapy before radiotherapy
NSCLC8 Gy × 3/12 Gy × 3Anti-PD-1Immunotherapy before radiotherapy

[i] NSCLC, non-small cell lung cancer; CTLA-4, cytotoxic T lymphocyte-associated antigen-4; PD-1, programmed cell death-1; PD-L1, programmed cell death 1 ligand 1.

However, immunotherapy also has defects such as cross-reactions. Specifically, while anti-CD47 eliminates tumor cells, it may accidentally injure the red blood cells that also carry CD47 on their surface (173), leading to anemia. Besides, the overlap and systemic toxicity of a combination of radiotherapy and immunotherapy are still difficult to deal with, although a reasonable combination can overcome immunosuppression and promote the generation of the abscopal effect (16). Moreover, immune-related adverse reactions are worrying. Even if radiotherapy can overcome the drug resistance of anti-PD-1 antibody, it does not have a long-lasting therapeutic effect on ~80% of patients with non-small cell lung cancer (NSCLC), due to tumor oxidative metabolism obstacles (174). The dosage and sequence of the combination of radiotherapy and immunotherapy for different tumors, or different combination options from different tumor radiotherapy and immunotherapy methods are currently unclear; therefore, it is necessary to further explore immunotherapy options and timings for different tumor types to find the best timing, dosage and sequence of the combination of radiotherapy and immunotherapy. Emerging methods and technologies may help to understand how to generate the abscopal effect and promote its incidence rate, which is beneficial to the treatment and prognosis of patients. These benefits are subsequently described in the present review.

New radiotherapy technologies are conducive to the abscopal effect. The development of radiotherapy technology has made a great progress. Compared with traditional radiotherapy methods, new technologies may be more conducive to the generation of the abscopal effect. Stereotactic body radiation therapy combined with immunotherapy is well tolerated and relatively safe, and there were cases of lung cancer as well as head and neck squamous cell carcinoma where the abscopal effect was generated by such a combination (163,166). In addition, technologies such as intensity-modulated radiation therapy, stereotactic ablative radiotherapy or proton therapy can change the range of radiotherapy according to the tumor size and greatly reduce radiation toxicity (3), thus overcoming the toxicity caused by combined therapy. Besides, high-dose radiation (HDR) brachytherapy can protect adjacent healthy tissues by bringing the emission source into the tumor tissue, thereby reducing radiotherapy-induced toxicity (175). Research has shown that a combination of HDR brachytherapy with anti-PD-1 or anti-CD137 antibodies can produce the abscopal effect (176). When these new radiotherapy approaches are combined with different immunotherapy methods, the best combination option and timing may be found, which may overcome the limitations caused by the toxicity of radiotherapy in the past and help to improve the current type of combination of radiotherapy and immunotherapy.

Triple therapy including radiotherapy and immunotherapy

As aforementioned, the combination of radiotherapy and immunotherapy can achieve in an improved way the antitumor effect and reduce the drug resistance to immunotherapy; however, such combination also has a limited effect on the generation of an abscopal effect sufficiently strong in certain tumors such as NSCLC. The underlying mechanism of such limitation may be the regulation of the ERK signaling pathway to act on Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2), which regulates tumor cell proliferation (177) and is the main effector mediating the downstream signal transduction of PD-1 in T cells (178). Previous research has shown that the triple therapy of SHP2 inhibitor, anti-PD-L1 antibody and radiotherapy can increase the ratio of M1/M2 cells and CTL/Treg lymphocytes to stimulate antitumor immunity (174). In addition, the oxidative phosphorylation (OXPHOS) of tumor mitochondria may be another cause of the aforementioned poor effect of the combination of radiotherapy and immunotherapy (179). The triple therapy of IACS-010759, an OXPHOS inhibitor, combined with anti-PD-1 antibody and radiotherapy can promote the abscopal effect (180) and resolve the problem of anti-PD-1 resistance in NSCLC. Other triple or quadruple therapies could be used to overcome the disadvantages of the traditional combination of radiotherapy and immunotherapy. For example, genetic ablation of the TGF-β signaling pathway components added to the conventional radioimmunotherapy could trigger a powerful antitumor response (63), as well as a combination of anti-PD-1 treatment after radiotherapy (181) and targeted suppression of antitumor immunity. Similarly, exosomes within the TME can also enhance the therapeutic effect, and ultimately promote the abscopal effect to prolong the survival time of the patient.

Oncolytic virus

In oncolytic immunotherapy, an oncolytic virus is often injected locally into tumors, which has a tropism for malignant tumor cells and can replicate in tumor cells to eventually promote their lysis (182). Oncolytic virus replication can induce the death of tumor immunogenic cells, send out immunological danger signals, promote tumors to produce TNF-α, and induce the body to produce strong immune effects (183), thereby enhancing the occurrence of the abscopal effect. The shrinkage of distal tumors after the local injection of an oncolytic virus has been reported, and the mechanism is similar to that of the abscopal effect (184). When oncolytic viruses are used in combination with immune checkpoint inhibitors and radiotherapy their effects can be enhanced. Specifically, the oncolytic adenovirus is currently one of the most promising oncolytic viruses (185). Recently, a patient with Hodgkin's lymphoma infected with the new coronavirus experienced systemic tumor regression. The reason may be that the coronavirus triggered antitumor immunity in his body (186); therefore, this novel coronavirus may also have the potential to be developed as an oncolytic virus to promote the abscopal effect.

Smart material technology

Smart material technology is divided into nanoparticles and intelligent radiotherapy biomaterials (187). Within nanoparticles, nanoparticle-delivered drugs have great potential for improving the antitumor immune effect. Nano-immunotherapy, which is the combination of nanoparticle-delivered drugs and immunotherapy, can be achieved in three different ways, and these nano-drugs are used to target cancer cells and the TME (58). When targeting cancer cells, nanoparticle-delivered drugs cause the immunogenic death of tumor cells and can be combined with immunotherapy to greatly promote antitumor immunity (188). Moreover, when combined with photodynamic radiotherapy to treat primary tumors, nanodrugs can promote the occurrence of abscopal effects (189). Calcium carbonate nanoparticles with anti-CD47 activity have been developed (190). When targeting the TME, nanodrugs such as antigen capture nanoparticles can capture TAAs to activate DCs, and thus promote the abscopal effect (191). Certain nanodrugs can also act on immunosuppressive molecules, such as IDO, TGF-β and IL-2 (192) to reshape the TME, which is beneficial for antitumor immunity.

The second type of smart material technology, smart radiotherapy biomaterials, also promotes the abscopal effect. For example, a hydrogel formed by alginate can capture the drug formed by the combination of 131I-labeled catalase and the immune adjuvant CpG, and the immune checkpoint inhibitor of the combination of the hydrogel and the drug can produce powerful antitumor immunity and the abscopal effect, which has been observed in experimental mice (193). Compared with traditional technologies, these new technologies have improved treatment methods to reduce toxicity towards normal tissues and/or lymphocytes, have improved targeting ability, are beneficial to patients, and can reduce the cost of treatment for patients.

6. Conclusions

Tumor metastasis has caused the suffering and mortality of >90% of patients with cancer. The abscopal effect can be used to combat tumor metastasis. The biggest advantage of this abscopal effect is the inhibition and elimination of distant and metastatic tumors. Therefore, by further studying the underlying mechanism and improved using of new technologies and methods to enhance the abscopal effect, an improved treatment plan for patients with cancer could be developed. Furthermore, new radiotherapy and immunotherapy approaches based on cells and exosomes that play a role in the abscopal effect are beneficial to increase the incidence of abscopal effects in clinical practice. Besides, its needs to be taken into consideration how to reduce the toxicity caused by treatment, relieve the suffering of patients, and reduce the cost of treatment. It is worth noting that microwave ablation has been found to induce the abscopal effect in clinical practice (194), and the effect of oncolytic viruses is similar to that of the abscopal effect. Therefore, investigating the mechanism of oncolytic viruses may help to find another way to promote the abscopal effect.

Availability of data and materials

Not applicable.

Authors' contributions

JW and GK wrote the manuscript. ZW and CL created the figures. JW and JL revised the manuscript. All authors read and approved the final version of the manuscript.

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 National Students' Innovation and Entrepreneurship Training Program (grant nos. S202210632275, S202210632129 and 202110632052).

References

1 

Chargari C, Deutsch E, Blanchard P, Gouy S, Martelli H, Guérin F, Dumas I, Bossi A, Morice P, Viswanathan AN and Haie-Meder C: Brachytherapy: An overview for clinicians. CA Cancer J Clin. 69:386–401. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Mohamad O, Tabuchi T, Nitta Y, Nomoto A, Sato A, Kasuya G, Makishima H, Choy H, Yamada S, Morishima T, et al: Risk of subsequent primary cancers after carbon ion radiotherapy, photon radiotherapy, or surgery for localised prostate cancer: A propensity score-weighted, retrospective, cohort study. Lancet Oncol. 20:674–685. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Kahalley LS, Peterson R, Ris MD, Janzen L, Okcu MF, Grosshans DR, Ramaswamy V, Paulino AC, Hodgson D, Mahajan A, et al: Superior intellectual outcomes after proton radiotherapy compared with photon radiotherapy for pediatric medulloblastoma. J Clin Oncol. 38:454–461. 2020. View Article : Google Scholar :

4 

Mole RH: Whole body irradiation; radiobiology or medicine? Br J Radiol. 26:234–241. 1953. View Article : Google Scholar : PubMed/NCBI

5 

Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L and Formenti SC: Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 58:862–870. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Tan AC, Ashley DM, López GY, Malinzak M, Friedman HS and Khasraw M: Management of glioblastoma: State of the art and future directions. CA Cancer J Clin. 70:299–312. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Ene CI, Kreuser SA, Jung M, Zhang H, Arora S, White Moyes K, Szulzewsky F, Barber J, Cimino PJ, Wirsching HG, et al: Anti-PD-L1 antibody direct activation of macrophages contributes to a radiation-induced abscopal response in glioblastoma. Neuro Oncol. 22:639–651. 2020. View Article : Google Scholar

8 

Lheureux S, Braunstein M and Oza AM: Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J Clin. 69:280–304. 2019.PubMed/NCBI

9 

Formenti SC, Rudqvist NP, Golden E, Cooper B, Wennerberg E, Lhuillier C, Vanpouille-Box C, Friedman K, Ferrari de Andrade L, Wucherpfennig KW, et al: Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med. 24:1845–1851. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Yang W, Zhang F, Deng H, Lin L, Wang S, Kang F, Yu G, Lau J, Tian R, Zhang M, et al: Smart nanovesicle-mediated immunogenic cell death through tumor microenvironment modulation for effective photodynamic immunotherapy. ACS Nano. 14:620–631. 2020. View Article : Google Scholar

11 

Hu ZI, McArthur HL and Ho AY: The abscopal effect of radiation therapy: What is it and how can we use it in breast cancer? Curr Breast Cancer Rep. 9:45–51. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Beyls C, Haustermans K, Deroose CM, Pans S, Vanbeckevoort D, Verslype C and Dekervel J: Could autoimmune disease contribute to the abscopal effect in metastatic hepatocellular carcinoma? Hepatology. 72:1152–1154. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Guan S, Wang H, Qi XH, Guo Q, Zhang HY, Liu H and Zhu BJ: Abscopal effect of local irradiation treatment for thymoma: A case report. Am J Transl Res. 12:2234–2240. 2020.PubMed/NCBI

14 

Chakravarty PK, Alfieri A, Thomas EK, Beri V, Tanaka KE, Vikram B and Guha C: Flt3-ligand administration after radiation therapy prolongs survival in a murine model of metastatic lung cancer. Cancer Res. 59:6028–6032. 1999.

15 

Camphausen K, Moses MA, Ménard C, Sproull M, Beecken WD, Folkman J and O'Reilly MS: Radiation abscopal antitumor effect is mediated through p53. Cancer Res. 63:1990–1993. 2003.PubMed/NCBI

16 

Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S and Formenti SC: Using immunotherapy to boost the abscopal effect. Nat Rev Cancer. 18:313–322. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Sexton RE, Mpilla G, Kim S, Philip PA and Azmi AS: Ras and exosome signaling. Semin Cancer Biol. 54:131–137. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Möller A and Lobb RJ: The evolving translational potential of small extracellular vesicles in cancer. Nat Rev Cancer. 20:697–709. 2020. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

He C, Li L, Wang L, Meng W, Hao Y and Zhu G: Exosome-mediated cellular crosstalk within the tumor microenvironment upon irradiation. Cancer Biol Med. 18:21–33. 2021. View Article : Google Scholar : PubMed/NCBI

21 

Hu X, Qiu Y, Zeng X and Wang H: Exosomes reveal the dual nature of radiotherapy in tumor immunology. Cancer Sci. 113:1105–1112. 2022. View Article : Google Scholar : PubMed/NCBI

22 

Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, Inghirami G, Coleman CN, Formenti SC and Demaria S: DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 8:156182017. View Article : Google Scholar : PubMed/NCBI

23 

Craig DJ, Nanavaty NS, Devanaboyina M, Stanbery L, Hamouda D, Edelman G, Dworkin L and Nemunaitis JJ: The abscopal effect of radiation therapy. Future Oncol. 17:1683–1694. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Zhao X, Hu S, Zeng L, Liu X, Song Y, Zhang Y, Chen Q, Bai Y, Zhang J, Zhang H, et al: Irradiation combined with PD-L1(-/-) and autophagy inhibition enhances the antitumor effect of lung cancer via cGAS-STING-mediated T cell activation. iScience. 25:1046902022. View Article : Google Scholar

25 

Wang J, Wang L, Lin Z, Tao L and Chen M: More efficient induction of antitumor T cell immunity by exosomes from CD40L gene-modified lung tumor cells. Mol Med Rep. 9:125–131. 2014. View Article : Google Scholar

26 

Choi D, Montermini L, Kim DK, Meehan B, Roth FP and Rak J: The impact of oncogenic EGFRvIII on the proteome of extracellular vesicles released from glioblastoma cells. Mol Cell Proteomics. 17:1948–1964. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Hsieh RC, Krishnan S, Wu RC, Boda AR, Liu A, Winkler M, Hsu WH, Lin SH, Hung MC, Chan LC, et al: ATR-mediated CD47 and PD-L1 up-regulation restricts radiotherapy-induced immune priming and abscopal responses in colorectal cancer. Sci Immunol. 7:eabl93302022. View Article : Google Scholar : PubMed/NCBI

28 

Feng M, Jiang W, Kim BYS, Zhang CC, Fu YX and Weissman IL: Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat Rev Cancer. 19:568–586. 2019. View Article : Google Scholar : PubMed/NCBI

29 

He S, Cheng J, Sun L, Wang Y, Wang C, Liu X, Zhang Z, Zhao M, Luo Y, Tian L, et al: HMGB1 released by irradiated tumor cells promotes living tumor cell proliferation via paracrine effect. Cell Death Dis. 9:6482018. View Article : Google Scholar : PubMed/NCBI

30 

Leonardi GC, Falzone L, Salemi R, Zanghì A, Spandidos DA, Mccubrey JA, Candido S and Libra M: Cutaneous melanoma: From pathogenesis to therapy (Review). Int J Oncol. 52:1071–1080. 2018.PubMed/NCBI

31 

Minton K: Predicting the anti-PD1 response. Nat Rev Immunol. 19:414–415. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Daassi D, Mahoney KM and Freeman GJ: The importance of exosomal PDL1 in tumour immune evasion. Nat Rev Immunol. 20:209–215. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Bennett F, Luxenberg D, Ling V, Wang IM, Marquette K, Lowe D, Khan N, Veldman G, Jacobs KA, Valge-Archer VE, et al: Program death-1 engagement upon TCR activation has distinct effects on costimulation and cytokine-driven proliferation: Attenuation of ICOS, IL-4, and IL-21, but not CD28, IL-7, and IL-15 responses. J Immunol. 170:711–718. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Yuan Y, Wang L, Ge D, Tan L, Cao B, Fan H and Xue L: Exosomal O-GlcNAc transferase from esophageal carcinoma stem cell promotes cancer immunosuppression through up-regulation of PD-1 in CD8(+) T cells. Cancer Lett. 500:98–106. 2021. View Article : Google Scholar

35 

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. 70:241–258. 2019.PubMed/NCBI

36 

Ye L, Zhang Q, Cheng Y, Chen X, Wang G, Shi M, Zhang T, Cao Y, Pan H, Zhang L, et al: Tumor-derived exosomal HMGB1 fosters hepatocellular carcinoma immune evasion by promoting TIM-1(+) regulatory B cell expansion. J Immunother Cancer. 6:1452018. View Article : Google Scholar : PubMed/NCBI

37 

Wang Y, Yi J, Chen X, Zhang Y, Xu M and Yang Z: The regulation of cancer cell migration by lung cancer cell-derived exosomes through TGF-β and IL-10. Oncol Lett. 11:1527–1530. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Andreola G, Rivoltini L, Castelli C, Huber V, Perego P, Deho P, Squarcina P, Accornero P, Lozupone F, Lugini L, et al: Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J Exp Med. 195:1303–1316. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Fuentes P, Sesé M, Guijar ro PJ, Emperador M, Sánchez-Redondo S, Peinado H, Hümmer S and Cajal SRY: Publisher Correction: ITGB3-mediated uptake of small extracellular vesicles facilitates intercellular communication in breast cancer cells. Nat Commun. 11:47302020. View Article : Google Scholar : PubMed/NCBI

40 

Ji Q, Zhou L, Sui H, Yang L, Wu X, Song Q, Jia R, Li R, Sun J, Wang Z, et al: Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat Commun. 11:12112020. View Article : Google Scholar : PubMed/NCBI

41 

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. 7:e14129092018. View Article : Google Scholar : PubMed/NCBI

42 

Ferguson Bennit HR, Gonda A, Kabagwira J, Oppegard L, Chi D, Licero Campbell J, De Leon M and Wall NR: Natural killer cell phenotype and functionality affected by exposure to extracellular survivin and lymphoma-derived exosomes. Int J Mol Sci. 22:12552021. View Article : Google Scholar : PubMed/NCBI

43 

Kitai Y, Kawasaki T, Sueyoshi T, Kobiyama K, Ishii KJ, Zou J, Akira S, Matsuda T and Kawai T: DNA-containing exosomes derived from cancer cells treated with topotecan activate a STING-Dependent pathway and reinforce antitumor immunity. J Immunol. 198:1649–1659. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Sun J, Jia H, Bao X, Wu Y, Zhu T, Li R and Zhao H: Tumor exosome promotes Th17 cell differentiation by transmitting the lncRNA CRNDE-h in colorectal cancer. Cell Death Dis. 12:1232021. View Article : Google Scholar : PubMed/NCBI

45 

Ding G, Zhou L, Qian Y, Fu M, Chen J, Chen J, Xiang J, Wu Z, Jiang G and Cao L: Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget. 6:29877–29888. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Zhao S, Mi Y, Guan B, Zheng B, Wei P, Gu Y, Zhang Z, Cai S, Xu Y, Li X, et al: Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J Hematol Oncol. 13:1562020. View Article : Google Scholar : PubMed/NCBI

47 

Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, Liu B, Su L and Qiu Z: Hypoxic tumor-derived exosomal miR-301a Mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer metastasis. Cancer Res. 78:4586–4598. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Chen X, Zhou J, Li X and Wang X, Lin Y and Wang X: Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 435:80–91. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Hurley JH: ESCRTs are everywhere. EMBO J. 34:2398–2407. 2015. View Article : Google Scholar : PubMed/NCBI

50 

de Araujo Farias V, O'Valle F, Serrano-Saenz S, Anderson P, Andrés E, López-Peñalver J, Tovar I, Nieto A, Santos A, Martín F, et al: Exosomes derived from mesenchymal stem cells enhance radiotherapy-induced cell death in tumor and metastatic tumor foci. Mol Cancer. 17:1222018. View Article : Google Scholar : PubMed/NCBI

51 

Stary V, Wolf B, Unterleuthner D, List J, Talic M, Laengle J, Beer A, Strobl J, Stary G, Dolznig H and Bergmann M: Short-course radiotherapy promotes pro-inflammatory macrophages via extracellular vesicles in human rectal cancer. J Immunother Cancer. 8:e0006672020. View Article : Google Scholar : PubMed/NCBI

52 

Ahn J, Xia T, Rabasa Capote A, Betancourt D and Barber GN: Extrinsic phagocyte-dependent STING signaling dictates the immunogenicity of dying cells. Cancer Cell. 33:862–873.e5. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Jiang MJ, Chen YY, Dai JJ, Gu DN, Mei Z, Liu FR, Huang Q and Tian L: Dying tumor cell-derived exosomal miR-194-5p potentiates survival and repopulation of tumor repopulating cells upon radiotherapy in pancreatic cancer. Mol Cancer. 19:682020. View Article : Google Scholar : PubMed/NCBI

54 

Khambu B, Huda N, Chen X, Antoine DJ, Li Y, Dai G, Köhler UA, Zong WX, Waguri S, Werner S, et al: HMGB1 promotes ductular reaction and tumorigenesis in autophagy-deficient livers. J Clin Invest. 128:2419–2435. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR and Ferguson TA: Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity. 29:21–32. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M and Formenti SC: Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology. 3:e285182014. View Article : Google Scholar : PubMed/NCBI

57 

Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G, et al: Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 334:1573–1577. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Shi Y and Lammers T: Combining nanomedicine and immunotherapy. Acc Chem Res. 52:1543–1554. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Lecciso M, Ocadlikova D, Sangaletti S, Trabanelli S, De Marchi E, Orioli E, Pegoraro A, Portararo P, Jandus C, Bontadini A, et al: ATP release from chemotherapy-treated dying leukemia cells elicits an immune suppressive effect by increasing regulatory T cells and tolerogenic dendritic cells. Front Immunol. 8:19182017. View Article : Google Scholar

60 

Yamaguchi H, Maruyama T, Urade Y and Nagata S: Immunosuppression via adenosine receptor activation by adenosine monophosphate released from apoptotic cells. Elife. 3:e021722014. View Article : Google Scholar : PubMed/NCBI

61 

Allard D, Allard B and Stagg J: On the mechanism of anti-CD39 immune checkpoint therapy. J Immunother Cancer. 8:e0001862020. View Article : Google Scholar : PubMed/NCBI

62 

Baghbani E, Noorolyai S, Shanehbandi D, Mokhtarzadeh A, Aghebati-Maleki L, Shahgoli VK, Brunetti O, Rahmani S, Shadbad MA, Baghbanzadeh A, et al: Regulation of immune responses through CD39 and CD73 in cancer: Novel checkpoints. Life Sci. 282:1198262021. View Article : Google Scholar : PubMed/NCBI

63 

Batlle E and Massagué J: Transforming growth factor-β signaling in immunity and cancer. Immunity. 50:924–940. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Formenti SC, Lee P, Adams S, Goldberg JD, Li X, Xie MW, Ratikan JA, Felix C, Hwang L, Faull KF, et al: Focal irradiation and systemic TGFβ blockade in metastatic breast cancer. Clin Cancer Res. 24:2493–2504. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Karapetyan L, Luke JJ and Davar D: Toll-Like Receptor 9 Agonists in Cancer. Onco Targets Ther. 13:10039–10060. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Vincent-Schneider H, Stumptner-Cuvelette P, Lankar D, Pain S, Raposo G, Benaroch P and Bonnerot C: Exosomes bearing HLA-DR1 molecules need dendritic cells to efficiently stimulate specific T cells. Int Immunol. 14:713–722. 2002. View Article : Google Scholar : PubMed/NCBI

67 

Xie F, Zhou X, Fang M, Li H, Su P, Tu Y, Zhang L and Zhou F: Extracellular vesicles in cancer immune microenvironment and cancer immunotherapy. Adv Sci (Weinh). 6:19017792019. View Article : Google Scholar : PubMed/NCBI

68 

Pitt JM, André F, Amigorena S, Soria JC, Eggermont A, Kroemer G and Zitvogel L: Dendritic cell-derived exosomes for cancer therapy. J Clin Invest. 126:1224–1232. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Quah BJ and O'Neill HC: Maturation of function in dendritic cells for tolerance and immunity. J Cell Mol Med. 9:643–654. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Pang G, Chen C, Liu Y, Jiang T, Yu H, Wu Y, Wang Y, Wang FJ, Liu Z and Zhang LW: Bioactive polysaccharide nanoparticles improve radiation-induced abscopal effect through manipulation of dendritic cells. ACS Appl Mater Interfaces. 11:42661–42670. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Accogli T, Bruchard M and Végran F: Modulation of CD4 T cell response according to tumor cytokine microenvironment. Cancers (Basel). 13:3732021. View Article : Google Scholar : PubMed/NCBI

72 

Shiokawa A, Kotaki R, Takano T, Nakajima-Adachi H and Hachimura S: Mesenteric lymph node CD11b(−) CD103(+) PD-L1High dendritic cells highly induce regulatory T cells. Immunology. 152:52–64. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Zhou F, Zhang GX and Rostami A: Distinct Role of IL-27 in Immature and LPS-Induced mature dendritic cell-mediated development of CD4(+) CD127(+)3G11(+) regulatory T cell subset. Front Immunol. 9:25622018. View Article : Google Scholar : PubMed/NCBI

74 

Shirasawa M, Yoshida T, Matsumoto Y, Shinno Y, Okuma Y, Goto Y, Horinouchi H, Yamamoto N, Watanabe SI, Ohe Y and Motoi N: Impact of chemoradiotherapy on the immune-related tumour microenvironment and efficacy of anti-PD-(L)1 therapy for recurrences after chemoradiotherapy in patients with unresectable locally advanced non-small cell lung cancer. Eur J Cancer. 140:28–36. 2020. View Article : Google Scholar : PubMed/NCBI

75 

Tang C, Wang X, Soh H, Seyedin S, Cortez MA, Krishnan S, Massarelli E, Hong D, Naing A, Diab A, et al: Combining radiation and immunotherapy: A new systemic therapy for solid tumors? Cancer Immunol Res. 2:831–838. 2014. View Article : Google Scholar : PubMed/NCBI

76 

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

77 

Wang J, Deng Z, Wang Z, Wu J, Gu T, Jiang Y and Li G: MicroRNA-155 in exosomes secreted from helicobacter pylori infection macrophages immunomodulates inflammatory response. Am J Transl Res. 8:3700–3709. 2016.PubMed/NCBI

78 

Wang P, Wang H, Huang Q, Peng C, Yao L, Chen H, Qiu Z, Wu Y, Wang L and Chen W: Exosomes from M1-Polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation. Theranostics. 9:1714–1727. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Deng F, Yan J, Lu J, Luo M, Xia P, Liu S, Wang X, Zhi F and Liu D: M2 Macrophage-Derived Exosomal miR-590-3p Attenuates DSS-Induced mucosal damage and promotes epithelial repair via the LATS1/YAP/β-Catenin Signalling Axis. J Crohns Colitis. 15:665–677. 2021. View Article : Google Scholar

80 

Wu J, Gao W, Tang Q, Yu Y, You W, Wu Z, Fan Y, Zhang L, Wu C, Han G, et al: M2 Macrophage-derived exosomes facilitate HCC metastasis by transferring αM β2 integrin to tumor cells. Hepatology. 73:1365–1380. 2021. View Article : Google Scholar

81 

Tavazoie MF, Pollack I, Tanqueco R, Ostendorf BN, Reis BS, Gonsalves FC, Kurth I, Andreu-Agullo C, Derbyshire ML, Posada J, et al: LXR/ApoE Activation restricts innate immune suppression in cancer. Cell. 172:825–840.e18. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Zheng P, Chen L, Yuan X, Luo Q, Liu Y, Xie G, Ma Y and Shen L: Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res. 36:532017. View Article : Google Scholar : PubMed/NCBI

83 

Hao Y, Yasmin-Karim S, Moreau M, Sinha N, Sajo E and Ngwa W: Enhancing radiotherapy for lung cancer using immunoadjuvants delivered in situ from new design radiotherapy biomaterials: A preclinical study. Phys Med Biol. 61:N697–n707. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Qian M, Wang S, Guo X, Wang J, Zhang Z, Qiu W, Gao X, Chen Z, Xu J, Zhao R, et al: Hypoxic glioma-derived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-κB pathways. Oncogene. 39:428–442. 2020. View Article : Google Scholar

85 

Kelly A, Gunaltay S, McEntee CP, Shuttleworth EE, Smedley C, Houston SA, Fenton TM, Levison S, Mann ER and Travis MA: Human monocytes and macrophages regulate immune tolerance via integrin αvβ8-mediated TGFβ activation. J Exp Med. 215:2725–2736. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Veremeyko T, Yung AWY, Dukhinova M, Kuznetsova IS, Pomytkin I, Lyundup A, Strekalova T, Barteneva NS and Ponomarev ED: Cyclic AMP pathway suppress autoimmune neuroinflammation by inhibiting functions of encephalitogenic CD4 T cells and enhancing M2 macrophage polarization at the site of inflammation. Front Immunol. 9:502018. View Article : Google Scholar : PubMed/NCBI

87 

Su B, Han H, Gong Y, Li X, Ji C, Yao J, Yang J, Hu W, Zhao W, Li J, et al: Let-7d inhibits intratumoral macrophage M2 polarization and subsequent tumor angiogenesis by targeting IL-13 and IL-10. Cancer Immunol Immunother. 70:1619–1634. 2021. View Article : Google Scholar

88 

Ivashkiv LB: IFNγ: Signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat Rev Immunol. 18:545–558. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Teresa Pinto A, Laranjeiro Pinto M, Patrícia Cardoso A, Monteiro C, Teixeira Pinto M, Filipe Maia A, Castro P, Figueira R, Monteiro A, Marques M, et al: Ionizing radiation modulates human macrophages towards a pro-inflammatory phenotype preserving their pro-invasive and pro-angiogenic capacities. Sci Rep. 6:187652016. View Article : Google Scholar : PubMed/NCBI

90 

Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, Pfirschke C, Voss RH, Timke C, Umansky L, et al: Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 24:589–602. 2013. View Article : Google Scholar : PubMed/NCBI

91 

Leblond MM, Pérès EA, Helaine C, Gérault AN, Moulin D, Anfray C, Divoux D, Petit E, Bernaudin M and Valable S: M2 macrophages are more resistant than M1 macrophages following radiation therapy in the context of glioblastoma. Oncotarget. 8:72597–72612. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Proctor DT, Huang J, Lama S, Albakr A, Van Marle G and Sutherland GR: Tumor-associated macrophage infiltration in meningioma. Neurooncol Adv. 1:vdz0182019.

93 

Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, Gupta R, Tsai JM, Sinha R, Corey D, et al: PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 545:495–499. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Zhao Y, Tao F, Jiang J, Chen L, Du J, Cheng X, He Q, Zhong S, Chen W, Wu X, et al: Tryptophan 2, 3-dioxygenase promotes proliferation, migration and invasion of ovarian cancer cells. Mol Med Rep. 23:4452021. View Article : Google Scholar :

95 

Suek N, Campesato LF, Merghoub T and Khalil DN: Targeted APC activation in cancer immunotherapy to enhance the abscopal effect. Front Immunol. 10:6042019. View Article : Google Scholar : PubMed/NCBI

96 

Zhou J, Li X, Wu X, Zhang T, Zhu Q and Wang X, Wang H, Wang K, Lin Y and Wang X: Exosomes Released from Tumor-Associated Macrophages Transfer miRNAs that induce a Treg/Th17 cell imbalance in epithelial ovarian cancer. Cancer Immunol Res. 6:1578–1592. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Jiang M, Liu X, Zhang D, Wang Y, Hu X, Xu F, Jin M, Cao F and Xu L: Celastrol treatment protects against acute ischemic stroke-induced brain injury by promoting an IL-33/ST2 axis-mediated microglia/macrophage M2 polarization. J Neuroinflammation. 15:782018. View Article : Google Scholar : PubMed/NCBI

98 

Wang C, Zhang C, Liu L, A X, Chen B, Li Y and Du J: Macrophage-Derived mir-155-Containing exosomes suppress fibroblast proliferation and promote fibroblast inflammation during cardiac injury. Mol Ther. 25:192–204. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Tan HY, Wang N, Zhang C, Chan YT, Yuen MF and Feng Y: Lysyl Oxidase-Like 4 fosters an immunosuppressive microenvironment during hepatocarcinogenesis. Hepatology. 73:2326–2341. 2021. View Article : Google Scholar

100 

Chen X, Zhang L, Zhang IY, Liang J, Wang H, Ouyang M, Wu S, da Fonseca ACC, Weng L, Yamamoto Y, et al: RAGE expression in tumor-associated macrophages promotes angiogenesis in glioma. Cancer Res. 74:7285–7297. 2014. View Article : Google Scholar : PubMed/NCBI

101 

Barnes TA and Amir E: HYPE or HOPE: The prognostic value of infiltrating immune cells in cancer. Br J Cancer. 118:e52018. View Article : Google Scholar : PubMed/NCBI

102 

Wang X, Shen H, Zhangyuan G, Huang R, Zhang W, He Q, Jin K, Zhuo H, Zhang Z, Wang J, et al: 14-3-3ζ delivered by hepatocellular carcinoma-derived exosomes impaired anti-tumor function of tumor-infiltrating T lymphocytes. Cell Death Dis. 9:1592018. View Article : Google Scholar

103 

Golstein P and Griffiths GM: An early history of T cell-mediated cytotoxicity. Nat Rev Immunol. 18:527–535. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Alonso R, Rodríguez MC, Pindado J, Merino E, Mérida I and Izquierdo M: Diacylglycerol kinase alpha regulates the secretion of lethal exosomes bearing Fas ligand during activation-induced cell death of T lymphocytes. J Biol Chem. 280:28439–28450. 2005. View Article : Google Scholar : PubMed/NCBI

105 

Cai Z, Yang F, Yu L, Yu Z, Jiang L, Wang Q, Yang Y, Wang L, Cao X and Wang J: Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J Immunol. 188:5954–5961. 2012. View Article : Google Scholar : PubMed/NCBI

106 

Lugini L, Cecchetti S, Huber V, Luciani F, Macchia G, Spadaro F, Paris L, Abalsamo L, Colone M, Molinari A, et al: Immune surveillance properties of human NK cell-derived exosomes. J Immunol. 189:2833–2842. 2012. View Article : Google Scholar : PubMed/NCBI

107 

van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde BJ, Knuth A and Boon T: A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. J Immunol. 178:2617–2621. 2007.PubMed/NCBI

108 

Torralba D, Baixauli F, Villarroya-Beltri C, Fernández-Delgado I, Latorre-Pellicer A, Acín-Pérez R, Martín-Cófreces NB, Jaso-Tamame ÁL, Iborra S, Jorge I, et al: Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts. Nat Commun. 9:26582018. View Article : Google Scholar : PubMed/NCBI

109 

Wang X, Shen H, He Q, Tian W, Xia A and Lu XJ: Exosomes derived from exhausted CD8+ T cells impaired the anticancer function of normal CD8+ T cells. J Med Genet. 56:29–31. 2019. View Article : Google Scholar

110 

Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I and Vale RD: T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 355:1428–1433. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Anderson AC, Joller N and Kuchroo VK: Lag-3, Tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity. 44:989–1004. 2016. View Article : Google Scholar : PubMed/NCBI

112 

Dimitrijević M, Arsenović-Ranin N, Kosec D, Bufan B, Nacka-Aleksić M, Pilipović I and Leposavić G: Sexual dimorphism in Th17/Treg axis in lymph nodes draining inflamed joints in rats with collagen-induced arthritis. Brain Behav Immun. 76:198–214. 2019. View Article : Google Scholar

113 

Asano T, Meguri Y, Yoshioka T, Kishi Y, Iwamoto M, Nakamura M, Sando Y, Yagita H, Koreth J, Kim HT, et al: PD-1 modulates regulatory T-cell homeostasis during low-dose interleukin-2 therapy. Blood. 129:2186–2197. 2017. View Article : Google Scholar : PubMed/NCBI

114 

Abbas AK, Trotta E, R Simeonov D, Marson A and Bluestone JA: Revisiting IL-2: Biology and therapeutic prospects. Sci Immunol. 3:eaat14822018. View Article : Google Scholar : PubMed/NCBI

115 

Jukić T, Jurin Martić A, Ivanković S, Antica M, Pavan Jukić D, Rotim C and Jurin M: The role of regulatory T lymphocytes in immune control of MC-2 fibrosarcoma. Acta Clin Croat. 59:351–358. 2020.

116 

Mailloux AW and Young MR: Regulatory T-cell trafficking: From thymic development to tumor-induced immune suppression. Crit Rev Immunol. 30:435–447. 2010. View Article : Google Scholar : PubMed/NCBI

117 

Hammami A, Allard D, Allard B and Stagg J: Targeting the adenosine pathway for cancer immunotherapy. Semin Immunol. 42:1013042019. View Article : Google Scholar : PubMed/NCBI

118 

Smyth LA, Ratnasothy K, Tsang JY, Boardman D, Warley A, Lechler R and Lombardi G: CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur J Immunol. 43:2430–2440. 2013. View Article : Google Scholar : PubMed/NCBI

119 

Aiello S, Rocchetta F, Longaretti L, Faravelli S, Todeschini M, Cassis L, Pezzuto F, Tomasoni S, Azzollini N, Mister M, et al: Extracellular vesicles derived from T regulatory cells suppress T cell proliferation and prolong allograft survival. Sci Rep. 7:115182017. View Article : Google Scholar : PubMed/NCBI

120 

Tung SL, Fanelli G, Matthews RI, Bazoer J, Letizia M, Vizcay-Barrena G, Faruqu FN, Philippeos C, Hannen R, Al-Jamal KT, et al: Regulatory T cell extracellular vesicles modify t-effector cell cytokine production and protect against human skin allograft damage. Front Cell Dev Biol. 8:3172020. View Article : Google Scholar : PubMed/NCBI

121 

Torri A, Carpi D, Bulgheroni E, Crosti MC, Moro M, Gruarin P, Rossi RL, Rossetti G, Di Vizio D, Hoxha M, et al: Extracellular MicroRNA signature of human helper T cell subsets in health and autoimmunity. J Biol Chem. 292:2903–2915. 2017. View Article : Google Scholar : PubMed/NCBI

122 

Tung SL, Boardman DA, Sen M, Letizia M, Peng Q, Cianci N, Dioni L, Carlin LM, Lechler R, Bollati V, et al: Regulatory T cell-derived extracellular vesicles modify dendritic cell function. Sci Rep. 8:60652018. View Article : Google Scholar : PubMed/NCBI

123 

Okoye IS, Coomes SM, Pelly VS, Czieso S, Papayannopoulos V, Tolmachova T, Seabra MC and Wilson MS: MicroRNA-containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity. 41:89–103. 2014. View Article : Google Scholar : PubMed/NCBI

124 

Sullivan JA, Tomita Y, Jankowska-Gan E, Lema DA, Arvedson MP, Nair A, Bracamonte-Baran W, Zhou Y, Meyer KK, Zhong W, et al: Treg-Cell-Derived IL-35-Coated extracellular vesicles promote infectious tolerance. Cell Rep. 30:1039–1051.e5. 2020. View Article : Google Scholar : PubMed/NCBI

125 

Eckert F, Schilbach K, Klumpp L, Bardoscia L, Sezgin EC, Schwab M, Zips D and Huber SM: Potential Role of CXCR4 targeting in the context of radiotherapy and immunotherapy of cancer. Front Immunol. 9:30182018. View Article : Google Scholar

126 

Zhou M, Luo C, Zhou Z, Li L and Huang Y: Improving anti-PD-L1 therapy in triple negative breast cancer by polymer-enhanced immunogenic cell death and CXCR4 blockade. J Control Release. 334:248–262. 2021. View Article : Google Scholar : PubMed/NCBI

127 

Kohno M, Murakami J, Wu L, Chan ML, Yun Z, Cho BCJ and de Perrot M: Foxp3(+) Regulatory T cell depletion after nonablative oligofractionated irradiation boosts the abscopal effects in murine malignant mesothelioma. J Immunol. 205:2519–2531. 2020. View Article : Google Scholar : PubMed/NCBI

128 

Anderson BE, McNiff JM, Matte C, Athanasiadis I, Shlomchik WD and Shlomchik MJ: Recipient CD4+ T cells that survive irradiation regulate chronic graft-versus-host disease. Blood. 104:1565–1573. 2004. View Article : Google Scholar : PubMed/NCBI

129 

Zhang T, Yu H, Ni C, Zhang T, Liu L, Lv Q, Zhang Z, Wang Z, Wu D, Wu P, et al: Hypofractionated stereotactic radiation therapy activates the peripheral immune response in operable stage I non-small-cell lung cancer. Sci Rep. 7:48662017. View Article : Google Scholar : PubMed/NCBI

130 

Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM and Weaver CT: Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 6:1123–1132. 2005. View Article : Google Scholar : PubMed/NCBI

131 

Ye ZJ, Zhou Q, Gu YY, Qin SM, Ma WL, Xin JB, Tao XN and Shi HZ: Generation and differentiation of IL-17-producing CD4+ T cells in malignant pleural effusion. J Immunol. 185:6348–6354. 2010. View Article : Google Scholar : PubMed/NCBI

132 

Zou W and Restifo NP: T(H)17 cells in tumour immunity and immunotherapy. Nat Rev Immunol. 10:248–256. 2010. View Article : Google Scholar : PubMed/NCBI

133 

Berkhout L, Barikbin R, Schiller B, Ravichandran G, Krech T, Neumann K, Sass G and Tiegs G: Deletion of tumour necrosis factor α receptor 1 elicits an increased TH17 immune response in the chronically inflamed liver. Sci Rep. 9:42322019. View Article : Google Scholar

134 

Campanati A, Orciani M, Lazzarini R, Ganzetti G, Consales V, Sorgentoni G, Di Primio R and Offidani A: TNF-α inhibitors reduce the pathological Th1-Th17/Th2 imbalance in cutaneous mesenchymal stem cells of psoriasis patients. Exp Dermatol. 26:319–324. 2017. View Article : Google Scholar

135 

Nalbant A: IL-17, IL-21, and IL-22 Cytokines of T Helper 17 cells in cancer. J Interferon Cytokine Res. 39:56–60. 2019. View Article : Google Scholar

136 

Chang SH: T helper 17 (Th17) cells and interleukin-17 (IL-17) in cancer. Arch Pharm Res. 42:549–559. 2019. View Article : Google Scholar : PubMed/NCBI

137 

Papadopoulou E, Nicolatou-Galitis O, Papassotiriou I, Linardou H, Karagianni A, Tsixlakis K, Tarampikou A, Michalakakou K, Vardas E and Bafaloukos D: The use of crevicular fluid to assess markers of inflammation and angiogenesis, IL-17 and VEGF, in patients with solid tumors receiving zoledronic acid and/or bevacizumab. Support Care Cancer. 28:177–184. 2020. View Article : Google Scholar

138 

Messmer MN, Netherby CS, Banik D and Abrams SI: Tumor-induced myeloid dysfunction and its implications for cancer immunotherapy. Cancer Immunol Immunother. 64:1–13. 2015. View Article : Google Scholar :

139 

Waigel S, Rendon BE, Lamont G, Richie J, Mitchell RA and Yaddanapudi K: MIF inhibition reverts the gene expression profile of human melanoma cell line-induced MDSCs to normal monocytes. Genom Data. 7:240–242. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Fenselau C and Ostrand-Rosenberg S: Molecular cargo in myeloid-derived suppressor cells and their exosomes. Cell Immunol. 359:1042582021. View Article : Google Scholar :

141 

Marvel D and Gabrilovich DI: Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J Clin Invest. 125:3356–3364. 2015. View Article : Google Scholar : PubMed/NCBI

142 

Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D and Carbone DP: Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 2:1096–1103. 1996. View Article : Google Scholar : PubMed/NCBI

143 

Horikawa N, Abiko K, Matsumura N, Hamanishi J, Baba T, Yamaguchi K, Yoshioka Y, Koshiyama M and Konishi I: Expression of vascular endothelial growth factor in ovarian cancer inhibits tumor immunity through the accumulation of myeloid-derived suppressor cells. Clin Cancer Res. 23:587–599. 2017. View Article : Google Scholar

144 

Hsieh CC, Hung CH, Chiang M, Tsai YC and He JT: Hepatic stellate cells enhance liver cancer progression by inducing myeloid-derived suppressor cells through interleukin-6 signaling. Int J Mol Sci. 20:50792019. View Article : Google Scholar : PubMed/NCBI

145 

Li L, Zhang J, Diao W, Wang D, Wei Y, Zhang CY and Zen K: MicroRNA-155 and MicroRNA-21 promote the expansion of functional myeloid-derived suppressor cells. J Immunol. 192:1034–1043. 2014. View Article : Google Scholar : PubMed/NCBI

146 

Meng G, Wei J, Wang Y, Qu D and Zhang J: miR-21 regulates immunosuppression mediated by myeloid-derived suppressor cells by impairing RUNX1-YAP interaction in lung cancer. Cancer Cell Int. 20:4952020. View Article : Google Scholar : PubMed/NCBI

147 

Safarzadeh E, Asadzadeh Z, Safaei S, Hatefi A, Derakhshani A, Giovannelli F, Brunetti O, Silvestris N and Baradaran B: MicroRNAs and lncRNAs-A new layer of myeloid-derived suppressor cells regulation. Front Immunol. 11:5723232020. View Article : Google Scholar : PubMed/NCBI

148 

Geis-Asteggiante L, Belew AT, Clements VK, Edwards NJ, Ostrand-Rosenberg S, El-Sayed NM and Fenselau C: Differential Content of Proteins, mRNAs, and miRNAs Suggests that MDSC and their exosomes may mediate distinct immune suppressive functions. J Proteome Res. 17:486–498. 2018. View Article : Google Scholar :

149 

Li T, Li M, Xu C, Xu X, Ding J, Cheng L and Ou R: miR-146a regulates the function of Th17 cell differentiation to modulate cervical cancer cell growth and apoptosis through NF-κB signaling by targeting TRAF6. Oncol Rep. 41:2897–2908. 2019.PubMed/NCBI

150 

Tian S, Song X, Wang Y, Wang X, Mou Y, Chen Q, Zhao H, Ma K, Wu Z, Yu H, et al: Chinese herbal medicine Baoyuan Jiedu decoction inhibits the accumulation of myeloid derived suppressor cells in pre-metastatic niche of lung via TGF-β/CCL9 pathway. Biomed Pharmacother. 129:1103802020. View Article : Google Scholar

151 

Mao Y, Sarhan D, Steven A, Seliger B, Kiessling R and Lundqvist A: Inhibition of tumor-derived prostaglandin-e2 blocks the induction of myeloid-derived suppressor cells and recovers natural killer cell activity. Clin Cancer Res. 20:4096–4106. 2014. View Article : Google Scholar : PubMed/NCBI

152 

Deng Z, Rong Y, Teng Y, Zhuang X, Samykutty A, Mu J, Zhang L, Cao P, Yan J, Miller D and Zhang HG: Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis. Oncogene. 36:639–651. 2017. View Article : Google Scholar :

153 

Zöller M, Zhao K, Kutlu N, Bauer N, Provaznik J, Hackert T and Schnölzer M: Immunoregulatory effects of myeloid-derived suppressor cell exosomes in mouse model of autoimmune alopecia areata. Front Immunol. 9:12792018. View Article : Google Scholar : PubMed/NCBI

154 

Grisaru-Tal S, Itan M, Klion AD and Munitz A: A new dawn for eosinophils in the tumour microenvironment. Nat Rev Cancer. 20:594–607. 2020. View Article : Google Scholar : PubMed/NCBI

155 

Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, Montabana E, Lang UE, Fu Q, Fong L and Blelloch R: Suppression of Exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 177:414–427.e13. 2019. View Article : Google Scholar : PubMed/NCBI

156 

Mesnil C, Raulier S, Paulissen G, Xiao X, Birrell MA, Pirottin D, Janss T, Starkl P, Ramery E, Henket M, et al: Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest. 126:3279–3295. 2016. View Article : Google Scholar : PubMed/NCBI

157 

Choi Y, Kim YM, Lee HR, Mun J, Sim S, Lee DH, Pham DL, Kim SH, Shin YS, Lee SW and Park HS: Eosinophil extracellular traps activate type 2 innate lymphoid cells through stimulating airway epithelium in severe asthma. Allergy. 75:95–103. 2020. View Article : Google Scholar

158 

da Silva JM, Moreira Dos Santos TP, Sobral LM, Queiroz-Junior CM, Rachid MA, Proudfoot AEI, Garlet GP, Batista AC, Teixeira MM, Leopoldino AM, et al: Relevance of CCL3/CCR5 axis in oral carcinogenesis. Oncotarget. 8:51024–51036. 2017. View Article : Google Scholar : PubMed/NCBI

159 

Hu Y, Chen Z, Jin L, Wang M and Liao W: Decreased expression of indolamine 2,3-dioxygenase in childhood allergic asthma and its inverse correlation with fractional concentration of exhaled nitric oxide. Ann Allergy Asthma Immunol. 119:429–434. 2017. View Article : Google Scholar : PubMed/NCBI

160 

Kratochvill F, Neale G, Haverkamp JM, Van de Velde LA, Smith AM, Kawauchi D, McEvoy J, Roussel MF, Dyer MA, Qualls JE and Murray PJ: TNF counterbalances the emergence of M2 tumor macrophages. Cell Rep. 12:1902–1914. 2015. View Article : Google Scholar : PubMed/NCBI

161 

Herrera FG, Bourhis J and Coukos G: Radiotherapy combination opportunities leveraging immunity for the next oncology practice. CA Cancer J Clin. 67:65–85. 2017. View Article : Google Scholar

162 

Cao M, Cabrera R, Xu Y, Liu C and Nelson D: Different radio-sensitivity of CD4(+)CD25(+) regulatory T cells and effector T cells to low dose gamma irradiation in vitro. Int J Radiat Biol. 87:71–80. 2011. View Article : Google Scholar

163 

Kareff SA, Lischalk JW, Krochmal R and Kim C: Abscopal effect in pulmonary carcinoid tumor following ablative stereotactic body radiation therapy: A case report. J Med Case Rep. 14:1772020. View Article : Google Scholar : PubMed/NCBI

164 

Ohmatsu K, Hashimoto Y, Kawanishi M, Ishii Y, Kono S, Kuribayashi S, Ariizumi S and Karasawa K: Abscopal complete regression of hepatocellular carcinoma with multiple pleural metastases. Int Cancer Conf J. 10:54–58. 2020. View Article : Google Scholar

165 

Hotta T, Okuno T, Nakao M, Amano Y, Isobe T and Tsubata Y: Reproducible abscopal effect in a patient with lung cancer who underwent whole-brain irradiation and atezolizumab administration. Thorac Cancer. 12:985–988. 2021. View Article : Google Scholar : PubMed/NCBI

166 

Choi JS, Sansoni ER, Lovin BD, Lindquist NR, Phan J, Mayo LL, Ferrarotto R and Su SY: Abscopal effect following immunotherapy and combined stereotactic body radiation therapy in recurrent metastatic head and neck squamous cell carcinoma: A report of two cases and literature review. Ann Otol Rhinol Laryngol. 129:517–522. 2020. View Article : Google Scholar

167 

Wang H, Lin X, Luo Y, Sun S, Tian X, Sun Y, Zhang S, Chen J, Zhang J, Liu X, et al: α-PD-L1 mAb enhances the abscopal effect of hypo-fractionated radiation by attenuating PD-L1 expression and inducing CD8(+) T-cell infiltration. Immunotherapy. 11:101–118. 2019. View Article : Google Scholar

168 

Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, Friedman K, Ponzo F, Babb JS, Goldberg J, et al: Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: A proof-of-principle trial. Lancet Oncol. 16:795–803. 2015. View Article : Google Scholar : PubMed/NCBI

169 

Thorne AH, Malo KN, Wong AJ, Nguyen TT, Cooch N, Reed C, Yan J, Broderick KE, Smith TRF, Masteller EL and Humeau L: Adjuvant screen identifies synthetic DNA-Encoding Flt3L and CD80 immunotherapeutics as candidates for enhancing anti-tumor T cell responses. Front Immunol. 11:3272020. View Article : Google Scholar : PubMed/NCBI

170 

Wennerberg E, Spada S, Rudqvist NP, Lhuillier C, Gruber S, Chen Q, Zhang F, Zhou XK, Gross SS, Formenti SC and Demaria S: CD73 blockade promotes dendritic cell infiltration of irradiated tumors and tumor rejection. Cancer Immunol Res. 8:465–478. 2020. View Article : Google Scholar : PubMed/NCBI

171 

Peluso MO, Adam A, Armet CM, Zhang L, O'Connor RW, Lee BH, Lake AC, Normant E, Chappel SC, Hill JA, et al: The Fully human anti-CD47 antibody SRF231 exerts dual-mechanism antitumor activity via engagement of the activating receptor CD32a. J Immunother Cancer. 8:e0004132020. View Article : Google Scholar : PubMed/NCBI

172 

Tsukui H, Horie H, Koinuma K, Ohzawa H, Sakuma Y, Hosoya Y, Yamaguchi H, Yoshimura K, Lefor AK, Sata N and Kitayama J: CD73 blockade enhances the local and abscopal effects of radiotherapy in a murine rectal cancer model. BMC Cancer. 20:4112020. View Article : Google Scholar : PubMed/NCBI

173 

Puro RJ, Bouchlaka MN, Hiebsch RR, Capoccia BJ, Donio MJ, Manning PT, Frazier WA, Karr RW and Pereira DS: Development of AO-176, a Next-Generation Humanized Anti-CD47 antibody with novel anticancer properties and negligible red blood cell binding. Mol Cancer Ther. 19:835–846. 2020. View Article : Google Scholar

174 

Chen D, Barsoumian HB, Yang L, Younes AI, Verma V, Hu Y, Menon H, Wasley M, Masropour F, Mosaffa S, et al: SHP-2 and PD-L1 inhibition combined with radiotherapy enhances systemic antitumor effects in an Anti-PD-1-Resistant model of non-small cell lung cancer. Cancer Immunol Res. 8:883–894. 2020. View Article : Google Scholar : PubMed/NCBI

175 

Muenkel J, Xu Z, Traughber BJ, Baig T, Xu K, Langmack C, Harris E and Podder TK: Feasibility of improving patient's safety with in vivo dose tracking in intracavitary and interstitial HDR brachytherapy. Brachytherapy. 20:353–360. 2021. View Article : Google Scholar

176 

Liu Y, Dong Y, Kong L, Shi F, Zhu H and Yu J: Abscopal effect of radiotherapy combined with immune checkpoint inhibitors. J Hematol Oncol. 11:1042018. View Article : Google Scholar : PubMed/NCBI

177 

Ahmed TA, Adamopoulos C, Karoulia Z, Wu X, Sachidanandam R, Aaronson SA and Poulikakos PI: SHP2 drives adaptive resistance to ERK signaling inhibition in molecularly defined subsets of ERK-Dependent tumors. Cell Rep. 26:65–78.e5. 2019. View Article : Google Scholar : PubMed/NCBI

178 

Strazza M, Adam K, Lerrer S, Straube J, Sandigursky S, Ueberheide B and Mor A: SHP2 Targets ITK Downstream of PD-1 to Inhibit T cell function. Inflammation. 44:1529–1539. 2021. View Article : Google Scholar : PubMed/NCBI

179 

Pustylnikov S, Costabile F, Beghi S and Facciabene A: Targeting mitochondria in cancer: Current concepts and immunotherapy approaches. Transl Res. 202:35–51. 2018. View Article : Google Scholar : PubMed/NCBI

180 

Chen D, Barsoumian HB, Fischer G, Yang L, Verma V, Younes AI, Hu Y, Masropour F, Klein K, Vellano C, et al: Combination treatment with radiotherapy and a novel oxidative phosphorylation inhibitor overcomes PD-1 resistance and enhances antitumor immunity. J Immunother Cancer. 8:e0002892020. View Article : Google Scholar : PubMed/NCBI

181 

Vanpouille-Box C, Diamond JM, Pilones KA, Zavadil J, Babb JS, Formenti SC, Barcellos-Hoff MH and Demaria S: TGFβ Is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res. 75:2232–2242. 2015. View Article : Google Scholar : PubMed/NCBI

182 

Liang Y, He J and Zhao Y: Modification of oncolytic adenovirus and its application in cancer therapy. Discov Med. 30:129–144. 2020.

183 

Havunen R, Santos JM, Sorsa S, Rantapero T, Lumen D, Siurala M, Airaksinen AJ, Cervera-Carrascon V, Tähtinen S, Kanerva A and Hemminki A: Abscopal effect in Non-injected tumors achieved with cytokine-armed oncolytic adenovirus. Mol Ther Oncolytics. 11:109–121. 2018. View Article : Google Scholar : PubMed/NCBI

184 

Kaufman HL, Amatruda T, Reid T, Gonzalez R, Glaspy J, Whitman E, Harrington K, Nemunaitis J, Zloza A, Wolf M and Senzer NN: Systemic versus local responses in melanoma patients treated with talimogene laherparepvec from a multi-institutional phase II study. J Immunother Cancer. 4:122016. View Article : Google Scholar : PubMed/NCBI

185 

Ono R, Takayama K, Sakurai F and Mizuguchi H: Efficient antitumor effects of a novel oncolytic adenovirus fully composed of species B adenovirus serotype 35. Mol Ther Oncolytics. 20:399–409. 2021. View Article : Google Scholar : PubMed/NCBI

186 

Challenor S and Tucker D: SARS-CoV-2-induced remission of Hodgkin lymphoma. Br J Haematol. 192:4152021. View Article : Google Scholar : PubMed/NCBI

187 

Ngwa W, Boateng F, Kumar R, Irvine DJ, Formenti S, Ngoma T, Herskind C, Veldwijk MR, Hildenbrand GL, Hausmann M, et al: Smart Radiation Therapy Biomaterials. Int J Radiat Oncol Biol Phys. 97:624–637. 2017. View Article : Google Scholar : PubMed/NCBI

188 

Zhao X, Yang K, Zhao R, Ji T, Wang X, Yang X, Zhang Y, Cheng K, Liu S, Hao J, et al: Inducing enhanced immunogenic cell death with nanocarrier-based drug delivery systems for pancreatic cancer therapy. Biomaterials. 102:187–197. 2016. View Article : Google Scholar : PubMed/NCBI

189 

Duan X, Chan C, Guo N, Han W, Weichselbaum RR and Lin W: Photodynamic therapy mediated by nontoxic core-shell nanoparticles synergizes with immune checkpoint blockade to elicit antitumor immunity and antimetastatic effect on breast cancer. J Am Chem Soc. 138:16686–16695. 2016. View Article : Google Scholar : PubMed/NCBI

190 

Chen Q, Wang C, Zhang X, Chen G, Hu Q, Li H, Wang J, Wen D, Zhang Y, Lu Y, et al: In situ sprayed bioresponsive immunotherapeutic gel for post-surgical cancer treatment. Nat Nanotechnol. 14:89–97. 2019. View Article : Google Scholar

191 

Min Y, Roche KC, Tian S, Eblan MJ, McKinnon KP, Caster JM, Chai S, Herring LE, Zhang L, Zhang T, et al: Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy. Nat Nanotechnol. 12:877–882. 2017. View Article : Google Scholar : PubMed/NCBI

192 

Zheng Y, Tang L, Mabardi L, Kumari S and Irvine DJ: Enhancing adoptive cell therapy of cancer through targeted delivery of small-molecule immunomodulators to internalizing or noninternalizing receptors. ACS Nano. 11:3089–3100. 2017. View Article : Google Scholar : PubMed/NCBI

193 

Chao Y, Xu L, Liang C, Feng L, Xu J, Dong Z, Tian L, Yi X, Yang K and Liu Z: Combined local immunostimulatory radioisotope therapy and systemic immune checkpoint blockade imparts potent antitumour responses. Nat Biomed Eng. 2:611–621. 2018. View Article : Google Scholar

194 

Xu H, Sun W, Kong Y, Huang Y, Wei Z, Zhang L, Liang J and Ye X: Immune abscopal effect of microwave ablation for lung metastases of endometrial carcinoma. J Cancer Res Ther. 16:1718–1721. 2020.

Related Articles

Journal Cover

January-2023
Volume 62 Issue 1

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
Kuang G, Wang Z, Luo C, Luo J and Wang J: Mechanism of exosomes in the tumor microenvironment in the abscopal effect (Review). Int J Oncol 62: 2, 2023
APA
Kuang, G., Wang, Z., Luo, C., Luo, J., & Wang, J. (2023). Mechanism of exosomes in the tumor microenvironment in the abscopal effect (Review). International Journal of Oncology, 62, 2. https://doi.org/10.3892/ijo.2022.5450
MLA
Kuang, G., Wang, Z., Luo, C., Luo, J., Wang, J."Mechanism of exosomes in the tumor microenvironment in the abscopal effect (Review)". International Journal of Oncology 62.1 (2023): 2.
Chicago
Kuang, G., Wang, Z., Luo, C., Luo, J., Wang, J."Mechanism of exosomes in the tumor microenvironment in the abscopal effect (Review)". International Journal of Oncology 62, no. 1 (2023): 2. https://doi.org/10.3892/ijo.2022.5450