Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of pathologically activated, mostly immature, myeloid cells that exert robust immunosuppressive functions. MDSCs expand during oncogenesis and have been linked to accelerated disease progression and resistance to treatment in both preclinical tumor models and patients with cancer. Thus, MDSCs stand out as promising targets for the development of novel immunotherapeutic regimens with superior efficacy. Here, we summarize accumulating preclinical and clinical evidence indicating that MDSCs also hamper the efficacy of radiotherapy (RT), as we critically discuss the potential of MDSC-targeting strategies as tools to achieve superior immunotherapeutic tumor control by RT in the clinic.

Myeloid-derived suppressor cells (MDSC) are a heterogeneous and highly plastic population of mostly immature myeloid cells (IMC) that expand systemically in the context of multiple pathologic conditions including cancer (1). In physiologic conditions, indeed, the IMCs that are released by the bone marrow (BM) as part of normal hematopoiesis migrate to peripheral organs and rapidly differentiate into granulocytes, macrophages or dendritic cells (DC; ref. 2). Conversely, in the context of indolent, chronic inflammation (as it occurs during cancer progression; ref. 3), BM-derived IMCs fail to differentiate and expand, both systemically and (at least in some malignancies) within the tumor microenvironment (TME; ref. 4). Moreover, tumor progression is frequently accompanied by at least some degree of extramedullary hematopoiesis, which also contributes to the expansion of the circulating pool of IMCs (5). Importantly, these IMCs represent a group of pathologically activated myeloid cells that mediate a plethora of immunosuppressive effects (4), ultimately underlying the widespread adoption of the term MDSCs (6).

Despite being developed as a cytostatic and cytotoxic agent, focal radiotherapy (RT)—at least when delivered according to specific fractionation regimens—mediates numerous immunostimulatory effects that actively contribute to tumor control (7). Consistent with this notion, accumulating evidence suggests that MDSCs actively counteract the therapeutic efficacy of RT, making them a promising target to develop novel combinatorial regimens that provide superior tumor control.

Here, we critically discuss preclinical and clinical data linking MDSC expansion to suppressed RT-driven immunostimulation, and we identify strategies that may potentially be harnessed to target MDSCs for enhancing tumor control by RT.

MDSCs were first reported and characterized for their immunosuppressive effects more than 40 years ago (8). It was only in the mid-2000s, however, that their morphologic, phenotypic, and functional heterogeneity began to emerge (9). Initially, two broad subpopulations of mouse MDSCs were identified: CD11b+Ly6C+Ly6G cells originating from mononuclear cells, which are now dubbed monocytic (M-) MDSCs and CD11b+Ly6ClowLy6G+ cells originating from low-density granulocytes, which are now referred to as granulocytic (G-) or polymorphonuclear (PMN-) MDSCs (9). In humans, M-MDSCs and G-MDSCs are commonly identified as CD33highCD11b+HLA-DR+CD14+CD15 and CD33midCD11b+HLA-DRCD14CD15+ cells, respectively (10). Alternatively, human MDSCs can be classified as G- based on a CD11b+CD14CD15+ or CD11b+CD14CD66b+ phenotype, or as M- based on a CD11b+CD14+HLA-DR−/loCD15 phenotype (11). Moreover, an early-stage (E-) population of MDSCs comprising even more immature progenitors of both the M- and G- type can be identified as CD33+HLA-DR and Lin (meaning these cells do not express any mature lineage marker, including CD3, CD14, CD15, CD19, and CD56; ref. 11).

More recently, fibrocytic (F-) MDSCs have been reported as a population of MDSCs emerging from umbilical cord blood precursors and expressing not only common MDSCs markers such as CD33, CD11b, CD14, and CD15, but also CD163, S100A8, IL1B, FN1, TLR4, and ICAM1 (12, 13). Conversely, F-MDSCs do not express pure fibrocyte markers including ACTA2, CCR7, and COL6A1 (12, 13). So far, F-MDSCs have mostly been found to mediate beneficial effects in nonmalignant disorders including type I diabetes (12), wound healing (14), and chronic obstructive pulmonary disorders (15), with the notable exception of an F-MDSC population with indoleamine 2,3-dioxygenase 1 (IDO1)-dependent immunosuppressive activity that has been identified in subjects with metastatic pediatric sarcomas (16). Finally, so-called eosinophilic (Eo-) MDSCs have been reported to emerge in the context of Staphylococcus aureus infection in mice as an immature cell population expressing MDSC markers as well as eosinophil markers such as SIGLECF and low IL5RA levels (17). Whether a human MDSC population resembling mouse Eo-MDSCs exists and what pathophysiologic functions it potentially plays remain to be clarified.

Both M- and G-MDSCs exert immunosuppressive functions largely (but not exclusively) by altering the microenvironment of immune effector cells (Fig. 1; ref. 1). Specifically, MDSCs can secrete multiple cytokines with immunosuppressive activity such as interleukin 4 (IL4), IL10, transforming growth factor beta 1 (TGFB1), colony stimulating factor 2 (CSF2, best known as GM-CSF), vascular endothelial growth factor A (VEGFA), as well as other bioactive factors that potently inhibit anticancer immunity, like prostaglandin E2 (PGE2) and L-kynurenine (1). Moreover, MDSCs actively compete with immune effector cells, most notably CD8+ cytotoxic T lymphocytes (CTL), for critical nutrients that are generally limited in the TME. These nutrients include not only glucose and fatty acids, but also amino acids like tryptophan, arginine, cysteine and cystine, a least in part as a consequence of IDO1 and arginase 1 (ARG1) overexpression (18). Finally, MDSCs (especially G-MDSCs) produce abundant amounts of reactive oxygen species (ROS) and reactive nitrogen species including nitric oxide (NO), which also contribute to establish an immunosuppressive microenvironment (19). That said, it is important to note that the specific immunosuppressive mechanisms used by MDSCs in different settings generally reflect their functional plasticity in response to microenvironmental cues (1).

Figure 1.

Main mechanisms of MDSC-dependent immunosuppression. Myeloid-derived suppressor cells (MDSC) exert multipronged immunosuppressive effects, mostly, but not exclusively, as they condition their microenvironment from both the metabolic and immunologic standpoints. In general, such alterations suppress the activity of immune effector cells including effector T (TEFF) cells, B lymphocytes, natural killer (NK) cells and dendritic cells (DC), as they expand or improve the functions of immunosuppressive cells including regulatory T (TREG) cells, regulatory B (BREG) cells, and M2-like tumor-associated macrophages (TAMs). Abbreviations: ARG1, arginase 1; H2O2, hydrogen peroxide; IDO1, indoleamine 2,3-dioxygenase 1; IL, interleukin; NO, nitric oxide; NOS2, nitric oxide synthase 2; NOX, NADPH oxidase; nTCR, nitrosylated TCR; ONOO, peroxynitrite anion; PD-1 (official name: PDCD1), programmed cell death 1; PD-L1 (official name: CD274); PGE2, prostaglandin E2; ROS, reactive oxygen species; STAT3, signal transducer and activator of transcription 3; TGFB1, transforming growth factor beta 1; VEGFA, vascular endothelial growth factor A.

Figure 1.

Main mechanisms of MDSC-dependent immunosuppression. Myeloid-derived suppressor cells (MDSC) exert multipronged immunosuppressive effects, mostly, but not exclusively, as they condition their microenvironment from both the metabolic and immunologic standpoints. In general, such alterations suppress the activity of immune effector cells including effector T (TEFF) cells, B lymphocytes, natural killer (NK) cells and dendritic cells (DC), as they expand or improve the functions of immunosuppressive cells including regulatory T (TREG) cells, regulatory B (BREG) cells, and M2-like tumor-associated macrophages (TAMs). Abbreviations: ARG1, arginase 1; H2O2, hydrogen peroxide; IDO1, indoleamine 2,3-dioxygenase 1; IL, interleukin; NO, nitric oxide; NOS2, nitric oxide synthase 2; NOX, NADPH oxidase; nTCR, nitrosylated TCR; ONOO, peroxynitrite anion; PD-1 (official name: PDCD1), programmed cell death 1; PD-L1 (official name: CD274); PGE2, prostaglandin E2; ROS, reactive oxygen species; STAT3, signal transducer and activator of transcription 3; TGFB1, transforming growth factor beta 1; VEGFA, vascular endothelial growth factor A.

Close modal

Such a diversified armamentarium of immunosuppressive tools confers MDSCs with the ability to:

  • (i) block the recruitment, proliferation, differentiation, and/or activation of CTLs as a consequence of nitrosylation-dependent chemokine inactivation (20), limited tryptophan, arginine, cysteine, and cystine availability (18, 21, 22), CD3, CD8, and TCR nitration or nitrosylation (23, 24), as well as NO-dependent DNA damage (25);

  • (ii) inhibit the maturation and activation of natural killer (NK) cells by triggering TGFB1 signaling (26, 27);

  • (iii) suppress DC-dependent CTL cross-priming via cystine depletion plus active IL10- and VEGFA-dependent mechanisms that result in quenched IL12 and IL23 secretion by DCs (18, 28–30);

  • (iv) inhibit antitumor B-cell responses by an ARG1- and TGFB1-dependent mechanism (31, 32);

  • (v) promote the expansion of CD4+CD25+FOXP3+ regulatory T (TREG) cells by reducing arginine availability coupled to IL10, VEGFA, and TGFB1 secretion (28, 33–36);

  • (vi) stimulate the immunosuppressive function of regulatory B (BREG) cells via exosomal PGE2 (37); and

  • (vii) engage in metabolic and IL10-dependent cross talks with tumor-associated macrophages (TAMs) to receive trophic signals and promote the expansion of immunosuppressive M2-like TAMs (38, 39).

Moreover, at least some MDSCs express coinhibitory ligands such as CD274 (best known as PD-L1) on their surface, hence inhibiting effector T (TEFF) cells and NK cells upon binding to the coinhibitory receptor programmed cell death 1 (PDCD1, best known as PD-1; refs. 40, 41), or upon transferring PD-L1 to other immunosuppressive cells such as BREG cells via exosomes (42).

Of note, some MDSC-derived mediators that actively promote immunosuppression such as PGE2, GM-CSF, and VEGFA can also be produced by (at least some) malignant cells, ultimately acting in an autocrine/paracrine manner to support the recruitment, expansion, and/or immunosuppressive activity of MDSCs themselves (43–46). Neoplastic cells (and notably the cancer stem cell compartment) can secrete various other factors that recruit MDSCs to the TME and support their immunosuppressive activity, including small metabolites like lactate (47). In turn, MDSCs can release stemness-preserving signals, ultimately delineating a vicious cycle whereby cancer stem cells and MDSCs support each other in the context of accrued tumor progression (48).

Importantly, many of these processes are sensitive to RT and have been mechanistically linked to RT-driven MDSC recruitment or expansion (see below). Moreover, RT is a potent inducer of hypoxia and consequent hypoxia-inducible factor 1 subunit alpha (HIF1α) activation in cancer cells (49). Although the precise impact of RT-driven hypoxia on MDSCs has not been directly investigated, HIF1α signaling in malignant cells has been consistently associated with MDSC accumulation in the TME and enhanced immunosuppression (40, 50). Along similar lines, ROS are important regulators of MDSC functions (51), but how ROS generation downstream of RT influences MDSC-dependent immunosuppression remains to be specifically assessed.

Focal RT has been used for the clinical management of various malignancies for over a century, reflecting considerable efficacy in multiple oncological indications as well as an extraordinary safety profile (52). The therapeutic activity of RT has classically been attributed to its ability to inflict direct and ROS-mediated damage to macromolecules, notably DNA and lipids (53). Besides the fact that modern technological platforms enable the delivery of high doses of RT to specific target volumes with extraordinary precision (hence maximally sparing healthy tissues), normal cells are more proficient than their malignant counterparts at repairing such macromolecular damage (54), offering a relatively large therapeutic window for clinical applications. That said, accumulating preclinical and clinical data challenge the traditional notion whereby RT purely operates as a cytostatic/cytotoxic agent as they highlight various RT-driven immunomodulatory pathways that ultimately influence therapeutic outcome (Fig. 2; refs. 7, 55).

Figure 2.

Immunomodulatory properties of RT. Depending on multiple parameters, including (but not limited to) dose and fractionation schedule, radiotherapy (RT) can mediate robust immunostimulatory effects as well as a pronounced immunosuppressive activity. On the one hand, upon interacting with cancer cells, RT can promote the expression of otherwise silenced potentially immunogenic antigens (Ag), the exposure of MHC class I molecules (MHC-I) and death receptors on the cell surface, as well as the release of immunostimulatory cytokines and damage-associated molecular patterns (DAMP), ultimately boosting the antigenicity, adjuvanticity, and immune susceptibility of malignant cells. On the other hand, RT can stimulate the expression or secretion of immunosuppressive factors by malignant cells, favor the recruitment of immunosuppressive cell populations to the tumor microenvironment (TME), and mediate cytotoxic effects on immune effector cells, ultimately establishing permissive conditions for immunoevasion. CALR, calreticulin; FAS, Fas cell-surface death receptor; IFN, interferon; IL, interleukin; INHBA, inhibin subunit beta A; MDSC, myeloid-derived suppressor cell; NK, natural killer; PD-L1 (official name: CD274); TAM, tumor-associated macrophage; TEFF, effector T; TGFB1, transforming growth factor beta 1; TRAIL-R2 (official name: TNFRSF10B), TNF receptor superfamily member 10b; TREG, regulatory T; VEGFA, vascular endothelial growth factor A.

Figure 2.

Immunomodulatory properties of RT. Depending on multiple parameters, including (but not limited to) dose and fractionation schedule, radiotherapy (RT) can mediate robust immunostimulatory effects as well as a pronounced immunosuppressive activity. On the one hand, upon interacting with cancer cells, RT can promote the expression of otherwise silenced potentially immunogenic antigens (Ag), the exposure of MHC class I molecules (MHC-I) and death receptors on the cell surface, as well as the release of immunostimulatory cytokines and damage-associated molecular patterns (DAMP), ultimately boosting the antigenicity, adjuvanticity, and immune susceptibility of malignant cells. On the other hand, RT can stimulate the expression or secretion of immunosuppressive factors by malignant cells, favor the recruitment of immunosuppressive cell populations to the tumor microenvironment (TME), and mediate cytotoxic effects on immune effector cells, ultimately establishing permissive conditions for immunoevasion. CALR, calreticulin; FAS, Fas cell-surface death receptor; IFN, interferon; IL, interleukin; INHBA, inhibin subunit beta A; MDSC, myeloid-derived suppressor cell; NK, natural killer; PD-L1 (official name: CD274); TAM, tumor-associated macrophage; TEFF, effector T; TGFB1, transforming growth factor beta 1; TRAIL-R2 (official name: TNFRSF10B), TNF receptor superfamily member 10b; TREG, regulatory T; VEGFA, vascular endothelial growth factor A.

Close modal

On the one hand, RT, at least when delivered according to specific fractionation schedules (56, 57), can elicit robust immunostimulatory effects. For instance, RT can increase the antigenicity of malignant cells (and hence their visibility to the host immune system) by favoring the exposure of MHC class I molecules on the cell surface (58) as well as the transcriptional derepression of otherwise silenced genes encoding for mutated MHC class I and II epitopes (59). Moreover, hypofractionated RT is particularly effective at increasing the adjuvanticity of cancer cells, i.e., at promoting the emission of immunostimulatory signals that support the recruitment and activation of antigen-presenting cells, ultimately enabling the cross-priming of tumor-targeting CTLs (60). These signals encompass immunogenic cell death (ICD)-associated damage-associated molecular patterns (DAMP), such as the surface-exposed calreticulin (CALR) and secreted ATP (61, 62), as well as proinflammatory cytokines such as type I interferon (IFN; refs. 63, 64). Finally, RT can promote the upregulation of death receptors such as Fas cell-surface death receptor (FAS) and tumor necrosis factor (TNF) receptor superfamily member 10b (TNFRSF10B, best known as TRAIL-R2 or DR5) on the surface of cancer cells, de facto increasing their susceptibility to lysis by activated CD8+ CTLs (59, 65). Abundant mechanistic data from preclinical tumor models and observational findings from clinical studies implicate most (if not all) these pathways in the therapeutic effects of RT.

On the other hand, RT can also elicit a plethora of immunosuppressive pathways. Thus, irradiated cancer cells can release immunosuppressive cytokines, such as TGFB1 (66), homodimeric inhibin subunit beta A (INHBA; ref. 67), IL6 (68), and IL10 (69), and express increased levels of PD-L1 on their surface (70, 71), ultimately inhibiting the activity of various immune effector cells potentially present in the TME. Moreover, RT can efficiently recruit and/or expand immunosuppressive cell populations including TREG cells (67, 72), M2-like TAMs (73, 74), and MDSCs (see below). Finally, RT can limit tumor-targeting immune responses by directly killing immune effector cells and causing at least some degree of vascular disruption coupled to local fibrosis and hypoxia, which altogether can largely compromise TEFF cell recruitment to the TME and activation (75–77). The relevance of these immunosuppressive pathways for the therapeutic effects of RT is substantiated by abundant mechanistic data in preclinical tumor models and emerging observational findings from clinical studies.

Importantly, the balance between RT-driven immunostimulation and immunosuppression, which ultimately dictates the efficacy of RT, at least in preclinical settings, is influenced by a variety of physical and biological parameters, including (but not limited to)

  • (i) RT dose, dose rate, fractionation, and administration schedule (in the context of combinatorial regimen; refs. 56, 57, 74, 78);

  • (ii) physical RT nature (i.e., which subatomic particles are used for irradiation) and linear energy transfer (i.e., the energy that an ionizing particle transfers to the material traversed per unit of distance; refs. 79–81);

  • (iii) tumor type, stage, anatomical localization, and baseline infiltration by immune cells (82, 83);

  • (iv) genetic, epigenetic, and immunologic intratumoral heterogeneity (84);

  • (v) local oxygenation (85); and

  • (vi) systemic microbial configuration (86).

These observations may provide an interpretative framework for apparently discrepant results emerging from studies investigating RT-dependent immunomodulation in both preclinical and clinical settings.

A growing preclinical literature illustrates the ability of RT to alter the abundance of circulating or tumor-infiltrating MDSCs in a therapeutically relevant manner in mouse models of cancer (Table 1).

Table 1.

RT and MDSCs in syngeneic mouse models of cancer.

Type of cancerModelIn vivo settingRT scheduleEffect on MDSCs after RTRef.
Breast cancer 4T1 cells Subcutaneous BALB/c 12 Gy × 2 - MDSC depletion upon RT, cyclophosphamide, PD-1 blockade and/or VISTA blockade (103
Breast cancer 4T1 cells Subcutaneous BALB/c 8 Gy × 3 - G-MDSC expansion in the spleen and TME (104
    - Decreased MDSC infiltration and FASL expression upon RT, PI3K inhibition, and PD-1 blockade  
Breast cancer TS/A cells Subcutaneous BALB/cAnN 8 Gy × 3 - G-MDSC recruitment upon RT, LTX-315, and CTLA4 blockade (72
Breast cancer TUBO cells Subcutaneous BALB/c 12 Gy × 1 - Limited effect on MDSC recruitment after 3 days (111
    - Depletion after 10 days with or without PD-L1 blockade  
CRC CT26 cells Subcutaneous BALB/c 30 Gy × 1 - Transient MDSC recruitment after 3 days and decreased MDSC infiltration after 14 days (110
   3 Gy × 10 - MDSC recruitment after 14 days upon depletion of CD8+ and CD4+ T cells  
CRC CT26 cells Subcutaneous BALB/c 2 Gy × 18 - MDSC recruitment after 14 days with 2 Gy × 18 (112
   8 Gy × 3   
   16.4 Gy × 1   
CRC MC38 cells Subcutaneous C57BL/6 20 Gy × 1 - M-MDSC recruitment with RT alone (102
    - MDSC depletion upon RT plus IFNAR1 or cGAMP and CCR2 blockade or Sting knockout  
    - Increased tumor control upon RT and CCR2 blockade or Ccr2 knockout  
CRC MC38 cells Subcutaneous C57BL/6 20 Gy × 1 - Increased RT efficiency and tumor control after MDSC depletion (111
Glioma CT2A cells Intercranial C57BL/6 3 Gy × 1.33 - M-MDSC recruitment with fractionated schedule (74
   4 Gy × 1   
Glioma CT2A cells Intercranial C57BL/6 2 Gy × 4 - Limited effect on MDSC recruitment (107
    - Depletion upon RT and injection of magnetic nanoparticles  
Glioma CT2A cells Intercranial C57BL/6 4 Gy × 1 - Decreased M-MDSC infiltration (108
HCC BNL-P2 cells Orthotopic 10 Gy × 1 - Limited effect on MDSC recruitment (106
  BALB/c  - Depletion upon RT and IL12 administration  
    - Increased tumor control upon RT and IL12 administration  
HCC H22 cells Subcutaneous C57BL/6 2.5 Gy × 16 - MDSC depletion in the tumor and peripheral blood (95
 Hepa1-6 cells ICR 4 Gy × 10 - Decreased plasma levels of MDSC-related cytokines  
   6 Gy × 6 + 4 Gy × 1   
   8 Gy × 5   
Lung cancer LL/2 cells Subcutaneous C57BL/6 4 Gy × 9 - MDSC recruitment (lower in AHFRT vs. CFRT) (105
   11.5 Gy × 2 - MDSC depletion in the spleen and peripheral blood 13 days after AHFRT  
    - PD-L1 overexpression (lower in AHFRT vs. CFRT)  
Lung cancer LLC cells Subcutaneous C57BL/6 20 Gy × 1 - Increased RT efficiency and tumor control after M-MDSC depletion upon RT, CGAMP and CCR2 blockade (102
Lung cancer LLC cells Subcutaneous C57BL/6 20 Gy × 1 - G-MDSC expansion in the tumor and peripheral blood with increased ARG1 activity and PD-L1 expression (98
    - Depletion upon RT and PDE5 inhibition  
    - Increased tumor control upon RT and PDE5 inhibition  
Lung cancer LLC cells Subcutaneous C57BL/6 10 Gy × 1 - MDSC recruitment (99
    - Depletion upon RT and LXR agonist  
    - Increased tumor control upon RT and LXR agonist  
Lung cancer LLC cells Subcutaneous C57BL/6 2 Gy × 3 - Decreased MDSC infiltration after RT and PD-L1 blockade (100
    - Increased tumor control upon RT and PD-L1 blockade  
Lung cancer LLC cells Subcutaneous C57BL/6 10 Gy × 4 - PD-L1 overexpression on MDSCs (101
    - Decreased MDSC infiltration after RT and PD-L1 blockade with or without VEGF blockade  
Lung cancer LLC cells Subcutaneous C57BL/6 6 Gy × 3 - IDO1+ MDSC recruitment in a dose-dependent manner (97
   12 Gy × 3 - Depletion upon RT plus IDO1 blockade  
Lung cancer LLC-OVA cells Subcutaneous C57BL/6 30 Gy × 1 - Decreased MDSC infiltration with or without SMAC mimetic (69
    - Partial restoration of MDSC infiltration upon RT and SMAC mimetic and IFNF blockade, CD8+ T-cell or TNF depletion  
Melanoma B16 cells Subcutaneous BALB/c 5 Gy × 1 - MDSC depletion in the bone marrow, peripheral blood, spleen, and tumor (higher in CIRT compared with XR) (109
 MelanA cells   - Decreased JAK2 and STAT3 expression in MDSCs (higher in CIRT compared with XR)  
 S91 cells     
Melanoma B16F10 cells Subcutaneous C57BL/6 4 Gy × 9 - MDSC recruitment (lower in AHFRT vs. CFRT) (105
   11.5 Gy × 2 - MDSC depletion in the spleen and peripheral blood 13 days after AHFRT  
    - PD-L1 overexpression (lower in AHFRT vs. CFRT)  
PDAC FC1242 cells Subcutaneous C57BL/6 8 Gy × 1 - MDSC recruitment and STAT3 activation in G-MDSCS (94
 PK5L1940 cells   - Decreased MDSC infiltration and STAT3 phosphorylation after RT and STAT3 blockade  
PDAC KC Endogenous C57BL/6 6 Gy × 3 - Tumor cell–derived lactate induced MDSC activation (47
    - Upregulation of S100a8, S100a9, Arg1, Nos2, Vegf, and Mmps in MDSCs  
PDAC Panc-02 cells Orthotopic C57BL/6 6 Gy × 3 - Tumor cell–derived lactate induced MDSCs activation (47
    - Upregulation of S100a8, S100a9, Arg1, Nos2, Vegf, and Mmps in MDSCs  
    - Increased tumor control by RT upon Ldha knockdown in tumor cells or Hif1a deletion in myeloid cells  
Prostate cancer DVL3 cells Subcutaneous C57BL/6J 2 Gy × 5 - Upregulation of an MDSC gene signature (90
Prostate cancer RM-1 cells Subcutaneous C57BL/6 3 Gy × 5 - MDSC expansion in tumor, peripheral blood, spleen, lymph node and lung (92
 RM-9 cells   - Depletion in tumor and spleen upon RT and CSF1R blockade  
 Myc-CaP cells     
Prostate cancer RM-1 cells Subcutaneous C57BL/6 FVB/NCrlBR 8 Gy × 1 - Rapid MDSC recruitment after 2 days, transient decrease after 8 days, and final increase after 14 days (91
 Myc-CaP cells  8 Gy × 2 - TREG cell recruitment upon RT and MDSC depletion  
Prostate cancer TRAMP-C1 cells Intramuscular C57BL/6J 8 Gy × 1 - MDSC expansion in tumor, peripheral blood and spleen at high RT dose (88
   25 Gy × 1 - G-MDSC aggregation in central necrotic and avascular hypoxic regions of irradiated tumors  
    - Increased RT efficiency and tumor control after G-MDSC depletion  
Prostate cancer TRAMP-C1 cells Orthotopic subcutaneous C57BL/6J 15 Gy × 1 - MDSC recruitment (more so in hormone resistant vs. hormone-sensitive model) (68, 89
 TRAM-HR cells   - Depletion upon RT and IL6 blockage, UVB irradiation or androgen deprivation therapy (87
Type of cancerModelIn vivo settingRT scheduleEffect on MDSCs after RTRef.
Breast cancer 4T1 cells Subcutaneous BALB/c 12 Gy × 2 - MDSC depletion upon RT, cyclophosphamide, PD-1 blockade and/or VISTA blockade (103
Breast cancer 4T1 cells Subcutaneous BALB/c 8 Gy × 3 - G-MDSC expansion in the spleen and TME (104
    - Decreased MDSC infiltration and FASL expression upon RT, PI3K inhibition, and PD-1 blockade  
Breast cancer TS/A cells Subcutaneous BALB/cAnN 8 Gy × 3 - G-MDSC recruitment upon RT, LTX-315, and CTLA4 blockade (72
Breast cancer TUBO cells Subcutaneous BALB/c 12 Gy × 1 - Limited effect on MDSC recruitment after 3 days (111
    - Depletion after 10 days with or without PD-L1 blockade  
CRC CT26 cells Subcutaneous BALB/c 30 Gy × 1 - Transient MDSC recruitment after 3 days and decreased MDSC infiltration after 14 days (110
   3 Gy × 10 - MDSC recruitment after 14 days upon depletion of CD8+ and CD4+ T cells  
CRC CT26 cells Subcutaneous BALB/c 2 Gy × 18 - MDSC recruitment after 14 days with 2 Gy × 18 (112
   8 Gy × 3   
   16.4 Gy × 1   
CRC MC38 cells Subcutaneous C57BL/6 20 Gy × 1 - M-MDSC recruitment with RT alone (102
    - MDSC depletion upon RT plus IFNAR1 or cGAMP and CCR2 blockade or Sting knockout  
    - Increased tumor control upon RT and CCR2 blockade or Ccr2 knockout  
CRC MC38 cells Subcutaneous C57BL/6 20 Gy × 1 - Increased RT efficiency and tumor control after MDSC depletion (111
Glioma CT2A cells Intercranial C57BL/6 3 Gy × 1.33 - M-MDSC recruitment with fractionated schedule (74
   4 Gy × 1   
Glioma CT2A cells Intercranial C57BL/6 2 Gy × 4 - Limited effect on MDSC recruitment (107
    - Depletion upon RT and injection of magnetic nanoparticles  
Glioma CT2A cells Intercranial C57BL/6 4 Gy × 1 - Decreased M-MDSC infiltration (108
HCC BNL-P2 cells Orthotopic 10 Gy × 1 - Limited effect on MDSC recruitment (106
  BALB/c  - Depletion upon RT and IL12 administration  
    - Increased tumor control upon RT and IL12 administration  
HCC H22 cells Subcutaneous C57BL/6 2.5 Gy × 16 - MDSC depletion in the tumor and peripheral blood (95
 Hepa1-6 cells ICR 4 Gy × 10 - Decreased plasma levels of MDSC-related cytokines  
   6 Gy × 6 + 4 Gy × 1   
   8 Gy × 5   
Lung cancer LL/2 cells Subcutaneous C57BL/6 4 Gy × 9 - MDSC recruitment (lower in AHFRT vs. CFRT) (105
   11.5 Gy × 2 - MDSC depletion in the spleen and peripheral blood 13 days after AHFRT  
    - PD-L1 overexpression (lower in AHFRT vs. CFRT)  
Lung cancer LLC cells Subcutaneous C57BL/6 20 Gy × 1 - Increased RT efficiency and tumor control after M-MDSC depletion upon RT, CGAMP and CCR2 blockade (102
Lung cancer LLC cells Subcutaneous C57BL/6 20 Gy × 1 - G-MDSC expansion in the tumor and peripheral blood with increased ARG1 activity and PD-L1 expression (98
    - Depletion upon RT and PDE5 inhibition  
    - Increased tumor control upon RT and PDE5 inhibition  
Lung cancer LLC cells Subcutaneous C57BL/6 10 Gy × 1 - MDSC recruitment (99
    - Depletion upon RT and LXR agonist  
    - Increased tumor control upon RT and LXR agonist  
Lung cancer LLC cells Subcutaneous C57BL/6 2 Gy × 3 - Decreased MDSC infiltration after RT and PD-L1 blockade (100
    - Increased tumor control upon RT and PD-L1 blockade  
Lung cancer LLC cells Subcutaneous C57BL/6 10 Gy × 4 - PD-L1 overexpression on MDSCs (101
    - Decreased MDSC infiltration after RT and PD-L1 blockade with or without VEGF blockade  
Lung cancer LLC cells Subcutaneous C57BL/6 6 Gy × 3 - IDO1+ MDSC recruitment in a dose-dependent manner (97
   12 Gy × 3 - Depletion upon RT plus IDO1 blockade  
Lung cancer LLC-OVA cells Subcutaneous C57BL/6 30 Gy × 1 - Decreased MDSC infiltration with or without SMAC mimetic (69
    - Partial restoration of MDSC infiltration upon RT and SMAC mimetic and IFNF blockade, CD8+ T-cell or TNF depletion  
Melanoma B16 cells Subcutaneous BALB/c 5 Gy × 1 - MDSC depletion in the bone marrow, peripheral blood, spleen, and tumor (higher in CIRT compared with XR) (109
 MelanA cells   - Decreased JAK2 and STAT3 expression in MDSCs (higher in CIRT compared with XR)  
 S91 cells     
Melanoma B16F10 cells Subcutaneous C57BL/6 4 Gy × 9 - MDSC recruitment (lower in AHFRT vs. CFRT) (105
   11.5 Gy × 2 - MDSC depletion in the spleen and peripheral blood 13 days after AHFRT  
    - PD-L1 overexpression (lower in AHFRT vs. CFRT)  
PDAC FC1242 cells Subcutaneous C57BL/6 8 Gy × 1 - MDSC recruitment and STAT3 activation in G-MDSCS (94
 PK5L1940 cells   - Decreased MDSC infiltration and STAT3 phosphorylation after RT and STAT3 blockade  
PDAC KC Endogenous C57BL/6 6 Gy × 3 - Tumor cell–derived lactate induced MDSC activation (47
    - Upregulation of S100a8, S100a9, Arg1, Nos2, Vegf, and Mmps in MDSCs  
PDAC Panc-02 cells Orthotopic C57BL/6 6 Gy × 3 - Tumor cell–derived lactate induced MDSCs activation (47
    - Upregulation of S100a8, S100a9, Arg1, Nos2, Vegf, and Mmps in MDSCs  
    - Increased tumor control by RT upon Ldha knockdown in tumor cells or Hif1a deletion in myeloid cells  
Prostate cancer DVL3 cells Subcutaneous C57BL/6J 2 Gy × 5 - Upregulation of an MDSC gene signature (90
Prostate cancer RM-1 cells Subcutaneous C57BL/6 3 Gy × 5 - MDSC expansion in tumor, peripheral blood, spleen, lymph node and lung (92
 RM-9 cells   - Depletion in tumor and spleen upon RT and CSF1R blockade  
 Myc-CaP cells     
Prostate cancer RM-1 cells Subcutaneous C57BL/6 FVB/NCrlBR 8 Gy × 1 - Rapid MDSC recruitment after 2 days, transient decrease after 8 days, and final increase after 14 days (91
 Myc-CaP cells  8 Gy × 2 - TREG cell recruitment upon RT and MDSC depletion  
Prostate cancer TRAMP-C1 cells Intramuscular C57BL/6J 8 Gy × 1 - MDSC expansion in tumor, peripheral blood and spleen at high RT dose (88
   25 Gy × 1 - G-MDSC aggregation in central necrotic and avascular hypoxic regions of irradiated tumors  
    - Increased RT efficiency and tumor control after G-MDSC depletion  
Prostate cancer TRAMP-C1 cells Orthotopic subcutaneous C57BL/6J 15 Gy × 1 - MDSC recruitment (more so in hormone resistant vs. hormone-sensitive model) (68, 89
 TRAM-HR cells   - Depletion upon RT and IL6 blockage, UVB irradiation or androgen deprivation therapy (87

Abbreviations: AHRT, ablative hypofractionated radiotherapy; CFRT, conventional fractionated radiotherapy; CIRT, carbon ion radiotherapy; CRC, colorectal carcinoma; G-, granulocytic; M-, monocytic; MDSC, monocyte-derived suppressor cell; NSCLC, non–small cell lung carcinoma; RT, radiotherapy; TREG, regulatory T; XR, X-ray.

Mouse TRAMP-C1 prostate tumors (including the hormone-resistant TRAMP-HR variant) established subcutaneously in immunocompetent syngeneic hosts drive a robust systemic expansion of M- and G-MDSCs upon exposure to single RT doses > 15 Gy (87–89). Such an expansion culminates with the intratumoral accumulation of MDSCs expressing immunosuppressive factors (i.e., ARG1 and IDO1) as well as matrix metallopeptidase 9 (MMP9), an extracellular enzyme that favors disease dissemination, especially in central, necrotic tumor areas (88). In this setting, tumor infiltration by MDSCs limits the efficacy of RT, as demonstrated by depletion experiments (88). Moreover, the ability of RT to recruit MDSCs to the tumor bed mechanistically involves IL6, which has also been linked to resistance to hormonotherapy (87), and can be prevented, at least partially, by UVB irradiation or vitamin D3 supplementation, correlating with (i) suppressed IL6 signaling, (ii) restored CD3+ T-cell infiltration, and (iii) increased therapeutic efficacy (87, 88). A similar systemic expansion and intratumoral accumulation of MDSCs has also been documented in immunocompetent mice bearing mouse DVL3, RM-1, RM-9, and Myc-Cap prostate tumors upon irradiation with 2 RT fractions of 8 Gy each (DVL3 and RM-1), 5 RT fractions of 3 Gy each (Myc-Cap, RM-1, and RM-9) and a single RT fraction of 8 Gy (Myc-Cap; refs. 90–92). In RM-1 tumors treated with 2 RT fractions of 8 Gy each, early M- and G-MDSC infiltration is followed by an influx of functional T cells, and MDSC depletion offers no therapeutic benefits because of compensatory TREG cell expansion (91). Conversely, RM-1 tumors responding to 5 RT fractions of 3 Gy each appear to recruit an MDSC population enriched in M-MDSCs, coupled to the intratumoral expansion of M2-like TAMs (92). In this latter setting, pharmacologic inhibition of colony stimulating factor 1 receptor (CSF1R), which is abundantly expressed by M-MDSCs (less so G-MDSCs) and M2-like TAMs (93), limits MDSC infiltration, downregulates the expression of ARG1 and MMP9 and ultimately improves the therapeutic efficacy of RT (92).

Mouse FC1242 and PK5L1940 pancreatic ductal adenocarcinomas (PDAC) established subcutaneously in immunocompetent syngeneic hosts recruit MDSCs (especially, but not exclusively G-MDSCs) upon irradiation with a single fraction of 8 Gy. This correlates with increased signal transducer and activator of transcription (STAT3) phosphorylation, which is critical for MDSC-dependent immunosuppression, and accrued Ki67 positivity, which reflects MDSC proliferation (94). Consistent with a mechanistic involvement, STAT3 inhibition with an antisense oligonucleotide blunts MDSC recruitment to irradiated PDACs, coupled to limited tumor infiltration by TREG cells, restored TEFF cell recruitment, and increased therapeutic efficacy (94). Similar findings have been documented with mouse Panc-02 PDACs as well as with endogenous PDAC models exposed to 3 RT fraction of 6 Gy each (47). In this setting, irradiation also promoted the secretion of lactate by cancer cells, which was ultimately found to promote the recruitment and immunosuppressive functions of MDSCs via a STAT3-dependent pathway involving mechanistic target of rapamycin (MTOR) and HIF1α (47). Accordingly, both the depletion of MDSCs with a Ly6G-targeting antibody and deletion of Hif1a in myeloid cells improved the therapeutic effects of RT (47). Apparently at odds with these findings, conditioned media from mouse Hepa1-6 and H22 hepatocellular carcinoma (HCC) cells exposed to single RT doses of 2.5, 4, 6, or 8 Gy block MDSC differentiation and proliferation via suppressed IL6 secretion and consequent STAT3 inhibition in MDSC precursors (95). Consistent with this notion, a total dose of 40 Gy delivered in 2.5, 4, 6, or 8 Gy per fraction to Hepa1-6 and H22 HCCs growing in immunocompetent syngeneic mice appears to mediate considerable therapeutic activity coupled to both systemic and intratumoral MDSC depletion (95). Whether lactate secretion upon irradiation with a single RT dose of 6 Gy is a prerogative of PDAC cells remains to be elucidated. Alternatively, the differential response of PDACs and HCCs to RT with respect to MDSCs may reflect pathways elicited by other components of the TME, such as cancer-associated fibroblasts (CAF). CAFs, which are abundant in PDAC lesions, can respond to RT by producing high amounts of MDSC chemoattractants like C–X–C motif chemokine ligand 1 (CXCL1; ref. 96).

Mouse Lewis lung carcinoma (LLC) tumors established in immunocompetent syngeneic hosts and exposed to 3 RT fraction of 6 or 12 Gy each recruit a population of MDSCs that expresses CD11b and IDO1, intermediate levels of Ly6G, but no CD106 and CD11c, as well as TREG cells (97). In this scenario, coadministration of a pharmacologic IDO1 inhibitor improves the efficacy of RT (delivered as 3 fractions of 12 Gy each) but fails to suppress MDSC recruitment (97). Conversely, blocking ARG1 or activating nuclear receptor subfamily 1 group H member 2 (NR1H2, best known as LXR) in the context of a single RT dose of 20 or 10 Gy, respectively, partially restores LLC infiltration by CD8+ CTLs as it extends the therapeutic benefits of RT (98, 99). Similar findings have been obtained by blocking PD-L1 in the context of 3 RT fractions of 2 Gy each or 4 RT fractions of 10 Gy each (100, 101). Moreover, neutralizing C–C motif chemokine ligand 2 (CCL2), blocking C–C motif chemokine receptor 2 (CCR2), or deleting Ccr2 efficiently prevents the RT-dependent infiltration of LLC tumors (and mouse MC38 colorectal carcinomas) by MDSCs (102). Ultimately, this enables superior therapeutic responses to RT plus a stimulator of interferon response cGAMP interactor (STING1) agonist (102). These findings, which mechanistically reflect CCL2 secretion downstream of STING1-dependent type I IFN signaling, nicely exemplify the complexity of RT-dependent immunomodulation and suggest that CCL2/CCR2 inhibition may represent a promising strategy to improve the efficacy of RT by offsetting MDSC expansion.

In preclinical models of breast cancer, including models of hormone receptor (HR)+ breast cancer (TS/A) and triple-negative breast cancer (4T1), RT schedules associated with good local disease control but suboptimal immunostimulation (3 fractions of 8 Gy each for TS/A and 4T1, and 2 fractions of 12 Gy for 4T1) appear to have limited effects on tumor infiltration by M- and G-MDSCs (72, 103, 104). In the TS/A model, improved immunostimulation by RT, as achieved by the coadministration of an oncolytic peptide and a cytotoxic T lymphocyte-associated protein 4 (CTLA4)-targeting immune-checkpoint inhibitor (ICI), results in the compensatory expansion of intratumoral MDSCs (72). Conversely, in the 4T1 model, improved overall survival (in the context of virtually unaffected local disease control), as achieved by combining RT with various combinatorial regimens involving a PI3K inhibitor, a V-set immunoregulatory receptor (best known as VISTA), a programmed cell death 1 (PDCD1, best known as PD-1) inhibitor, and/or low-dose cyclophosphamide, is associated with profound MDSC depletion, at least in part reflecting abundant VISTA expression by tumor-infiltrating MDSCs (103, 104). Along these lines, mouse LL/2 lung carcinomas and B16F10 melanomas respond better to a total RT dose of 23 Gy delivered in 2 fractions 8 days apart from each other than they do to a total dose of 36 Gy delivered in 9 daily fractions (but only so in immunocompetent hosts), correlating with superior MDSC depletion, ARG1 and kinase insert domain receptor (the main VEGFA receptor) expression by MDSCs, limited VEGFA secretion by cancer cells, restored CTL infiltration as well as activation of a systemic immune responses that can delay the growth of the same tumors implanted contralaterally shortly after irradiation (105).

A single RT dose of 10 Gy mediates poor therapeutic effects against large BNL-P2 HCCs established subcutaneously or orthotopically in immunocompetent syngeneic mice as it fails to deplete intratumoral MDSCs (106). Conversely, RT plus intratumoral administration of a viral vector encoding IL12 enables superior disease control coupled to MDSC depletion and restored tumor infiltration by active CTLs and NK cells (106). Similar findings correlating the therapeutic effects of RT with M-MDSC (and M2-like TAM) depletion have been obtained in orthotopic models of glioma (CT-2A spheroids) treated with 4 RT fractions of 2 Gy each (107) as well as single RT doses of 2, 4, or 8 Gy (74, 108). In this latter setting, 3 RT fractions of 1.33 Gy was less efficient than an equivalent biologically effective dose (BED) of 4 Gy in a single fraction, and this correlated with increased intratumoral MDSC and M2-like TAM abundance (74). These findings provide additional support to the influence of fractionation on the immunobiological effects of RT. Of note, at least in preclinical models of melanoma (B16, MelanA, and S91), the ability of a single RT dose of 5 Gy to deplete MDSCs is considerably ameliorated (correlating with improved CTL activation and disease control) when carbon ions are used instead of X-rays, at least in part reflecting superior STAT3 inhibition (109). These data highlight the important of physical parameters other than dose and fraction on the effects of RT.

Finally, it is interesting to note that in mouse models of breast cancer (TUBO), colorectal carcinoma (CT26 and MC38), and lung carcinoma (LLC) exposed to single RT doses of 12 Gy (TUBO and MC38) or 30 Gy (CT26, MC38, and LLC), intratumoral G- and M-MDSC depletion linked to (at least partial) therapeutic efficacy mechanistically depends on the activation of a tumor-targeting adaptive immune response involving DC-dependent cross-presentation (110), CD4+ T cell–dependent help (110), as well as TNF and IFNγ secretion by CD8+ CTLs, ultimately resulting in apoptotic MDSC death (69, 110, 111). In the CT26 model, 3 RT doses of 8 Gy as well as 18 RT fractions of 2 Gy each mediate superior therapeutic activity than a single RT fraction of 16.4 Gy, but whereas hypofractionation enables the recruitment of CD8+ CTLs to the tumor bed, conventional fractionation causes pronounced intratumoral MDSC expansion (112). In most of these settings, improved tumor control can be achieved by adding immunostimulatory agents including ICIs targeting PD-1, PD-L1, or T-cell immunoreceptor with Ig and ITIM domains (TIGIT; refs. 69, 111, 112). Conversely, delivering 10 RT fractions of 3 Gy each after the initial dose of 30 Gy compromises therapeutic efficacy in the CT26 model, and this correlates with recovered MDSC infiltration and CTL depletion (110). Whether alternative fractionation schedules can be successfully used as maintenance regimens in this setting remains to be investigated.

Taken together, these observations highlight the complex relationship between RT and numerical as well as functional MDSC alterations in preclinical tumor models as they provide important mechanistic insights toward the development of therapeutic approaches harnessing RT and MDSC-targeting strategies. Specifically, it seems that the ability of RT to influence systemic MDSC expansion, recruitment, and immunosuppressive activity varies considerably with total dose and fractionation schedule, with hypofractionated regimens standing out as superior approaches for MDSC depletion (at least when total dose is clinically relevant). That said, only a few studies have comparatively assessed different fractionation schedules of the same BED (74, 105, 112), highlighting the need for additional work to deconvolute the impact of RT dose and fractionation on MDSCs.

A few clinical studies have investigated the impact of RT on the abundance of circulating or tumor-infiltrating MDSCs in patients with cancer (Table 2).

Table 2.

RT and MDSCs in patients with cancer.

Type of cancerNo of patientsRT scheduleaNoteEffect on MDSCs after RTRef.
Cervical cancer 10 2 Gy × 23 Combined with platinum-based chemotherapy and BT - M-MDSC expansion in 9 of 10 patients (122
  7 Gy × 3–4    
HNSCC 70 Gy Combined with platinum-based chemotherapy - MDSC expansion in 1 of 3 patients (116
HNSCC 15 2–2.2 Gy × 30–25 Combined with cisplatin and/or cetuximab - G-MDSC expansion, correlating with IL6 levels and arginase activity (121
    - PD-L1 overexpression on MDSCs  
HNSCC 20 70 Gy With or without platinum-based chemotherapy - MDSC expansion in 14 of 20 patients (120
HNSCC 43 <2 Gy × 30 No combination therapy - MDSC depletion (125
    - Higher levels of MDSCs correlating with decreased overall survival  
LARC 13 5 Gy × 5 SCRT - MDSC depletion after 5 weeks (124
    - Lower levels of M-MDSCs corresponding to poor responders to SCRT  
Melanoma 9.5 Gy × 3 No combination therapy - Decrease in MDSCs (126
NSCLC 2.75 Gy × 24 Combined with cisplatin - MDSC expansion types 4 and 7 (115
NSCLC 7.5 Gy × 8 SRBT - G-MDSC expansion after 72 hours (113
  12.5 Gy × 4  - Progressive decrease of MDSCs after 6 months  
NSCLC 14 60–66 Gy Combined with platinum-based chemotherapy - MDSC expansion in 5 of 14 patients (116
    - MDSC depletion in 5 of 14 patients  
OSCC 45 60–66 Gy No combination therapy - MDSC expansion (118
    - PD-L1 overexpression on MDSCs  
OSCC 248 60–66 Gy Adjuvant RT after surgery - PD-L1 overexpression on MDSCs, correlating with high IL6 levels (119
PDAC 9 Gy × 3 SRBT - MDSC expansion (94
  10 Gy × 3    
  11 Gy × 3    
Pharyngeal cancer 75 >66 Gy Combined with chemotherapy - PD-L1 overexpression on MDSCs, correlating with high IL6 levels (119
SCLC 4 Gy Combined with platinum-based chemotherapy - MDSC expansion in 2 of 3 patients (116
Type of cancerNo of patientsRT scheduleaNoteEffect on MDSCs after RTRef.
Cervical cancer 10 2 Gy × 23 Combined with platinum-based chemotherapy and BT - M-MDSC expansion in 9 of 10 patients (122
  7 Gy × 3–4    
HNSCC 70 Gy Combined with platinum-based chemotherapy - MDSC expansion in 1 of 3 patients (116
HNSCC 15 2–2.2 Gy × 30–25 Combined with cisplatin and/or cetuximab - G-MDSC expansion, correlating with IL6 levels and arginase activity (121
    - PD-L1 overexpression on MDSCs  
HNSCC 20 70 Gy With or without platinum-based chemotherapy - MDSC expansion in 14 of 20 patients (120
HNSCC 43 <2 Gy × 30 No combination therapy - MDSC depletion (125
    - Higher levels of MDSCs correlating with decreased overall survival  
LARC 13 5 Gy × 5 SCRT - MDSC depletion after 5 weeks (124
    - Lower levels of M-MDSCs corresponding to poor responders to SCRT  
Melanoma 9.5 Gy × 3 No combination therapy - Decrease in MDSCs (126
NSCLC 2.75 Gy × 24 Combined with cisplatin - MDSC expansion types 4 and 7 (115
NSCLC 7.5 Gy × 8 SRBT - G-MDSC expansion after 72 hours (113
  12.5 Gy × 4  - Progressive decrease of MDSCs after 6 months  
NSCLC 14 60–66 Gy Combined with platinum-based chemotherapy - MDSC expansion in 5 of 14 patients (116
    - MDSC depletion in 5 of 14 patients  
OSCC 45 60–66 Gy No combination therapy - MDSC expansion (118
    - PD-L1 overexpression on MDSCs  
OSCC 248 60–66 Gy Adjuvant RT after surgery - PD-L1 overexpression on MDSCs, correlating with high IL6 levels (119
PDAC 9 Gy × 3 SRBT - MDSC expansion (94
  10 Gy × 3    
  11 Gy × 3    
Pharyngeal cancer 75 >66 Gy Combined with chemotherapy - PD-L1 overexpression on MDSCs, correlating with high IL6 levels (119
SCLC 4 Gy Combined with platinum-based chemotherapy - MDSC expansion in 2 of 3 patients (116

Abbreviations: BT, brachytherapy; G-, granulocytic; HNSCC, head and neck squamous cell carcinoma; LARC, locally advanced rectal cancer; M-, monocytic; MDSC, monocyte-derived suppressor cell; NSCLC, non–small cell lung carcinoma; OSCC, oral squamous cell carcinoma; PDAC, pancreatic ductal adenocarcinoma; RT, radiotherapy; SCRT, short-course radiotherapy; SCLC, small cell lung carcinoma.

aTotal dose unless otherwise specified.

No differences in circulating M- and G-MDSCs were detected in 7 patients with non–small cell lung carcinoma (NSCLC) 3 days, 1 month, 3 months, and 6 months after stereotactic body radiotherapy (SBRT) with 8 fractions of 7.5 Gy each or 4 fractions of 12.5 Gy each (113). In a distinct cohort of 18 subjects with NSCLC, the circulating levels of 10 specific MDSC subsets (114) at baseline did not differ from those of 8 healthy donors, and they were not altered by carboplatin–pemetrexed (n = 7) or carboplatin–vinorelbine (n = 5) chemotherapy (115). However, patients receiving a 5-week cycle of concurrent chemoradiotherapy (24 daily RT fractions of 2.75 Gy each) preceded by low-dose cisplatin (n = 6) did exhibit a slight increase in circulating CD14+HLA-DRlow and CD14+CD33+HLA-DRlow MDSCs that was paralleled by a decrease in blood-borne lymphocytes (115). Similar findings have been documented in a cohort of 20 patients with small cell lung carcinoma (SCLC), NSCLC, or head and neck squamous cell carcinoma (HNSCC) treated with total RT doses of 45, 60 to 66, or 70 Gy, respectively, in the context of cisplatin-based chemotherapy (116). In this setting, >20% elevations in circulating MDSCs were detected in 8 of 20 patients. This correlated with an increase in blood-borne TREG cells and was associated with suppressed T-cell responses against the tumor-associated antigen telomerase reverse transcriptase (TERT; ref. 116). Of note, the negative correlation between circulating MDSCs and TERT-targeting immunity also appears to hold true and have negative prognostic value in other cancer types (e.g., anal squamous cell carcinoma) and irrespective of RT (117).

An expansion in circulating CD11b+HLA-DRCD33+CD14+ MDSCs coupled to PD-L1 upregulation on the MDSC surface has also been documented in a cohort of 45 patients with oral squamous cell carcinoma (OSCC) 2 weeks after adjuvant RT at a total dose of 60 to 66 Gy, especially when OSCC cells expressed stem cell markers that have been linked with MDSC expansion in preclinical cancer models (46, 118). Similar results have been obtained in a partially overlapping cohort of 96 patients with HNSCC (encompassing OSCC and pharyngeal cancer) who received adjuvant RT at a total dose greater than or equal to 66 Gy for definite RT, especially in subjects with high circulating levels of IL6 (119); and in an independent cohort of 20 subjects with HNSCC who received a 7-week course of curative-intent RT (median dose 70 Gy) with or without chemotherapy (120). Like subjects with NSCLC, patients with (most often HPV+) HNSCC receiving a total RT dose of 66 to 70 Gy (in 2–2.2 Gy/fraction RT over 7 weeks in the context of cisplatin- or cetuximab-based chemotherapy) exhibited a progressive accumulation of circulating CD14CD15+CD33+ G-MDSCs (but not M-MDSCs), correlating with increased blood-borne IL6 levels, increased ARG1 activity in the circulation, STAT3 activation in the CD33+ compartment (but not in M- or G-MDSCs), as well as accrued PD-L1 expression on both M- and G-MDSCs (121). Interestingly, most of these changes largely resolved 5 weeks after RT interruption (121). Similarly, 9 of 10 patients with cervical carcinoma experienced a considerable expansion of circulating CD3CD19CD1aHLA-DRCD14+CD15 M-MDSCs coupled to PD-1 upregulation on CD4+ T cells and impaired T-cell reactivity upon receiving 23 RT fractions of 2 Gy each, but these alterations resolved 6 to 9 weeks after RT cessation (122), suggesting a transient nature for RT-driven MDSC elevation (at least in these oncological settings). An increase in circulating MDSCs with active STAT3 has also been documented in 7 patients with PDAC receiving RT in 3 fractions of 9, 10, or 11 Gy as part of disease management (94). In this context, however, blood-borne IL6 levels remained unchanged, while both VEGFA and TGFB1 increased after treatment, suggesting an IL6-independent mechanism of expansion (94).

In a cohort of 62 subjects with locally advanced rectal cancer (LARC) receiving long-course neoadjuvant RT (a total dose of 40–45 Gy delivered to the pelvis in 25 to 28 fractions over 5 weeks, with/or without a boost of 5.4 Gy to the primary tumor) followed by radical surgery, tumor-infiltrating MDSCs did not exhibit consistent numerical alterations post-RT across patients and were not statistically associated with progression-free survival or overall survival (OS; ref. 123). However, patients with low MDSCs at baseline had a higher chance to exhibit good disease control at surgical resection (123). Conversely, in a distinct cohort of 13 patients with LARC receiving short-course neoadjuvant RT (a total dose of 25 Gy delivered to the pelvis in 5 daily fractions over 1 week) patients with poor disease control at surgical resection experienced a major decrease in circulating HLA-DRCD11b+CD15CD14+CD33+ M-MDSCs and (less so) LinHLA-DRCD11b+CD15+CD14CD33+ G-MDSCs 5 weeks from RT initiation, correlating with an increase in PD-1-expressing TREG cells (124). Also in this setting, MDSC depletion driven by RT rapidly subsided after therapy (124). Along similar lines, circulating MDSCs were decreased in 43 patients with HCC receiving a total RT dose <60 Gy in conventional 2-Gy fractions over 4 to 6 weeks, and posttreatment MDSC abundance negatively correlated with OS in both univariate and multivariate Cox regression analyses (125). Moreover, palliative RT with a total dose of 28.5 Gy delivered in 3 fractions caused disease regression not only to the irradiated paraspinal lesion but also to nonirradiated lymphadenopathy and splenic lesions in a patient with melanoma progressing on CTLA4 blockage, which was paralleled by a trend for transient circulating MDSC depletion (126).

Altogether, these findings lend further support to the notion that the immunomodulatory effects of RT on MDSC abundance and function exhibit a high degree of context dependency, which considerably complicates the development of MDSC-targeting strategies to improve RT efficacy in the clinic (see below).

The abundant literature discussed herein lend strong support to the notion that both the abundance and immunosuppressive activity of MDSCs can be altered by focal RT, which ultimately affects therapeutic outcome. Specifically, the suboptimal depletion and the expansion of MDSCs after RT have been linked with incomplete therapeutic responses in both preclinical tumor models and patients with cancer. Thus, MDSCs stand out as promising targets to ameliorate the efficacy of RT, as demonstrated (at least in preclinical settings) via a variety of pharmacologic interventions or genetic approaches. Of note, many pharmacologic agents that can be harnessed to directly or indirectly target MDSC-dependent immunosuppression are clinically available or currently under clinical development, although none of them has been conceived to specifically interfere with MDSCs.

Clinically approved agents that (at least based on preclinical data) target MDSCs or their functions include, but are not limited to, the IL6 inhibitor tocilizumab, which is currently licensed for use in patients with moderately to severely active rheumatoid arthritis (127), the MTOR inhibitor everolimus, which is commonly used to prevent transplant rejection as well as for the management of multiple neoplasms (128), various anticancer tyrosine kinase inhibitors that (specifically or alongside other kinases) block CSF1R (e.g., pexidartinib, dasatinib, and sunitinib; ref. 129), multiple chemotherapeutics that directly kill MDSCs (e.g., cyclophosphamide; ref. 130), the phosphodiesterase 5 (PDE5) inhibitor tadalafil (131), as well as a variety of ICIs targeting PD-1 (i.e., nivolumab and pembrolizumab) and PD-L1 (i.e., avelumab, atezolizumab, and durvalumab; ref. 132). MDSC-targeting drugs that are still under clinical development encompass IDO1 inhibitors (133) and CCR2 inhibitors (134), as well as VISTA- and TIGIT-targeting ICIs (132). Despite such an ample therapeutic armamentarium, however, none of these agents is used in combination with RT as part of routine clinical protocols for patients with cancer, and only a few are being/have been formally evaluated for their capacity to ameliorate the efficacy of RT in clinical trials (55, 135), generally with rather deceiving results (136). As a notable exception, durvalumab has been shown to cooperate efficiently with SBRT as a neoadjuvant approach for patients with potentially resectable early-stage NSCLC (71). However, whether such a positive outcome involves MDSC depletion remains to be elucidated. Of note, several interventional clinical trials currently recruiting participants are assessing the therapeutic potential of RT in combination with one (or more) agent(s) that (at least based on preclinical data) can deplete or inhibit MDSCs (source www.clinicaltrials.gov). Most of these studies, however, involve FDA-approved (rather than investigational) agents and are not designed to ascertain the potential role of MDSC inhibition. As an exception, the rationale for testing tadalafil together with chemoradiation in patients with stage III–IV astrocytoma reflects the ability of this agent to inhibit MDSCs (NCT04757662). In addition, researchers conducting three clinical trials testing SBRT in patients with advanced malignancies are planning to monitor patients for circulating immune cell subsets including MDSCs (NCT04073745, NCT03348748, and NCT04068649).

In summary, although MDSCs stand out as promising targets to extend the clinical benefits of RT, additional research is needed for translating such a promise into a clinical reality. At least in part, this reflects the extraordinary plasticity of MDSCs, their multipronged immunosuppressive activity, and the existence of mechanisms that drive compensatory myelopoiesis upon MDSC depletion (4). Moreover, currently available preclinical data fail to identify a common denominator for the interaction between RT and MDSCs, which considerably complicates the development of therapeutic strategies that can be rapidly translated to clinical testing. Although RT dose and fractionation have been shown to influence the impact of RT on MDSCs by a few comparative studies (74, 105, 112), various other parameters remain to be comparatively analyzed in this respect. Such potential confounders include, but are not limited to:

  • (i) model and anatomic localization (most studies discussed herein relied on mouse cancer cell lines established subcutaneously in immunocompetent syngeneic hosts, which generally do not recapitulate the immunobiology of human malignancies as closely as orthotopically implanted or endogenous tumors; ref. 137);

  • (ii) time (in the preclinical literature that we summarized, MDSCs were most often quantified 8 to 14 days after RT initiation, with little attention given to early time points);

  • (iii) RT-related parameters beyond dose and fractionation (many studies fail to provide detailed information on particle type, dose rate, and linear energy transfer, which may all influence the biological effects of RT); and

  • (iv) MDSC identification (besides the use of surface markers that are not necessarily identical across different studies, the actual ability of MDSCs expanding or contracting after RT to mediate immunosuppressive functions has rarely been tested with ex vivo functional assays).

Despite these and other incognita, we surmise that obtaining additional insights into MDSC-dependent immunosuppression in adequate preclinical tumor models that consider the aforementioned points, developing strategies that target nonredundant MDSC functions, and selecting patients with prominent MDSC expansion/activity may ultimately unlock the full therapeutic potential of MDSC-targeting strategies as partners for RT in clinical cancer management.

D.I. Gabrilovich reports other support from AstraZeneca outside the submitted work. L. Galluzzi reports grants from Lytix and Phosplatin, personal fees from Boehringer Ingelheim, AstraZeneca, OmniSEQ, The Longevity Labs, Inzen, Luke Heller TECPR2 Foundation, Sotio, and Onxeo outside the submitted work. No disclosures were reported by the other authors.

C. Jiménez-Cortegana is supported by a Margarita Salas fellowship from the University of Sevilla (Spain). The L. Galluzzi lab is supported by a Breakthrough Level 2 grant from the US DoD BRCP (#BC180476P1), by the 2019 Laura Ziskin Prize in Translational Research (#ZP-6177, PI: Formenti) from the Stand Up to Cancer (SU2C), by a Mantle Cell Lymphoma Research Initiative (MCL-RI, PI: Chen-Kiang) grant from the Leukemia and Lymphoma Society, by a startup grant from the Dept. of Radiation Oncology at Weill Cornell Medicine (New York), by a Rapid Response Grant from the Functional Genomics Initiative (New York), by industrial collaborations with Lytix Biopharma (Oslo, Norway) and Phosplatin (New York), and by donations from Phosplatin (New York), the Luke Heller TECPR2 Foundation (Boston), Sotio a.s. (Prague, Czech Republic), Onxeo (Paris, France), Ricerchiamo (Brescia), Lytix Biopharma (Oslo, Norway), and Noxopharm (Chatswood, Australia).

1.
Veglia
F
,
Sanseviero
E
,
Gabrilovich
DI
.
Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity
.
Nat Rev Immunol
2021
;
21
:
485
98
.
2.
Álvarez-Errico
D
,
Vento-Tormo
R
,
Sieweke
M
,
Ballestar
E
.
Epigenetic control of myeloid cell differentiation, identity and function
.
Nat Rev Immunol
2015
;
15
:
7
17
.
3.
Coussens
LM
,
Zitvogel
L
,
Palucka
AK
.
Neutralizing tumor-promoting chronic inflammation: a magic bullet?
Science
2013
;
339
:
286
91
.
4.
Grover
A
,
Sanseviero
E
,
Timosenko
E
,
Gabrilovich
DI
.
Myeloid-derived suppressor cells: a propitious road to clinic
.
Cancer Discov
2021
;
11
:
2693
706
.
5.
Talmadge
JE
,
Gabrilovich
DI
.
History of myeloid-derived suppressor cells
.
Nat Rev Cancer
2013
;
13
:
739
52
.
6.
Gabrilovich
DI
,
Bronte
V
,
Chen
SH
,
Colombo
MP
,
Ochoa
A
,
Ostrand-Rosenberg
S
, et al
.
The terminology issue for myeloid-derived suppressor cells
.
Cancer Res
2007
;
67
:
425
;
author reply 6
.
7.
Rodriguez-Ruiz
ME
,
Vitale
I
,
Harrington
KJ
,
Melero
I
,
Galluzzi
L
.
Immunological impact of cell death signaling driven by radiation on the tumor microenvironment
.
Nat Immunol
2020
;
21
:
120
34
.
8.
Bennett
JA
,
Rao
VS
,
Mitchell
MS
.
Systemic bacillus Calmette-Guerin (BCG) activates natural suppressor cells
.
Proc Natl Acad Sci U S A
1978
;
75
:
5142
4
.
9.
Movahedi
K
,
Guilliams
M
,
Van den Bossche
J
,
Van den Bergh
R
,
Gysemans
C
,
Beschin
A
, et al
.
Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity
.
Blood
2008
;
111
:
4233
44
.
10.
Cassetta
L
,
Baekkevold
ES
,
Brandau
S
,
Bujko
A
,
Cassatella
MA
,
Dorhoi
A
, et al
.
Deciphering myeloid-derived suppressor cells: isolation and markers in humans, mice and non-human primates
.
Cancer Immunol Immunother
2019
;
68
:
687
97
.
11.
Bronte
V
,
Brandau
S
,
Chen
SH
,
Colombo
MP
,
Frey
AB
,
Greten
TF
, et al
.
Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards
.
Nat Commun
2016
;
7
:
12150
.
12.
Zoso
A
,
Mazza
EM
,
Bicciato
S
,
Mandruzzato
S
,
Bronte
V
,
Serafini
P
, et al
.
Human fibrocytic myeloid-derived suppressor cells express IDO and promote tolerance via Treg-cell expansion
.
Eur J Immunol
2014
;
44
:
3307
19
.
13.
Mazza
EM
,
Zoso
A
,
Mandruzzato
S
,
Bronte
V
,
Serafini
P
,
Inverardi
L
, et al
.
Gene expression profiling of human fibrocytic myeloid-derived suppressor cells (f-MDSCs)
.
Genom Data
2014
;
2
:
389
92
.
14.
Ou
L
,
Shi
Y
,
Dong
W
,
Liu
C
,
Schmidt
TJ
,
Nagarkatti
P
, et al
.
Kruppel-like factor KLF4 facilitates cutaneous wound healing by promoting fibrocyte generation from myeloid-derived suppressor cells
.
J Invest Dermatol
2015
;
135
:
1425
34
.
15.
Wright
AK
,
Newby
C
,
Hartley
RA
,
Mistry
V
,
Gupta
S
,
Berair
R
, et al
.
Myeloid-derived suppressor cell-like fibrocytes are increased and associated with preserved lung function in chronic obstructive pulmonary disease
.
Allergy
2017
;
72
:
645
55
.
16.
Zhang
H
,
Maric
I
,
DiPrima
MJ
,
Khan
J
,
Orentas
RJ
,
Kaplan
RN
, et al
.
Fibrocytes represent a novel MDSC subset circulating in patients with metastatic cancer
.
Blood
2013
;
122
:
1105
13
.
17.
Goldmann
O
,
Beineke
A
,
Medina
E
.
Identification of a novel subset of myeloid-derived suppressor cells during chronic staphylococcal infection that resembles immature eosinophils
.
J Infect Dis
2017
;
216
:
1444
51
.
18.
Srivastava
MK
,
Sinha
P
,
Clements
VK
,
Rodriguez
P
,
Ostrand-Rosenberg
S
.
Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine
.
Cancer Res
2010
;
70
:
68
77
.
19.
Sies
H
,
Jones
DP
.
Reactive oxygen species (ROS) as pleiotropic physiological signalling agents
.
Nat Rev Mol Cell Biol
2020
;
21
:
363
83
.
20.
Molon
B
,
Ugel
S
,
Del Pozzo
F
,
Soldani
C
,
Zilio
S
,
Avella
D
, et al
.
Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells
.
J Exp Med
2011
;
208
:
1949
62
.
21.
Rodriguez
PC
,
Quiceno
DG
,
Ochoa
AC
.
L-arginine availability regulates T-lymphocyte cell-cycle progression
.
Blood
2007
;
109
:
1568
73
.
22.
Bantug
GR
,
Galluzzi
L
,
Kroemer
G
,
Hess
C
.
The spectrum of T cell metabolism in health and disease
.
Nat Rev Immunol
2018
;
18
:
19
34
.
23.
Brito
C
,
Naviliat
M
,
Tiscornia
AC
,
Vuillier
F
,
Gualco
G
,
Dighiero
G
, et al
.
Peroxynitrite inhibits T lymphocyte activation and proliferation by promoting impairment of tyrosine phosphorylation and peroxynitrite-driven apoptotic death
.
J Immunol
1999
;
162
:
3356
66
.
24.
Nagaraj
S
,
Gupta
K
,
Pisarev
V
,
Kinarsky
L
,
Sherman
S
,
Kang
L
, et al
.
Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer
.
Nat Med
2007
;
13
:
828
35
.
25.
Cartwright
ANR
,
Suo
S
,
Badrinath
S
,
Kumar
S
,
Melms
J
,
Luoma
A
, et al
.
Immunosuppressive myeloid cells induce nitric oxide-dependent DNA damage and p53 pathway activation in CD8(+) T cells
.
Cancer Immunol Res
2021
;
9
:
470
85
.
26.
Li
H
,
Han
Y
,
Guo
Q
,
Zhang
M
,
Cao
X
.
Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1
.
J Immunol
2009
;
182
:
240
9
.
27.
Tauriello
DVF
,
Sancho
E
,
Batlle
E
.
Overcoming TGFβ-mediated immune evasion in cancer
.
Nat Rev Cancer
2022
;
22
:
25
-
44
.
28.
Hu
CE
,
Gan
J
,
Zhang
RD
,
Cheng
YR
,
Huang
GJ
.
Up-regulated myeloid-derived suppressor cell contributes to hepatocellular carcinoma development by impairing dendritic cell function
.
Scand J Gastroenterol
2011
;
46
:
156
64
.
29.
Ohm
JE
,
Shurin
MR
,
Esche
C
,
Lotze
MT
,
Carbone
DP
,
Gabrilovich
DI
.
Effect of vascular endothelial growth factor and FLT3 ligand on dendritic cell generation in vivo
.
J Immunol
1999
;
163
:
3260
8
.
30.
Gabrilovich
D
,
Ishida
T
,
Oyama
T
,
Ran
S
,
Kravtsov
V
,
Nadaf
S
, et al
.
Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo
.
Blood
1998
;
92
:
4150
66
.
31.
Vito
A
,
Salem
O
,
El-Sayes
N
,
MacFawn
IP
,
Portillo
AL
,
Milne
K
, et al
.
Immune checkpoint blockade in triple negative breast cancer influenced by B cells through myeloid-derived suppressor cells
.
Commun Biol
2021
;
4
:
859
.
32.
Wang
Y
,
Schafer
CC
,
Hough
KP
,
Tousif
S
,
Duncan
SR
,
Kearney
JF
, et al
.
Myeloid-derived suppressor cells impair B cell responses in lung cancer through IL-7 and STAT5
.
J Immunol
2018
;
201
:
278
95
.
33.
Lucca
LE
,
Dominguez-Villar
M
.
Modulation of regulatory T cell function and stability by co-inhibitory receptors
.
Nat Rev Immunol
2020
;
20
:
680
93
.
34.
Hoechst
B
,
Ormandy
LA
,
Ballmaier
M
,
Lehner
F
,
Krüger
C
,
Manns
MP
, et al
.
A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells
.
Gastroenterology
2008
;
135
:
234
43
.
35.
Serafini
P
,
Mgebroff
S
,
Noonan
K
,
Borrello
I
.
Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells
.
Cancer Res
2008
;
68
:
5439
49
.
36.
Terme
M
,
Pernot
S
,
Marcheteau
E
,
Sandoval
F
,
Benhamouda
N
,
Colussi
O
, et al
.
VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer
.
Cancer Res
2013
;
73
:
539
49
.
37.
Wu
X
,
Zhu
D
,
Tian
J
,
Tang
X
,
Guo
H
,
Ma
J
, et al
.
Granulocytic myeloid-derived suppressor cell exosomal prostaglandin E2 ameliorates collagen-induced arthritis by enhancing IL-10(+) B cells
.
Front Immunol
2020
;
11
:
588500
.
38.
Vitale
I
,
Manic
G
,
Coussens
LM
,
Kroemer
G
,
Galluzzi
L
.
Macrophages and metabolism in the tumor microenvironment
.
Cell Metab
2019
;
30
:
36
50
.
39.
Sinha
P
,
Clements
VK
,
Bunt
SK
,
Albelda
SM
,
Ostrand-Rosenberg
S
.
Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response
.
J Immunol
2007
;
179
:
977
83
.
40.
Noman
MZ
,
Desantis
G
,
Janji
B
,
Hasmim
M
,
Karray
S
,
Dessen
P
, et al
.
PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation
.
J Exp Med
2014
;
211
:
781
90
.
41.
Green
KA
,
Cook
WJ
,
Green
WR
.
Myeloid-derived suppressor cells in murine retrovirus-induced AIDS inhibit T- and B-cell responses in vitro that are used to define the immunodeficiency
.
J Virol
2013
;
87
:
2058
71
.
42.
Lee-Chang
C
,
Rashidi
A
,
Miska
J
,
Zhang
P
,
Pituch
KC
,
Hou
D
, et al
.
Myeloid-derived suppressive cells promote B cell-mediated immunosuppression via transfer of PD-L1 in glioblastoma
.
Cancer Immunol Res
2019
;
7
:
1928
43
.
43.
Sinha
P
,
Clements
VK
,
Fulton
AM
,
Ostrand-Rosenberg
S
.
Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells
.
Cancer Res
2007
;
67
:
4507
13
.
44.
Jayaraman
P
,
Parikh
F
,
Lopez-Rivera
E
,
Hailemichael
Y
,
Clark
A
,
Ma
G
, et al
.
Tumor-expressed inducible nitric oxide synthase controls induction of functional myeloid-derived suppressor cells through modulation of vascular endothelial growth factor release
.
J Immunol
2012
;
188
:
5365
76
.
45.
Wang
Y
,
Ding
Y
,
Guo
N
,
Wang
S
.
MDSCs: key criminals of tumor pre-metastatic niche formation
.
Front Immunol
2019
;
10
:
172
.
46.
Liu
C
,
Qiang
J
,
Deng
Q
,
Xia
J
,
Deng
L
,
Zhou
L
, et al
.
ALDH1A1 activity in tumor-initiating cells remodels myeloid-derived suppressor cells to promote breast cancer progression
.
Cancer Res
2021
;
81
:
5919
34
.
47.
Yang
X
,
Lu
Y
,
Hang
J
,
Zhang
J
,
Zhang
T
,
Huo
Y
, et al
.
Lactate-modulated immunosuppression of myeloid-derived suppressor cells contributes to the radioresistance of pancreatic cancer
.
Cancer Immunol Res
2020
;
8
:
1440
51
.
48.
Bayik
D
,
Lathia
JD
.
Cancer stem cell-immune cell crosstalk in tumour progression
.
Nat Rev Cancer
2021
;
21
:
526
36
.
49.
Telarovic
I
,
Wenger
RH
,
Pruschy
M
.
Interfering with tumor hypoxia for radiotherapy optimization
.
J Exp Clin Cancer Res
2021
;
40
:
197
.
50.
Deng
J
,
Li
J
,
Sarde
A
,
Lines
JL
,
Lee
YC
,
Qian
DC
, et al
.
Hypoxia-induced VISTA promotes the suppressive function of myeloid-derived suppressor cells in the tumor microenvironment
.
Cancer Immunol Res
2019
;
7
:
1079
90
.
51.
Ohl
K
,
Tenbrock
K
.
Reactive oxygen species as regulators of MDSC-mediated immune suppression
.
Front Immunol
2018
;
9
:
2499
.
52.
De Ruysscher
D
,
Niedermann
G
,
Burnet
NG
,
Siva
S
,
Lee
AWM
,
Hegi-Johnson
F
.
Radiotherapy toxicity
.
Nat Rev Dis Primers
2019
;
5
:
13
.
53.
Pilié
PG
,
Tang
C
,
Mills
GB
,
Yap
TA
.
State-of-the-art strategies for targeting the DNA damage response in cancer
.
Nat Rev Clin Oncol
2019
;
16
:
81
104
.
54.
Scully
R
,
Panday
A
,
Elango
R
,
Willis
NA
.
DNA double-strand break repair-pathway choice in somatic mammalian cells
.
Nat Rev Mol Cell Biol
2019
;
20
:
698
714
.
55.
Petroni
G
,
Cantley
LC
,
Santambrogio
L
,
Formenti
SC
,
Galluzzi
L
.
Radiotherapy as a tool to elicit clinically actionable signalling pathways in cancer
.
Nat Rev Clin Oncol
2022
;
19
:
114
31
.
56.
Petroni
G
,
Buqué
A
,
Yamazaki
T
,
Bloy
N
,
Liberto
MD
,
Chen-Kiang
S
, et al
.
Radiotherapy delivered before CDK4/6 inhibitors mediates superior therapeutic effects in ER(+) breast cancer
.
Clin Cancer Res
2021
;
27
:
1855
63
.
57.
Petroni
G
,
Galluzzi
L
.
Impact of treatment schedule on the efficacy of cytostatic and immunostimulatory agents
.
Oncoimmunology
2021
;
10
:
1889101
.
58.
Zebertavage
LK
,
Alice
A
,
Crittenden
MR
,
Gough
MJ
.
Transcriptional upregulation of NLRC5 by radiation drives STING- and interferon-independent MHC-I expression on cancer cells and T cell cytotoxicity
.
Sci Rep
2020
;
10
:
7376
.
59.
Lhuillier
C
,
Rudqvist
NP
,
Yamazaki
T
,
Zhang
T
,
Charpentier
M
,
Galluzzi
L
, et al
.
Radiotherapy-exposed CD8+ and CD4+ neoantigens enhance tumor control
.
J Clin Invest
2021
;
131
:
e138740
.
60.
Galluzzi
L
,
Vitale
I
,
Warren
S
,
Adjemian
S
,
Agostinis
P
,
Martinez
AB
, et al
.
Consensus guidelines for the definition, detection and interpretation of immunogenic cell death
.
J Immunother Cancer
2020
;
8
:
e000337
.
61.
Fucikova
J
,
Spisek
R
,
Kroemer
G
,
Galluzzi
L
.
Calreticulin and cancer
.
Cell Res
2021
;
31
:
5
16
.
62.
Kepp
O
,
Bezu
L
,
Yamazaki
T
,
Di Virgilio
F
,
Smyth
MJ
,
Kroemer
G
, et al
.
ATP and cancer immunosurveillance
.
EMBO J
2021
;
40
:
e108130
.
63.
Formenti
SC
,
Rudqvist
NP
,
Golden
E
,
Cooper
B
,
Wennerberg
E
,
Lhuillier
C
, et al
.
Radiotherapy induces responses of lung cancer to CTLA-4 blockade
.
Nat Med
2018
;
24
:
1845
51
.
64.
Yamazaki
T
,
Kirchmair
A
,
Sato
A
,
Buqué
A
,
Rybstein
M
,
Petroni
G
, et al
.
Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy
.
Nat Immunol
2020
;
21
:
1160
71
.
65.
DeSelm
C
,
Palomba
ML
,
Yahalom
J
,
Hamieh
M
,
Eyquem
J
,
Rajasekhar
VK
, et al
.
Low-dose radiation conditioning enables CAR T cells to mitigate antigen escape
.
Mol Ther
2018
;
26
:
2542
52
.
66.
Vanpouille-Box
C
,
Diamond
JM
,
Pilones
KA
,
Zavadil
J
,
Babb
JS
,
Formenti
SC
, et al
.
TGFβ is a master regulator of radiation therapy-induced antitumor immunity
.
Cancer Res
2015
;
75
:
2232
42
.
67.
De Martino
M
,
Daviaud
C
,
Diamond
JM
,
Kraynak
J
,
Alard
A
,
Formenti
SC
, et al
.
Activin a promotes regulatory T-cell-mediated immunosuppression in irradiated breast cancer
.
Cancer Immunol Res
2021
;
9
:
89
102
.
68.
Wu
CT
,
Chen
MF
,
Chen
WC
,
Hsieh
CC
.
The role of IL-6 in the radiation response of prostate cancer
.
Radiat Oncol
2013
;
8
:
159
.
69.
Tao
Z
,
McCall
NS
,
Wiedemann
N
,
Vuagniaux
G
,
Yuan
Z
,
Lu
B
.
SMAC mimetic debio 1143 and ablative radiation therapy synergize to enhance antitumor immunity against lung cancer
.
Clin Cancer Res
2019
;
25
:
1113
24
.
70.
Lan
Y
,
Moustafa
M
,
Knoll
M
,
Xu
C
,
Furkel
J
,
Lazorchak
A
, et al
.
Simultaneous targeting of TGF-β/PD-L1 synergizes with radiotherapy by reprogramming the tumor microenvironment to overcome immune evasion
.
Cancer Cell
2021
;
39
:
1388
403
.
71.
Altorki
NK
,
McGraw
TE
,
Borczuk
AC
,
Saxena
A
,
Port
JL
,
Stiles
BM
, et al
.
Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial
.
Lancet Oncol
2021
;
22
:
824
35
.
72.
Yamazaki
T
,
Wennerberg
E
,
Hensler
M
,
Buqué
A
,
Kraynak
J
,
Fucikova
J
, et al
.
LTX-315-enabled, radiotherapy-boosted immunotherapeutic control of breast cancer by NK cells
.
Oncoimmunology
2021
;
10
:
1962592
.
73.
Almahariq
MF
,
Quinn
TJ
,
Kesarwani
P
,
Kant
S
,
Miller
CR
,
Chinnaiyan
P
.
Inhibition of colony-stimulating factor-1 receptor enhances the efficacy of radiotherapy and reduces immune suppression in glioblastoma
.
In Vivo
2021
;
35
:
119
29
.
74.
Riva
M
,
Wouters
R
,
Nittner
D
,
Ceuster
J
,
Sterpin
E
,
Giovannoni
R
, et al
.
Radiation dose-escalation and dose-fractionation modulate the immune microenvironment, cancer stem cells and vasculature in experimental high-grade gliomas
.
J Neurosurg Sci
2020
.
doi: 10.23736/s0390-5616.20.05060-2
.
75.
Taylor
CT
,
Colgan
SP
.
Regulation of immunity and inflammation by hypoxia in immunological niches
.
Nat Rev Immunol
2017
;
17
:
774
85
.
76.
Davidson
S
,
Coles
M
,
Thomas
T
,
Kollias
G
,
Ludewig
B
,
Turley
S
, et al
.
Fibroblasts as immune regulators in infection, inflammation and cancer
.
Nat Rev Immunol
2021
;
21
:
704
17
.
77.
Ziani
L
,
Buart
S
,
Chouaib
S
,
Thiery
J
.
Hypoxia increases melanoma-associated fibroblasts immunosuppressive potential and inhibitory effect on T cell-mediated cytotoxicity
.
Oncoimmunology
2021
;
10
:
1950953
.
78.
Sofia Vala
I
,
Martins
LR
,
Imaizumi
N
,
Nunes
RJ
,
Rino
J
,
Kuonen
F
, et al
.
Low doses of ionizing radiation promote tumor growth and metastasis by enhancing angiogenesis
.
PLoS One
2010
;
5
:
e11222
.
79.
Mirjolet
C
,
Nicol
A
,
Limagne
E
,
Mura
C
,
Richard
C
,
Morgand
V
, et al
.
Impact of proton therapy on antitumor immune response
.
Sci Rep
2021
;
11
:
13444
.
80.
Marcus
D
,
Lieverse
RIY
,
Klein
C
,
Abdollahi
A
,
Lambin
P
,
Dubois
LJ
, et al
.
Charged particle and conventional radiotherapy: current implications as partner for immunotherapy
.
Cancers
2021
;
13
:
1468
.
81.
Huang
Y
,
Dong
Y
,
Zhao
J
,
Zhang
L
,
Kong
L
,
Lu
JJ
.
Comparison of the effects of photon, proton and carbon-ion radiation on the ecto-calreticulin exposure in various tumor cell lines
.
Ann Transl Med
2019
;
7
:
542
.
82.
De Martino
M
,
Vanpouille-Box
C
,
Galluzzi
L
.
Immunological barriers to immunotherapy in primary and metastatic breast cancer
.
EMBO Mol Med
2021
;
13
:
e14393
.
83.
Galon
J
,
Bruni
D
.
Approaches to treat immune hot, altered and cold tumours with combination immunotherapies
.
Nat Rev Drug Discov
2019
;
18
:
197
218
.
84.
Vitale
I
,
Shema
E
,
Loi
S
,
Galluzzi
L
.
Intratumoral heterogeneity in cancer progression and response to immunotherapy
.
Nat Med
2021
;
27
:
212
24
.
85.
Dewhirst
MW
,
Cao
Y
,
Moeller
B
.
Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response
.
Nat Rev Cancer
2008
;
8
:
425
37
.
86.
Shiao
SL
,
Kershaw
KM
,
Limon
JJ
,
You
S
,
Yoon
J
,
Ko
EY
, et al
.
Commensal bacteria and fungi differentially regulate tumor responses to radiation therapy
.
Cancer Cell
2021
;
39
:
1202
13
.
87.
Wu
CT
,
Huang
YC
,
Chen
WC
,
Chen
MF
.
Effect of 1α,25-Dihydroxyvitamin D3 on the radiation response in prostate cancer: association with IL-6 signaling
.
Front Oncol
2021
;
11
:
619365
.
88.
Fu
SY
,
Chen
FH
,
Wang
CC
,
Yu
CF
,
Chiang
CS
,
Hong
JH
.
Role of myeloid-derived suppressor cells in high-dose-irradiated TRAMP-C1 tumors: a therapeutic target and an index for assessing tumor microenvironment
.
Int J Radiat Oncol Biol Phys
2021
;
109
:
1547
58
.
89.
Wu
CT
,
Chen
WC
,
Chen
MF
.
The response of prostate cancer to androgen deprivation and irradiation due to immune modulation
.
Cancers
2018
;
11
:
20
.
90.
Haughey
CM
,
Mukherjee
D
,
Steele
RE
,
Popple
A
,
Dura-Perez
L
,
Pickard
A
, et al
.
Investigating radiotherapy response in a novel syngeneic model of prostate cancer
.
Cancers
2020
;
12
:
2804
.
91.
Lin
L
,
Kane
N
,
Kobayashi
N
,
Kono
EA
,
Yamashiro
JM
,
Nickols
NG
, et al
.
High-dose per fraction radiotherapy induces both antitumor immunity and immunosuppressive responses in prostate tumors
.
Clin Cancer Res
2021
;
27
:
1505
15
.
92.
Xu
J
,
Escamilla
J
,
Mok
S
,
David
J
,
Priceman
S
,
West
B
, et al
.
CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer
.
Cancer Res
2013
;
73
:
2782
94
.
93.
Lee
SE
,
Lim
JY
,
Kim
TW
,
Ryu
DB
,
Park
SS
,
Jeon
YW
, et al
.
Different role of circulating myeloid-derived suppressor cells in patients with multiple myeloma undergoing autologous stem cell transplantation
.
J Immunother Cancer
2019
;
7
:
35
.
94.
Oweida
AJ
,
Mueller
AC
,
Piper
M
,
Milner
D
,
Van Court
B
,
Bhatia
S
, et al
.
Response to radiotherapy in pancreatic ductal adenocarcinoma is enhanced by inhibition of myeloid-derived suppressor cells using STAT3 anti-sense oligonucleotide
.
Cancer Immunol Immunother
2021
;
70
:
989
1000
.
95.
Chen
J
,
Wang
Z
,
Ding
Y
,
Huang
F
,
Huang
W
,
Lan
R
, et al
.
Hypofractionated irradiation suppressed the off-target mouse hepatocarcinoma growth by inhibiting myeloid-derived suppressor cell-mediated immune suppression
.
Front Oncol
2020
;
10
:
4
.
96.
Zhang
H
,
Yue
J
,
Jiang
Z
,
Zhou
R
,
Xie
R
,
Xu
Y
, et al
.
CAF-secreted CXCL1 conferred radioresistance by regulating DNA damage response in a ROS-dependent manner in esophageal squamous cell carcinoma
.
Cell Death Dis
2017
;
8
:
e2790
.
97.
Li
A
,
Barsoumian
HB
,
Schoenhals
JE
,
Caetano
MS
,
Wang
X
,
Menon
H
, et al
.
IDO1 inhibition overcomes radiation-induced “rebound immune suppression” by Reducing Numbers of IDO1-expressing myeloid-derived suppressor cells in the tumor microenvironment
.
Int J Radiat Oncol Biol Phys
2019
;
104
:
903
12
.
98.
Zhang Md
J
,
Zhang Md
L
,
Yang Md
Y
,
Liu Md
Q
,
Ma Md
H
,
Huang Md
A
, et al
.
Polymorphonuclear-MDSCs facilitate tumor regrowth after radiation by suppressing CD8(+) T cells
.
Int J Radiat Oncol Biol Phys
2021
;
109
:
1533
46
.
99.
Liang
H
,
Shen
X
.
LXR activation radiosensitizes non-small cell lung cancer by restricting myeloid-derived suppressor cells
.
Biochem Biophys Res Commun
2020
;
528
:
330
5
.
100.
Gong
X
,
Li
X
,
Jiang
T
,
Xie
H
,
Zhu
Z
,
Zhou
F
, et al
.
Combined radiotherapy and anti-PD-L1 antibody synergistically enhances antitumor effect in non-small cell lung cancer
.
J Thorac Oncol
2017
;
12
:
1085
97
.
101.
Chen
JL
,
Pan
CK
,
Huang
YS
,
Tsai
CY
,
Wang
CW
,
Lin
YL
, et al
.
Evaluation of antitumor immunity by a combination treatment of high-dose irradiation, anti-PDL1, and anti-angiogenic therapy in murine lung tumors
.
Cancer Immunol Immunother
2021
;
70
:
391
404
.
102.
Liang
H
,
Deng
L
,
Hou
Y
,
Meng
X
,
Huang
X
,
Rao
E
, et al
.
Host STING-dependent MDSC mobilization drives extrinsic radiation resistance
.
Nat Commun
2017
;
8
:
1736
.
103.
Pilones
KA
,
Hensler
M
,
Daviaud
C
,
Kraynak
J
,
Fucikova
J
,
Galluzzi
L
, et al
.
Converging focal radiation and immunotherapy in a preclinical model of triple negative breast cancer: contribution of VISTA blockade
.
Oncoimmunology
2020
;
9
:
1830524
.
104.
Han
MG
,
Jang
BS
,
Kang
MH
,
Na
D
,
Kim
IA
.
PI3Kγδ inhibitor plus radiation enhances the antitumour immune effect of PD-1 blockade in syngenic murine breast cancer and humanised patient-derived xenograft model
.
Eur J Cancer
2021
;
157
:
450
63
.
105.
Lan
J
,
Li
R
,
Yin
LM
,
Deng
L
,
Gui
J
,
Chen
BQ
, et al
.
Targeting myeloid-derived suppressor cells and programmed death ligand 1 confers therapeutic advantage of ablative hypofractionated radiation therapy compared with conventional fractionated radiation therapy
.
Int J Radiat Oncol Biol Phys
2018
;
101
:
74
87
.
106.
Wu
CJ
,
Tsai
YT
,
Lee
IJ
,
Wu
PY
,
Lu
LS
,
Tsao
WS
, et al
.
Combination of radiation and interleukin 12 eradicates large orthotopic hepatocellular carcinoma through immunomodulation of tumor microenvironment
.
Oncoimmunology
2018
;
7
:
e1477459
.
107.
Wu
C
,
Muroski
ME
,
Miska
J
,
Lee-Chang
C
,
Shen
Y
,
Rashidi
A
, et al
.
Repolarization of myeloid derived suppressor cells via magnetic nanoparticles to promote radiotherapy for glioma treatment
.
Nanomedicine
2019
;
16
:
126
37
.
108.
Riva
M
,
Wouters
R
,
Sterpin
E
,
Giovannoni
R
,
Boon
L
,
Himmelreich
U
, et al
.
Radiotherapy, temozolomide, and antiprogrammed cell death protein 1 treatments modulate the immune microenvironment in experimental high-grade glioma
.
Neurosurgery
2021
;
88
:
E205
e15
.
109.
Zhou
H
,
Yang
P
,
Li
H
,
Zhang
L
,
Li
J
,
Zhang
T
, et al
.
Carbon ion radiotherapy boosts anti-tumour immune responses by inhibiting myeloid-derived suppressor cells in melanoma-bearing mice
.
Cell Death Discov
2021
;
7
:
332
.
110.
Filatenkov
A
,
Baker
J
,
Mueller
AM
,
Kenkel
J
,
Ahn
GO
,
Dutt
S
, et al
.
Ablative tumor radiation can change the tumor immune cell microenvironment to induce durable complete remissions
.
Clin Cancer Res
2015
;
21
:
3727
39
.
111.
Deng
L
,
Liang
H
,
Burnette
B
,
Beckett
M
,
Darga
T
,
Weichselbaum
RR
, et al
.
Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice
.
J Clin Invest
2014
;
124
:
687
95
.
112.
Grapin
M
,
Richard
C
,
Limagne
E
,
Boidot
R
,
Morgand
V
,
Bertaut
A
, et al
.
Optimized fractionated radiotherapy with anti-PD-L1 and anti-TIGIT: a promising new combination
.
J Immunother Cancer
2019
;
7
:
160
.
113.
Navarro-Martín
A
,
Galiana
IL
,
Berenguer Frances
MA
,
Cacicedo
J
,
Cañas Cortés
R
,
Comas Anton
S
, et al
.
Preliminary study of the effect of stereotactic body radiotherapy (SBRT) on the immune system in lung cancer patients unfit for surgery: immunophenotyping analysis
.
Int J Mol Sci
2018
;
19
:
3963
.
114.
Mandruzzato
S
,
Brandau
S
,
Britten
CM
,
Bronte
V
,
Damuzzo
V
,
Gouttefangeas
C
, et al
.
Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: results from an interim study
.
Cancer Immunol Immunother
2016
;
65
:
161
9
.
115.
Talebian Yazdi
M
,
Schinkelshoek
MS
,
Loof
NM
,
Taube
C
,
Hiemstra
PS
,
Welters
MJ
, et al
.
Standard radiotherapy but not chemotherapy impairs systemic immunity in non-small cell lung cancer
.
Oncoimmunology
2016
;
5
:
e1255393
.
116.
Boustani
J
,
Joseph
ELM
,
Martin
E
,
Benhmida
S
,
Lecoester
B
,
Tochet
F
, et al
.
Cisplatin-based chemoradiation decreases telomerase-specific CD4 TH1 response but increases immune suppressive cells in peripheral blood
.
BMC Immunol
2021
;
22
:
38
.
117.
Spehner
L
,
Kim
S
,
Vienot
A
,
François
E
,
Buecher
B
,
Adotevi
O
, et al
.
Anti-telomerase CD4(+) Th1 immunity and monocytic-myeloid-derived-suppressor cells are associated with long-term efficacy achieved by docetaxel, cisplatin, and 5-fluorouracil (DCF) in advanced anal squamous cell carcinoma: translational study of epitopes-HPV01 and 02 trials
.
Int J Mol Sci
2020
;
21
:
6838
.
118.
Tsai
MS
,
Chen
WC
,
Lai
CH
,
Chen
YY
,
Chen
MF
.
Epigenetic therapy regulates the expression of ALDH1 and immunologic response: relevance to the prognosis of oral cancer
.
Oral Oncol
2017
;
73
:
88
96
.
119.
Tsai
MS
,
Chen
WC
,
Lu
CH
,
Chen
MF
.
The prognosis of head and neck squamous cell carcinoma related to immunosuppressive tumor microenvironment regulated by IL-6 signaling
.
Oral Oncol
2019
;
91
:
47
55
.
120.
Sridharan
V
,
Margalit
DN
,
Lynch
SA
,
Severgnini
M
,
Zhou
J
,
Chau
NG
, et al
.
Definitive chemoradiation alters the immunologic landscape and immune checkpoints in head and neck cancer
.
Br J Cancer
2016
;
115
:
252
60
.
121.
Sampath
S
,
Won
H
,
Massarelli
E
,
Li
M
,
Frankel
P
,
Vora
N
, et al
.
Combined modality radiation therapy promotes tolerogenic myeloid cell populations and STAT3-related gene expression in head and neck cancer patients
.
Oncotarget
2018
;
9
:
11279
90
.
122.
van Meir
H
,
Nout
RA
,
Welters
MJ
,
Loof
NM
,
de Kam
ML
,
van Ham
JJ
, et al
.
Impact of (chemo)radiotherapy on immune cell composition and function in cervical cancer patients
.
Oncoimmunology
2017
;
6
:
e1267095
.
123.
Teng
F
,
Meng
X
,
Kong
L
,
Mu
D
,
Zhu
H
,
Liu
S
, et al
.
Tumor-infiltrating lymphocytes, forkhead box P3, programmed death ligand-1, and cytotoxic T lymphocyte-associated antigen-4 expressions before and after neoadjuvant chemoradiation in rectal cancer
.
Transl Res
2015
;
166
:
721
32
.
124.
Napolitano
M
,
D'Alterio
C
,
Cardone
E
,
Trotta
AM
,
Pecori
B
,
Rega
D
, et al
.
Peripheral myeloid-derived suppressor and T regulatory PD-1 positive cells predict response to neoadjuvant short-course radiotherapy in rectal cancer patients
.
Oncotarget
2015
;
6
:
8261
70
.
125.
Wang
D
,
An
G
,
Xie
S
,
Yao
Y
,
Feng
G
.
The clinical and prognostic significance of CD14(+)HLA-DR(-/low) myeloid-derived suppressor cells in hepatocellular carcinoma patients receiving radiotherapy
.
Tumour Biol
2016
;
37
:
10427
33
.
126.
Postow
MA
,
Callahan
MK
,
Barker
CA
,
Yamada
Y
,
Yuan
J
,
Kitano
S
, et al
.
Immunologic correlates of the abscopal effect in a patient with melanoma
.
N Engl J Med
2012
;
366
:
925
31
.
127.
Choy
EH
,
De Benedetti
F
,
Takeuchi
T
,
Hashizume
M
,
John
MR
,
Kishimoto
T
.
Translating IL-6 biology into effective treatments
.
Nat Rev Rheumatol
2020
;
16
:
335
45
.
128.
Janku
F
,
Yap
TA
,
Meric-Bernstam
F
.
Targeting the PI3K pathway in cancer: are we making headway?
Nat Rev Clin Oncol
2018
;
15
:
273
91
.
129.
Petroni
G
,
Buqué
A
,
Zitvogel
L
,
Kroemer
G
,
Galluzzi
L
.
Immunomodulation by targeted anticancer agents
.
Cancer Cell
2021
;
39
:
310
45
.
130.
Galluzzi
L
,
Humeau
J
,
Buqué
A
,
Zitvogel
L
,
Kroemer
G
.
Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors
.
Nat Rev Clin Oncol
2020
;
17
:
725
41
.
131.
Serafini
P
,
Meckel
K
,
Kelso
M
,
Noonan
K
,
Califano
J
,
Koch
W
, et al
.
Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function
.
J Exp Med
2006
;
203
:
2691
702
.
132.
Sharma
P
,
Siddiqui
BA
,
Anandhan
S
,
Yadav
SS
,
Subudhi
SK
,
Gao
J
, et al
.
The next decade of immune checkpoint therapy
.
Cancer Discov
2021
;
11
:
838
57
.
133.
Platten
M
,
Nollen
EAA
,
Röhrig
UF
,
Fallarino
F
,
Opitz
CA
.
Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond
.
Nat Rev Drug Discov
2019
;
18
:
379
401
.
134.
Masuda
T
,
Noda
M
,
Kogawa
T
,
Kitagawa
D
,
Hayashi
N
,
Jomori
T
, et al
.
Phase I dose-escalation trial to repurpose propagermanium, an oral CCL2 inhibitor, in patients with breast cancer
.
Cancer Sci
2020
;
111
:
924
31
.
135.
Grassberger
C
,
Ellsworth
SG
,
Wilks
MQ
,
Keane
FK
,
Loeffler
JS
.
Assessing the interactions between radiotherapy and antitumour immunity
.
Nat Rev Clin Oncol
2019
;
16
:
729
45
.
136.
Colman
H
,
Raizer
JJ
,
Walbert
T
,
Plotkin
SR
,
Chamberlain
MC
,
Wong
ET
, et al
.
Phase 1b/2 study of pexidartinib (PEX) in combination with radiation therapy (XRT) and temozolomide (TMZ) in newly diagnosed glioblastoma
.
J Clin Oncol
2018
;
36
:
2015
.
137.
Buque
A
,
Galluzzi
L
.
Modeling tumor immunology and immunotherapy in mice
.
Trends Cancer
2018
;
4
:
599
601
.