Next Article in Journal
Human Epidermal Growth Factor Receptor-3 Expression Is Regulated at Transcriptional Level in Breast Cancer Settings by Junctional Adhesion Molecule-A via a Pathway Involving Beta-Catenin and FOXA1
Next Article in Special Issue
Immunosuppressive Cell Subsets and Factors in Myeloid Leukemias
Previous Article in Journal
Quantitative FDG PET Assessment for Oncology Therapy
Previous Article in Special Issue
Hydrogen-Peroxide Synthesis and LDL-Uptake Controls Immunosuppressive Properties in Monocyte-Derived Dendritic Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor Immune Evasion Induced by Dysregulation of Erythroid Progenitor Cells Development

by
Tomasz M. Grzywa
1,2,3,
Magdalena Justyniarska
1,
Dominika Nowis
3,* and
Jakub Golab
1,*
1
Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
2
Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
3
Laboratory of Experimental Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(4), 870; https://doi.org/10.3390/cancers13040870
Submission received: 20 January 2021 / Revised: 13 February 2021 / Accepted: 15 February 2021 / Published: 19 February 2021
(This article belongs to the Special Issue Mechanisms of Tumor Immune Evasion)

Abstract

:

Simple Summary

Tumor immune evasion is one of the hallmarks of tumor progression that enables tumor growth despite the activity of the host immune system. It is mediated by various types of cells. Recently, immature red blood cells called erythroid progenitor cells (EPCs) were identified as regulators of the immune response in cancer. EPCs expand in cancer as a result of dysregulated erythropoiesis and potently suppress the immune response. Thus, targeting dysregulated EPC differentiation appears to be a promising therapeutic strategy.

Abstract

Cancer cells harness normal cells to facilitate tumor growth and metastasis. Within this complex network of interactions, the establishment and maintenance of immune evasion mechanisms are crucial for cancer progression. The escape from the immune surveillance results from multiple independent mechanisms. Recent studies revealed that besides well-described myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs) or regulatory T-cells (Tregs), erythroid progenitor cells (EPCs) play an important role in the regulation of immune response and tumor progression. EPCs are immature erythroid cells that differentiate into oxygen-transporting red blood cells. They expand in the extramedullary sites, including the spleen, as well as infiltrate tumors. EPCs in cancer produce reactive oxygen species (ROS), transforming growth factor β (TGF-β), interleukin-10 (IL-10) and express programmed death-ligand 1 (PD-L1) and potently suppress T-cells. Thus, EPCs regulate antitumor, antiviral, and antimicrobial immunity, leading to immune suppression. Moreover, EPCs promote tumor growth by the secretion of growth factors, including artemin. The expansion of EPCs in cancer is an effect of the dysregulation of erythropoiesis, leading to the differentiation arrest and enrichment of early-stage EPCs. Therefore, anemia treatment, targeting ineffective erythropoiesis, and the promotion of EPC differentiation are promising strategies to reduce cancer-induced immunosuppression and the tumor-promoting effects of EPCs.

Graphical Abstract

1. Introduction

Cancer immunotherapy has strongly changed the therapeutic landscape in clinical oncology, leading to significant improvements in cancer patients survival [1]. However, despite the induction of durable responses in an unprecedented percentage of cancer patients, the majority still do not respond to the treatment and eventually progress to refractory disease. There are several defined causes of immunotherapy resistance, including low tumor mutational burden [2], impaired antigen presentation by the major histocompatibility complex (MHC) proteins [3], loss of interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) pathway genes [4,5], as well as the development of immunosuppressive tumor microenvironment (TME) [6,7].
TME is composed of many types of cells that regulate tumor growth and progression [8]. The role of regulatory T-cells (Tregs) [9], myeloid-derived suppressor cells (MDSCs) [10], tumor-associated macrophages (TAMs) [11], tumor-associated neutrophils (TANs) [12], and cancer-associated fibroblasts (CAFs) [13] in the regulation of anti-tumor immune response has been established by many years of research (Table 1). Recent reports point to another population of cells, i.e., erythroid progenitor cells (EPCs), that regulate local and systemic immunity in cancer. These cells use similar mechanisms to immune cells and are crucial in the regulation of immune response and cancer progression.
In this review, we discuss the role of the dysregulation of erythropoiesis by cancer cells to induce immune evasion and promote cancer progression.

2. Regulation of Erythropoiesis

The differentiation of hematopoietic stem cells (HSCs) to erythroid cells is a stepwise process strictly regulated by multiple intrinsic and extrinsic factors (Table 2), which results in the production of over 2 × 1011 red blood cells (RBCs) per day and allows for the maintenance of erythroid homeostasis [44,45,46,47,48]. This complex net of interactions provides adequate production of RBCs depending on the body’s needs. Insufficient oxygen supply to the peripheral tissues resulting in hypoxia is a key trigger of increased erythropoiesis, which is regulated by the increased production of erythropoietin (EPO) in the kidney peritubular fibroblasts and liver interstitial cells and hepatocytes [49].
HSCs reside in a unique niche that is created and regulated by various cell types, growth factors, and chemokines [69]. The commitment of HSCs to erythroid lineage begins with the differentiation to a multipotent megakaryocyte–erythroid progenitor cell (MEP), followed by a bust-forming unit-erythroid (BFU-E) and colony-forming unit-erythroid (CFU-E). During terminal erythropoiesis, CFU-E differentiates into proerythroblasts, basophilic erythroblasts, polychromatic erythroblasts, and orthochromatic erythroblasts that expel their nuclei and generate reticulocytes [70]. Reticulocytes are released to the circulation, where they mature to RBCs within a few days. In healthy humans, erythroblasts represent about 20–30% of nucleated cells in the bone marrow [71,72].
The first steps of erythropoiesis are regulated by hematopoietic cytokines including stem cell factor (SCF), interleukin 3 (IL-3), insulin-like growth factor 1 (IGF-1), and granulocyte-macrophage colony-stimulating factor (GM-CSF) [73,74,75]. Further erythroid cell differentiation is regulated mainly by EPO [45,76,77]. The impairment of steady-state erythropoiesis triggers stress erythropoiesis that maintains erythroid homeostasis. Stress erythropoiesis is regulated by additional factors including hypoxia, bone morphogenetic protein 4 (BMP4), Hedgehog, glucocorticoids, and peroxisome proliferator-activated receptor α (PPAR-α) [78,79].
Cell lineage specification is regulated through defined transcriptional programs. It is well established that a zinc-finger transcription factor GATA1 is a master transcriptional regulator of differentiation toward erythroid lineage [80]. It is induced at the very early stages of erythropoiesis and is responsible for the regulation of all known erythroid genes [80]. Thus, GATA1 is necessary for erythropoiesis and its lack cannot be compensated as Gata1 knockout mice fail to generate mature RBCs [81]. Therefore, the cleavage of GATA1 is a key mechanism of erythropoiesis regulation. GATA1 is cleaved by caspases, primarily caspase-3, which is activated in the nucleus of terminally differentiating erythroid cells to enable maturation to RBCs [80,82,83]. Nonetheless, the activation of caspases and GATA1 degradation at earlier stages of differentiation induces differentiation arrest and apoptosis. Therefore, GATA1 is protected from degradation in early-stage EPCs by EPO signaling, p19INK4d cyclin-dependent kinase inhibitor, and HSP70 protein chaperone [76,82,84].

3. Erythroid Progenitor Cells as Immune Regulators

EPCs are predominantly erythroblasts and reticulocytes that differentiate into mature RBCs. EPCs are characterized by the expression of transferrin receptor 1 (CD71) and glycophorin A (CD235a) in humans, and CD71 and TER119 in mice [85]. For many years, EPCs were considered to be solely erythrocytes precursors, without any other significant functions in the human body. However, recent studies revealed the importance of the previously neglected role of EPCs.
Immunomodulatory functions of EPCs were described for the first time in neonates, which are characterized by a physiological enrichment of EPCs in extramedullary sites, including the spleen, liver, and peripheral blood [86]. Neonatal EPCs express arginase-2 (ARG2), L-arginine degrading enzyme, and secrete transforming growth factor β (TGF-β), leading to the suppression of cytokine production by myeloid cells [86] and the promotion of T-cell differentiation toward Tregs cells [87]. Despite initial hypotheses that only neonatal EPCs have significant immunoregulatory properties [86], further research expanded our knowledge and revealed that these properties are a general feature of EPCs. The regulation of immune cells by erythroid cells was described for EPCs induced by pregnancy [88], systemic inflammation [89], HIV infection [90], COVID-19 [91], and anemia [92].
EPCs in different conditions modulate immune response via various mechanisms (Table 3). Recent studies also demonstrated that EPCs that expand during cancer progression possess significant immunomodulatory properties and promote tumor growth.

4. The Role of Erythroid Progenitor Cells in Cancer

Cancer progression is associated with the suppression of immune response that enables tumor growth and leads to increased susceptibility to infections in patients with advanced disease [96]. It is caused by the remodeling of the immune cell landscape that impairs not only a local anti-tumor response, but also systemic antibacterial and antiviral immunity [97]. Cancer cells and tumor-associated stromal cells reprogram hematopoiesis and promote the polarization of immune cells toward suppressive phenotypes. In cancer, the spleen is a key organ of extramedullary hematopoiesis and is responsible for the production of suppressive immune cells [98]. It is well established that cancer dysregulates hematopoiesis to generate MDSCs that suppress antitumor response [10,99]. However, during tumor progression, immune cells in the murine spleen are vastly outnumbered by another type of cells, EPCs [41,62]. Moreover, substantial EPC expansion is observed in the peripheral blood and the liver of tumor-bearing mice and cancer patients. EPCs also infiltrate murine and human tumors, and their frequency in TME is much higher than that of MDSCs or Treg cells [41,42,43,62].
Similar to neonatal counterparts, EPCs induced by cancer were found to potently suppress immune response (Figure 1). The proliferation and cytotoxicity of CD8+ T-cells, as well as the proliferation of CD4+ T-cells and TH1 differentiation, are inhibited by tumor-induced murine EPCs [41]. In murine models, the depletion of EPCs with anti-CD71 antibody inhibits tumor growth [43]. Likewise, EPCs isolated from peripheral blood of cancer patients or human tumors potently suppress T-cell proliferation and the production of IFN-γ via paracrine and direct cell-to-cell contact manner [41,42].
Erythropoiesis is a continuous process by which erythroid cells change their characteristics to differentiate into specialized oxygen-transporting RBCs. The transcriptional profile [41,42,100,101,102,103,104,105,106,107] and cell proteome [106,108,109,110,111] substantially change during erythroid maturation. Growing evidence indicates that the role of EPCs in cancer changes with maturation (Table 4). During differentiation, EPCs lose expression of CD45, a pan-leukocyte marker [112]. Therefore, CD45 may be used as a marker of early-stage EPCs [41]. In tumor-bearing mice, CD45+ EPCs constitute over 40% of EPCs and are predominantly responsible for the immunosuppressive effects of EPCs [41]. These early-stage EPCs were found to potently suppress T-cells, in contrast to more mature CD45 erythroid cells [41,62]. In mice, the suppressive capacity of CD45+ EPCs falls between Tregs and MDSCs [41], but in humans, CD45+ EPCs are even more potent immunosuppressors than both Tregs and MDSCs [42].
Transcriptional analysis revealed a close resemblance between CD45+ EPCs and MDSCs and enrichment in the reactive oxygen species (ROS) pathway in CD45+ EPCs [41]. Early stage CD45+ EPCs have upregulated expression of NADPH oxidase (NOX) family members [41,42], crucial ROS-generating NADPH oxidases [113]. As a result, they have increased ROS levels compared to CD45 counterparts [41,42]. Although ROS are required for T-cell activation, excessive ROS levels impair T-cell immunity [114]. Thus, ROS are a well-established mechanism of T-cell suppression by MDSCs [115]. Similarly, EPCs were found to suppress T-cells in a ROS-dependent manner. Apocynin, an NADPH oxidase inhibitor, as well as N-acetylcysteine, an ROS scavenger, diminished the suppressive effects of EPCs [41,42,116]. The infiltration of EPCs to TME probably contributes to the high ROS levels triggering oxidative stress. High ROS levels in TME impair the functions of tumor-infiltrating lymphocytes and dendritic cells, while promoting the recruitment and accumulation of Tregs and MDSCs [117]. However, ROS inhibition did not restore T-cell functions completely [41]. Further studies revealed that CD45+ EPCs induced by cancer use multiple additional immunoregulatory mechanisms, including IL-10, TGF-β, and PD-1/PD-L1 [42,43]. Thus, the immunoregulatory functions of EPCs rely on many mechanisms identified for immunosuppressive cells in TME (Table 1).
It seems that EPCs impair both anti-tumor immunity and systemic immune response to pathogens. In mice, CD45+ EPCs potently inhibit the antigen-specific response of tumor-infiltrating cytotoxic T-cells [41]. The transfer of CD45+ EPCs into tumor-bearing mice accelerated tumor growth [41], confirming the suppression of anti-tumor response by EPCs. Likewise, CD45+ EPCs suppressed proliferation and cytokine production by tumor-infiltrating T-cells from cancer patients [42].
Importantly, the expansion of EPCs in cancer is most remarkable in the spleen (Table 3), which is the largest secondary lymphoid organ involved in the development of systemic immune response to blood-borne antigens [118]. Similarly to neonates that are also characterized by the expansion of EPCs in the spleen [86], adult tumor-bearing mice have increased susceptibility to viral and bacterial infections compared to healthy mice [41]. Ex vivo, EPCs suppressed antigen-specific cytotoxic T-cells [41]. In vivo, the depletion of EPCs with anti-CD71 antibody rescued the suppressed proliferation of virus-specific CD8+ T-cells, restoring anti-viral immunity in tumor-bearing mice while the transfer of CD45+ EPCs potentiated the suppression of immune response [41]. In humans, anemic cancer patients have higher EPC numbers and increased Epstein–Barr viral (EBV) load due to suppressed anti-viral immunity [41]. These latter findings suggest that as in mice, EPCs suppress a systemic immune response in cancer patients.
It was suggested that the suppressive properties of EPCs may be restricted to stress erythropoiesis-induced EPCs. However, CD45+ EPCs isolated from the spleen, liver as well as bone marrow of the tumor-bearing mice suppressed T-cells to a similar extent [41]. Moreover, steady-state EPCs from human bone marrow also suppress T-cells [92]. Nonetheless, there are significant differences in the expression of immunomodulatory molecules, including PD-L1, between EPCs isolated from the bone marrow, spleen, and TME [43]. This suggests that the differences in EPC properties may result from stimulation with some factors, presumably cytokines or TME components, which may enhance or diminish the immunosuppressive properties of EPCs.

Tumor-Promoting Role of CD45 EPCs

The majority of tumor-induced EPCs are CD45 [41,62]. While early-stage CD45+ EPCs potently suppress the immune response, more mature CD45 EPCs lack this capacity (Table 4). However, the transfer of CD45 EPCs also promotes tumor growth and decreases the survival of tumor-bearing mice [62].
These tumor-induced splenic CD45 EPCs were called Ter-Cells [62]. They are a population of late-stage EPCs as they have a high nucleus/cytoplasm ratio, scant cytoplasm, dense chromatin, and few organelles, as well as lacking the expression of the major histocompatibility complex (MHC) class I [62], a marker of mature erythroid cells [119]. In contrast to early-stage EPCs, CD45 EPCs do not influence T-cell proliferation, dendritic cell activation, and cytokine secretion as well as fail to induce Tregs [62]. Moreover, CD45 EPCs have very low or undetectable levels of immune-related mediators, including IL-10, TGF-β, IL-4, prostaglandin E2 (PGE-2), and ROS [41,42,62]. Therefore, CD45 EPCs do not promote tumor growth by inhibiting the anti-tumor response.
Transcriptional analysis revealed marked overexpression of a neurotrophic factor artemin in CD45 EPCs [62]. The physiological role of artemin involves the regulation of neuronal survival, maintenance, and differentiation [120]. Artemin also has protumorigenic activity and promotes cancer cell survival, proliferation, migration, and invasiveness [62,121,122,123]. In murine models, it promotes tumor growth and accelerates disease progression [62]. Artemin activates the glial cell-derived neurotrophic factor (GDNF) family receptor alpha-3 (GFRα3) and its co-receptor RET on cancer cells. Downstream signaling of artemin promotes the phosphorylation of extracellular signal-regulated kinase (ERK), protein kinase B (AKT), and caspase-9, promoting proliferation and invasiveness, while preventing apoptosis in tumor cells, even induced by the therapy [62]. The same effects are exerted by artemin-secreting CD45 EPCs. Thus, the reduction in EPC expansion reduces the increase in the artemin concentration in the serum and decreases tumor growth [62]. Artemin-expressing CD45 EPCs were also detected in the spleens of patients with hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC) [62,123], which suggests their role in cancer patients.
Moreover, these differences are also manifested by their localization. While immunomodulatory early-stage EPCs accumulate in the spleen and intensively infiltrate TME, tumor-promoting late-stage EPCs are detected mainly in the spleen where they secrete artemin into circulation [41,42,62,123]. Collectively, early-stage and late-stage EPCs differ substantially regarding their gene expression profile, level of immunomodulatory mediators, and their role in promoting cancer progression (Table 5).

5. Expansion of Erythroid Progenitor Cells

EPCs predominantly occupy niches in the bone marrow where they differentiate into RBCs. However, EPC frequency in the steady-state bone marrow is relatively low, especially when compared to mature erythrocytes. In healthy individuals, EPCs are not detected in extramedullary sites, besides a small percentage of reticulocytes in peripheral blood [124]. However, under several conditions, EPCs substantially expand in the bone marrow as well as in extramedullary sites.
The expansion of EPCs is physiological in neonates and during pregnancy [88,93,125,126]. In neonates, EPCs accumulate in the extramedullary sites due to insufficient bone marrow erythropoiesis during the first days of life [125,126]. During pregnancy, extramedullary erythropoiesis enables the production of sufficient numbers of erythrocytes [88]. Moreover, the expansion of EPCs is also observed in anemic patients as a mechanism increasing oxygen transport [92,127]. Recent studies revealed that extramedullary erythropoiesis and EPC expansion may also be a part of the inflammatory response [128,129,130]. A recent analysis of blood transcriptome revealed that the signature of immature erythroid cells is also associated with severe respiratory syncytial virus (RSV) infection, pharmacological immunosuppression, and late-stage cancer [131].
In tumor-bearing mice, EPCs expand during tumor progression in many extramedullary organs (Table 6), predominantly the spleen, liver, and peripheral blood, as well as infiltrate tumors [41,43,62]. In humans, EPCs were detected in the spleen, TME, and peripheral blood of cancer patients [41,62,123,131]. In general, anemia severity correlates with the frequency of EPCs [41]. In some cases, the expansion of EPCs in peripheral blood is so substantial that it causes a so-called leukoerythroblastic reaction [132,133].

6. Cancer-Induced Dysregulation of Erythropoiesis

The main cause of EPC expansion is the increase in EPO concentrations in response to anemia. However, the mechanisms of EPC induction by cancer are complex and rely on multiple components that dysregulate erythropoiesis (Figure 2), leading to ineffective erythropoiesis, characterized by erythroid differentiation arrest and increased apoptosis of erythroid cells, and is a feature of various diseases, including β-thalassemia [135]. Importantly, emerging evidence suggests that cancers not only induce potent EPC expansion, but also arrest their development at the earliest stages of differentiation. This leads to the suppression of immune response driven by EPCs, which are potent but physiologically transient immunosuppressors [92].

6.1. Dysregulation of Hematopoietic Stem and Progenitor Cells Differentiation

The dysregulation of erythropoiesis by cancer begins at the first stage of hematopoiesis. Malignant hematological cells suppress hematopoietic stem and progenitor cells (HSPCs) in the bone marrow, limiting their differentiation and inducing a quiescent state. This suppression is mediated by various mechanisms, including SCF [51], TNF-α [136], arginase [137], TGF-β [138,139,140], and stanniocalcin 1 [141]. On the other site, HSPCs are enriched in the extramedullary sites, predominantly the spleen and the circulation of cancer patients, and are myeloid-biased to generate suppressive myeloid cells [142,143,144,145,146]. Cytokines and growth factors secreted by cancer cells and TME force hematopoiesis to the generation and maintenance of immunosuppressive cells that promote tumor growth [144]. Increased numbers of HSPCs in the circulation correlate with advanced tumor stage and decreased progression-free survival in cancer patients [142,146]. In cancer, TNF-α secreted by activated T-cells increases the proliferation of HSPCs and induces emergency myelopoiesis in the bone marrow [143]. Nonetheless, despite strong myeloid polarization, HSPCs in the extramedullary sites, including the spleen, also exhibit increased capacity to differentiate into BFU-E in tumor-bearing mice compared to healthy mice [144].

6.2. Disruption of Hematopoietic Stem and Progenitor Cells Niche

Cancer cells also directly impair HSPCs’ maintenance and differentiation by disrupting their niche, resulting in the loss of quiescence and stemness of HSPCs [51,147]. This phenomenon is the most prominent for hematological malignancies that primarily develop in the bone marrow and outcompete native HSPC niches [148]. Nonetheless, solid tumors were also reported to disrupt the HSPCs’ niche. Melanoma cells secrete vascular endothelial growth factor (VEGF), which reduces available vascular niches in bone marrow, promoting HSC mobilization [149]. Moreover, tumor-secreted exosomes educate bone marrow cells toward a pro-metastatic phenotype [150] and promote the production of pro-inflammatory cytokines by mesenchymal stem cells to support cancer cell growth while suppressing HSPCs [151].

6.3. Suppression of Erythroid Differentiation of Hematopoiesis Stem Cells

The differentiation of HSCs is skewed towards myelopoiesis by cancer. Thus, the potential of erythroid differentiation of HSCs is commonly suppressed, especially in the bone marrow. EPC precursors, MEPs, are the most suppressed progenitor cells in the bone marrow of mice with hematological malignancies [57,58,136,140,152,153,154]. In plasma cell myeloma, malignant cell infiltration correlates negatively with hemoglobin concentration, but not with leukocytes or platelet counts, which suggests the selective impairment of erythropoiesis by malignant cells [155]. This suppression is partially compensated by the increased proliferation of early-stage EPCs in cancer [57]. Moreover, erythroid progenitors are activated in extramedullary sites, including the spleen [154].

6.4. Chronic Erythropoietin Production

EPC expansion is triggered primarily by EPO, which promotes the survival, proliferation, and differentiation of EPCs [156]. EPO induces the expansion of highly proliferative early-stage EPCs [157]. Normally, this response is rapid and EPO concentration quickly decreases after the induction of EPC expansion, which results in RBC generation [158]. However, when the erythropoietic response is insufficient to rescue anemia, EPO is produced constantly. This results in the substantial expansion of early-stage EPCs in the bone marrow as well as in the extramedullary sites. In cancer, EPO is secreted predominantly in response to tissue hypoxia resulting from anemia.
Moreover, tumors may directly trigger the upregulation of EPO production in a vascular endothelial growth factor (VEGF)-dependent mechanism. Vascular endothelial growth factor (VEGF) is a growth factor produced by malignant and stromal cells in TME to induce neovascularization, vessel remodeling [159], and to modulate antitumor immune response [160]. VEGF concentration is substantially increased in the plasma of cancer patients [161]. Importantly, VEGF stimulates EPO secretion by splenic stromal cells expressing platelet-derived growth factor receptor β (PDGFR-β) [162]. Increased VEGF concentration in plasma leads to the increased reticulocyte index in the circulation and expansion of early-stage EPCs in the bone marrow and the spleen [163].

6.5. Induction of Erythroid Cell Apoptosis

Another mechanism of erythropoiesis dysregulation is the direct induction of erythroid cell apoptosis by cancer cells. Apoptosis induced by death receptor Fas (CD95) and Fas ligand (FasL, CD95L, or CD178) interaction is a critical negative regulatory axis of erythropoiesis [76,82]. These negative signals can be overcome by high EPO concentrations that promote EPC expansion during erythropoietic stress response [164].
Cancer cells secrete multiple factors that induce the expression of death receptors on EPCs, increasing their susceptibility to apoptosis [59,63]. Thus, EPCs from cancer patients have significantly upregulated Fas receptor [59,63]. Moreover, ligands for cell death receptors are commonly overexpressed by malignant cells [59,63,165]. The activation of death receptors triggers the activation of caspases that cleave GATA1 transcription factors, leading to the maturation arrest or apoptosis of EPCs [166]. Maturation arrest caused by GATA1 degradation results in the accumulation of EPCs at the earliest stages of differentiation [155,166]. Moreover, GATA1 downregulation decreases the induction of anti-apoptotic proteins, including Bcl-xL and Bcl-2 [167,168]. The enhanced loss of erythroid precursors due to apoptosis leads to compensatory mechanisms and, consequently, higher percentages of early erythroblasts in the bone marrow of cancer patients [59].
Similar to FasL, malignant cells have an increased level of TNF-related apoptosis-inducing ligand (TRAIL) [59,63,169]. EPCs are characterized by the physiological expression of TRAIL receptors [170]. Their stimulation by TRAIL on cancer cells induces differentiation arrest caused by the activation of caspases and the induction of ERK1/ERK2 signaling [82,170,171].

6.6. Transforming Growth Factor β

TGF-β is an important cytokine that promotes tumor growth and immune evasion [172]. Its concentration is substantially increased in the TME and serum of cancer patients [62,140,172,173,174]. Overactivation of the TGF-β pathway affects not only cells in TME, but also hematopoietic cells. Indeed, in cancer patients TGF-β signaling is the most dysregulated signaling pathway in HSPCs, which leads to impaired hematopoiesis, especially erythropoiesis [140]. In erythroid cells, TGF-β potently inhibits proliferation and self-renewal, but at a low concentration it may accelerate the differentiation of late-stage EPCs by promoting mitophagy [83,175,176,177,178].
TGF-β induces the maturation arrest of early-stage EPCs by noncanonical activation of p38, which in turn triggers GATA1 degradation [57,58,140]. Moreover, TGF-β activates SMAD2 and SMAD3 via the type III TGF-β receptor, which is transiently upregulated in early-stage EPCs [178]. Indeed, EPCs in tumor-bearing mice have overactivated SMAD2 and SMAD3 [62]. Accordingly, tumor-induced expansion of EPCs is substantially reduced in Smad3-deficient mice [62]. Moreover, EPC expansion may be prevented by the treatment with neutralizing antibody against TGF-β [62]. The ability to induce EPCs is decreased in mice bearing TGF-β-deficient tumor cells; however, not completely [62]. These findings confirm that TGF-β secreted by tumor cells and also by non-malignant cells in TME is a key factor inducing EPC expansion in cancer via SMAD signaling.
Another mechanism by which TGF-β impairs erythropoiesis involves IL-33, a member of the IL-1 superfamily of cytokines. Tumor-secreted TGF-β induces the expression of IL-33 in TME [179]. Indeed, an increased concentration of IL-33 has been reported in different types of cancer and often correlates with poor prognosis [180]. Notably, IL-33 potently inhibits the differentiation of EPCs at early stages by NF-κB activation and the inhibition of signaling pathways downstream of erythropoietin receptor (EPO-R) [181].
Other members of the TGF-β superfamily, including growth differentiation factor 11 (GDF11, also known as BMP11) and GDF15, have a similar role in the regulation of erythropoiesis. GDF11 induces the differentiation arrest of early-stage EPCs by the activation of SMAD2 and SMAD3 pathways, inhibiting terminal differentiation [182,183,184]. In myelodysplastic syndrome (MDS) patients, GDF11 serum concentration is negatively correlated with late erythropoiesis [185]. Erythropoiesis is also suppressed by GDF15, which modulates iron metabolism [186].
On the other side, some members of the BMP pathway, including BMP4, are crucial regulators of stress erythropoiesis and initiate the differentiation and expansion of EPCs, enabling erythropoietic response [128,187,188].

6.7. Iron Restriction

Iron is an important trace element required for many biological processes, including the heme synthesis [78,189]. Thus, its metabolism is regulated by multiple proteins including iron-transporting transferrin, iron-storing ferritin, and ferroportin responsible for iron export from the cell [190]. Absolute iron deficiency is detected in over 40% of cancer patients [191]. Notably, iron restriction selectively impairs erythroid cell differentiation, but not granulocytic nor megakaryocytic progenitors [61,192,193,194]. Iron is a metabolic checkpoint that restrains the expansion of EPCs triggered by EPO in the case of insufficient iron availability. Iron restriction downregulates the crucial control element of the EPO receptor, Scribble, preventing further EPC maturation [61]. Moreover, iron control of EPC differentiation is mediated by an aconitase-associated regulatory pathway that compromises heme production and modulates EPO signaling [194]. This results in profound changes in the gene expression profile, including the downregulation of GATA1 and its target genes, leading to the impairment of EPC maturation with the differentiation arrest of early EPCs [192,193,194,195].
EPCs can obtain and concentrate iron with exceptional efficacy [196]. Nonetheless, cancer cells and nonmalignant cells in TME are also characterized by increased iron metabolism [197,198]. Cancer cells overexpress CD71 and compete with the EPCs for transferrin-bound iron [199]. Moreover, cells in TME, especially macrophages, accumulate iron, leading to its sequestration from EPCs and exaggerating iron deficiency [165].

6.8. Pro-Inflammatory Cytokine-Driven Erythropoiesis Impairment

Anemia of inflammation (also referred to as anemia of chronic disease) is associated with systemic inflammation, which is one of the hallmarks of cancer and is primarily caused by altered iron distribution [200,201]. Inflammation activates the inflammasome, which triggers enzymatic activation of caspases [202]. Inflammasome assembly in HSPCs leads to the GATA1 cleavage by caspase-1, which favors myelopoiesis over erythropoiesis and suppresses terminal erythropoiesis, leading to the maturation arrest of EPCs [203]. In mice expressing active KrasG12D, the activation of inflammasome leads to myeloproliferation and anemia with a compensatory expansion of EPCs in peripheral blood [204]. In this model, anemia as well as EPC expansion are reduced after pharmacological inflammasome inhibition [204].
Chronic inflammation inhibits the late-stage differentiation of EPCs, leading to the maturation arrest of the early-stage EPCs, which is mediated by various cytokines [205]. One of the critical mediators of inflammation is interferon γ (IFN-γ) [206], which also potently impairs erythropoiesis, leading to anemia [207]. Erythroid cells stimulated with IFN-γ have increased levels of pro-apoptotic caspases, induced differentiation arrest, and triggered apoptosis [208,209]. Moreover, IFN-γ upregulates the expression of Fas on EPCs, increasing their susceptibility to apoptosis in vivo [210]. Additionally, IFN-γ induces the expression of a key regulator of myeloid differentiation, PU.1, in EPCs [207]. During physiological erythropoiesis, the expression of PU.1 is downregulated due to the inhibitory effects on GATA1 functions and erythroid cell differentiation [211,212,213]. Thus, chronic IFN-γ production results in decreased erythropoietic activity in the bone marrow, but increased myelopoietic activity [207]. Moreover, IFN-γ reduces RBC life span and increases macrophage erythrophagocytosis, aggravating anemia and stimulating EPC expansion [207].
Similar suppressive effects on erythropoiesis have been described for another pro-inflammatory cytokine, TNF-α. Cancer patients are characterized by the chronic production of TNF-α, which promotes immune escape and tumor progression [214]. TNF-α induces the maturation arrest of early-stage EPCs and promotes their apoptosis [82,215,216,217,218]. This effect is mediated by the p55 TNF receptor and the activation of caspases [82,215]. TNF-α also upregulates p38 MAPK in EPCs, which phosphorylates acetylated GATA1, promoting its degradation [219,220]. Moreover, TNF-α upregulates PU.1 and GATA2 in HSPCs, which antagonize erythroid cell differentiation [221].
Likewise, the maturation arrest of EPCs at early stages and the inhibition of EPC proliferation are also triggered by other proinflammatory cytokines that are overexpressed in cancer, including IL-1 [222], IL-6 [223] or IL-12 [224].

6.9. Cancer-Secreted Chemokines

Erythropoiesis is also influenced by dysregulated chemokine profiles in the bone marrow plasma and serum of cancer patients. One of these chemokines is CCL3, which is upregulated in the majority of patients with hematopoietic malignancies [57,58] and a subset of patients with solid tumors [225]. CCL3 suppresses erythroid differentiation by p38 activation via CCR1, and this leads to the degradation of GATA1 [57,58,226]. On the other side, chemokines may also promote erythropoiesis by recruiting monocyte-derived macrophages to create erythroblastic islands in the extramedullary sites [227].

6.10. Induction of Extramedullary Stress Erythropoiesis

Suppressed bone marrow steady-state erythropoiesis is a hallmark of inflammation and is caused by the production of pro-inflammatory cytokines and iron sequestration [130,207,228,229]. Suppression steady-state erythropoiesis is often observed in patients with hematological malignancies [136,140,152,153] and solid tumors [230]. As a consequence, stress-erythropoiesis is activated in extramedullary sites to maintain erythroid homeostasis [129]. EPO secreted in response to anemia promotes the formation of erythroblastic islands in the spleen followed by the extensive proliferation of erythroid cells [228,231]. Multiple inflammatory cytokines that suppress erythropoiesis in the bone marrow simultaneously induce stress erythropoiesis. This effect was reported for IFN-γ [207], TNF-α [229], IL-1β [229,232], IL-6 [223], and G-CSF [233]. Notably, extramedullary stress erythropoiesis may be also induced by other factors, including ultraviolet B (UVB) exposure, tumor-promoting environmental stress [43], and chronic stress [234,235], which often accompany cancer.
Importantly, EPCs in extramedullary sites may still exhibit differentiation arrest that results in the enrichment of early-stage EPCs [207]. Thus, early-stage EPC fraction is increased in the spleen of tumor-bearing mice compared with acute anemic mice that also have induced stress erythropoiesis [41].

6.11. Chemotherapy-Induced Impairment of Erythropoiesis

The myelosuppressive effects of chemotherapy are another cause of anemia in cancer [166]. Importantly, early-stage EPCs are especially sensitive to the cytotoxic effects of chemotherapeutic agents, while late-stage EPCs are more resistant [236]. EPC apoptosis triggered by chemotherapy is induced by caspase activation and can be prevented by the SCF-mediated up-regulation of anti-apoptotic proteins Bcl-2 and Bcl-XL [236]. Therefore, it was suggested that SCF may be used in the supportive therapy of chemotherapy-treated cancer patients [237]. This approach may diminish the development of anemia, which would cause the extensive expansion of EPCs after treatment.

7. Modulation of EPCs to Inhibit Their Tumor-Promoting Effects

The development of strategies that modulate the immune response in cancer patients is of great clinical interest. The modulation of immunosuppressive and tumor growth-promoting EPC mechanisms is a promising approach to diminish their negative role. Moreover, treating anemia to prevent EPC expansion as well as targeting ineffective erythropoiesis may causally decrease the tumor-promoting effects of EPCs.

7.1. Modulation of EPCs Immunosuppressive Mechanisms

7.1.1. Reactive Oxygen Species

The production of ROS by EPCs is a key mechanism of immune suppression as ROS scavengers substantially rescue T-cell function suppressed by both murine and human EPCs [41,42,116]. Several antioxidant-based therapies were demonstrated to have potent antitumor effects in preclinical studies [238]. However, further studies revealed that antioxidants may accelerate tumor progression and promote metastasis [239,240]. Therefore, current studies focus on increasing rather than decreasing ROS levels in TME due to increased vulnerability to oxidative stress-induced apoptosis [241]. Indeed, ROS-generating agents or inhibitors of antioxidant systems are efficient in preclinical studies; however, they are without satisfactory results in clinical trials [242]. Thus, more research is required to determine the clinical utility of ROS-based therapies in cancer.

7.1.2. IL-10

IL-10 was considered for many years as a potent anti-inflammatory cytokine. Accordingly, EPCs were found to secrete IL-10, which suppresses T-cells [42]. However, many studies in this field demonstrated that its role in cancer is more complex than initially envisioned [243,244,245,246]. Intriguingly, IL-10-based therapy, including pegylated IL-10 (Pegilodecakin), is much more efficient than therapies neutralizing IL-10 effects [246,247,248].

7.1.3. PD-L1/PD-1 Axis

Targeting immune checkpoints has revolutionized clinical oncology. Monoclonal antibodies targeting PD-L1 or PD-1 reverse the inhibitory signals triggered by the PD-L1/PD-1 axis and enhance antitumor immune response [249,250,251]. PD-L1 is also expressed by murine and human tumor-induced EPCs [43]. Interestingly, the expression of PD-L1 is higher in stress erythropoiesis EPCs compared to steady-state EPCs in the bone marrow, and it reaches the highest levels in tumor-infiltrating EPCs [43]. Although an exact role of the PD-L1/PD-1 axis in the EPC-mediated suppression of immune response was not assessed, it seems that immune checkpoint inhibitors may at least partially diminish their tumor-promoting effects.

7.1.4. TGF-β

The production of TGF-β in the TME is crucial to induce and maintain its immunosuppressive character [172]. TGF-β is produced by various types of cells, including EPCs [42]. The inhibition of SMAD signaling induced by TGF-β rescues T-cell proliferation and the production of IFN-γ suppressed by EPCs [42]. Therefore, modulating TGF-β signaling is a promising strategy to attenuate immune evasion induced by tumor-associated cells, including EPCs. Indeed, several anti-TGF-β-based immunotherapies were shown to be effective in preclinical studies, especially in combination with immune checkpoint inhibitors [252,253,254,255]. Therefore, targeting TGF-β signaling is a promising approach to suppress the tumor-promoting effects of EPCs.

7.2. Anti-Artemin Therapy

Late-stage EPCs promote tumor growth and invasiveness via the secretion of artemin [62,123]. Anti-artemin neutralizing antibody inhibits tumor growth and increases the survival of tumor-bearing mice [62]. Anti-artemin therapy is also currently tested for the treatment of cystitis-induced bladder hyperalgesia [256]. However, the clinical utility of targeting artemin or its signaling as the modulation of tumor growth-promoting effects of EPCs is unknown.

7.3. Treating Anemia to Prevent EPC Expansion

Since the modulation of EPCs’ tumor-promoting mechanisms is rather ineffective, a decrease in EPC expansion and the induction of their differentiation is a promising strategy. The correction of anemia in cancer patients is one of the strategies to prevent EPC expansion. Most anemic patients have iron deficiency (ID) [191]; therefore, the determination of iron status and treatment is recommended for cancer patients according to the guidelines [66,257,258]. Current European Society for Medical Oncology (ESMO) guidelines of anemia management in cancer patients are presented in Figure 3.
Impaired iron status can be diagnosed by total iron-binding capacity (TIBC), transferrin saturation (TSAT), or serum ferritin (SF) levels tests. TSAT enables the determination of iron status available for erythropoiesis, and its low levels together with high SF (>100 ng/mL) suggest functional iron deficiency (FID) [66]. Iron should be supplemented intravenously or orally for patients with low ferritin and without anemia of inflammation (CRP < 5 mg/L) [66].
However, anemia in cancer is not commonly caused by absolute ID, but rather results from iron sequestration (functional ID) [66,200,201]. In this group of patients, iron-replenishing strategies may not be effective. Therefore, targeting iron metabolisms with hepcidin antagonists to modulate the hepcidin-ferroportin axis is a promising treatment option [66,259]. Moreover, several novel therapies are currently under investigation for cancer-associated anemia, including ascorbic acid, androgens, BMP2 and BMP6 antagonists, as well as activin traps [66].

7.4. Targeting Ineffective Erythropoiesis to Decrease EPC Expansion

Modulating EPC expansion by promoting EPC differentiation is a novel therapeutic strategy to diminish the tumor-promoting effects of EPCs (Figure 4).
TGF-β is a key negative regulator of erythropoiesis, which triggers differentiation arrest and promotes the expansion of EPCs [62]. TGF-β inhibitors stimulate EPC differentiation in vitro [178,260]. In murine models, the anti-TGF-β antibody inhibits tumor growth and prevents the expansion of CD45 EPCs [62]. Moreover, SMAD inhibitors rescue T-cell proliferation and IFN-γ production suppressed by EPCs [42]. Therefore, the modulation of TGF-β and SMAD2/3 signaling is a promising approach to promote erythroid cell maturation and to diminish their suppressive effects [261], despite the lack of potent antitumor effects of monotherapy in clinical trials [262].
Myelodysplastic syndromes (MDS) are characterized by the impairment of erythroid cell differentiation with maturation arrest at the early stage [263,264]. In murine MDS models, TGF-β superfamily ligand-trapping fusion protein ACE-536 promotes erythroid maturation by binding GDF11, inhibiting SMAD2/3 signaling, and promoting late-stage erythropoiesis, reducing rescue anemia [182]. Similar effects are exerted by the compound ACE-011, which promotes terminal erythropoiesis and prevents EPC expansion in β-thalassemia [184]. In clinical trials, luspatercept (ACE-536) and sotatercept (ACE-011) reduced the severity of anemia in patients with MDS and β-thalassemia [265,266,267]. Clinical trials of sotatercept in cancer patients showed that it may also be effective for the treatment of chemotherapy-induced anemia [268].
Moreover, it was suggested that targeting BMP signaling may be beneficial for anemia of inflammation [269]. The inhibition of BMP downregulates IL-6 signaling and decreases hepcidin levels, resulting in the restoration of erythropoiesis suppressed by inflammation [270,271].
Caspase-1 activation by inflammasome is one of the mechanisms skewing the differentiation of HSPCs toward myeloid cells [203]. Thus, the inhibition of caspase-1 results in the upregulation of GATA1 and the rescue of inflammation-induced anemia [203]. Moreover, caspase inhibitors trigger the differentiation of EPCs after the induction of maturation arrest by FasL or TNF-α [82]. Several caspase-1 inhibitors are available, including a potent and selective inhibitor, VX-765 [272]. Therefore, it is of great interest to evaluate the effects of caspase inhibitors on EPC expansion and differentiation in cancer.
P38 MAPK is an important pathway regulating erythropoiesis. P38 is activated by multiple inflammatory signals and restrains EPC differentiation by GATA1 degradation [220] and by suppressing the silencing of Bim, which triggers apoptosis [273]. Moreover, p38 functions as an oncogenic kinase with a complex role in cancer [274]. Therefore, p38 inhibition may simultaneously have an antitumor effect and promote EPC maturation. Notably, p38 inhibition enhances EPC maturation in an EPO-independent manner [273], but also decreases the production of endogenous EPO under stress conditions [275], suggesting that p38 inhibitor therapy should probably be combined with ESAs.
In erythroid cells, EPO triggers EPO-R and association with cytoplasmic Janus kinase 2 (JAK2), a crucial signal transducer [276]. The overactivation of JAK2, most commonly caused by V617F mutation, is associated with myeloproliferative neoplasms, including polycythemia vera [277]. Nonetheless, increased JAK2 activity may also be caused by high levels of EPO caused by anemia and chronic hypoxia, a state observed in β-thalassemia and cancer. In β-thalassemia, JAK2 inhibitors decrease ineffective erythropoiesis, prevent the expansion of EPCs, and reduce splenomegaly [278]. JAK2 inhibitors, including ruxolitinib and fedratinib, are approved for the treatment of patients with MPNs [279]. Moreover, targeting JAKs is a promising therapeutic strategy for the treatment of different types of cancer [280]. Whether JAKs inhibitors may decrease the expansion of EPCs in cancer patients remains unknown.
The mechanistic target of rapamycin (mTOR) is a central protein kinase orchestrating cell growth, metabolism, and immune response. Thus, mTOR is widely tested in clinical trials as a target for cancer therapy [281]. Importantly, mTOR inhibition rescues EPC differentiation under ineffective erythropoiesis by inducing the cell cycle exit of early-stage EPCs [282]. Moreover, mTOR inhibition in EPCs may be triggered by Forkhead-box-class-O3 (FoxO3) [282]. The activation of FoxO3 by resveratrol induces early erythroid maturation and decreases their proliferation, resulting in a reduction in ineffective erythropoiesis [283].
Erythropoiesis is also regulated by serotonin (5-HT) [284,285]. Dysregulated tryptophan metabolism with an enhanced kynurenine pathway is common in cancer, and is increasingly being recognized as a viable metabolic pathway regulating immune response [286]. A skew towards the kynurenine pathway leads to decreased serotonin (5-HT) concentrations, resulting in the impaired differentiation and decreased survival of EPCs [284,285]. The upregulation of 5-HT triggered by EPO is crucial to protect EPCs from apoptosis at the CFU-E-to-proerythroblast transition checkpoint [284]. Pharmacological increase in 5-HT with fluoxetine, a selective serotonin reuptake inhibitor (SSRI), rescues anemia [284]. Therefore, targeting the 5-HT axis in EPCs with either SSRI or kynurenine pathway inhibitors may diminish the tumor-promoting role of immature erythroid cells.
Enasidenib is an Food and Drug Administration (FDA)-approved, first-in-class preferential inhibitor of mutated isocitrate dehydrogenase 2 (IDH2) that promotes the differentiation of acute myeloid leukemia blasts [287]. Interestingly, enasidenib was found to act independently of IDH2 on EPCs. Enasidenib potently promotes erythroid differentiation through the modulation of protoporphyrin IX (PPIX) accumulation and hemoglobin production in late-stage EPCs [288]. As a result, increased hemoglobin concentration and RBC transfusion independence were reported for enasidenib-treated patients [287,289].
Some studies suggested that natural compounds may decrease EPC expansion. Dangguibuxue decoction (DGBX), a traditional Chinese medicine, abolishes EPC accumulation, promotes their differentiation, and rescues anemia, leading to the activation of anti-tumor immune response and a decrease in tumor growth [290]. Moreover, a recent study revealed that vitamin C has a critical role in the regulation of late-stage erythropoiesis and is able to rescue ineffective erythropoiesis [291].

7.5. Splenectomy

In cancer, the spleen becomes a central organ of extramedullary hematopoiesis, responsible for the generation of suppressive cells including EPCs and myeloid cells [292]. Therefore, it was suggested that splenectomy could be beneficial for cancer patients. In preclinical models, splenectomy inhibits tumor growth and prolongs the survival of tumor-bearing mice [62]. It also abolishes the induction of EPC expansion in extramedullary sites [62]. Similarly, splenectomy leads to the depletion of MDSCs, enhancing the activation of antitumor immunity [293]. Intriguingly, splenectomy before tumor inoculation or during tumor progression attenuates the decrease in RBC count and hemoglobin concentration, alleviating anemia [62].
However, clinical data are much less promising. Randomized trials showed that splenectomy in cancer patients not only has no advantages, but is also associated with increased perioperative morbidity [294,295,296]. Therefore, more preclinical and clinical studies are required to evaluate the effects of splenectomy in cancer.

8. Clinical Consequences of Tumor-Induced Anemia and EPC Expansion

Anemia is very common in cancer patients. Its prevalence differs from 30–90% depending on the type of cancer as well as the diagnostic criteria. It substantially decreases the quality of life of cancer patients [296,297]. Moreover, anemia is associated with shorter survival for patients with different types of cancer and a 65% overall increase in the risk of mortality compared to non-anemic cancer patients [191,298,299]. Importantly, severe anemia is associated with hypoxia in the TME of both primary and metastatic tumors [132], which is a known driver of aggressive tumor phenotype [300].
Clinical outcomes of EPC expansion are still unclear. In cancer patients, EPC expansion is the most prominent in individuals with moderate or severe anemia [41]. Moreover, the expansion of CD45+ EPCs correlates with a higher EBV load and suppressed T-cell response against the major antigenic EBV proteins, LMP2 and EBNA1 [41]. A recent study also demonstrated that CD45 EPCs may have clinical significance. In PDAC patients, the counts of CD45 EPCs in the spleen are increased compared with noncancerous pancreatic tumors or benign pancreatic masses [123]. High CD45 EPC counts predicted poor prognosis and were associated with larger tumor size and lymph node metastases [123]. Moreover, increased serum artemin concentrations, as well as increased expression of its receptors, correlate with poor prognosis in cancer patients [62,123]. Collectively, these observations demonstrate that in cancer patients, early-stage CD45+ EPCs may suppress the immune response, and late-stage CD45 EPCs may promote tumor growth by the secretion of artemin. Nonetheless, more research is required to accurately dissect the clinical role of EPCs in cancer patients.

9. Conclusions

In recent years, we have expanded our knowledge regarding the mechanisms of tumor evasion induced by dysregulation in hematopoiesis. The initial assumption that cancer only significantly regulates myelopoiesis turned out to be an oversimplification. Emerging evidence demonstrates that harnessing erythroid lineage cells together with megakaryocytes and platelets [301,302] is critical for cancer progression and immune evasion. EPCs promote tumor growth by either suppressing anti-tumor immune response or secreting growth factors, depending on the developmental stage. EPCs share many similarities with well-described suppressive cells of the immune system and use the same mechanisms to regulate the immune response.
However, several issues remain unclear and need to be investigated. First, despite differences in the expression of the immunomodulatory molecules [43], factors regulating the immunosuppressive properties of EPCs are unknown. Presumably, tumor-secreted cytokines and TME may potentiate the tumor-promoting role of EPCs. Moreover, it remains elusive whether and, if so, what factors promote the recruitment of EPCs to TME. Additionally, it is not known what are the interactions between EPCs and other cells in TME, including MDSCs, TAMs or NK cells, and comprehensive studies on the role of EPCs in TME are currently limited by technological advances. Recently, transcriptional profiling at a single-cell level has revolutionized our understanding of the complexity of cell interactions in TME, and indicates further research directions [303]. However, most of the protocols involve extensive hypotonic lysis of red blood cells [304], which drastically reduces the number of EPCs [305], excluding them from analyses of TME networks. Therefore, there is a great need to define the whole landscape of TME that includes EPCs.
Future research should focus on the comprehensive characterization of immunomodulatory mechanisms of EPCs and their regulation to better understand their function in tumor immune evasion and to enable targeting them in immunotherapy. It remains unknown whether EPCs in cancer may induce Treg differentiation, similar to their neonatal counterparts [87]. Similarly, regardless of a well-established role of EPCs in neonates [86,94], the regulation of myeloid cell response by EPCs in cancer is unknown. Moreover, erythroid cells were reported to produce IL-1β, IL-2, IL-4, IL-6, IFN-γ, and TNF-α [306]; however, their role in immune regulation by EPCs remains unknown.
It needs to be determined whether and how EPCs contribute to the clinical outcome of cancer patients undergoing various types of treatment, including immunotherapy. It was reported that EPCs may contribute to the drug resistance of cancer cells [116]. Therefore, targeting EPCs or their effector mechanisms in combination with other therapies may improve therapeutic effectiveness. Finally, the development and clinical testing of agents that could rescue erythroid maturation under cancer-induced EPC differentiation arrest are of great interest. Until then, anemia treatment is the best strategy to reduce EPC expansion and differentiation arrest, as well as to minimalize the tumor-promoting role of EPCs and to improve the survival and quality of life of cancer patients.

Author Contributions

Conceptualization, T.M.G.; writing—Original draft preparation, T.M.G., M.J.; writing—Review and editing, T.M.G., D.N., J.G.; Visualization, T.M.G.; Supervision, D.N., J.G.; Funding Acquisition, D.N., J.G., T.M.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants UMO-2019/35/B/NZ6/00540 from the National Science Center in Poland (D.N.), 013/RID/2018/19 (Regionalna Inicjatywa Doskonałości, project budget 12,000,000 PLN) from the Polish Ministry of Education and Science (J.G.) and 1M19/M/MG2/N/20 from the Medical University of Warsaw (T.M.G.). Figures were created with Biorender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
  2. Samstein, R.M.; Lee, C.H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef] [PubMed]
  3. Rodig, S.J.; Gusenleitner, D.; Jackson, D.G.; Gjini, E.; Giobbie-Hurder, A.; Jin, C.; Chang, H.; Lovitch, S.B.; Horak, C.; Weber, J.S.; et al. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [Green Version]
  4. Gao, J.; Shi, L.Z.; Zhao, H.; Chen, J.; Xiong, L.; He, Q.; Chen, T.; Roszik, J.; Bernatchez, C.; Woodman, S.E.; et al. Loss of IFN-γ Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell 2016, 167, 397–404. [Google Scholar] [CrossRef] [Green Version]
  5. Kearney, C.J.; Vervoort, S.J.; Hogg, S.J.; Ramsbottom, K.M.; Freeman, A.J.; Lalaoui, N.; Pijpers, L.; Michie, J.; Brown, K.K.; Knight, D.A.; et al. Tumor immune evasion arises through loss of TNF sensitivity. Sci. Immunol. 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  6. Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef]
  7. Van Elsas, M.J.; van Hall, T.; van der Burg, S.H. Future Challenges in Cancer Resistance to Immunotherapy. Cancers 2020, 12, 935. [Google Scholar] [CrossRef] [Green Version]
  8. Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [Green Version]
  9. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef] [PubMed]
  10. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [Green Version]
  11. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  12. Shaul, M.E.; Fridlender, Z.G. Tumour-associated neutrophils in patients with cancer. Nat. Rev. Clin. Oncol. 2019, 16, 601–620. [Google Scholar] [CrossRef]
  13. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [Green Version]
  14. Stewart, C.A.; Metheny, H.; Iida, N.; Smith, L.; Hanson, M.; Steinhagen, F.; Leighty, R.M.; Roers, A.; Karp, C.L.; Müller, W.; et al. Interferon-dependent IL-10 production by Tregs limits tumor Th17 inflammation. J. Clin. Invest. 2013, 123, 4859–4874. [Google Scholar] [CrossRef]
  15. Busse, D.; de la Rosa, M.; Hobiger, K.; Thurley, K.; Flossdorf, M.; Scheffold, A.; Höfer, T. Competing feedback loops shape IL-2 signaling between helper and regulatory T lymphocytes in cellular microenvironments. Proc. Natl. Acad. Sci. USA 2010, 107, 3058–3063. [Google Scholar] [CrossRef] [Green Version]
  16. Yuan, X.L.; Chen, L.; Li, M.X.; Dong, P.; Xue, J.; Wang, J.; Zhang, T.T.; Wang, X.A.; Zhang, F.M.; Ge, H.L.; et al. Elevated expression of Foxp3 in tumor-infiltrating Treg cells suppresses T-cell proliferation and contributes to gastric cancer progression in a COX-2-dependent manner. Clin. Immunol. 2010, 134, 277–288. [Google Scholar] [CrossRef]
  17. Sundström, P.; Stenstad, H.; Langenes, V.; Ahlmanner, F.; Theander, L.; Ndah, T.G.; Fredin, K.; Börjesson, L.; Gustavsson, B.; Bastid, J.; et al. Regulatory T Cells from Colon Cancer Patients Inhibit Effector T-cell Migration through an Adenosine-Dependent Mechanism. Cancer Immunol. Res. 2016, 4, 183–193. [Google Scholar] [CrossRef] [Green Version]
  18. Grzywa, T.M.; Sosnowska, A.; Matryba, P.; Rydzynska, Z.; Jasinski, M.; Nowis, D.; Golab, J. Myeloid Cell-Derived Arginase in Cancer Immune Response. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef]
  19. Yu, J.; Du, W.; Yan, F.; Wang, Y.; Li, H.; Cao, S.; Yu, W.; Shen, C.; Liu, J.; Ren, X. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J. Immunol. 2013, 190, 3783–3797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Della Chiesa, M.; Carlomagno, S.; Frumento, G.; Balsamo, M.; Cantoni, C.; Conte, R.; Moretta, L.; Moretta, A.; Vitale, M. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood 2006, 108, 4118–4125. [Google Scholar] [CrossRef]
  21. Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. 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, 781–790. [Google Scholar] [CrossRef]
  22. Huang, B.; Pan, P.Y.; Li, Q.; Sato, A.I.; Levy, D.E.; Bromberg, J.; Divino, C.M.; Chen, S.H. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006, 66, 1123–1131. [Google Scholar] [CrossRef] [Green Version]
  23. Pan, P.Y.; Ma, G.; Weber, K.J.; Ozao-Choy, J.; Wang, G.; Yin, B.; Divino, C.M.; Chen, S.H. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010, 70, 99–108. [Google Scholar] [CrossRef] [Green Version]
  24. Srivastava, M.K.; Sinha, P.; Clements, V.K.; Rodriguez, P.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010, 70, 68–77. [Google Scholar] [CrossRef] [Green Version]
  25. Kusmartsev, S.; Nefedova, Y.; Yoder, D.; Gabrilovich, D.I. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 2004, 172, 989–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Lu, T.; Ramakrishnan, R.; Altiok, S.; Youn, J.-I.; Cheng, P.; Celis, E.; Pisarev, V.; Sherman, S.; Sporn, M.B.; Gabrilovich, D. Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J. Clin. Invest. 2011, 121, 4015–4029. [Google Scholar] [CrossRef] [Green Version]
  27. Gordon, S.R.; Maute, R.L.; Dulken, B.W.; Hutter, G.; George, B.M.; McCracken, M.N.; Gupta, R.; Tsai, J.M.; Sinha, R.; Corey, D.; et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017, 545, 495–499. [Google Scholar] [CrossRef]
  28. Rodriguez, P.C.; Quiceno, D.G.; Zabaleta, J.; Ortiz, B.; Zea, A.H.; Piazuelo, M.B.; Delgado, A.; Correa, P.; Brayer, J.; Sotomayor, E.M.; et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004, 64, 5839–5849. [Google Scholar] [CrossRef] [Green Version]
  29. Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.T.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 Blocks CD8+ T Cell-Dependent Responses to Chemotherapy by Suppressing IL-12 Expression in Intratumoral Dendritic Cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Kaplanov, I.; Carmi, Y.; Kornetsky, R.; Shemesh, A.; Shurin, G.V.; Shurin, M.R.; Dinarello, C.A.; Voronov, E.; Apte, R.N. Blocking IL-1β reverses the immunosuppression in mouse breast cancer and synergizes with anti–PD-1 for tumor abrogation. Proc. Natl. Acad. Sci. USA 2019, 116, 1361–1369. [Google Scholar] [CrossRef] [Green Version]
  31. Chittezhath, M.; Dhillon, M.K.; Lim, J.Y.; Laoui, D.; Shalova, I.N.; Teo, Y.L.; Chen, J.; Kamaraj, R.; Raman, L.; Lum, J.; et al. Molecular Profiling Reveals a Tumor-Promoting Phenotype of Monocytes and Macrophages in Human Cancer Progression. Immunity 2014, 41, 815–829. [Google Scholar] [CrossRef] [Green Version]
  32. Vom Berg, J.; Vrohlings, M.; Haller, S.; Haimovici, A.; Kulig, P.; Sledzinska, A.; Weller, M.; Becher, B. Intratumoral IL-12 combined with CTLA-4 blockade elicits T cell-mediated glioma rejection. J. Exp. Med. 2013, 210, 2803–2811. [Google Scholar] [CrossRef] [Green Version]
  33. Kratochvill, F.; Neale, G.; Haverkamp, J.M.; Van de Velde, L.-A.; Smith, A.M.; Kawauchi, D.; McEvoy, J.; Roussel, M.F.; Dyer, M.A.; Qualls, J.E.; et al. TNF Counterbalances the Emergence of M2 Tumor Macrophages. Cell Rep. 2015, 12, 1902–1914. [Google Scholar] [CrossRef] [Green Version]
  34. Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.; Jonkers, J.; et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef]
  35. Michaeli, J.; Shaul, M.E.; Mishalian, I.; Hovav, A.H.; Levy, L.; Zolotriov, L.; Granot, Z.; Fridlender, Z.G. Tumor-associated neutrophils induce apoptosis of non-activated CD8 T-cells in a TNFα and NO-dependent mechanism, promoting a tumor-supportive environment. Oncoimmunology 2017, 6, e1356965. [Google Scholar] [CrossRef]
  36. Wang, T.T.; Zhao, Y.L.; Peng, L.S.; Chen, N.; Chen, W.; Lv, Y.P.; Mao, F.Y.; Zhang, J.Y.; Cheng, P.; Teng, Y.S.; et al. Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF-PD-L1 pathway. Gut 2017, 66, 1900–1911. [Google Scholar] [CrossRef] [Green Version]
  37. Nazareth, M.R.; Broderick, L.; Simpson-Abelson, M.R.; Kelleher, R.J., Jr.; Yokota, S.J.; Bankert, R.B. Characterization of human lung tumor-associated fibroblasts and their ability to modulate the activation of tumor-associated T cells. J. Immunol. 2007, 178, 5552–5562. [Google Scholar] [CrossRef] [Green Version]
  38. Lakins, M.A.; Ghorani, E.; Munir, H.; Martins, C.P.; Shields, J.D. Cancer-associated fibroblasts induce antigen-specific deletion of CD8+ T Cells to protect tumour cells. Nat. Commun. 2018, 9, 948. [Google Scholar] [CrossRef]
  39. Cheng, Y.; Li, H.; Deng, Y.; Tai, Y.; Zeng, K.; Zhang, Y.; Liu, W.; Zhang, Q.; Yang, Y. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 2018, 9, 422. [Google Scholar] [CrossRef]
  40. Kumar, V.; Donthireddy, L.; Marvel, D.; Condamine, T.; Wang, F.; Lavilla-Alonso, S.; Hashimoto, A.; Vonteddu, P.; Behera, R.; Goins, M.A.; et al. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017, 32, 654–668. [Google Scholar] [CrossRef] [Green Version]
  41. Zhao, L.; He, R.; Long, H.; Guo, B.; Jia, Q.; Qin, D.; Liu, S.-Q.; Wang, Z.; Xiang, T.; Zhang, J.; et al. Late-stage tumors induce anemia and immunosuppressive extramedullary erythroid progenitor cells. Nat. Med. 2018, 24, 1536–1544. [Google Scholar] [CrossRef]
  42. Chen, J.; Qiao, Y.-D.; Li, X.; Xu, J.-L.; Ye, Q.-J.; Jiang, N.; Zhang, H.; Wu, X.-Y. Intratumoral CD45+ CD71+ erythroid cells induce immune tolerance and predict tumor recurrence in hepatocellular carcinoma. Cancer Lett. 2020. [Google Scholar] [CrossRef]
  43. Sano, Y.; Yoshida, T.; Choo, M.-K.; Jiménez-Andrade, Y.; Hill, K.R.; Georgopoulos, K.; Park, J.M. Multiorgan Signaling Mobilizes Tumor-Associated Erythroid Cells Expressing Immune Checkpoint Molecules. Mol. Cancer Res. 2020. [Google Scholar] [CrossRef]
  44. Hurwitz, S.N.; Jung, S.K.; Kurre, P. Hematopoietic stem and progenitor cell signaling in the niche. Leukemia 2020, 34, 3136–3148. [Google Scholar] [CrossRef]
  45. Peter, V.; Guntram, B.; Igor, T.; Iris, Z.U.; Ulrich, G.; Reinhard, S.; Karl, S.; Wolfgang, F.; Peter, B.; Michael, P.; et al. Normal and pathological erythropoiesis in adults: From gene regulation to targeted treatment concepts. Haematologica 2018, 103, 1593–1603. [Google Scholar] [CrossRef]
  46. Hattangadi, S.M.; Wong, P.; Zhang, L.; Flygare, J.; Lodish, H.F. From stem cell to red cell: Regulation of erythropoiesis at multiple levels by multiple proteins, RNAs, and chromatin modifications. Blood 2011, 118, 6258–6268. [Google Scholar] [CrossRef] [Green Version]
  47. Oburoglu, L.; Romano, M.; Taylor, N.; Kinet, S. Metabolic regulation of hematopoietic stem cell commitment and erythroid differentiation. Curr. Opin. Hematol. 2016, 23, 198–205. [Google Scholar] [CrossRef]
  48. Koury, M.J. Tracking erythroid progenitor cells in times of need and times of plenty. Exp. Hematol. 2016, 44, 653–663. [Google Scholar] [CrossRef]
  49. Haase, V.H. Regulation of erythropoiesis by hypoxia-inducible factors. Blood Rev. 2013, 27, 41–53. [Google Scholar] [CrossRef] [Green Version]
  50. Lee, W.-C.; Hsu, P.-Y.; Hsu, H.-Y. Stem cell factor produced by tumor cells expands myeloid-derived suppressor cells in mice. Sci. Rep. 2020, 10, 11257. [Google Scholar] [CrossRef]
  51. Colmone, A.; Amorim, M.; Pontier, A.L.; Wang, S.; Jablonski, E.; Sipkins, D.A. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science 2008, 322, 1861–1865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Mouchemore, K.A.; Anderson, R.L.; Hamilton, J.A. Neutrophils, G-CSF and their contribution to breast cancer metastasis. FEBS J. 2018, 285, 665–679. [Google Scholar] [CrossRef] [Green Version]
  53. Dentelli, P.; Rosso, A.; Olgasi, C.; Camussi, G.; Brizzi, M.F. IL-3 is a novel target to interfere with tumor vasculature. Oncogene 2011, 30, 4930–4940. [Google Scholar] [CrossRef] [Green Version]
  54. Kasper, C.; Terhaar, A.; Fosså, A.; Welt, A.; Seeber, S.; Nowrousian, M.R. Recombinant human erythropoietin in the treatment of cancer-related anaemia. Eur. J. Haematol. 1997, 58, 251–256. [Google Scholar] [CrossRef]
  55. Zhang, Y.; Wei, Y.; Liu, D.; Liu, F.; Li, X.; Pan, L.; Pang, Y.; Chen, D. Role of growth differentiation factor 11 in development, physiology and disease. Oncotarget 2017, 8, 81604–81616. [Google Scholar] [CrossRef] [Green Version]
  56. Bashir, M.; Damineni, S.; Mukherjee, G.; Kondaiah, P. Activin-A signaling promotes epithelial–mesenchymal transition, invasion, and metastatic growth of breast cancer. NPJ Breast Cancer 2015, 1, 15007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wang, Y.; Gao, A.; Zhao, H.; Lu, P.; Cheng, H.; Dong, F.; Gong, Y.; Ma, S.; Zheng, Y.; Zhang, H.; et al. Leukemia cell infiltration causes defective erythropoiesis partially through MIP-1α/CCL3. Leukemia 2016, 30, 1897–1908. [Google Scholar] [CrossRef]
  58. Liu, L.; Yu, Z.; Cheng, H.; Mao, X.; Sui, W.; Deng, S.; Wei, X.; Lv, J.; Du, C.; Xu, J.; et al. Multiple myeloma hinders erythropoiesis and causes anaemia owing to high levels of CCL3 in the bone marrow microenvironment. Sci. Rep. 2020, 10, 20508. [Google Scholar] [CrossRef]
  59. Silvestris, F.; Cafforio, P.; Tucci, M.; Dammacco, F. Negative regulation of erythroblast maturation by Fas-L+/TRAIL+ highly malignant plasma cells: A major pathogenetic mechanism of anemia in multiple myeloma. Blood 2002, 99, 1305–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Hexner, E.O.; Serdikoff, C.; Jan, M.; Swider, C.R.; Robinson, C.; Yang, S.; Angeles, T.; Emerson, S.G.; Carroll, M.; Ruggeri, B.; et al. Lestaurtinib (CEP701) is a JAK2 inhibitor that suppresses JAK2/STAT5 signaling and the proliferation of primary erythroid cells from patients with myeloproliferative disorders. Blood 2008, 111, 5663–5671. [Google Scholar] [CrossRef]
  61. Khalil, S.; Delehanty, L.; Grado, S.; Holy, M.; White, Z., III; Freeman, K.; Kurita, R.; Nakamura, Y.; Bullock, G.; Goldfarb, A. Iron modulation of erythropoiesis is associated with Scribble-mediated control of the erythropoietin receptor. J. Exp. Med. 2017, 215, 661–679. [Google Scholar] [CrossRef]
  62. Han, Y.; Liu, Q.; Hou, J.; Gu, Y.; Zhang, Y.; Chen, Z.; Fan, J.; Zhou, W.; Qiu, S.; Zhang, Y.; et al. Tumor-Induced Generation of Splenic Erythroblast-like Ter-Cells Promotes Tumor Progression. Cell 2018, 173, 634–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Silvestris, F.; Tucci, M.; Cafforio, P.; Dammacco, F. Fas-L up-regulation by highly malignant myeloma plasma cells: Role in the pathogenesis of anemia and disease progression. Blood 2001, 97, 1155–1164. [Google Scholar] [CrossRef] [Green Version]
  64. Gilreath, J.A.; Stenehjem, D.D.; Rodgers, G.M. Diagnosis and treatment of cancer-related anemia. Am. J. Hematol. 2014, 89, 203–212. [Google Scholar] [CrossRef] [PubMed]
  65. Zowczak, M.; Iskra, M.; Torliński, L.; Cofta, S. Analysis of serum copper and zinc concentrations in cancer patients. Biol. Trace Elem. Res. 2001, 82, 1–8. [Google Scholar] [CrossRef]
  66. Gilreath, J.A.; Rodgers, G.M. How I treat cancer-associated anemia. Blood 2020, 136, 801–813. [Google Scholar] [CrossRef]
  67. Pinnix, Z.K.; Miller, L.D.; Wang, W.; D’Agostino, R., Jr.; Kute, T.; Willingham, M.C.; Hatcher, H.; Tesfay, L.; Sui, G.; Di, X.; et al. Ferroportin and iron regulation in breast cancer progression and prognosis. Sci. Transl. Med. 2010, 2, 43ra56. [Google Scholar] [CrossRef]
  68. Vela, D.; Vela-Gaxha, Z. Differential regulation of hepcidin in cancer and non-cancer tissues and its clinical implications. Exp. Mol. Med. 2018, 50, e436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Mendelson, A.; Frenette, P.S. Hematopoietic stem cell niche maintenance during homeostasis and regeneration. Nat. Med. 2014, 20, 833–846. [Google Scholar] [CrossRef] [Green Version]
  70. Mei, Y.; Liu, Y.; Ji, P. Understanding terminal erythropoiesis: An update on chromatin condensation, enucleation, and reticulocyte maturation. Blood Rev. 2020. [Google Scholar] [CrossRef] [PubMed]
  71. Bain, B.J. The bone marrow aspirate of healthy subjects. Br. J. Haematol. 1996, 94, 206–209. [Google Scholar] [CrossRef] [PubMed]
  72. Parmentier, S.; Kramer, M.; Weller, S.; Schuler, U.; Ordemann, R.; Rall, G.; Schaich, M.; Bornhäuser, M.; Ehninger, G.; Kroschinsky, F. Reevaluation of reference values for bone marrow differential counts in 236 healthy bone marrow donors. Ann. Hematol. 2020, 99, 2723–2729. [Google Scholar] [CrossRef]
  73. Goodman, J.W.; Hall, E.A.; Miller, K.L.; Shinpock, S.G. Interleukin 3 promotes erythroid burst formation in “serum-free” cultures without detectable erythropoietin. Proc. Natl. Acad. Sci. USA 1985, 82, 3291–3295. [Google Scholar] [CrossRef] [Green Version]
  74. Emerson, S.G.; Thomas, S.; Ferrara, J.L.; Greenstein, J.L. Developmental regulation of erythropoiesis by hematopoietic growth factors: Analysis on populations of BFU-E from bone marrow, peripheral blood, and fetal liver. Blood 1989, 74, 49–55. [Google Scholar] [CrossRef] [Green Version]
  75. Muta, K.; Krantz, S.B.; Bondurant, M.C.; Wickrema, A. Distinct roles of erythropoietin, insulin-like growth factor I, and stem cell factor in the development of erythroid progenitor cells. J. Clin. Invest. 1994, 94, 34–43. [Google Scholar] [CrossRef] [Green Version]
  76. De Maria, R.; Testa, U.; Luchetti, L.; Zeuner, A.; Stassi, G.; Pelosi, E.; Riccioni, R.; Felli, N.; Samoggia, P.; Peschle, C. Apoptotic role of Fas/Fas ligand system in the regulation of erythropoiesis. Blood 1999, 93, 796–803. [Google Scholar] [CrossRef]
  77. Bhoopalan, S.V.; Huang, L.J.; Weiss, M.J. Erythropoietin regulation of red blood cell production: From bench to bedside and back. F1000Research 2020, 9. [Google Scholar] [CrossRef]
  78. Muckenthaler, M.U.; Rivella, S.; Hentze, M.W.; Galy, B. A Red Carpet for Iron Metabolism. Cell 2017, 168, 344–361. [Google Scholar] [CrossRef] [Green Version]
  79. Lee, H.-Y.; Gao, X.; Barrasa, M.I.; Li, H.; Elmes, R.R.; Peters, L.L.; Lodish, H.F. PPAR-α and glucocorticoid receptor synergize to promote erythroid progenitor self-renewal. Nature 2015, 522, 474–477. [Google Scholar] [CrossRef] [Green Version]
  80. Gutiérrez, L.; Caballero, N.; Fernández-Calleja, L.; Karkoulia, E.; Strouboulis, J. Regulation of GATA1 levels in erythropoiesis. IUBMB Life 2020, 72, 89–105. [Google Scholar] [CrossRef] [PubMed]
  81. Pevny, L.; Simon, M.C.; Robertson, E.; Klein, W.H.; Tsai, S.F.; D’Agati, V.; Orkin, S.H.; Costantini, F. Erythroid differentiation in chimaeric mice blocked by a targeted mutation in the gene for transcription factor GATA-1. Nature 1991, 349, 257–260. [Google Scholar] [CrossRef]
  82. De Maria, R.; Zeuner, A.; Eramo, A.; Domenichelli, C.; Bonci, D.; Grignani, F.; Srinivasula, S.M.; Alnemri, E.S.; Testa, U.; Peschle, C. Negative regulation of erythropoiesis by caspase-mediated cleavage of GATA-1. Nature 1999, 401, 489–493. [Google Scholar] [CrossRef]
  83. Han, X.; Zhang, J.; Peng, Y.; Peng, M.; Chen, X.; Chen, H.; Song, J.; Hu, X.; Ye, M.; Li, J.; et al. Unexpected role for p19INK4d in posttranscriptional regulation of GATA1 and modulation of human terminal erythropoiesis. Blood 2017, 129, 226–237. [Google Scholar] [CrossRef] [Green Version]
  84. Ribeil, J.-A.; Zermati, Y.; Vandekerckhove, J.; Cathelin, S.; Kersual, J.; Dussiot, M.; Coulon, S.; Cruz Moura, I.; Zeuner, A.; Kirkegaard-Sørensen, T.; et al. Hsp70 regulates erythropoiesis by preventing caspase-3-mediated cleavage of GATA-1. Nature 2007, 445, 102–105. [Google Scholar] [CrossRef]
  85. Elahi, S. Neglected Cells: Immunomodulatory Roles of CD71+ Erythroid Cells. Trends Immunol. 2019, 40, 181–185. [Google Scholar] [CrossRef] [PubMed]
  86. Elahi, S.; Ertelt, J.M.; Kinder, J.M.; Jiang, T.T.; Zhang, X.; Xin, L.; Chaturvedi, V.; Strong, B.S.; Qualls, J.E.; Steinbrecher, K.A.; et al. Immunosuppressive CD71+ erythroid cells compromise neonatal host defence against infection. Nature 2013, 504, 158–162. [Google Scholar] [CrossRef] [Green Version]
  87. Shahbaz, S.; Bozorgmehr, N.; Koleva, P.; Namdar, A.; Jovel, J.; Fava, R.A.; Elahi, S. CD71+VISTA+ erythroid cells promote the development and function of regulatory T cells through TGF-β. PLoS Biol. 2018, 16, e2006649. [Google Scholar] [CrossRef] [Green Version]
  88. Delyea, C.; Bozorgmehr, N.; Koleva, P.; Dunsmore, G.; Shahbaz, S.; Huang, V.; Elahi, S. CD71+ Erythroid Suppressor Cells Promote Fetomaternal Tolerance through Arginase-2 and PDL-1. J. Immunol. 2018, 200, 4044–4058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Shim, Y.A.; Weliwitigoda, A.; Campbell, T.; Dosanjh, M.; Johnson, P. Splenic erythroid progenitors decrease TNF-α production by macrophages and reduce systemic inflammation in a mouse model of T cell-induced colitis. Eur. J. Immunol. 2020. [Google Scholar] [CrossRef]
  90. Namdar, A.; Dunsmore, G.; Shahbaz, S.; Koleva, P.; Xu, L.; Jovel, J.; Houston, S.; Elahi, S. CD71+ Erythroid Cells Exacerbate HIV-1 Susceptibility, Mediate trans-Infection, and Harbor Infective Viral Particles. mBio 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  91. Bernardes, J.P.; Mishra, N.; Tran, F.; Bahmer, T.; Best, L.; Blase, J.I.; Bordoni, D.; Franzenburg, J.; Geisen, U.; Josephs-Spaulding, J.; et al. Longitudinal Multi-omics Analyses Identify Responses of Megakaryocytes, Erythroid Cells, and Plasmablasts as Hallmarks of Severe COVID-19. Immunity 2020, 53, 1296–1314. [Google Scholar] [CrossRef]
  92. Grzywa, T.M.; Sosnowska, A.; Rydzynska, Z.; Lazniewski, M.; Plewczynski, D.; Klicka, K.; Malecka, M.; Rodziewicz-Lurzynska, A.; Ciepiela, O.; Justyniarska, M.; et al. Potent but transient immunosuppression of T-cells is a general feature of erythroid progenitor cells. bioRxiv 2021. [Google Scholar] [CrossRef]
  93. Dunsmore, G.; Koleva, P.; Ghobakhloo, N.; Sutton, R.; Ambrosio, L.; Meng, X.; Hotte, N.; Nguyen, V.; Madsen, K.L.; Dieleman, L.A.; et al. Lower Abundance and Impaired Function of CD71+ Erythroid Cells in Inflammatory Bowel Disease Patients during Pregnancy. J. Crohns Colitis 2019, 13, 230–244. [Google Scholar] [CrossRef]
  94. Elahi, S.; Vega-López, M.A.; Herman-Miguel, V.; Ramírez-Estudillo, C.; Mancilla-Ramírez, J.; Motyka, B.; West, L.; Oyegbami, O. CD71+ Erythroid Cells in Human Neonates Exhibit Immunosuppressive Properties and Compromise Immune Response Against Systemic Infection in Neonatal Mice. Front. Immunol. 2020, 11, 597433. [Google Scholar] [CrossRef]
  95. Shahbaz, S.; Xu, L.; Osman, M.; Sligl, W.; Shields, J.; Joyce, M.; Tyrrell, L.; Oyegbami, O.; Elahi, S. Erythroid precursors and progenitors suppress adaptive immunity and get invaded by SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
  96. Lindsey Robert, B.; Sankar, S.; Michael, A.; Gayle, B.; Bernard, C.C.; Corey, C.; Brenda, C.; Erik, R.D.; Ashley Morris, E.; Alison, G.F.; et al. Prevention and Treatment of Cancer-Related Infections, Version 2.2016, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2016, 14, 882–913. [Google Scholar] [CrossRef]
  97. Allen, B.M.; Hiam, K.J.; Burnett, C.E.; Venida, A.; DeBarge, R.; Tenvooren, I.; Marquez, D.M.; Cho, N.W.; Carmi, Y.; Spitzer, M.H. Systemic dysfunction and plasticity of the immune macroenvironment in cancer models. Nat. Med. 2020, 26, 1125–1134. [Google Scholar] [CrossRef]
  98. Wu, C.; Hua, Q.; Zheng, L. Generation of Myeloid Cells in Cancer: The Spleen Matters. Front. Immunol. 2020, 11, 1126. [Google Scholar] [CrossRef]
  99. Strauss, L.; Guarneri, V.; Gennari, A.; Sica, A. Implications of metabolism-driven myeloid dysfunctions in cancer therapy. Cell. Mol. Immunol. 2020. [Google Scholar] [CrossRef]
  100. Gillespie, M.A.; Palii, C.G.; Sanchez-Taltavull, D.; Shannon, P.; Longabaugh, W.J.R.; Downes, D.J.; Sivaraman, K.; Espinoza, H.M.; Hughes, J.R.; Price, N.D.; et al. Absolute Quantification of Transcription Factors Reveals Principles of Gene Regulation in Erythropoiesis. Mol. Cell 2020. [Google Scholar] [CrossRef]
  101. An, X.; Schulz, V.P.; Li, J.; Wu, K.; Liu, J.; Xue, F.; Hu, J.; Mohandas, N.; Gallagher, P.G. Global transcriptome analyses of human and murine terminal erythroid differentiation. Blood 2014, 123, 3466–3477. [Google Scholar] [CrossRef] [Green Version]
  102. Li, J.; Hale, J.; Bhagia, P.; Xue, F.; Chen, L.; Jaffray, J.; Yan, H.; Lane, J.; Gallagher, P.G.; Mohandas, N.; et al. Isolation and transcriptome analyses of human erythroid progenitors: BFU-E and CFU-E. Blood 2014, 124, 3636–3645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yan, H.; Hale, J.; Jaffray, J.; Li, J.; Wang, Y.; Huang, Y.; An, X.; Hillyer, C.; Wang, N.; Kinet, S.; et al. Developmental differences between neonatal and adult human erythropoiesis. Am. J. Hematol. 2018, 93, 494–503. [Google Scholar] [CrossRef]
  104. Yang, Y.; Wang, H.; Chang, K.-H.; Qu, H.; Zhang, Z.; Xiong, Q.; Qi, H.; Cui, P.; Lin, Q.; Ruan, X.; et al. Transcriptome dynamics during human erythroid differentiation and development. Genomics 2013, 102, 431–441. [Google Scholar] [CrossRef] [Green Version]
  105. Shi, L.; Lin, Y.-H.; Sierant, M.C.; Zhu, F.; Cui, S.; Guan, Y.; Sartor, M.A.; Tanabe, O.; Lim, K.-C.; Engel, J.D. Developmental transcriptome analysis of human erythropoiesis. Hum. Mol. Genet. 2014, 23, 4528–4542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Liu, X.; Zhang, Y.; Ni, M.; Cao, H.; Signer, R.A.J.; Li, D.; Li, M.; Gu, Z.; Hu, Z.; Dickerson, K.E.; et al. Regulation of mitochondrial biogenesis in erythropoiesis by mTORC1-mediated protein translation. Nat. Cell Biol. 2017, 19, 626–638. [Google Scholar] [CrossRef] [Green Version]
  107. Huang, P.; Zhao, Y.; Zhong, J.; Zhang, X.; Liu, Q.; Qiu, X.; Chen, S.; Yan, H.; Hillyer, C.; Mohandas, N.; et al. Putative regulators for the continuum of erythroid differentiation revealed by single-cell transcriptome of human BM and UCB cells. Proc. Natl. Acad. Sci. USA 2020, 201915085. [Google Scholar] [CrossRef]
  108. Gautier, E.-F.; Ducamp, S.; Leduc, M.; Salnot, V.; Guillonneau, F.; Dussiot, M.; Hale, J.; Giarratana, M.-C.; Raimbault, A.; Douay, L.; et al. Comprehensive Proteomic Analysis of Human Erythropoiesis. Cell Rep. 2016, 16, 1470–1484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Amon, S.; Meier-Abt, F.; Gillet, L.C.; Dimitrieva, S.; Theocharides, A.P.A.; Manz, M.G.; Aebersold, R. Sensitive Quantitative Proteomics of Human Hematopoietic Stem and Progenitor Cells by Data-independent Acquisition Mass Spectrometry. Mol. Cell Proteom. 2019, 18, 1454–1467. [Google Scholar] [CrossRef] [Green Version]
  110. Brand, M.; Ranish, J.A.; Kummer, N.T.; Hamilton, J.; Igarashi, K.; Francastel, C.; Chi, T.H.; Crabtree, G.R.; Aebersold, R.; Groudine, M. Dynamic changes in transcription factor complexes during erythroid differentiation revealed by quantitative proteomics. Nat. Struct. Mol. Biol. 2004, 11, 73–80. [Google Scholar] [CrossRef]
  111. Jassinskaja, M.; Johansson, E.; Kristiansen, T.A.; Åkerstrand, H.; Sjöholm, K.; Hauri, S.; Malmström, J.; Yuan, J.; Hansson, J. Comprehensive Proteomic Characterization of Ontogenic Changes in Hematopoietic Stem and Progenitor Cells. Cell Rep. 2017, 21, 3285–3297. [Google Scholar] [CrossRef] [Green Version]
  112. Mello, F.V.; Land, M.G.P.; Costa, E.S.; Teodósio, C.; Sanchez, M.-L.; Bárcena, P.; Peres, R.T.; Pedreira, C.E.; Alves, L.R.; Orfao, A. Maturation-associated gene expression profiles during normal human bone marrow erythropoiesis. Cell Death Discov. 2019, 5, 69. [Google Scholar] [CrossRef] [Green Version]
  113. Panday, A.; Sahoo, M.K.; Osorio, D.; Batra, S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cell. Mol. Immunol. 2015, 12, 5–23. [Google Scholar] [CrossRef] [Green Version]
  114. Franchina, D.G.; Dostert, C.; Brenner, D. Reactive Oxygen Species: Involvement in T Cell Signaling and Metabolism. Trends Immunol. 2018, 39, 489–502. [Google Scholar] [CrossRef]
  115. Ohl, K.; Tenbrock, K. Reactive Oxygen Species as Regulators of MDSC-Mediated Immune Suppression. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  116. Xia, W.; Sainan, Y.; Xin, P.; Silian, H.; Zailin, Y.; Xiaomin, S.; Wen, C.; Yong, Z. CD45+ Erythroid Progenitor Cell Contribute to Antiangiogenic Drug Resistance Through Reactive Oxygen Species in Lymphoma. Res. Sq. 2020. [Google Scholar] [CrossRef]
  117. Kotsafti, A.; Scarpa, M.; Castagliuolo, I.; Scarpa, M. Reactive Oxygen Species and Antitumor Immunity-From Surveillance to Evasion. Cancers 2020, 12, 1748. [Google Scholar] [CrossRef]
  118. Lewis, S.M.; Williams, A.; Eisenbarth, S.C. Structure and function of the immune system in the spleen. Sci. Immunol. 2019, 4, eaau6085. [Google Scholar] [CrossRef]
  119. Imai, T.; Ishida, H.; Suzue, K.; Taniguchi, T.; Okada, H.; Shimokawa, C.; Hisaeda, H. Cytotoxic activities of CD8+ T cells collaborate with macrophages to protect against blood-stage murine malaria. Elife 2015, 4. [Google Scholar] [CrossRef]
  120. Ilieva, M.; Nielsen, J.; Korshunova, I.; Gotfryd, K.; Bock, E.; Pankratova, S.; Michel, T.M. Artemin and an Artemin-Derived Peptide, Artefin, Induce Neuronal Survival, and Differentiation Through Ret and NCAM. Front. Mol. Neurosci. 2019, 12. [Google Scholar] [CrossRef] [Green Version]
  121. Wang, J.; Wang, H.; Cai, J.; Du, S.; Xin, B.; Wei, W.; Zhang, T.; Shen, X. Artemin regulates CXCR4 expression to induce migration and invasion in pancreatic cancer cells through activation of NF-κB signaling. Exp. Cell Res. 2018, 365, 12–23. [Google Scholar] [CrossRef]
  122. Song, Z.; Yang, F.; Du, H.; Li, X.; Liu, J.; Dong, M.; Xu, X. Role of artemin in non-small cell lung cancer. Thorac. Cancer 2018, 9, 555–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Li, T.-J.; Li, H.; Zhang, W.-H.; Xu, S.-S.; Jiang, W.; Li, S.; Gao, H.-L.; Han, X.; Xu, H.-X.; Wu, C.-T.; et al. Human splenic TER cells: A relevant prognostic factor acting via the artemin-GFRα3-ERK pathway in pancreatic ductal adenocarcinoma. Int. J. Cancer 2020. [Google Scholar] [CrossRef]
  124. Dzierzak, E.; Philipsen, S. Erythropoiesis: Development and differentiation. Cold Spring Harb. Perspect. Med. 2013, 3, a011601. [Google Scholar] [CrossRef]
  125. Ileana, C.; Sjaak, P. Flicking the switch: Adult hemoglobin expression in erythroid cells derived from cord blood and human induced pluripotent stem cells. Haematologica 2014, 99, 1647–1649. [Google Scholar] [CrossRef] [Green Version]
  126. Baron, M.H.; Isern, J.; Fraser, S.T. The embryonic origins of erythropoiesis in mammals. Blood 2012, 119, 4828–4837. [Google Scholar] [CrossRef] [Green Version]
  127. Riley, R.S.; Ben-Ezra, J.M.; Goel, R.; Tidwell, A. Reticulocytes and reticulocyte enumeration. J. Clin. Lab. Anal. 2001, 15, 267–294. [Google Scholar] [CrossRef]
  128. Paulson, R.F.; Hariharan, S.; Little, J.A. Stress erythropoiesis: Definitions and models for its study. Exp. Hematol. 2020, 89, 43–54. [Google Scholar] [CrossRef]
  129. Paulson, R.F.; Ruan, B.; Hao, S.; Chen, Y. Stress Erythropoiesis is a Key Inflammatory Response. Cells 2020, 9, 634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Jackson, A.; Nanton, M.R.; O’Donnell, H.; Akue, A.D.; McSorley, S.J. Innate immune activation during Salmonella infection initiates extramedullary erythropoiesis and splenomegaly. J. Immunol. 2010, 185, 6198–6204. [Google Scholar] [CrossRef] [Green Version]
  131. Rinchai, D.; Altman, M.C.; Konza, O.; Hässler, S.; Martina, F.; Toufiq, M.; Garand, M.; Kabeer, B.S.A.; Palucka, K.; Mejias, A.; et al. Definition of erythroid cell-positive blood transcriptome phenotypes associated with severe respiratory syncytial virus infection. Clin. Transl. Med. 2020, 10, e244. [Google Scholar] [CrossRef]
  132. Tabares Calvache, E.; Tabares Calvache, A.D.; Faulhaber, G.A.M. Systematic review about etiologic association to the leukoerythroblastic reaction. Int. J. Lab. Hematol. 2020, 42, 495–500. [Google Scholar] [CrossRef]
  133. Delsol, G.; Guiu-Godfrin, B.; Guiu, M.; Pris, J.; Corberand, J.; Fabre, J. Leukoerythroblastosis and cancer frequency, prognosis, and physiopathologic significance. Cancer 1979, 44, 1009–1013. [Google Scholar] [CrossRef]
  134. Kornblau, S.M.; Cohen, A.C.; Soper, D.; Huang, Y.-W.; Cesano, A. Age-related changes of healthy bone marrow cell signaling in response to growth factors provide insight into low risk MDS. Cytom. Part B Clin. Cytom. 2014, 86, 383–396. [Google Scholar] [CrossRef]
  135. Oikonomidou, P.R.; Rivella, S. What can we learn from ineffective erythropoiesis in thalassemia? Blood Rev. 2018, 32, 130–143. [Google Scholar] [CrossRef] [PubMed]
  136. Manso, B.A.; Zhang, H.; Mikkelson, M.G.; Gwin, K.A.; Secreto, C.R.; Ding, W.; Parikh, S.A.; Kay, N.E.; Medina, K.L. Bone marrow hematopoietic dysfunction in untreated chronic lymphocytic leukemia patients. Leukemia 2019, 33, 638–652. [Google Scholar] [CrossRef] [PubMed]
  137. Mussai, F.; De Santo, C.; Abu-Dayyeh, I.; Booth, S.; Quek, L.; McEwen-Smith, R.M.; Qureshi, A.; Dazzi, F.; Vyas, P.; Cerundolo, V. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood 2013, 122, 749–758. [Google Scholar] [CrossRef] [Green Version]
  138. Stefanie, G.; Manuel, R.-P.; Paul, J.; Annemarie, K.; Felix, B.; Julian, G.; Christoph, Z.; Ulrich, G.; Guido, K.; Frank, L.; et al. Transforming growth factor β1-mediated functional inhibition of mesenchymal stromal cells in myelodysplastic syndromes and acute myeloid leukemia. Haematologica 2018, 103, 1462–1471. [Google Scholar] [CrossRef] [Green Version]
  139. Gong, Y.; Zhao, M.; Yang, W.; Gao, A.; Yin, X.; Hu, L.; Wang, X.; Xu, J.; Hao, S.; Cheng, T.; et al. Megakaryocyte-derived excessive transforming growth factor β1 inhibits proliferation of normal hematopoietic stem cells in acute myeloid leukemia. Exp. Hematol. 2018, 60, 40–46. [Google Scholar] [CrossRef] [Green Version]
  140. Bruns, I.; Cadeddu, R.-P.; Brueckmann, I.; Fröbel, J.; Geyh, S.; Büst, S.; Fischer, J.C.; Roels, F.; Wilk, C.M.; Schildberg, F.A.; et al. Multiple myeloma–related deregulation of bone marrow–derived CD34+ hematopoietic stem and progenitor cells. Blood 2012, 120, 2620–2630. [Google Scholar] [CrossRef] [Green Version]
  141. Waclawiczek, A.; Hamilton, A.; Rouault-Pierre, K.; Abarrategi, A.; Albornoz, M.G.; Miraki-Moud, F.; Bah, N.; Gribben, J.; Fitzgibbon, J.; Taussig, D.; et al. Mesenchymal niche remodeling impairs hematopoiesis via stanniocalcin 1 in acute myeloid leukemia. J. Clin. Invest. 2020, 130, 3038–3050. [Google Scholar] [CrossRef]
  142. Wu, W.-C.; Sun, H.-W.; Chen, H.-T.; Liang, J.; Yu, X.-J.; Wu, C.; Wang, Z.; Zheng, L. Circulating hematopoietic stem and progenitor cells are myeloid-biased in cancer patients. Proc. Natl. Acad. Sci. USA 2014, 111, 4221–4226. [Google Scholar] [CrossRef] [Green Version]
  143. Al Sayed, M.F.; Amrein, M.A.; Bührer, E.D.; Huguenin, A.-L.; Radpour, R.; Riether, C.; Ochsenbein, A.F. T-cell–Secreted TNFα Induces Emergency Myelopoiesis and Myeloid-Derived Suppressor Cell Differentiation in Cancer. Cancer Res. 2019, 79, 346–359. [Google Scholar] [CrossRef] [Green Version]
  144. Wu, C.; Ning, H.; Liu, M.; Lin, J.; Luo, S.; Zhu, W.; Xu, J.; Wu, W.-C.; Liang, J.; Shao, C.-K.; et al. Spleen mediates a distinct hematopoietic progenitor response supporting tumor-promoting myelopoiesis. J. Clin. Invest. 2018, 128, 3425–3438. [Google Scholar] [CrossRef] [Green Version]
  145. Engblom, C.; Pfirschke, C.; Pittet, M.J. The role of myeloid cells in cancer therapies. Nat. Rev. Cancer 2016, 16, 447–462. [Google Scholar] [CrossRef]
  146. Giles, A.J.; Reid, C.M.; Evans, J.D.; Murgai, M.; Vicioso, Y.; Highfill, S.L.; Kasai, M.; Vahdat, L.; Mackall, C.L.; Lyden, D.; et al. Activation of Hematopoietic Stem/Progenitor Cells Promotes Immunosuppression Within the Pre–metastatic Niche. Cancer Res. 2016, 76, 1335–1347. [Google Scholar] [CrossRef] [Green Version]
  147. Sugiyama, T.; Kohara, H.; Noda, M.; Nagasawa, T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity 2006, 25, 977–988. [Google Scholar] [CrossRef] [Green Version]
  148. Glait-Santar, C.; Desmond, R.; Feng, X.; Bat, T.; Chen, J.; Heuston, E.; Mizukawa, B.; Mulloy, J.C.; Bodine, D.M.; Larochelle, A.; et al. Functional Niche Competition Between Normal Hematopoietic Stem and Progenitor Cells and Myeloid Leukemia Cells. Stem Cells 2015, 33, 3635–3642. [Google Scholar] [CrossRef] [Green Version]
  149. O’Donnell, R.K.; Falcon, B.; Hanson, J.; Goldstein, W.E.; Perruzzi, C.; Rafii, S.; Aird, W.C.; Benjamin, L.E. VEGF-A/VEGFR Inhibition Restores Hematopoietic Homeostasis in the Bone Marrow and Attenuates Tumor Growth. Cancer Res. 2016, 76, 517–524. [Google Scholar] [CrossRef] [Green Version]
  150. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [Green Version]
  151. Li, X.; Wang, S.; Zhu, R.; Li, H.; Han, Q.; Zhao, R.C. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J. Hematol. Oncol. 2016, 9, 42. [Google Scholar] [CrossRef] [Green Version]
  152. Zhang, B.; Ho, Y.W.; Huang, Q.; Maeda, T.; Lin, A.; Lee, S.U.; Hair, A.; Holyoake, T.L.; Huettner, C.; Bhatia, R. Altered microenvironmental regulation of leukemic and normal stem cells in chronic myelogenous leukemia. Cancer Cell 2012, 21, 577–592. [Google Scholar] [CrossRef] [Green Version]
  153. Lopes, M.; Duarte, T.L.; Teles, M.J.; Mosteo, L.; Chacim, S.; Aguiar, E.; Pereira-Reis, J.; Oliveira, M.; Silva, A.M.N.; Gonçalves, N.; et al. Loss of erythroblasts in acute myeloid leukemia causes iron redistribution with clinical implications. bioRxiv 2020. [Google Scholar] [CrossRef]
  154. Cheng, H.; Hao, S.; Liu, Y.; Pang, Y.; Ma, S.; Dong, F.; Xu, J.; Zheng, G.; Li, S.; Yuan, W.; et al. Leukemic marrow infiltration reveals a novel role for Egr3 as a potent inhibitor of normal hematopoietic stem cell proliferation. Blood 2015, 126, 1302–1313. [Google Scholar] [CrossRef]
  155. Bouchnita, A.; Eymard, N.; Moyo, T.K.; Koury, M.J.; Volpert, V. Bone marrow infiltration by multiple myeloma causes anemia by reversible disruption of erythropoiesis. Am. J. Hematol. 2016, 91, 371–378. [Google Scholar] [CrossRef] [Green Version]
  156. Haase, V.H. Hypoxic regulation of erythropoiesis and iron metabolism. Am. J. Physiol. Ren. Physiol. 2010, 299, F1–F13. [Google Scholar] [CrossRef] [Green Version]
  157. Dev, A.; Fang, J.; Sathyanarayana, P.; Pradeep, A.; Emerson, C.; Wojchowski, D.M. During EPO or anemia challenge, erythroid progenitor cells transit through a selectively expandable proerythroblast pool. Blood 2010, 116, 5334–5346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Jelkmann, W. Regulation of erythropoietin production. J. Physiol. 2011, 589, 1251–1258. [Google Scholar] [CrossRef] [PubMed]
  159. Viallard, C.; Larrivée, B. Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis 2017, 20, 409–426. [Google Scholar] [CrossRef]
  160. Yang, J.; Yan, J.; Liu, B. Targeting VEGF/VEGFR to Modulate Antitumor Immunity. Front. Immunol. 2018, 9, 978. [Google Scholar] [CrossRef] [Green Version]
  161. Kut, C.; Mac Gabhann, F.; Popel, A.S. Where is VEGF in the body? A meta-analysis of VEGF distribution in cancer. Br. J. Cancer 2007, 97, 978–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Xue, Y.; Lim, S.; Yang, Y.; Wang, Z.; Jensen, L.D.E.; Hedlund, E.-M.; Andersson, P.; Sasahara, M.; Larsson, O.; Galter, D.; et al. PDGF-BB modulates hematopoiesis and tumor angiogenesis by inducing erythropoietin production in stromal cells. Nat. Med. 2012, 18, 100–110. [Google Scholar] [CrossRef]
  163. Greenwald, A.C.; Licht, T.; Kumar, S.; Oladipupo, S.S.; Iyer, S.; Grunewald, M.; Keshet, E. VEGF expands erythropoiesis via hypoxia-independent induction of erythropoietin in noncanonical perivascular stromal cells. J. Exp. Med. 2019, 216, 215–230. [Google Scholar] [CrossRef] [Green Version]
  164. Liu, Y.; Pop, R.; Sadegh, C.; Brugnara, C.; Haase, V.H.; Socolovsky, M. Suppression of Fas-FasL coexpression by erythropoietin mediates erythroblast expansion during the erythropoietic stress response in vivo. Blood 2006, 108, 123–133. [Google Scholar] [CrossRef] [PubMed]
  165. Bordini, J.; Bertilaccio, M.T.; Ponzoni, M.; Fermo, I.; Chesi, M.; Bergsagel, P.L.; Camaschella, C.; Campanella, A. Erythroblast apoptosis and microenvironmental iron restriction trigger anemia in the VK*MYC model of multiple myeloma. Haematologica 2015, 100, 834–841. [Google Scholar] [CrossRef] [Green Version]
  166. Testa, U. Apoptotic mechanisms in the control of erythropoiesis. Leukemia 2004, 18, 1176–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Gregory, T.; Yu, C.; Ma, A.; Orkin, S.H.; Blobel, G.A.; Weiss, M.J. GATA-1 and erythropoietin cooperate to promote erythroid cell survival by regulating bcl-xL expression. Blood 1999, 94, 87–96. [Google Scholar] [CrossRef]
  168. Tanaka, H.; Matsumura, I.; Nakajima, K.; Daino, H.; Sonoyama, J.; Yoshida, H.; Oritani, K.; Machii, T.; Yamamoto, M.; Hirano, T.; et al. GATA-1 blocks IL-6-induced macrophage differentiation and apoptosis through the sustained expression of cyclin D1 and bcl-2 in a murine myeloid cell line M1. Blood 2000, 95, 1264–1273. [Google Scholar] [CrossRef]
  169. Spierings, D.C.J.; de Vries, E.G.E.; Timens, W.; Groen, H.J.M.; Boezen, H.M.; de Jong, S. Expression of TRAIL and TRAIL Death Receptors in Stage III Non-Small Cell Lung Cancer Tumors. Clin. Cancer Res. 2003, 9, 3397–3405. [Google Scholar]
  170. Secchiero, P.; Melloni, E.; Heikinheimo, M.; Mannisto, S.; Di Pietro, R.; Iacone, A.; Zauli, G. TRAIL regulates normal erythroid maturation through an ERK-dependent pathway. Blood 2004, 103, 517–522. [Google Scholar] [CrossRef] [Green Version]
  171. Zamai, L.; Secchiero, P.; Pierpaoli, S.; Bassini, A.; Papa, S.; Alnemri, E.S.; Guidotti, L.; Vitale, M.; Zauli, G. TNF-related apoptosis-inducing ligand (TRAIL) as a negative regulator of normal human erythropoiesis. Blood 2000, 95, 3716–3724. [Google Scholar]
  172. Batlle, E.; Massagué, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef] [PubMed]
  173. González-Santiago, A.E.; Mendoza-Topete, L.A.; Sánchez-Llamas, F.; Troyo-Sanromán, R.; Gurrola-Díaz, C.M. TGF-β1 serum concentration as a complementary diagnostic biomarker of lung cancer: Establishment of a cut-point value. J. Clin. Lab. Anal. 2011, 25, 238–243. [Google Scholar] [CrossRef] [PubMed]
  174. Shirai, Y.; Kawata, S.; Tamura, S.; Ito, N.; Tsushima, H.; Takaishi, K.; Kiso, S.; Matsuzawa, Y. Plasma transforming growth factor-beta 1 in patients with hepatocellular carcinoma. Comparison with chronic liver diseases. Cancer 1994, 73, 2275–2279. [Google Scholar] [CrossRef]
  175. Zermati, Y.; Fichelson, S.; Valensi, F.; Freyssinier, J.M.; Rouyer-Fessard, P.; Cramer, E.; Guichard, J.; Varet, B.; Hermine, O. Transforming growth factor inhibits erythropoiesis by blocking proliferation and accelerating differentiation of erythroid progenitors. Exp. Hematol. 2000, 28, 885–894. [Google Scholar] [CrossRef]
  176. Kuhikar, R.; Khan, N.; Philip, J.; Melinkeri, S.; Kale, V.; Limaye, L. Transforming growth factor β1 accelerates and enhances in vitro red blood cell formation from hematopoietic stem cells by stimulating mitophagy. Stem Cell Res. Ther. 2020, 11, 71. [Google Scholar] [CrossRef]
  177. Akel, S.; Petrow-Sadowski, C.; Laughlin, M.J.; Ruscetti, F.W. Neutralization of autocrine transforming growth factor-beta in human cord blood CD34+CD38Lin cells promotes stem-cell-factor-mediated erythropoietin-independent early erythroid progenitor development and reduces terminal differentiation. Stem Cells 2003, 21, 557–567. [Google Scholar] [CrossRef] [PubMed]
  178. Gao, X.; Lee, H.Y.; da Rocha, E.L.; Zhang, C.; Lu, Y.F.; Li, D.; Feng, Y.; Ezike, J.; Elmes, R.R.; Barrasa, M.I.; et al. TGF-β inhibitors stimulate red blood cell production by enhancing self-renewal of BFU-E erythroid progenitors. Blood 2016, 128, 2637–2641. [Google Scholar] [CrossRef] [PubMed]
  179. Taniguchi, S.; Elhance, A.; Van Duzer, A.; Kumar, S.; Leitenberger, J.J.; Oshimori, N. Tumor-initiating cells establish an IL-33–TGF-β niche signaling loop to promote cancer progression. Science 2020, 369, eaay1813. [Google Scholar] [CrossRef]
  180. Fournié, J.-J.; Poupot, M. The Pro-tumorigenic IL-33 Involved in Antitumor Immunity: A Yin and Yang Cytokine. Front. Immunol. 2018, 9, 2506. [Google Scholar] [CrossRef] [Green Version]
  181. Swann, J.W.; Koneva, L.A.; Regan-Komito, D.; Sansom, S.N.; Powrie, F.; Griseri, T. IL-33 promotes anemia during chronic inflammation by inhibiting differentiation of erythroid progenitors. J. Exp. Med. 2020, 217, e20200164. [Google Scholar] [CrossRef] [PubMed]
  182. Suragani, R.N.V.S.; Cadena, S.M.; Cawley, S.M.; Sako, D.; Mitchell, D.; Li, R.; Davies, M.V.; Alexander, M.J.; Devine, M.; Loveday, K.S.; et al. Transforming growth factor-β superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat. Med. 2014, 20, 408–414. [Google Scholar] [CrossRef] [PubMed]
  183. Martinez, P.A.; Li, R.; Ramanathan, H.N.; Bhasin, M.; Pearsall, R.S.; Kumar, R.; Suragani, R.N.V.S. Smad2/3-pathway ligand trap luspatercept enhances erythroid differentiation in murine β-thalassaemia by increasing GATA-1 availability. J. Cell. Mol. Med. 2020, 24, 6162–6177. [Google Scholar] [CrossRef] [PubMed]
  184. Dussiot, M.; Maciel, T.T.; Fricot, A.; Chartier, C.; Negre, O.; Veiga, J.; Grapton, D.; Paubelle, E.; Payen, E.; Beuzard, Y.; et al. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia. Nat. Med. 2014, 20, 398–407. [Google Scholar] [CrossRef] [PubMed]
  185. Han, Y.; Wang, H.; Shao, Z. GDF11 Increased in Patients with Myelodysplastic Syndrome. Blood 2015, 126, 5224. [Google Scholar] [CrossRef]
  186. Tanno, T.; Bhanu, N.V.; Oneal, P.A.; Goh, S.H.; Staker, P.; Lee, Y.T.; Moroney, J.W.; Reed, C.H.; Luban, N.L.; Wang, R.H.; et al. High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Nat. Med. 2007, 13, 1096–1101. [Google Scholar] [CrossRef]
  187. Lenox, L.E.; Perry, J.M.; Paulson, R.F. BMP4 and Madh5 regulate the erythroid response to acute anemia. Blood 2005, 105, 2741–2748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Perry, J.M.; Harandi, O.F.; Paulson, R.F. BMP4, SCF, and hypoxia cooperatively regulate the expansion of murine stress erythroid progenitors. Blood 2007, 109, 4494–4502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Kim, A.; Nemeth, E. New insights into iron regulation and erythropoiesis. Curr. Opin. Hematol. 2015, 22, 199–205. [Google Scholar] [CrossRef]
  190. Clara, C.; Antonella, N.; Laura, S. Iron metabolism and iron disorders revisited in the hepcidin era. Haematologica 2020, 105, 260–272. [Google Scholar] [CrossRef] [Green Version]
  191. Ludwig, H.; Müldür, E.; Endler, G.; Hübl, W. Prevalence of iron deficiency across different tumors and its association with poor performance status, disease status and anemia. Ann. Oncol. 2013, 24, 1886–1892. [Google Scholar] [CrossRef]
  192. Liu, S.; Bhattacharya, S.; Han, A.; Suragani, R.N.; Zhao, W.; Fry, R.C.; Chen, J.J. Haem-regulated eIF2alpha kinase is necessary for adaptive gene expression in erythroid precursors under the stress of iron deficiency. Br. J. Haematol. 2008, 143, 129–137. [Google Scholar] [CrossRef] [Green Version]
  193. Masahiro, K.; Hiroki, K.; Hiroshi, H.; Ari, I.-N.; Tohru, F.; Akihiko, M.; Yukihiro, I.; Kenji, I.; Wataru, H.; Naohisa, T.; et al. Iron-heme-Bach1 axis is involved in erythroblast adaptation to iron deficiency. Haematologica 2017, 102, 454–465. [Google Scholar] [CrossRef] [Green Version]
  194. Bullock, G.C.; Delehanty, L.L.; Talbot, A.-L.; Gonias, S.L.; Tong, W.-H.; Rouault, T.A.; Dewar, B.; Macdonald, J.M.; Chruma, J.J.; Goldfarb, A.N. Iron control of erythroid development by a novel aconitase-associated regulatory pathway. Blood 2010, 116, 97–108. [Google Scholar] [CrossRef] [Green Version]
  195. Shaw, G.C.; Cope, J.J.; Li, L.; Corson, K.; Hersey, C.; Ackermann, G.E.; Gwynn, B.; Lambert, A.J.; Wingert, R.A.; Traver, D.; et al. Mitoferrin is essential for erythroid iron assimilation. Nature 2006, 440, 96–100. [Google Scholar] [CrossRef]
  196. Schranzhofer, M.; Schifrer, M.; Cabrera, J.A.; Kopp, S.; Chiba, P.; Beug, H.; Müllner, E.W. Remodeling the regulation of iron metabolism during erythroid differentiation to ensure efficient heme biosynthesis. Blood 2006, 107, 4159–4167. [Google Scholar] [CrossRef]
  197. Brown, R.A.M.; Richardson, K.L.; Kabir, T.D.; Trinder, D.; Ganss, R.; Leedman, P.J. Altered Iron Metabolism and Impact in Cancer Biology, Metastasis, and Immunology. Front. Oncol. 2020, 10, 476. [Google Scholar] [CrossRef]
  198. Pfeifhofer-Obermair, C.; Tymoszuk, P.; Petzer, V.; Weiss, G.; Nairz, M. Iron in the Tumor Microenvironment—Connecting the Dots. Front. Oncol. 2018, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Daniels, T.R.; Delgado, T.; Rodriguez, J.A.; Helguera, G.; Penichet, M.L. The transferrin receptor part I: Biology and targeting with cytotoxic antibodies for the treatment of cancer. Clin. Immunol. 2006, 121, 144–158. [Google Scholar] [CrossRef] [PubMed]
  200. Ganz, T. Anemia of Inflammation. N. Engl. J. Med. 2019, 381, 1148–1157. [Google Scholar] [CrossRef]
  201. Weiss, G.; Ganz, T.; Goodnough, L.T. Anemia of inflammation. Blood 2019, 133, 40–50. [Google Scholar] [CrossRef] [Green Version]
  202. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 36. [Google Scholar] [CrossRef]
  203. Tyrkalska, S.D.; Pérez-Oliva, A.B.; Rodríguez-Ruiz, L.; Martínez-Morcillo, F.J.; Alcaraz-Pérez, F.; Martínez-Navarro, F.J.; Lachaud, C.; Ahmed, N.; Schroeder, T.; Pardo-Sánchez, I.; et al. Inflammasome Regulates Hematopoiesis through Cleavage of the Master Erythroid Transcription Factor GATA1. Immunity 2019, 51, 50–63. [Google Scholar] [CrossRef] [PubMed]
  204. Hamarsheh, S.; Osswald, L.; Saller, B.S.; Unger, S.; De Feo, D.; Vinnakota, J.M.; Konantz, M.; Uhl, F.M.; Becker, H.; Lübbert, M.; et al. Oncogenic KrasG12D causes myeloproliferation via NLRP3 inflammasome activation. Nat. Commun. 2020, 11, 1659. [Google Scholar] [CrossRef] [Green Version]
  205. Prince, O.D.; Langdon, J.M.; Layman, A.J.; Prince, I.C.; Sabogal, M.; Mak, H.H.; Berger, A.E.; Cheadle, C.; Chrest, F.J.; Yu, Q.; et al. Late stage erythroid precursor production is impaired in mice with chronic inflammation. Haematologica 2012, 97, 1648–1656. [Google Scholar] [CrossRef] [PubMed]
  206. Ivashkiv, L.B. IFNγ: Signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 545–558. [Google Scholar] [CrossRef]
  207. Libregts, S.F.; Gutiérrez, L.; de Bruin, A.M.; Wensveen, F.M.; Papadopoulos, P.; van Ijcken, W.; Ozgür, Z.; Philipsen, S.; Nolte, M.A. Chronic IFN-γ production in mice induces anemia by reducing erythrocyte life span and inhibiting erythropoiesis through an IRF-1/PU.1 axis. Blood 2011, 118, 2578–2588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Dai, C.; Krantz, S.B. Interferon γ Induces Upregulation and Activation of Caspases 1, 3, and 8 to Produce Apoptosis in Human Erythroid Progenitor Cells. Blood 1999, 93, 3309–3316. [Google Scholar] [CrossRef]
  209. Wang, C.Q.; Udupa, K.B.; Lipschitz, D.A. Interferon-γ exerts its negative regulatory effect primarily on the earliest stages of murine erythroid progenitor cell development. J. Cell. Physiol. 1995, 162, 134–138. [Google Scholar] [CrossRef]
  210. Dai, C.-H.; Price, J.O.; Brunner, T.; Krantz, S.B. Fas Ligand Is Present in Human Erythroid Colony-Forming Cells and Interacts with Fas Induced by Interferon γ to Produce Erythroid Cell Apoptosis. Blood 1998, 91, 1235–1242. [Google Scholar] [CrossRef] [Green Version]
  211. Zhang, P.; Zhang, X.; Iwama, A.; Yu, C.; Smith, K.A.; Mueller, B.U.; Narravula, S.; Torbett, B.E.; Orkin, S.H.; Tenen, D.G. PU. 1 inhibits GATA-1 function and erythroid differentiation by blocking GATA-1 DNA binding. Blood 2000, 96, 2641–2648. [Google Scholar] [CrossRef]
  212. Yamada, T.; Kondoh, N.; Matsumoto, M.; Yoshida, M.; Maekawa, A.; Oikawa, T. Overexpression of PU.1 induces growth and differentiation inhibition and apoptotic cell death in murine erythroleukemia cells. Blood 1997, 89, 1383–1393. [Google Scholar] [CrossRef]
  213. Burda, P.; Laslo, P.; Stopka, T. The role of PU.1 and GATA-1 transcription factors during normal and leukemogenic hematopoiesis. Leukemia 2010, 24, 1249–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Montfort, A.; Colacios, C.; Levade, T.; Andrieu-Abadie, N.; Meyer, N.; Ségui, B. The TNF Paradox in Cancer Progression and Immunotherapy. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  215. Rusten, L.S.; Jacobsen, S.E. Tumor necrosis factor (TNF)-alpha directly inhibits human erythropoiesis in vitro: Role of p55 and p75 TNF receptors. Blood 1995, 85, 989–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Tsopra, O.A.; Ziros, P.G.; Lagadinou, E.D.; Symeonidis, A.; Kouraklis-Symeonidis, A.; Thanopoulou, E.; Angelopoulou, M.K.; Vassilakopoulos, T.P.; Pangalis, G.A.; Zoumbos, N.C. Disease-related anemia in chronic lymphocytic leukemia is not due to intrinsic defects of erythroid precursors: A possible pathogenetic role for tumor necrosis factor-alpha. Acta Haematol. 2009, 121, 187–195. [Google Scholar] [CrossRef]
  217. Papadaki, H.A.; Kritikos, H.D.; Valatas, V.; Boumpas, D.T.; Eliopoulos, G.D. Anemia of chronic disease in rheumatoid arthritis is associated with increased apoptosis of bone marrow erythroid cells: Improvement following anti–tumor necrosis factor-α antibody therapy. Blood 2002, 100, 474–482. [Google Scholar] [CrossRef] [PubMed]
  218. Jacobs-Helber, S.M.; Roh, K.-H.; Bailey, D.; Dessypris, E.N.; Ryan, J.J.; Chen, J.; Wickrema, A.; Barber, D.L.; Dent, P.; Sawyer, S.T. Tumor necrosis factor-alpha expressed constitutively in erythroid cells or induced by erythropoietin has negative and stimulatory roles in normal erythropoiesis and erythroleukemia. Blood 2003, 101, 524–531. [Google Scholar] [CrossRef]
  219. Bibikova, E.; Youn, M.-Y.; Danilova, N.; Ono-Uruga, Y.; Konto-Ghiorghi, Y.; Ochoa, R.; Narla, A.; Glader, B.; Lin, S.; Sakamoto, K.M. TNF-mediated inflammation represses GATA1 and activates p38 MAP kinase in RPS19-deficient hematopoietic progenitors. Blood 2014, 124, 3791–3798. [Google Scholar] [CrossRef] [PubMed]
  220. Hernandez-Hernandez, A.; Ray, P.; Litos, G.; Ciro, M.; Ottolenghi, S.; Beug, H.; Boyes, J. Acetylation and MAPK phosphorylation cooperate to regulate the degradation of active GATA-1. EMBO J. 2006, 25, 3264–3274. [Google Scholar] [CrossRef] [Green Version]
  221. Manso, B.A.; Krull, J.E.; Gwin, K.A.; Lothert, P.K.; Welch, B.M.; Novak, A.J.; Parikh, S.A.; Kay, N.E.; Medina, K.L. Chronic lymphocytic leukemia B-cell-derived TNFα impairs bone marrow myelopoiesis. iScience 2021, 24, 101994. [Google Scholar] [CrossRef] [PubMed]
  222. Schooley, J.C.; Kullgren, B.; Allison, A.C. Inhibition by interleukin-1 of the action of erythropoietin on erythroid precursors and its possible role in the pathogenesis of hypoplastic anaemias. Br. J. Haematol. 1987, 67, 11–17. [Google Scholar] [CrossRef] [PubMed]
  223. Chou, D.B.; Sworder, B.; Bouladoux, N.; Roy, C.N.; Uchida, A.M.; Grigg, M.; Robey, P.G.; Belkaid, Y. Stromal-derived IL-6 alters the balance of myeloerythroid progenitors during Toxoplasma gondii infection. J. Leukoc. Biol. 2012, 92, 123–131. [Google Scholar] [CrossRef] [Green Version]
  224. Gołab, J.; Zagozdzon, R.; Stokłosa, T.; Jakóbisiak, M.; Pojda, Z.; Machaj, E. Erythropoietin prevents the development of interleukin-12-induced anemia and thrombocytopenia but does not decrease its antitumor activity in mice. Blood 1998, 91, 4387–4388. [Google Scholar] [CrossRef]
  225. De la Fuente López, M.; Landskron, G.; Parada, D.; Dubois-Camacho, K.; Simian, D.; Martinez, M.; Romero, D.; Roa, J.C.; Chahuán, I.; Gutiérrez, R.; et al. The relationship between chemokines CCL2, CCL3, and CCL4 with the tumor microenvironment and tumor-associated macrophage markers in colorectal cancer. Tumor Biol. 2018, 40, 1010428318810059. [Google Scholar] [CrossRef] [Green Version]
  226. Buck, I.; Morceau, F.; Cristofanon, S.; Heintz, C.; Chateauvieux, S.; Reuter, S.; Dicato, M.; Diederich, M. Tumor necrosis factor α inhibits erythroid differentiation in human erythropoietin-dependent cells involving p38 MAPK pathway, GATA-1 and FOG-1 downregulation and GATA-2 upregulation. Biochem. Pharmacol. 2008, 76, 1229–1239. [Google Scholar] [CrossRef]
  227. Liao, C.; Prabhu, K.S.; Paulson, R.F. Monocyte-derived macrophages expand the murine stress erythropoietic niche during the recovery from anemia. Blood 2018, 132, 2580–2593. [Google Scholar] [CrossRef] [Green Version]
  228. Millot, S.; Andrieu, V.; Letteron, P.; Lyoumi, S.; Hurtado-Nedelec, M.; Karim, Z.; Thibaudeau, O.; Bennada, S.; Charrier, J.L.; Lasocki, S.; et al. Erythropoietin stimulates spleen BMP4-dependent stress erythropoiesis and partially corrects anemia in a mouse model of generalized inflammation. Blood 2010, 116, 6072–6081. [Google Scholar] [CrossRef] [PubMed]
  229. Bennett, L.F.; Liao, C.; Quickel, M.D.; Yeoh, B.S.; Vijay-Kumar, M.; Hankey-Giblin, P.; Prabhu, K.S.; Paulson, R.F. Inflammation induces stress erythropoiesis through heme-dependent activation of SPI-C. Sci. Signal. 2019, 12, eaap7336. [Google Scholar] [CrossRef]
  230. Corazza, F.; Beguin, Y.; Bergmann, P.; André, M.; Ferster, A.; Devalck, C.; Fondu, P.; Buyse, M.; Sariban, E. Anemia in children with cancer is associated with decreased erythropoietic activity and not with inadequate erythropoietin production. Blood 1998, 92, 1793–1798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Chen, Y.; Xiang, J.; Qian, F.; Diwakar, B.T.; Ruan, B.; Hao, S.; Prabhu, K.S.; Paulson, R.F. Epo receptor signaling in macrophages alters the splenic niche to promote erythroid differentiation. Blood 2020, 136, 235–246. [Google Scholar] [CrossRef]
  232. Song, X.; Krelin, Y.; Dvorkin, T.; Bjorkdahl, O.; Segal, S.; Dinarello, C.A.; Voronov, E.; Apte, R.N. CD11b+/Gr-1+ Immature Myeloid Cells Mediate Suppression of T Cells in Mice Bearing Tumors of IL-1β-Secreting Cells. J. Immunol. 2005, 175, 8200–8208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Jing, W.; Guo, X.; Qin, F.; Li, Y.; Wang, G.; Bi, Y.; Jin, X.; Han, L.; Dong, X.; Zhao, Y. G-CSF shifts erythropoiesis from bone marrow into spleen in the setting of systemic inflammation. Life Sci. Alliance 2021, 4, e202000737. [Google Scholar] [CrossRef] [PubMed]
  234. McKim, D.B.; Yin, W.; Wang, Y.; Cole, S.W.; Godbout, J.P.; Sheridan, J.F. Social Stress Mobilizes Hematopoietic Stem Cells to Establish Persistent Splenic Myelopoiesis. Cell Rep. 2018, 25, 2552–2562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Alamo, I.G.; Kannan, K.B.; Loftus, T.J.; Ramos, H.; Efron, P.A.; Mohr, A.M. Severe trauma and chronic stress activates extramedullary erythropoiesis. J. Trauma Acute Care Surg. 2017, 83, 144–150. [Google Scholar] [CrossRef]
  236. Zeuner, A.; Pedini, F.; Signore, M.; Testa, U.; Pelosi, E.; Peschle, C.; De Maria, R. Stem cell factor protects erythroid precursor cells from chemotherapeutic agents via up-regulation of BCL-2 family proteins. Blood 2003, 102, 87–93. [Google Scholar] [CrossRef] [Green Version]
  237. Bartucci, M.; Dattilo, R.; Martinetti, D.; Todaro, M.; Zapparelli, G.; Di Virgilio, A.; Biffoni, M.; De Maria, R.; Zeuner, A. Prevention of Chemotherapy-Induced Anemia and Thrombocytopenia by Constant Administration of Stem Cell Factor. Clin. Cancer Res. 2011, 17, 6185–6191. [Google Scholar] [CrossRef] [Green Version]
  238. Gao, P.; Zhang, H.; Dinavahi, R.; Li, F.; Xiang, Y.; Raman, V.; Bhujwalla, Z.M.; Felsher, D.W.; Cheng, L.; Pevsner, J.; et al. HIF-dependent antitumorigenic effect of antioxidants in vivo. Cancer Cell 2007, 12, 230–238. [Google Scholar] [CrossRef] [Green Version]
  239. Le Gal, K.; Ibrahim, M.X.; Wiel, C.; Sayin, V.I.; Akula, M.K.; Karlsson, C.; Dalin, M.G.; Akyürek, L.M.; Lindahl, P.; Nilsson, J.; et al. Antioxidants can increase melanoma metastasis in mice. Sci. Transl. Med. 2015, 7, 308re308. [Google Scholar] [CrossRef]
  240. Sayin, V.I.; Ibrahim, M.X.; Larsson, E.; Nilsson, J.A.; Lindahl, P.; Bergo, M.O. Antioxidants Accelerate Lung Cancer Progression in Mice. Sci. Transl. Med. 2014, 6, 221ra215. [Google Scholar] [CrossRef]
  241. Reczek, C.R.; Chandel, N.S. The Two Faces of Reactive Oxygen Species in Cancer. Annu. Rev. Cancer Biol. 2017, 1, 79–98. [Google Scholar] [CrossRef]
  242. Firczuk, M.; Bajor, M.; Graczyk-Jarzynka, A.; Fidyt, K.; Goral, A.; Zagozdzon, R. Harnessing altered oxidative metabolism in cancer by augmented prooxidant therapy. Cancer Lett. 2020, 471, 1–11. [Google Scholar] [CrossRef] [PubMed]
  243. Naing, A.; Infante, J.R.; Papadopoulos, K.P.; Chan, I.H.; Shen, C.; Ratti, N.P.; Rojo, B.; Autio, K.A.; Wong, D.J.; Patel, M.R.; et al. PEGylated IL-10 (Pegilodecakin) Induces Systemic Immune Activation, CD8+ T Cell Invigoration and Polyclonal T Cell Expansion in Cancer Patients. Cancer Cell 2018, 34, 775–791. [Google Scholar] [CrossRef] [Green Version]
  244. Vahl, J.M.; Friedrich, J.; Mittler, S.; Trump, S.; Heim, L.; Kachler, K.; Balabko, L.; Fuhrich, N.; Geppert, C.-I.; Trufa, D.I.; et al. Interleukin-10-regulated tumour tolerance in non-small cell lung cancer. Br. J. Cancer 2017, 117, 1644–1655. [Google Scholar] [CrossRef] [Green Version]
  245. Qiao, J.; Liu, Z.; Dong, C.; Luan, Y.; Zhang, A.; Moore, C.; Fu, K.; Peng, J.; Wang, Y.; Ren, Z.; et al. Targeting Tumors with IL-10 Prevents Dendritic Cell-Mediated CD8+ T Cell Apoptosis. Cancer Cell 2019, 35, 901–915. [Google Scholar] [CrossRef]
  246. Oft, M. IL-10: Master switch from tumor-promoting inflammation to antitumor immunity. Cancer Immunol. Res. 2014, 2, 194–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Mumm, J.B.; Emmerich, J.; Zhang, X.; Chan, I.; Wu, L.; Mauze, S.; Blaisdell, S.; Basham, B.; Dai, J.; Grein, J.; et al. IL-10 elicits IFNγ-dependent tumor immune surveillance. Cancer Cell 2011, 20, 781–796. [Google Scholar] [CrossRef] [Green Version]
  248. Oft, M. Immune regulation and cytotoxic T cell activation of IL-10 agonists—Preclinical and clinical experience. Semin. Immunol. 2019, 44, 101325. [Google Scholar] [CrossRef] [PubMed]
  249. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [Green Version]
  250. Gong, J.; Chehrazi-Raffle, A.; Reddi, S.; Salgia, R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: A comprehensive review of registration trials and future considerations. J. Immunother. Cancer 2018, 6, 8. [Google Scholar] [CrossRef]
  251. Chamoto, K.; Hatae, R.; Honjo, T. Current issues and perspectives in PD-1 blockade cancer immunotherapy. Int. J. Clin. Oncol. 2020, 25, 790–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  253. Li, S.; Liu, M.; Do, M.H.; Chou, C.; Stamatiades, E.G.; Nixon, B.G.; Shi, W.; Zhang, X.; Li, P.; Gao, S.; et al. Cancer immunotherapy via targeted TGF-β signalling blockade in TH cells. Nature 2020, 587, 121–125. [Google Scholar] [CrossRef] [PubMed]
  254. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Groeneveldt, C.; van Hall, T.; van der Burg, S.H.; Ten Dijke, P.; van Montfoort, N. Immunotherapeutic Potential of TGF-β Inhibition and Oncolytic Viruses. Trends Immunol. 2020, 41, 406–420. [Google Scholar] [CrossRef]
  256. DeBerry, J.J.; Saloman, J.L.; Dragoo, B.K.; Albers, K.M.; Davis, B.M. Artemin Immunotherapy Is Effective in Preventing and Reversing Cystitis-Induced Bladder Hyperalgesia via TRPA1 Regulation. J. Pain 2015, 16, 628–636. [Google Scholar] [CrossRef] [Green Version]
  257. Bohlius, J.; Bohlke, K.; Lazo-Langner, A. Management of Cancer-Associated Anemia With Erythropoiesis-Stimulating Agents: ASCO/ASH Clinical Practice Guideline Update. J. Oncol. Pract. 2019, 15, 399–402. [Google Scholar] [CrossRef] [Green Version]
  258. Aapro, M.; Beguin, Y.; Bokemeyer, C.; Dicato, M.; Gascón, P.; Glaspy, J.; Hofmann, A.; Link, H.; Littlewood, T.; Ludwig, H.; et al. Management of anaemia and iron deficiency in patients with cancer: ESMO Clinical Practice Guidelines. Ann. Oncol. 2018, 29, iv96–iv110. [Google Scholar] [CrossRef]
  259. Katsarou, A.; Pantopoulos, K. Hepcidin Therapeutics. Pharmaceuticals 2018, 11, 127. [Google Scholar] [CrossRef] [Green Version]
  260. Zhou, L.; Nguyen, A.N.; Sohal, D.; Ying Ma, J.; Pahanish, P.; Gundabolu, K.; Hayman, J.; Chubak, A.; Mo, Y.; Bhagat, T.D.; et al. Inhibition of the TGF-beta receptor I kinase promotes hematopoiesis in MDS. Blood 2008, 112, 3434–3443. [Google Scholar] [CrossRef] [Green Version]
  261. Verma, A.; Suragani, R.N.V.S.; Aluri, S.; Shah, N.; Bhagat, T.D.; Alexander, M.J.; Komrokji, R.; Kumar, R. Biological basis for efficacy of activin receptor ligand traps in myelodysplastic syndromes. J. Clin. Invest. 2020, 130, 582–589. [Google Scholar] [CrossRef]
  262. Teixeira, A.F.; ten Dijke, P.; Zhu, H.-J. On-Target Anti-TGF-β Therapies Are Not Succeeding in Clinical Cancer Treatments: What Are Remaining Challenges? Front. Cell Dev. Biol. 2020, 8. [Google Scholar] [CrossRef]
  263. Hu, J.; Liu, J.; Xue, F.; Halverson, G.; Reid, M.; Guo, A.; Chen, L.; Raza, A.; Galili, N.; Jaffray, J.; et al. Isolation and functional characterization of human erythroblasts at distinct stages: Implications for understanding of normal and disordered erythropoiesis in vivo. Blood 2013, 121, 3246–3253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Elvarsdóttir, E.M.; Mortera-Blanco, T.; Dimitriou, M.; Bouderlique, T.; Jansson, M.; Hofman, I.J.F.; Conte, S.; Karimi, M.; Sander, B.; Douagi, I.; et al. A three-dimensional in vitro model of erythropoiesis recapitulates erythroid failure in myelodysplastic syndromes. Leukemia 2020, 34, 271–282. [Google Scholar] [CrossRef] [Green Version]
  265. Fenaux, P.; Platzbecker, U.; Mufti, G.J.; Garcia-Manero, G.; Buckstein, R.; Santini, V.; Díez-Campelo, M.; Finelli, C.; Cazzola, M.; Ilhan, O.; et al. Luspatercept in Patients with Lower-Risk Myelodysplastic Syndromes. N. Engl. J. Med. 2020, 382, 140–151. [Google Scholar] [CrossRef] [PubMed]
  266. Cappellini, M.D.; Viprakasit, V.; Taher, A.T.; Georgiev, P.; Kuo, K.H.M.; Coates, T.; Voskaridou, E.; Liew, H.-K.; Pazgal-Kobrowski, I.; Forni, G.L.; et al. A Phase 3 Trial of Luspatercept in Patients with Transfusion-Dependent β-Thalassemia. N. Engl. J. Med. 2020, 382, 1219–1231. [Google Scholar] [CrossRef]
  267. Cappellini, M.D.; Porter, J.; Origa, R.; Forni, G.L.; Voskaridou, E.; Galactéros, F.; Taher, A.T.; Arlet, J.-B.; Ribeil, J.-A.; Garbowski, M.; et al. Sotatercept, a novel transforming growth factor β ligand trap, improves anemia in β-thalassemia: A phase II, open-label, dose-finding study. Haematologica 2019, 104, 477–484. [Google Scholar] [CrossRef]
  268. Raftopoulos, H.; Laadem, A.; Hesketh, P.J.; Goldschmidt, J.; Gabrail, N.; Osborne, C.; Ali, M.; Sherman, M.L.; Wang, D.; Glaspy, J.A.; et al. Sotatercept (ACE-011) for the treatment of chemotherapy-induced anemia in patients with metastatic breast cancer or advanced or metastatic solid tumors treated with platinum-based chemotherapeutic regimens: Results from two phase 2 studies. Support. Care Cancer 2016, 24, 1517–1525. [Google Scholar] [CrossRef] [Green Version]
  269. Morrell, N.W.; Bloch, D.B.; ten Dijke, P.; Goumans, M.-J.T.H.; Hata, A.; Smith, J.; Yu, P.B.; Bloch, K.D. Targeting BMP signalling in cardiovascular disease and anaemia. Nat. Rev. Cardiol. 2016, 13, 106–120. [Google Scholar] [CrossRef] [Green Version]
  270. Steinbicker, A.U.; Sachidanandan, C.; Vonner, A.J.; Yusuf, R.Z.; Deng, D.Y.; Lai, C.S.; Rauwerdink, K.M.; Winn, J.C.; Saez, B.; Cook, C.M.; et al. Inhibition of bone morphogenetic protein signaling attenuates anemia associated with inflammation. Blood 2011, 117, 4915–4923. [Google Scholar] [CrossRef] [Green Version]
  271. Mayeur, C.; Kolodziej, S.A.; Wang, A.; Xu, X.; Lee, A.; Yu, P.B.; Shen, J.; Bloch, K.D.; Bloch, D.B. Oral administration of a bone morphogenetic protein type I receptor inhibitor prevents the development of anemia of inflammation. Haematologica 2015, 100, e68–e71. [Google Scholar] [CrossRef] [Green Version]
  272. Wannamaker, W.; Davies, R.; Namchuk, M.; Pollard, J.; Ford, P.; Ku, G.; Decker, C.; Charifson, P.; Weber, P.; Germann, U.A.; et al. (S)-1-((S)-2-{[1 -(4-amino-3-chloro-phenyl)-methanoyl]-amino}-3,3-dimethyl-butanoyl)-pyrrolidine-2-carboxylic acid ((2R,3S)-2-ethoxy-5-oxo-tetrahydro-furan-3-yl)-amide (VX-765), an orally available selective interleukin (IL)-converting enzyme/caspase-1 inhibitor, exhibits potent anti-inflammatory activities by inhibiting the release of IL-1beta and IL-18. J. Pharm. Exp. Ther. 2007, 321, 509–516. [Google Scholar] [CrossRef]
  273. Hu, P.; Nebreda, A.R.; Hanenberg, H.; Kinnebrew, G.H.; Ivan, M.; Yoder, M.C.; Filippi, M.-D.; Broxmeyer, H.E.; Kapur, R. P38α/JNK signaling restrains erythropoiesis by suppressing Ezh2-mediated epigenetic silencing of Bim. Nat. Commun. 2018, 9, 3518. [Google Scholar] [CrossRef]
  274. Martínez-Limón, A.; Joaquin, M.; Caballero, M.; Posas, F.; de Nadal, E. The p38 Pathway: From Biology to Cancer Therapy. Int. J. Mol. Sci. 2020, 21, 1913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Tamura, K.; Sudo, T.; Senftleben, U.; Dadak, A.M.; Johnson, R.; Karin, M. Requirement for p38α in Erythropoietin Expression: A Role for Stress Kinases in Erythropoiesis. Cell 2000, 102, 221–231. [Google Scholar] [CrossRef] [Green Version]
  276. Kuhrt, D.; Wojchowski, D.M. Emerging EPO and EPO receptor regulators and signal transducers. Blood 2015, 125, 3536–3541. [Google Scholar] [CrossRef] [Green Version]
  277. James, C.; Ugo, V.; Le Couédic, J.-P.; Staerk, J.; Delhommeau, F.; Lacout, C.; Garçon, L.; Raslova, H.; Berger, R.; Bennaceur-Griscelli, A.; et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005, 434, 1144–1148. [Google Scholar] [CrossRef]
  278. Libani, I.V.; Guy, E.C.; Melchiori, L.; Schiro, R.; Ramos, P.; Breda, L.; Scholzen, T.; Chadburn, A.; Liu, Y.; Kernbach, M.; et al. Decreased differentiation of erythroid cells exacerbates ineffective erythropoiesis in beta-thalassemia. Blood 2008, 112, 875–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  279. Talpaz, M.; Kiladjian, J.-J. Fedratinib, a newly approved treatment for patients with myeloproliferative neoplasm-associated myelofibrosis. Leukemia 2021, 35, 1–17. [Google Scholar] [CrossRef]
  280. Hosseini, A.; Gharibi, T.; Marofi, F.; Javadian, M.; Babaloo, Z.; Baradaran, B. Janus kinase inhibitors: A therapeutic strategy for cancer and autoimmune diseases. J. Cell. Physiol. 2020, 235, 5903–5924. [Google Scholar] [CrossRef]
  281. Hua, H.; Kong, Q.; Zhang, H.; Wang, J.; Luo, T.; Jiang, Y. Targeting mTOR for cancer therapy. J. Hematol. Oncol. 2019, 12, 71. [Google Scholar] [CrossRef] [PubMed]
  282. Zhang, X.; Campreciós, G.; Rimmelé, P.; Liang, R.; Yalcin, S.; Mungamuri, S.K.; Barminko, J.; D’Escamard, V.; Baron, M.H.; Brugnara, C.; et al. FOXO3-mTOR metabolic cooperation in the regulation of erythroid cell maturation and homeostasis. Am. J. Hematol. 2014, 89, 954–963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Franco, S.S.; De Falco, L.; Ghaffari, S.; Brugnara, C.; Sinclair, D.A.; Matte, A.; Iolascon, A.; Mohandas, N.; Bertoldi, M.; An, X.; et al. Resveratrol accelerates erythroid maturation by activation of FoxO3 and ameliorates anemia in beta-thalassemic mice. Haematologica 2014, 99, 267–275. [Google Scholar] [CrossRef]
  284. Sibon, D.; Coman, T.; Rossignol, J.; Lamarque, M.; Kosmider, O.; Bayard, E.; Fouquet, G.; Rignault, R.; Topçu, S.; Bonneau, P.; et al. Enhanced Renewal of Erythroid Progenitors in Myelodysplastic Anemia by Peripheral Serotonin. Cell Rep. 2019, 26, 3246–3256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Amireault, P.; Hatia, S.; Bayard, E.; Bernex, F.; Collet, C.; Callebert, J.; Launay, J.-M.; Hermine, O.; Schneider, E.; Mallet, J.; et al. Ineffective erythropoiesis with reduced red blood cell survival in serotonin-deficient mice. Proc. Natl. Acad. Sci. USA 2011, 108, 13141–13146. [Google Scholar] [CrossRef] [Green Version]
  286. Platten, M.; Nollen, E.A.A.; Röhrig, U.F.; Fallarino, F.; Opitz, C.A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug Discov. 2019, 18, 379–401. [Google Scholar] [CrossRef]
  287. Stein, E.M.; DiNardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef]
  288. Dutta, R.; Zhang, T.Y.; Köhnke, T.; Thomas, D.; Linde, M.; Gars, E.; Stafford, M.; Kaur, S.; Nakauchi, Y.; Yin, R.; et al. Enasidenib drives human erythroid differentiation independently of isocitrate dehydrogenase 2. J. Clin. Invest. 2020, 130, 1843–1849. [Google Scholar] [CrossRef]
  289. Stein, E.M.; DiNardo, C.D.; Fathi, A.T.; Pollyea, D.A.; Stone, R.M.; Altman, J.K.; Roboz, G.J.; Patel, M.R.; Collins, R.; Flinn, I.W.; et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood 2019, 133, 676–687. [Google Scholar] [CrossRef] [Green Version]
  290. Li, C.; Zhu, F.; Xu, C.; Xiao, P.; Wen, J.; Zhang, X.; Wu, B. Dangguibuxue decoction abolishes abnormal accumulation of erythroid progenitor cells induced by melanoma. J. Ethnopharmacol. 2019, 242, 112035. [Google Scholar] [CrossRef]
  291. Gonzalez-Menendez, P.; Romano, M.; Yan, H.; Deshmukh, R.; Papoin, J.; Oburoglu, L.; Daumur, M.; Dumé, A.S.; Phadke, I.; Mongellaz, C.; et al. An IDH1-vitamin C crosstalk drives human erythroid development by inhibiting pro-oxidant mitochondrial metabolism. Cell Rep. 2021, 34, 108723. [Google Scholar] [CrossRef]
  292. Steenbrugge, J.; De Jaeghere, E.A.; Meyer, E.; Denys, H.; De Wever, O. Splenic Hematopoietic and Stromal Cells in Cancer Progression. Cancer Res. 2021, 81, 27–34. [Google Scholar] [CrossRef] [PubMed]
  293. Levy, L.; Mishalian, I.; Bayuch, R.; Zolotarov, L.; Michaeli, J.; Fridlender, Z.G. Splenectomy inhibits non-small cell lung cancer growth by modulating anti-tumor adaptive and innate immune response. OncoImmunology 2015, 4, e998469. [Google Scholar] [CrossRef] [Green Version]
  294. Sano, T.; Sasako, M.; Mizusawa, J.; Yamamoto, S.; Katai, H.; Yoshikawa, T.; Nashimoto, A.; Ito, S.; Kaji, M.; Imamura, H.; et al. Randomized Controlled Trial to Evaluate Splenectomy in Total Gastrectomy for Proximal Gastric Carcinoma. Ann. Surg. 2017, 265, 277–283. [Google Scholar] [CrossRef] [PubMed]
  295. Fallah, J.; Olszewski, A.J. Diagnostic and therapeutic splenectomy for splenic lymphomas: Analysis of the National Cancer Data Base. Hematology 2019, 24, 378–386. [Google Scholar] [CrossRef] [Green Version]
  296. Yu, W.; Choi, G.S.; Chung, H.Y. Randomized clinical trial of splenectomy versus splenic preservation in patients with proximal gastric cancer. Br. J. Surg. 2006, 93, 559–563. [Google Scholar] [CrossRef]
  297. Crawford, J.; Cella, D.; Cleeland, C.S.; Cremieux, P.Y.; Demetri, G.D.; Sarokhan, B.J.; Slavin, M.B.; Glaspy, J.A. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002, 95, 888–895. [Google Scholar] [CrossRef]
  298. Caro, J.J.; Salas, M.; Ward, A.; Goss, G. Anemia as an independent prognostic factor for survival in patients with cancer: A systemic, quantitative review. Cancer 2001, 91, 2214–2221. [Google Scholar] [CrossRef]
  299. Ludwig, H.; Van Belle, S.; Barrett-Lee, P.; Birgegård, G.; Bokemeyer, C.; Gascón, P.; Kosmidis, P.; Krzakowski, M.; Nortier, J.; Olmi, P.; et al. The European Cancer Anaemia Survey (ECAS): A large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur. J. Cancer 2004, 40, 2293–2306. [Google Scholar] [CrossRef] [PubMed]
  300. Petrova, V.; Annicchiarico-Petruzzelli, M.; Melino, G.; Amelio, I. The hypoxic tumour microenvironment. Oncogenesis 2018, 7, 10. [Google Scholar] [CrossRef] [PubMed]
  301. Haemmerle, M.; Stone, R.L.; Menter, D.G.; Afshar-Kharghan, V.; Sood, A.K. The Platelet Lifeline to Cancer: Challenges and Opportunities. Cancer Cell 2018, 33, 965–983. [Google Scholar] [CrossRef] [Green Version]
  302. Lucotti, S.; Muschel, R.J. Platelets and Metastasis: New Implications of an Old Interplay. Front. Oncol. 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  303. Fan, J.; Slowikowski, K.; Zhang, F. Single-cell transcriptomics in cancer: Computational challenges and opportunities. Exp. Mol. Med. 2020, 52, 1452–1465. [Google Scholar] [CrossRef] [PubMed]
  304. Slyper, M.; Porter, C.B.M.; Ashenberg, O.; Waldman, J.; Drokhlyansky, E.; Wakiro, I.; Smillie, C.; Smith-Rosario, G.; Wu, J.; Dionne, D.; et al. A single-cell and single-nucleus RNA-Seq toolbox for fresh and frozen human tumors. Nat. Med. 2020, 26, 792–802. [Google Scholar] [CrossRef] [PubMed]
  305. Wynn, J.L.; Scumpia, P.O.; Stocks, B.T.; Romano-Keeler, J.; Alrifai, M.W.; Liu, J.-H.; Kim, A.S.; Alford, C.E.; Matta, P.; Weitkamp, J.-H.; et al. Neonatal CD71+ Erythroid Cells Do Not Modify Murine Sepsis Mortality. J. Immunol. 2015, 195, 1064–1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Sennikov, S.V.; Injelevskaya, T.V.; Krysov, S.V.; Silkov, A.N.; Kovinev, I.B.; Dyachkova, N.J.; Zenkov, A.N.; Loseva, M.I.; Kozlov, V.A. Production of hemo- and immunoregulatory cytokines by erythroblast antigen+ and glycophorin A+ cells from human bone marrow. BMC Cell Biol. 2004, 5, 39. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The role of erythroid progenitor cells (EPCs) in cancer. During disease progression, EPCs expand in the extramedullary sites, including the spleen. Moreover, EPCs are abundant in the peripheral blood of cancer patients and infiltrate the tumor microenvironment. Early-stage CD45+ EPCs use (1) reactive oxygen species (ROS), (2) interleukin-10 (IL-10), (3) transforming growth factor β (TGF-β), and (4) programmed death-ligand 1 (PD-L1) to modulate the immune response. EPCs inhibit (A) T-cell activation, (B) production of interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α), (C) T-cell proliferation, and (D) cytotoxicity of CD8+ T-cells. More mature CD45 EPCs regulate cancer progression by (5) secretion of a neurotropic factor, artemin. These late-stage EPCs, called Ter-cells, promote (E) tumor cell migration and invasiveness as well as (F) tumor growth and cell proliferation.
Figure 1. The role of erythroid progenitor cells (EPCs) in cancer. During disease progression, EPCs expand in the extramedullary sites, including the spleen. Moreover, EPCs are abundant in the peripheral blood of cancer patients and infiltrate the tumor microenvironment. Early-stage CD45+ EPCs use (1) reactive oxygen species (ROS), (2) interleukin-10 (IL-10), (3) transforming growth factor β (TGF-β), and (4) programmed death-ligand 1 (PD-L1) to modulate the immune response. EPCs inhibit (A) T-cell activation, (B) production of interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α), (C) T-cell proliferation, and (D) cytotoxicity of CD8+ T-cells. More mature CD45 EPCs regulate cancer progression by (5) secretion of a neurotropic factor, artemin. These late-stage EPCs, called Ter-cells, promote (E) tumor cell migration and invasiveness as well as (F) tumor growth and cell proliferation.
Cancers 13 00870 g001
Figure 2. Mechanisms of erythropoiesis dysregulation in cancer. Expansion of early-stage EPCs is caused by (A) chronic erythropoietin (EPO) production. However, EPCs are unable to generate mature red blood cells (RBCs) due to increased apoptosis and differentiation arrest. EPCs apoptosis is triggered by (B) FasL/Fas and (C) TRAIL-TRAIL-R interaction between EPCs and cancer cells. Differentiation arrest of early-stage EPCs is caused by (D) transforming growth factor β (TGF-β), (E) iron restriction, (F) pro-inflammatory cytokines, and (G) cancer-secreted chemokines. Inhibited maturation is an effect of GATA1 degradation mediated by caspase-3 or p38 activation. (H) Bone marrow steady-state erythropoiesis is suppressed by inflammation and triggers stress erythropoiesis and expansion of EPCs in extramedullary sites.
Figure 2. Mechanisms of erythropoiesis dysregulation in cancer. Expansion of early-stage EPCs is caused by (A) chronic erythropoietin (EPO) production. However, EPCs are unable to generate mature red blood cells (RBCs) due to increased apoptosis and differentiation arrest. EPCs apoptosis is triggered by (B) FasL/Fas and (C) TRAIL-TRAIL-R interaction between EPCs and cancer cells. Differentiation arrest of early-stage EPCs is caused by (D) transforming growth factor β (TGF-β), (E) iron restriction, (F) pro-inflammatory cytokines, and (G) cancer-secreted chemokines. Inhibited maturation is an effect of GATA1 degradation mediated by caspase-3 or p38 activation. (H) Bone marrow steady-state erythropoiesis is suppressed by inflammation and triggers stress erythropoiesis and expansion of EPCs in extramedullary sites.
Cancers 13 00870 g002
Figure 3. Management of anemia in cancer patients according to ESMO guidelines [258]. Additionally to TSAT and SF, the percentage of hypochromic cells (%HYPO) > 5% and reticulocytes hemoglobin content (CHr) < 28 pg can be used to determine impaired iron status. ID can be treated with oral iron only if ferritin < 30 ng/mL and CRP < 5 mg/L. CRP—C-reactive protein, ESA—erythropoiesis-stimulating agent, Hb—hemoglobin, i.v.—intravenous, ID—iron deficiency, RBCs—red blood cells, SF—serum ferritin, TSAT—transferrin saturation.
Figure 3. Management of anemia in cancer patients according to ESMO guidelines [258]. Additionally to TSAT and SF, the percentage of hypochromic cells (%HYPO) > 5% and reticulocytes hemoglobin content (CHr) < 28 pg can be used to determine impaired iron status. ID can be treated with oral iron only if ferritin < 30 ng/mL and CRP < 5 mg/L. CRP—C-reactive protein, ESA—erythropoiesis-stimulating agent, Hb—hemoglobin, i.v.—intravenous, ID—iron deficiency, RBCs—red blood cells, SF—serum ferritin, TSAT—transferrin saturation.
Cancers 13 00870 g003
Figure 4. Targeting EPC expansion as a novel therapeutic strategy. Cancer-induced dysregulation of erythropoiesis resulting in the differentiation arrest of EPCs and their expansion may be diminished by different agents. Inhibitors of (1) transforming growth factor β (TGF-β) and (2) BMP signaling rescue maturation arrest. (3) Caspases inhibitors inhibit GATA1 cleavage triggered by the inflammasome. (4) P38 inhibitors promote differentiation and decrease the apoptosis of EPCs. (5) JAK inhibitors decrease activation of EPO-induced signaling, which decreases EPCs expansion. (6) mTOR inhibitors and inducers of FoxO3 promote differentiation of early-stage EPCs. (7) Increasing serotonin (5-HT) concentration with either selective serotonin reuptake inhibitors (SSRIs) or inhibitors of the kynurenine metabolism pathway promotes differentiation of EPCs. (8) Enasidenib, an inhibitor of mutated IDH2 promotes terminal differentiation of EPCs.
Figure 4. Targeting EPC expansion as a novel therapeutic strategy. Cancer-induced dysregulation of erythropoiesis resulting in the differentiation arrest of EPCs and their expansion may be diminished by different agents. Inhibitors of (1) transforming growth factor β (TGF-β) and (2) BMP signaling rescue maturation arrest. (3) Caspases inhibitors inhibit GATA1 cleavage triggered by the inflammasome. (4) P38 inhibitors promote differentiation and decrease the apoptosis of EPCs. (5) JAK inhibitors decrease activation of EPO-induced signaling, which decreases EPCs expansion. (6) mTOR inhibitors and inducers of FoxO3 promote differentiation of early-stage EPCs. (7) Increasing serotonin (5-HT) concentration with either selective serotonin reuptake inhibitors (SSRIs) or inhibitors of the kynurenine metabolism pathway promotes differentiation of EPCs. (8) Enasidenib, an inhibitor of mutated IDH2 promotes terminal differentiation of EPCs.
Cancers 13 00870 g004
Table 1. Immunomodulatory cells in cancer and their mechanisms of immune regulation.
Table 1. Immunomodulatory cells in cancer and their mechanisms of immune regulation.
CellsMechanismsEffectsRef
Regulatory T-cells (Tregs)IL-10T-cell suppression[14]
IL-2 consumptionT-cell suppression[15]
COX-2 and PGE2T-cell suppression[16]
AdenosineT-cell suppression[17]
Myeloid-derived suppressor cells (MDSCs)ARG1T-cell suppression[18]
IDOT-cell suppression
Tregs induction
NK cell suppression
[19,20]
PD-L1/PD-1T-cell suppression[21]
IL-10Tregs induction[22]
TGF-βTregs induction[22]
CD40/CD40LTregs activation[23]
Depletion of cystine and cysteineT-cell suppression[24]
ROST-cell suppression[25]
Free radical peroxynitriteResistance to cytotoxic T-cells[26]
Tumor associated macrophages (TAMs)PD-L1/PD-1Decreased phagocytosis[27]
ARG1T-cell suppression[28]
IL-10T-cell suppression[29]
IL-1βMDSC infiltration
Induction of the protumor phenotype
[30,31]
IL-12Induction of T-cell response[32]
TNF-αInduction of anti-tumor response[33]
Tumor associated neutrophils (TANs)ARG1T-cell suppression[18,28]
NOST-cell suppression
T-cell apoptosis
[34,35]
PD-L1/PD-1T-cell suppression[36]
Cancer associated fibroblasts (CAFs)PD-L1/PD-1T-cell suppression[37]
FasL, PD-L2T-cell suppression[38]
IL-6Induction of PD-L1+ TANs[39]
ChemokinesMDSC infiltration[40]
Erythroid progenitor cells (EPCs)ROST-cell suppression[41,42]
IL-10T-cell suppression[42]
PD-L1/PD-1T-cell suppression[43]
TGF-βT-cell suppression[42]
ARG1—arginase 1, COX-2—cyclooxygenase-2, FasL—Fas ligand (CD95L, CD178), IDO—Indoleamine-pyrrole 2,3-dioxygenase, IL—interleukin, NK—natural killer, NOS—nitric oxide synthase, PD-1—programmed cell death 1, PD-L1—programmed death-ligand 1, PGE2—Prostaglandin E2, ROS—reactive oxygen species, TGF-β—transforming growth factor β, TNF-α—tumor necrosis factor α.
Table 2. Regulation of erythropoiesis.
Table 2. Regulation of erythropoiesis.
FactorRole in ErythropoiesisDysregulation in CancerReferences
SCFGrowth factors regulating early stages of erythropoiesisProduction in TME
Increased serum concentration
[50,51]
G-CSF[52]
IL-3[53]
EPOGrowth factors regulating late stages of erythropoiesisIncreased serum concentration[54]
GDF11Production in TME[55]
Activin AProduction in TME[56]
GATA1Crucial TFs regulating erythropoiesisDecreased expression in EPCs in cancer[57,58,59]
STAT5Increased in EPCs in MPNs
Decreased in EPCs in iron deficiency
[60,61]
MCL-1Survival factors for erythroid cells
BCL-xL
HSP70
TGF-βNegative regulators of erythropoiesisProduction in TME
Increased concentration
[62]
SMAD signalingIncreased level in EPCs in cancer[62]
FasLHigh expression on cancer cells[59,63]
FasIncreased level in EPCs in cancer[59,63]
Vitamin B12Essential vitamins, trace elements, and iron-metabolism proteinsDecreased in a subset of patients[64]
Folic AcidDecreased in a subset of patients[64]
CopperIncreased concentration[65]
IronDecreased in a subset of patients[66]
FerritinDecreased or increased[66]
TransferrinDecreased in a subset of patients[66]
FerroportinDecreased expression[67]
HepcidinIncreased concentration[68]
MPN—myeloproliferative neoplasm, TF—transcription factor, TME—tumor microenvironment.
Table 3. Mechanisms of immunomodulatory functions of EPCs.
Table 3. Mechanisms of immunomodulatory functions of EPCs.
SourceMechanismEffectMouseHumansRef.
NeonatesARG2↓cytokine production bymyeloid cells++[86,93]
TGF-β↑Tregs differentiation++[87]
ROS↓cytokine production bymyeloid cells
↓cytokine production by T-cells
-+[94]
PD-1/PD-L1↓cytokine production by T-cells++[88]
PregnancyARG2↓cytokine production bymyeloid cells++[24,93]
TGF-β↑Tregs differentiationn.d.+[93]
ROS↓cytokine production bymyeloid cells
↓cytokine production by T-cells
n.d.+[93]
PD-1/PD-L1↓cytokine production by T-cells++[88]
Inflammatory diseasesEPCs phagocytosis↓cytokine production byred pulp macrophages+n.d.[89]
HIV-infected patientsROS↑HIV replication in T-cells
↑HIV trans-infection
n.d.+[90]
COVID-19 patientsARG1↓cytokine production by T-cells
↓T-cell proliferation
n.d.+[95]
ARG2↓cytokine production by T-cells
↓T-cell proliferation
n.d.+[95]
ROS↓cytokine production by T-cells
↓T-cell proliferation
n.d.+[95]
AnemiaARG1↓cytokine production by T-cells
↓T-cell proliferation
-+[92]
ARG2↓cytokine production by T-cells
↓T-cell proliferation
++[92]
ROS↓cytokine production by T-cells
↓T-cell proliferation
++[92]
CancerTGF-β↓T-cells proliferation↓cytokine production by T-cellsn.d.+[42]
ROS↓T-cell proliferation↓cytokine production by T-cells++[41,42]
PD-L1/PD-1↓cytokine production by T-cells++[43]
IL-10↓T-cell proliferation↓cytokine production by T-cellsn.d.+[42]
↑—promoted, ↓—suppressed, n.d.—no data, - — no role, +—reported mechanism.
Table 4. Differences in immune-related mediators between early-stage and late-stage EPCs [41,42,43,62].
Table 4. Differences in immune-related mediators between early-stage and late-stage EPCs [41,42,43,62].
FeatureEarly-Stage EPCs (CD45+)Late-Stage EPCs (CD45-)
ROS level
IL-10
TGF-β
ROS pathway
IL-10 pathway
TGF-β pathway
PD-1/PD-L1n.d.n.d.
ARG2n.d.n.d.
↑—increased, ↓—decreased, n.d.—no data.
Table 5. Different role of early-stage and late-stage EPCs in cancer [41,42,43,62,116,123].
Table 5. Different role of early-stage and late-stage EPCs in cancer [41,42,43,62,116,123].
ProcessEarly-Stage EPCs (CD45+)Late-Stage EPCs (CD45-)
T-cell proliferation↓ suppressed↔ no effect
Production of IFN-γ by T-cells↓ suppressed↔ no effect
Production of TNF-α by T-cells↓ suppressed↔ no effect
CD8+ T-cells cytotoxicity↓ suppressed↔ no effect
Dendritic cells activationn.d.↔ no effect
Production of IL-6 and IL-12 by dendritic cellsn.d.↔ no effect
Tregs inductionn.d.↔ no effect
Anti-tumor immune response↓ suppressed↔ no effect
Activation of signaling pathways in tumor cellsn.d.↑ promoted
Regulation of cancer cell metabolism↑ promotedn.d.
Tumor cells proliferationn.d.↑ promoted
Tumor cells invasivenessn.d.↑ promoted
Tumor growth↑ promoted↑ promoted
↑—promoted, ↓—suppressed, ↔—no effect, n.d.—no data.
Table 6. The frequency of EPCs in tumor-bearing mice and cancer patients.
Table 6. The frequency of EPCs in tumor-bearing mice and cancer patients.
OrganMiceHumansRef.
HealthyTumor-BearingHealthyCancer Patient
Peripheral blood5%60%0.13%2–4.25%[41,42]
Spleen5%20–50%0.02%0.15%[41,43,62,123]
Bone marrow15–20%55%14%n.d.[41,43,134]
Liver10%2–30%2.5%10%[41,42,62]
Lymph node1%1%n.dn.d[41,62]
Tumor-2–10%-10%[41,42,62]
n.d.—no data, - — not applicable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Grzywa, T.M.; Justyniarska, M.; Nowis, D.; Golab, J. Tumor Immune Evasion Induced by Dysregulation of Erythroid Progenitor Cells Development. Cancers 2021, 13, 870. https://doi.org/10.3390/cancers13040870

AMA Style

Grzywa TM, Justyniarska M, Nowis D, Golab J. Tumor Immune Evasion Induced by Dysregulation of Erythroid Progenitor Cells Development. Cancers. 2021; 13(4):870. https://doi.org/10.3390/cancers13040870

Chicago/Turabian Style

Grzywa, Tomasz M., Magdalena Justyniarska, Dominika Nowis, and Jakub Golab. 2021. "Tumor Immune Evasion Induced by Dysregulation of Erythroid Progenitor Cells Development" Cancers 13, no. 4: 870. https://doi.org/10.3390/cancers13040870

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop