You are on page 1of 4

ARTICLE IN PRESS

Cancer Letters ■■ (2015) ■■–■■

Contents lists available at ScienceDirect

Cancer Letters
j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / c a n l e t

Mini-review

Expansion and functions of myeloid-derived suppressor cells in the


tumor microenvironment
Peng Qu, Li-zhen Wang, P. Charles Lin *
National Cancer Institute, National Institutes of Health, Frederick, MD, USA

A R T I C L E I N F O A B S T R A C T

Keywords:
Myeloid derived suppressor cells (MDSCs) are a group of immature myeloid cells accumulated in most
Myeloid-derived suppressor cells
cancer patients and mouse tumor models. MDSCs suppress host immune response and concurrently
Tumor microenvironment
angiogenesis
promote tumor angiogenesis, thereby promote tumor growth and progression. In this review, we discuss
immune suppression recent progresses in expansion and activity of tumor MDSCs, and describe new findings about immu-
M-MDSC nosuppressive function of different subtypes of MDSCs in cancer. We also discussed tumor angiogenic
PMN-MDSC activities and pro-tumor invasion/metastatic roles of MDSCs in tumor progression.
© 2015 Published by Elsevier Ireland Ltd.

Introduction been studied in the context of peripheral circulation, spleen and bone
marrow from tumor mouse models [10].
MDSCs are heterogeneous population of myeloid lineage cells
including myeloid progenitors and immature myeloid cells (IMCs).
MDSCs are highly increased in humans and mouse models with Expansion and activation of MDSCs in tumor bearing hosts
various pathological disorders. They exert potent immune-
suppressive functions and are important negative regulators of MDSCs are highly elevated in cancer patients as well as tumor
immune responses in chronic inflammation and cancer [1–3]. In bearing animal models. They possess strong immune suppressive
various cancer patients, MDSCs might be used as a prognostic marker activities. What regulates MDSC expansion and activation in tumor
and potential therapeutic targets toward elimination of their im- conditions is an important question that has been investigated ex-
munosuppressive activity and enhancement of anti-tumor immune tensively in the past. Studies have shown that many secreted factors,
responses [4–7]. such as growth factors and microbial products, are released in the
MDSCs are defined as Gr-1 + CD11b+ cells in mice. They are further tumor microenvironments. These factors activate the expansion or
divided into two subsets using antibodies that distinguish between production of MDSCs [11]. Among them, granulocyte-macrophage
Ly-6C and Ly-6G: co-expression of CD11b with Ly-6G+/Ly-6Chigh as colony stimulating factor (GM-CSF), G-CSF, IL-6, prostaglandin-E2
monocytic lineage cells (M-MDSCs) whereas CD11b with Ly-6G+/ (PGE2), cyclooxygenase 2 (Cox2) and VEGF seem to play a major role
Ly-6Clow as granulocytic lineage cells (PMN-MDSCs). Both subsets [12–17]. Accordingly, clinical studies demonstrate that the numbers
are elevated in tumor-bearing mice, although the higher increase of CD45 + lin−HLA-DR− human MDSCs decrease with a treatment
is primarily observed in PMN-MDSCs [8]. In humans, three sub- of bevacizumab, a monoclonal antibody directed against VEGF-A,
groups of MDSCs have been reported on the basis of their ability on patients with lung, breast and colorectal cancers [18]. Cytokines
to mediate immune suppression using a variety of different inducing MDSCs acted on a common molecular pathway and the
markers such as CD11b, CD14 and CD33. Human PMN-MDSCs immune-regulatory activity of both tumor-induced and BM-
are CD14-CD11b + CD33 + CD15+, and human M-MDSCs are derived MDSCs is shown entirely dependent on the C/EBPβ
CD14 + HLA-DR-/low cells. The third sub group of human MDSCs is transcription factor [10].
Lin-HLA-DR-CD33+ cells [9]. Human MDSCs are enumerated pri- Some of these factors activate STAT transcription factors (STAT3,
marily in peripheral blood from cancer patients and characterized STAT5, STAT6) in MDSCs, which function as important signal trans-
by immature myeloid cell surface markers. On the other hand, the ducers in the expansion and activation of MDSCs [19]. The
markers for murine MDSC are CD11b and Gr-1. Murine MDSCs have development of MDSCs is also regulated by interferon regulatory
factor 8 (IRF-8). MDSC-inducing factors such as G-CSF and GM-
CSF downregulate IRF-8 via STAT3- and STAT5-dependent pathways.
The levels of IRF-8 in MDSCs in cancer patients decline with in-
creasing MDSC frequency, implicating IRF-8 as a negative regulator
* Corresponding author. Tel.: +1 301 228 4688; fax: +1 301 846 7017. in human MDSC biology [19]. Our studies also support the impor-
E-mail address: p.lin@nih.gov (P.C. Lin). tance of STAT3 in MDSC expansion. We show that myeloid specific

http://dx.doi.org/10.1016/j.canlet.2015.10.022
0304-3835/© 2015 Published by Elsevier Ireland Ltd.

Please cite this article in press as: Peng Qu, Li-zhen Wang, P. Charles Lin, Expansion and functions of myeloid-derived suppressor cells in the tumor microenvironment, Cancer Letters
(2015), doi: 10.1016/j.canlet.2015.10.022
ARTICLE IN PRESS
2 P. Qu et al./Cancer Letters ■■ (2015) ■■–■■

expression of either apoptosis inhibitor 6 (Api6) or metalloprotease-


12 (MMP12) activates STAT3 in these cells and consequently leads
to systemic expansion of MDSCs. Inhibition of STAT3 in vitro abol-
ishes the proliferation and suppressive activity of MDSCs [20,21].
Moreover, factors that promote myeloid-lineage cell differentia-
tion reduce MDSC population expansion [22]. A growing number
of studies have demonstrated a strong correlation between the levels
of MDSCs in blood of cancer patients and tumor stage and meta-
static status [23–25].

Regulatory functions of MDSCs in the tumor


microenvironment

The pro tumor functions of MDSCs are at least two folds: (1)
immune suppression through inhibition of functional T cells and
NK cells as well as induction of Treg expansion; (2) promoting tumor
angiogenesis and tumor invasion/metastasis via production of an-
giogenic factors and proteases in tumor tissues.
Fig. 1. The regulation mechanisms of MDSCs in the tumor microenvironment. PMN-
MDSCs can take up, process and present antigens to antigen-specific T cells. Through
Immune suppressive activities of MDSCs cell–cell contact, PMN-MDSCs induce nitration of the T-cell receptors (TCR) on the
surface of T cells. T cells become unresponsive to antigen specific stimulation.
As reflected in its name, a major function of MDSC is potent M-MDSCs are differentiated into tumor associated macrophages (TAM) that inhibit
T and NK cell immune responses. Both PMN-MDSCs and M-MDSCs produce reac-
immune suppressive activity via targeting other immune cells, such
tive oxygen species (ROS) and nitric oxide (NO) which inhibit T and NK cells in a
as T cells in tumor conditions [26–30]. MDSCs also suppress pro- non-specific manner. MDSCs induce Treg expansion via secretion of IL-10 and TGF-
liferation and cytokine secretion in both T lymphocytes and Natural β. In some cancer models, the induction of Treg by MDSCs is associated with the
Killer (NK) cells, as well as induction of apoptosis in T cell subsets expression of cytotoxic lymphocyte antigen 4 (CTLA4). Tregs inhibit T and NK cells.
[31]. Many factors have been implicated in MDSC-mediated immune MDSCs contribute to tumor angiogenesis by production of factors such as MMP9,
CCL2, VEGF and BV8. TGF-β signaling in MDSCs plays an important role in tumor
suppression, including inducible nitric oxide synthase (iNOS),
metastasis.
arginase-1 (Arg1), Cox-2, PGE2 and TGF-β [32–35]. MDSCs deliver
the suppressive functions through a combination of multiple
molecules.
In general, PMN-MDSCs are more prevalent in the tumor mi- TCR and inhibition of TCR signaling [42]. PMN-MDSCs also down-
croenvironment, however, M-MDSCs have much stronger immune regulate the ζ chain of TCR to suppress T cell function [43].
suppressive activity [9]. The suppressive function of M-MDSCs is Moreover, MDSCs induce Tregs expansion to promote tumor pro-
associated with the metabolism of L-arginine. Both Arg1 and iNOS gression through different mechanisms in various tumor models.
use L-arginine as a common substrate to produce urea or NO that In an ovarian cancer model, the induction of Tregs by MDSCs is as-
blocks T cell proliferation, differentiation and cytokine production sociated with the expression of cytotoxic lymphocyte 4 antigen
[33]. In addition to direct signaling effects through NO, iNOS can (CTLA4), while MDSCs induce Treg expansion via secretion of
also decreases the available levels of its substrate L-arginine, a process IL-10/TGF-β in colon tumor models [44]. Studies also find that ex-
that can be further accelerated by the metabolic activities of Arg- pression of the immune stimulatory receptor CD40 on MDSCs is
1. Up-regulation of Arg1 activity leads to a depletion of L-arginine required for MDSCs to suppress T-cell and induce Treg accumula-
from the pathological microenvironment, which leads to a reduc- tion in tumor models [9,45]. MDSC can directly impact on the
tion of TCR expression and T cell proliferation [36]. This metabolic prevalence of Tregs through IFNγ dependent production of IL-10 or
targeting is not unique to L-arginine, and studies have identified ad- iNOS [46]. Thus, MDSCs participate in both the control of T cell re-
ditional metabolic targets utilized in MDSC-mediated immune sponses and the induction of Tregs capable of potentiating long-
suppression, such as cystine and cysteine [37]. Moreover, M-MDSCs term immune suppression.
are also capable of differentiating into tumor associated mac-
rophages (TAMs), which promote T cell apoptosis and lead to Tumor angiogenic and pro-tumor invasion/metastatic activities of
immune suppression [9]. Both M-MDSCs and TAMs suppress CD8+ MDSCs
T cell functions in a non-specific manner at the tumor site. M-MDSCs
produce high levels of Arg1 and NO, whereas TAMs up-regulate the In addition to suppressing host immune functions within the
expression of either Arg1 or iNOS, but not of both proteins, depen- tumor microenvironment, MDSCs also promote invasion and me-
dent on the tumor microenvironment [38]. TAMs also inhibit T cell tastasis of tumor cells. In tumor-bearing hosts, MDSCs are increased
function by secreting cytokines such as IL-1β and TGF-β [39] (Fig. 1). in lymph organs or in other organs, including lungs and livers, where
PMN-MDSCs utilize various mechanisms to subdue the host MDSCs could facilitate tumor metastasis to these organ sites [10,47].
immune response. They produce TGF-β, a potent immune suppres- Preclinical evidence has shown that hypoxic breast cancer cells
sor [34,40]. PMN-MDSCs also have the ability to suppress immune produce carbonic anhydrase IX (CAIX) and G-CSF to drive mobili-
responses in an antigen-specific manner. They can take up, process zation of PMN-MDSCs to the breast cancer lung metastatic niches
and present antigens to antigen-specific T cells [1]. Or through cell– [48].
cell contact, PMN-MDSCs induce nitration of T-cell receptors (TCR), It was found that the TGF-β signaling pathway is associated with
which makes T cells unresponsive to antigen stimulation [11]. ROS tumor cell motility, invasion and metastasis [49]. MDSCs produce
is another major factor responsible for PMN-MDSCs mediated large numbers of MMPs and TGF-β1, which has a profound impact
immune suppression. Inhibition of ROS production using an up- on tumor progression and metastasis through modulation of tumor
stream inhibitor abolished PMN-MDSC mediated suppression in vitro vascularization and tumor cell invasion. TGF-β signaling in MDSCs
[41]. The combination of ROS and NO suppresses CD8+ T cell re- of tumor-bearing hosts is fundamentally important for tumor me-
sponses by producing peroxynitrite, which resulted in nitration of tastasis. In a breast cancer mouse model, treatment with TGF-β

Please cite this article in press as: Peng Qu, Li-zhen Wang, P. Charles Lin, Expansion and functions of myeloid-derived suppressor cells in the tumor microenvironment, Cancer Letters
(2015), doi: 10.1016/j.canlet.2015.10.022
ARTICLE IN PRESS
P. Qu et al./Cancer Letters ■■ (2015) ■■–■■ 3

neutralization antibody significantly decreased the number of MDSCs metastasis. Targeting MDSCs may offer a therapeutic approach to
in tumor tissues and premetastatic lung through induction of MDSCs cancer patients.
apoptosis [50]. Genetic deletion of Tgfbr2 specifically in MDSCs dra-
matically blocks their suppressive functions and decreases tumor
metastasis (Fig. 1) [51–53]. Interestingly, enhanced metastasis in the Acknowledgment
absence of tumor cell response to TGF- β stimulation has been shown
to involve increased chemokine production resulting in recruit- The work was supported by the Intramural Research Program
ment of pro-metastatic MDSCs to the tumor microenvironment at at the NCI, NIH.
the leading invasive edge [54]. These data suggest that TGF- β is a
major regulator in MDSCs-mediated pro-metastasis in the tumor
Conflict of interest
microenvironment.
Additionally, MDSCs secrete factors to promote tumor angio-
The authors declare no competing financial interests.
genesis, a step essential for tumor growth and progression [55]. The
pro-angiogenic function of MDSCs is mediated in part through in-
duction of MMP9, a protease that degrades extracellular matrix References
(ECM). As a result, it frees VEGF deposited in the matrix, which leads
to activation of VEGF receptors on endothelium and angiogenesis [1] D.I. Gabrilovich, S. Nagaraj, Myeloid-derived suppressor cells as regulators of
[55]. Activation of STAT3 in MDSCs increases the production of VEGF the immune system, Nat. Rev. Immunol. 9 (2009) 162–174.
and FGF, both of which are potent angiogenic factors and pro- [2] L. Dolcetti, I. Marigo, B. Mantelli, E. Peranzoni, P. Zanovello, V. Bronte, Myeloid-
derived suppressor cell role in tumor-related inflammation, Cancer Lett. 267
motes tumor angiogenesis [56]. Studies have examined the (2008) 216–225.
mechanisms of tumor refractoriness to VEGF blockade in synge- [3] A. Arina, V. Bronte, Myeloid-derived suppressor cell impact on endogenous and
neic mouse tumor models. Interestingly, MDSCs mediated adoptively transferred T cells, Curr. Opin. Immunol. 33 (2015) 120–125.
[4] D. Di Mitri, A. Toso, A. Alimonti, Molecular pathways: targeting tumor-infiltrating
angiogenesis contributes to refractoriness to anti-VEGF tumor therapy myeloid-derived suppressor cells for cancer therapy, Clin. Cancer Res. 21 (2015)
in mouse models [9,57,58]. Our results reveal that MDSCs in the 3108–3112.
tumor microenvironment produced CCL2, which is known to func- [5] R.F. Gabitass, N.E. Annels, D.D. Stocken, H.A. Pandha, G.W. Middleton, Elevated
myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer
tion as an angiogenic factor. Blocking CCL2 produced by MDSCs led
are an independent prognostic factor and are associated with significant
to decreased endothelial cell migration [59]. Together, these studies elevation of the Th2 cytokine interleukin-13, Cancer Immunol. Immunother.
suggest that MDSCs promote tumor angiogenesis by production of 60 (2011) 1419–1430.
[6] S. Ugel, F. Delpozzo, G. Desantis, F. Papalini, F. Simonato, N. Sonda, et al.,
factors that alter the tumor microenvironment in a pro-angiogenic
Therapeutic targeting of myeloid-derived suppressor cells, Curr. Opin.
manner (Fig. 1). Pharmacol. 9 (2009) 470–481.
[7] A.J. Montero, C.M. Diaz-Montero, C.E. Kyriakopoulos, V. Bronte, S. Mandruzzato,
Emerging role of miRNA in MDSC biology Myeloid-derived suppressor cells in cancer patients: a clinical perspective, J.
Immunother. 35 (2012) 107–115.
[8] J.I. Youn, S. Nagaraj, M. Collazo, D.I. Gabrilovich, Subsets of myeloid-derived
There is emerging evidence that microRNAs (miRNAs) play im- suppressor cells in tumor-bearing mice, J. Immunol. 181 (2008) 5791–5802.
portant roles in the activation and function of MDSCs. miRNAs are [9] D. Marvel, D.I. Gabrilovich, Myeloid-derived suppressor cells in the tumor
microenvironment: expect the unexpected, J. Clin. Invest. 25 (9) (2015)
small non-coding RNA molecules which are conserved in eukary- 3356–3364.
otic organisms and are paired with complementary sequences within [10] P. Qu, K.C. Boelte, P.C. Lin, Negative regulation of myeloid-derived suppressor
mRNA molecules, thus resulting in translational repression or target cells in cancer, Immunol. Invest. 41 (2012) 562–580.
[11] D.I. Gabrilovich, S. Ostrand-Rosenberg, V. Bronte, Coordinated regulation of
degradation [60–62]. Studies have identified many miRNAs, includ- myeloid cells by tumours, Nat. Rev. Immunol. 12 (2012) 253–268.
ing miR-17-5p, miR-20a, miR-21, miR-494, miR-125b, miR-155, miR- [12] N. Obermajer, R. Muthuswamy, J. Lesnock, R.P. Edwards, P. Kalinski, Positive
223 and miR-146a, which target transcription factors important for feedback between PGE2 and COX2 redirects the differentiation of human
dendritic cells toward stable myeloid-derived suppressor cells, Blood 118 (2011)
the development and differentiation of MDSCs [63,64].
5498–5505.
A study shows that both miR-17-5p and miR-20a promote the [13] I. Marigo, E. Bosio, S. Solito, C. Mesa, A. Fernandez, L. Dolcetti, et al., Tumor-
suppressive function of PMN-MDSCs, but not M-MDSCs, through induced tolerance and immune suppression depend on the C/EBPbeta
transcription factor, Immunity 32 (2010) 790–802.
targeting STAT3 [65]. Tumor-associated factors downregulate the
[14] S.K. Bunt, L. Yang, P. Sinha, V.K. Clements, J. Leips, S. Ostrand-Rosenberg, Reduced
expression of miR-17-5p and miR-20a, and consequently enhance inflammation in the tumor microenvironment delays the accumulation of
the suppressive function of PMN-MDSCs. Thus, miR-17-5p and miR- myeloid-derived suppressor cells and limits tumor progression, Cancer Res. 67
20a might potentially be useful targets in immunotherapeutic (2007) 10019–10026.
[15] S.K. Bunt, V.K. Clements, E.M. Hanson, P. Sinha, S. Ostrand-Rosenberg,
strategies to inhibit PMN-MDSCs via reducing STAT3 expression. The Inflammation enhances myeloid-derived suppressor cell cross-talk by signaling
roles of miR-155 and miR-21 on MDSCs are also associated with through Toll-like receptor 4, J. Leukoc. Biol. 85 (2009) 996–1004.
STAT3 activation via targeting SHIP-1 and PTEN, respectively [66]. [16] P. Sinha, V.K. Clements, A.M. Fulton, S. Ostrand-Rosenberg, Prostaglandin E2
promotes tumor progression by inducing myeloid-derived suppressor cells,
Even though those miRNAs regulate MDSC properties to create a Cancer Res. 67 (2007) 4507–4513.
microenvironment suitable for cancer formation and develop- [17] L. Wu, H. Du, Y. Li, P. Qu, C. Yan, Signal transducer and activator of transcription
ment, most of our knowledge of miRNA on MDSCs comes from 3 (Stat3C) promotes myeloid-derived suppressor cell expansion and immune
suppression during lung tumorigenesis, Am. J. Pathol. 179 (2011) 2131–2141.
murine studies. There is limited clinical information about the role [18] T. Osada, G. Chong, R. Tansik, T. Hong, N. Spector, R. Kumar, et al., The effect of
and regulation of miRNA on MDSCs in cancer patients. The signif- anti-VEGF therapy on immature myeloid cell and dendritic cells in cancer
icance of miRNAs for the expansion and function of MDSCs in cancer patients, Cancer Immunol. Immunother. 57 (2008) 1115–1124.
[19] J.D. Waight, C. Netherby, M.L. Hensen, A. Miller, Q. Hu, S. Liu, et al., Myeloid-
patients are largely unknown and hence require further investiga- derived suppressor cell development is regulated by a STAT/IRF-8 axis, J. Clin.
tion [67]. Invest. 123 (2013) 4464–4478.
[20] P. Qu, H. Du, Y. Li, C. Yan, Myeloid-specific expression of Api6/AIM/Sp alpha
induces systemic inflammation and adenocarcinoma in the lung, J. Immunol.
Conclusion
182 (2009) 1648–1659.
[21] P. Qu, C. Yan, H. Du, Matrix metalloproteinase 12 overexpression in myeloid
MDSCs are heterogeneous myeloid cells that are highly el- lineage cells plays a key role in modulating myelopoiesis, immune suppression,
evated in cancer patients and tumor bearing animal models. These and lung tumorigenesis, Blood 117 (2011) 4476–4489.
[22] S. Kusmartsev, F. Cheng, B. Yu, Y. Nefedova, E. Sotomayor, R. Lush, et al.,
cells promote tumor growth and progression through immune sup- All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing
pression as well as enhanced tumor angiogenesis and invasion/ mice and improves the effect of vaccination, Cancer Res. 63 (2003) 4441–4449.

Please cite this article in press as: Peng Qu, Li-zhen Wang, P. Charles Lin, Expansion and functions of myeloid-derived suppressor cells in the tumor microenvironment, Cancer Letters
(2015), doi: 10.1016/j.canlet.2015.10.022
ARTICLE IN PRESS
4 P. Qu et al./Cancer Letters ■■ (2015) ■■–■■

[23] S. Solito, I. Marigo, L. Pinton, V. Damuzzo, S. Mandruzzato, V. Bronte, Myeloid- [45] P.Y. Pan, G. Ma, K.J. Weber, J. Ozao-Choy, G. Wang, B. Yin, et al., Immune
derived suppressor cell heterogeneity in human cancers, Ann. N. Y. Acad. Sci. stimulatory receptor CD40 is required for T-cell suppression and T regulatory
1319 (2014) 47–65. cell activation mediated by myeloid-derived suppressor cells in cancer, Cancer
[24] K.R. Jordan, R.N. Amaria, O. Ramirez, E.B. Callihan, D. Gao, M. Borakove, et al., Res. 70 (2010) 99–108.
Myeloid-derived suppressor cells are associated with disease progression and [46] B. Huang, P.Y. Pan, Q. Li, A.I. Sato, D.E. Levy, J. Bromberg, et al., Gr-1+CD115+
decreased overall survival in advanced-stage melanoma patients, Cancer immature myeloid suppressor cells mediate the development of tumor-induced
Immunol. Immunother. 62 (2013) 1711–1722. T regulatory cells and T-cell anergy in tumor-bearing host, Cancer Res. 66 (2006)
[25] S. Nagaraj, D.I. Gabrilovich, Myeloid-derived suppressor cells in human cancer, 1123–1131.
Cancer J. 16 (2010) 348–353. [47] I. Younos, M. Donkor, T. Hoke, A. Dafferner, H. Samson, S. Westphal, et al., Tumor-
[26] V. Bronte, P. Serafini, A. Mazzoni, D.M. Segal, P. Zanovello, L-arginine metabolism and organ-dependent infiltration by myeloid-derived suppressor cells, Int.
in myeloid cells controls T-lymphocyte functions, Trends Immunol. 24 (2003) Immunopharmacol. 11 (2011) 816–826.
302–306. [48] S.C. Chafe, Y. Lou, J. Sceneay, M. Vallejo, M.J. Hamilton, P.C. McDonald, et al.,
[27] S. Kusmartsev, S. Nagaraj, D.I. Gabrilovich, Tumor-associated CD8+ T cell Carbonic anhydrase IX promotes myeloid-derived suppressor cell mobilization
tolerance induced by bone marrow-derived immature myeloid cells, J. Immunol. and establishment of a metastatic niche by stimulating G-CSF production, Cancer
175 (2005) 4583–4592. Res. 75 (2015) 996–1008.
[28] P. Serafini, I. Borrello, V. Bronte, Myeloid suppressor cells in cancer: recruitment, [49] L. Yang, Y. Pang, H.L. Moses, TGF-beta and immune cells: an important regulatory
phenotype, properties, and mechanisms of immune suppression, Semin. Cancer axis in the tumor microenvironment and progression, Trends Immunol. 31
Biol. 16 (2006) 53–65. (2010) 220–227.
[29] A. Mazzoni, V. Bronte, A. Visintin, J.H. Spitzer, E. Apolloni, P. Serafini, et al., [50] Z. Li, Y. Pang, S.K. Gara, B.R. Achyut, C. Heger, P.K. Goldsmith, et al., Gr-1+CD11b+
Myeloid suppressor lines inhibit T cell responses by an NO-dependent cells are responsible for tumor promoting effect of TGF-beta in breast cancer
mechanism, J. Immunol. 168 (2002) 689–695. progression, Int. J. Cancer 131 (2012) 2584–2595.
[30] I. Marigo, L. Dolcetti, P. Serafini, P. Zanovello, V. Bronte, Tumor-induced tolerance [51] L. Yang, TGFbeta and cancer metastasis: an inflammation link, Cancer Metastasis
and immune suppression by myeloid derived suppressor cells, Immunol. Rev. Rev. 29 (2010) 263–271.
222 (2008) 162–179. [52] Y. Pang, S.K. Gara, B.R. Achyut, Z. Li, H.H. Yan, C.P. Day, et al., TGF-beta signaling
[31] V. Bronte, M. Wang, W.W. Overwijk, D.R. Surman, F. Pericle, S.A. Rosenberg, et al., in myeloid cells is required for tumor metastasis, Cancer Discov. 3 (2013)
Apoptotic death of CD8+ T lymphocytes after immunization: induction of a 936–951.
suppressive population of Mac-1+/Gr-1+ cells, J. Immunol. 161 (1998) 5313– [53] M. Bodogai, K. Moritoh, C. Lee-Chang, C.M. Hollander, C.A. Sherman-Baust, R.P.
5320. Wersto, et al., Immunosuppressive and prometastatic functions of myeloid-
[32] H. Li, Y. Han, Q. Guo, M. Zhang, X. Cao, Cancer-expanded myeloid-derived derived suppressive cells rely upon education from tumor-associated B cells,
suppressor cells induce anergy of NK cells through membrane-bound TGF-beta Cancer Res. 75 (17) (2015) 3456–3465.
1, J. Immunol. 182 (2009) 240–249. [54] B. Bierie, H.L. Moses, Transforming growth factor beta (TGF-beta) and
[33] C. Bogdan, Regulation of lymphocytes by nitric oxide, Methods Mol. Biol. 677 inflammation in cancer, Cytokine Growth Factor Rev. 21 (2010) 49–59.
(2011) 375–393. [55] L. Yang, L.M. DeBusk, K. Fukuda, B. Fingleton, B. Green-Jarvis, Y. Shyr, et al.,
[34] L. Yang, J. Huang, X. Ren, A.E. Gorska, A. Chytil, M. Aakre, et al., Abrogation of Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing
TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells host directly promotes tumor angiogenesis, Cancer Cell 6 (2004) 409–421.
that promote metastasis, Cancer Cell 13 (2008) 23–35. [56] M. Kujawski, M. Kortylewski, H. Lee, A. Herrmann, H. Kay, H. Yu, Stat3 mediates
[35] K.H. Parker, D.W. Beury, S. Ostrand-Rosenberg, Myeloid-derived suppressor cells: myeloid cell-dependent tumor angiogenesis in mice, J. Clin. Invest. 118 (2008)
critical cells driving immune suppression in the tumor microenvironment, Adv. 3367–3377.
Cancer Res. 128 (2015) 95–139. [57] P. Serafini, C. De Santo, I. Marigo, S. Cingarlini, L. Dolcetti, G. Gallina, et al.,
[36] P.C. Rodriguez, A.H. Zea, A.C. Ochoa, Mechanisms of tumor evasion from Derangement of immune responses by myeloid suppressor cells, Cancer
the immune response, Cancer Chemother. Biol. Response Modif. 21 (2003) Immunol. Immunother. 53 (2004) 64–72.
351–364. [58] A.S. Chung, J. Lee, N. Ferrara, Targeting the tumour vasculature: insights from
[37] M.K. Srivastava, P. Sinha, V.K. Clements, P. Rodriguez, S. Ostrand-Rosenberg, physiological angiogenesis, Nat. Rev. Cancer 10 (2010) 505–514.
Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine [59] K.C. Boelte, L.E. Gordy, S. Joyce, M.A. Thompson, L. Yang, P.C. Lin, Rgs2 mediates
and cysteine, Cancer Res. 70 (2010) 68–77. pro-angiogenic function of myeloid derived suppressor cells in the tumor
[38] G. Solinas, S. Schiarea, M. Liguori, M. Fabbri, S. Pesce, L. Zammataro, et al., microenvironment via upregulation of MCP-1, PLoS ONE 6 (2011) e18534.
Tumor-conditioned macrophages secrete migration-stimulating factor: a new [60] V. Ambros, The functions of animal microRNAs, Nature 431 (2004) 350–355.
marker for M2-polarization, influencing tumor cell motility, J. Immunol. 185 [61] D.P. Bartel, MicroRNAs: target recognition and regulatory functions, Cell 136
(2010) 642–652. (2009) 215–233.
[39] S. Kusmartsev, D.I. Gabrilovich, Effect of tumor-derived cytokines and growth [62] R.M. O’Connell, D.S. Rao, A.A. Chaudhuri, D. Baltimore, Physiological and
factors on differentiation and immune suppressive features of myeloid cells in pathological roles for microRNAs in the immune system, Nat. Rev. Immunol.
cancer, Cancer Metastasis Rev. 25 (2006) 323–331. 10 (2010) 111–122.
[40] S. Ostrand-Rosenberg, P. Sinha, Myeloid-derived suppressor cells: linking [63] R.M. O’Connell, K.D. Taganov, M.P. Boldin, G. Cheng, D. Baltimore, MicroRNA-155
inflammation and cancer, J. Immunol. 182 (2009) 4499–4506. is induced during the macrophage inflammatory response, Proc. Natl. Acad. Sci.
[41] C.A. Corzo, M.J. Cotter, P. Cheng, F. Cheng, S. Kusmartsev, E. Sotomayor, et al., U.S.A. 104 (2007) 1604–1609.
Mechanism regulating reactive oxygen species in tumor-induced myeloid- [64] A.A. Chaudhuri, A.Y. So, N. Sinha, W.S. Gibson, K.D. Taganov, R.M. O’Connell,
derived suppressor cells, J. Immunol. 182 (2009) 5693–5701. et al., MicroRNA-125b potentiates macrophage activation, J. Immunol. 187
[42] S. Nagaraj, D.I. Gabrilovich, Myeloid-derived suppressor cells, Adv. Exp. Med. (2011) 5062–5068.
Biol. 601 (2007) 213–223. [65] M. Zhang, Q. Liu, S. Mi, X. Liang, Z. Zhang, X. Su, et al., Both miR-17-5p and
[43] A.V. Ezernitchi, I. Vaknin, L. Cohen-Daniel, O. Levy, E. Manaster, A. Halabi, et al., miR-20a alleviate suppressive potential of myeloid-derived suppressor cells by
TCR zeta down-regulation under chronic inflammation is mediated by myeloid modulating STAT3 expression, J. Immunol. 186 (2011) 4716–4724.
suppressor cells differentially distributed between various lymphatic organs, [66] L. Li, J. Zhang, W. Diao, D. Wang, Y. Wei, C.Y. Zhang, et al., MicroRNA-155 and
J. Immunol. 177 (2006) 4763–4772. MicroRNA-21 promote the expansion of functional myeloid-derived suppressor
[44] R. Yang, Z. Cai, Y. Zhang, W.H. Yutzy 4th, K.F. Roby, R.B. Roden, CD80 in immune cells, J. Immunol. 192 (2014) 1034–1043.
suppression by mouse ovarian carcinoma-associated Gr-1+CD11b+ myeloid cells, [67] S. Chen, Y. Zhang, T.M. Kuzel, B. Zhang, Regulating tumor myeloid-derived
Cancer Res. 66 (2006) 6807–6815. suppressor cells by microRNAs, Cancer Cell Microenviron. 2 (2015) pii: e637.

Please cite this article in press as: Peng Qu, Li-zhen Wang, P. Charles Lin, Expansion and functions of myeloid-derived suppressor cells in the tumor microenvironment, Cancer Letters
(2015), doi: 10.1016/j.canlet.2015.10.022

You might also like