523
Views
17
CrossRef citations to date
0
Altmetric
Review

Myeloid-derived suppressor cells: their role in the pathophysiology of hematologic malignancies and potential as therapeutic targets

, &
Pages 2251-2263 | Received 26 Aug 2014, Accepted 09 Nov 2014, Published online: 21 Jan 2015

References

  • Talmadge JE. Pathways mediating the expansion and immunosuppressive activity of myeloid-derived suppressor cells and their relevance to cancer therapy. Clin Cancer Res 2007;13:5243–5248.
  • Sonnenfeld A. Leukamische reaktiones bei carcinoma. Zeitschrift f Klin Med 1929:111.
  • Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by tumors. Adv Immunol 2006;90:51–81.
  • Talmadge JE, Gabrilovich DI. History of myeloid-derived suppressor cells. Nat Rev Cancer 2013;13:739–752.
  • Goh C, Narayanan S, Hahn YS. Myeloid-derived suppressor cells: the dark knight or the joker in viral infections? Immunol Rev 2013;255:210–221.
  • Tadmor T, Attias D, Polliack A. Myeloid-derived suppressor cells—their role in haemato-oncological malignancies and other cancers and possible implications for therapy. Br J Haematol 2011; 153:557–567.
  • Donkor MK, Lahue E, Hoke TA, et al. Mammary tumor heterogeneity in the expansion of myeloid-derived suppressor cells. Int Immunopharmacol 2009;9:937–948.
  • Diaz-Montero CM, Salem ML, Nishimura MI, et al. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother 2009;58:49–59.
  • Liu Y, van Ginderachter JA, Brys L, et al. Nitric oxide-independent CTL suppression during tumor progression: association with arginase-producing (M2) myeloid cells. J Immunol 2003;170:5064–5074.
  • Corzo CA, Cotter MJ, Cheng P, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol 2009;182:5693–5701.
  • Schmid MC, Varner JA. Myeloid cells in the tumor microenvironment: modulation of tumor angiogenesis and tumor inflammation. J Oncol 2010;2010:201026.
  • Sawant A, Deshane J, Jules J, et al. Myeloid-derived suppressor cells function as novel osteoclast progenitors enhancing bone loss in breast cancer. Cancer Res 2013;73:672–682.
  • Abe F, Dafferner AJ, Donkor M, et al. Myeloid-derived suppressor cells in mammary tumor progression in FVB Neu transgenic mice. Cancer Immunol Immunother 2010;59:47–62.
  • Whiteside TL. Tricks tumors use to escape from immune control. Oral Oncol 2009;45:e119–e123.
  • Montero AJ, Diaz-Montero CM, Kyriakopoulos CE, et al. Myeloid-derived suppressor cells in cancer patients: a clinical perspective. J Immunother 2012;35:107–115.
  • Poschke I, Kiessling R. On the armament and appearances of human myeloid-derived suppressor cells. Clin Immunol 2012;144: 250–268.
  • Rashid OM, Nagahashi M, Ramachandran S, et al. Resection of the primary tumor improves survival in metastatic breast cancer by reducing overall tumor burden. Surgery 2013;153:771–778.
  • Salvadori S, Martinelli G, Zier K. Resection of solid tumors reverses T cell defects and restores protective immunity. J Immunol 2000;164:2214–2220.
  • Predina JD, Kapoor V, Judy BF, et al. Cytoreduction surgery reduces systemic myeloid suppressor cell populations and restores intratumoral immunotherapy effectiveness. J Hematol Oncol 2012;5:34.
  • Mazzoni A, Bronte V, Visintin A, et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol 2002;168:689–695.
  • Rodriguez PC, Quiceno DG, Zabaleta J, 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.
  • Dumitru CA, Moses K, Trellakis S, et al. Neutrophils and granulocytic myeloid-derived suppressor cells: immunophenotyping, cell biology and clinical relevance in human oncology. Cancer Immunol Immunother 2012;61:1155–1167.
  • Rodriguez PC, Ernstoff MS, Hernandez C, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res 2009;69: 1553–1560.
  • Kusmartsev S, Cheng F, Yu B, et al. All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination. Cancer Res 2003;63:4441–4449.
  • Kusmartsev S, Su Z, Heiser A, et al. Reversal of myeloid cell-mediated immunosuppression in patients with metastatic renal cell carcinoma. Clin Cancer Res 2008;14:8270–8278.
  • Nefedova Y, Fishman M, Sherman S, et al. Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res 2007;67:11021–11028.
  • Lee JM, Seo JH, Kim YJ, et al. The restoration of myeloid-derived suppressor cells as functional antigen-presenting cells by NKT cell help and all-trans-retinoic acid treatment. Int J Cancer 2012;131: 741–751.
  • Iclozan C, Antonia S, Chiappori A, et al. Therapeutic regulation of myeloid-derived suppressor cells and immune response to cancer vaccine in patients with extensive stage small cell lung cancer. Cancer Immunol Immunother 2013;62:909–918.
  • Young MR, Wright MA, Vellody K, et al. Skewed differentiation of bone marrow CD34 + cells of tumor bearers from dendritic toward monocytic cells, and the redirection of differentiation toward dendritic cells by 1alpha,25-dihydroxyvitamin D3. Int J Immunopharmacol 1999;21:675–688.
  • Kulbersh JS, Day TA, Gillespie MB, et al. 1alpha,25-Dihydroxyvitamin D(3) to skew intratumoral levels of immune inhibitory CD34(+) progenitor cells into dendritic cells. Otolaryngol Head Neck Surg 2009;140:235–240.
  • Walsh JE, Clark A-M, Day TA, et al. Use of α,25-dihydroxyvitamin D3 treatment to stimulate immune infiltration into head and neck squamous cell carcinoma. Hum Immunol 2010;71:659–665.
  • Walker DD, Reeves TD, de Costa AM, et al. Immunological modulation by 1alpha,25-dihydroxyvitamin D3 in patients with squamous cell carcinoma of the head and neck. Cytokine 2012;58: 448–454.
  • Le HK, Graham L, Cha E, et al. Gemcitabine directly inhibits myeloid derived suppressor cells in BALB/c mice bearing 4T1 mammary carcinoma and augments expansion of T cells from tumor-bearing mice. Int Immunopharmacol 2009;9:900–909.
  • Tomihara K, Fuse H, Heshiki W, et al. Gemcitabine chemotherapy induces phenotypic alterations of tumor cells that facilitate antitumor T cell responses in a mouse model of oral cancer. Oral Oncol 2014;50:457–467.
  • Vincent J, Mignot G, Chalmin F, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res 2010;70:3052–3061.
  • Mikyskova R, Indrova M, Simova J, et al. Genetically modified tumour vaccines producing IL-12 augment chemotherapy of HPV16-associated tumours with gemcitabine. Oncol Rep 2011;25:1683–1689.
  • Esaki S, Goshima F, Kimura H, et al. Enhanced antitumoral activity of oncolytic herpes simplex virus with gemcitabine using colorectal tumor models. Int J Cancer 2013;132:1592–1601.
  • Yanagimoto H, Shiomi H, Satoi S, et al. A phase II study of personalized peptide vaccination combined with gemcitabine for non-resectable pancreatic cancer patients. Oncol Rep 2010;24:795–801.
  • Annels NE, Shaw VE, Gabitass RF, et al. The effects of gemcitabine and capecitabine combination chemotherapy and of low-dose adjuvant GM-CSF on the levels of myeloid-derived suppressor cells in patients with advanced pancreatic cancer. Cancer Immunol Immunother 2014;63:175–183.
  • Montero AJ, Diaz-Montero CM, Deutsch YE, et al. Phase 2 study of neoadjuvant treatment with NOV-002 in combination with doxorubicin and cyclophosphamide followed by docetaxel in patients with HER-2 negative clinical stage II-IIIc breast cancer. Breast Cancer Res Treat 2012;132:215–223.
  • Xin H, Zhang C, Herrmann A, et al. Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res 2009;69:2506–2513.
  • Ko JS, Zea AH, Rini BI, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res 2009;15:2148–2157.
  • Pan PY, Wang GX, Yin B, et al. Reversion of immune tolerance in advanced malignancy: modulation of myeloid-derived suppressor cell development by blockade of stem-cell factor function. Blood 2008;111:219–228.
  • Ozao-Choy J, Ma G, Kao J, et al. The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res 2009;69:2514–2522.
  • Bose A, Taylor J, Alber S, et al. Sunitinib facilitates the activation and recruitment of therapeutic anti-tumor immunity in concert with specific vaccination. Int J Cancer 2011;129:2158–2170.
  • Amato RJ, Hawkins RE, Kaufman HL, et al. Vaccination of metastatic renal cancer patients with MVA-5T4: a randomized, double-blind, placebo-controlled phase III study. Clin Cancer Res 2010;16:5539–5547.
  • Farsaci B, Higgins JP, Hodge JW. Consequence of dose scheduling of sunitinib on host immune response elements and vaccine combination therapy. Int J Cancer 2012;130:1948–1959.
  • Gabrilovich D, Ishida T, Oyama T, et al. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood 1998;92:4150–4166.
  • Kusmartsev S, Eruslanov E, Kubler H, et al. Oxidative stress regulates expression of VEGFR1 in myeloid cells: link to tumor-induced immune suppression in renal cell carcinoma. J Immunol 2008;181: 346–353.
  • Fricke I, Mirza N, Dupont J, et al. Vascular endothelial growth factor-trap overcomes defects in dendritic cell differentiation but does not improve antigen-specific immune responses. Clin Cancer Res 2007;13:4840–4848.
  • Schilling B, Sucker A, Griewank K, et al. Vemurafenib reverses immunosuppression by myeloid derived suppressor cells. Int J Cancer 2013;133:1653–1663.
  • Sumimoto H, Imabayashi F, Iwata T, et al. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med 2006;203:1651–1656.
  • Xu J, Escamilla J, Mok S, et al. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res 2013;73:2782–2794.
  • Priceman SJ, Sung JL, Shaposhnik Z, et al. Targeting distinct tumor-infiltrating myeloid cells by inhibiting CSF-1 receptor: combating tumor evasion of antiangiogenic therapy. Blood 2010; 115:1461–1471.
  • Mok S, Koya RC, Tsui C, et al. Inhibition of CSF-1 receptor improves the antitumor efficacy of adoptive cell transfer immunotherapy. Cancer Res 2014;74:153–161.
  • Ries Carola H, Cannarile Michael A, Hoves S, et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 2014;25:846–859.
  • Chalmin F, Ladoire S, Mignot G, et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest 2010;120:457–471.
  • Filipazzi P, Valenti R, Huber V, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 2007;25:2546–2553.
  • Kujawski M, Kortylewski M, Lee H, et al. Stat3 mediates myeloid cell-dependent tumor angiogenesis in mice. J Clin Invest 2008;118:3367–3377.
  • Poschke I, Mougiakakos D, Hansson J, et al. Immature immunosuppressive CD14 + HLA-DR-/low cells in melanoma patients are Stat3hi and overexpress CD80, CD83, and DC-sign. Cancer Res 2010;70:4335–4345.
  • Nefedova Y, Cheng P, Gilkes D, et al. Activation of dendritic cells via inhibition of Jak2/STAT3 signaling. J Immunol 2005;175: 4338–4346.
  • Rodan GA, Fleisch HA. Bisphosphonates: mechanisms of action. J Clin Invest 1996;97:2692–2696.
  • Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell 2002;109:625–637.
  • Melani C, Sangaletti S, Barazzetta FM, et al. Amino-biphosphonate mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res 2007;67: 11438–11446.
  • Porembka MR, Mitchem JB, Belt BA, et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunol Immunother 2012;61:1373–1385.
  • Serafini P, Meckel K, Kelso M, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med 2006;203: 2691–2702.
  • Noonan KA, Ghosh N, Rudraraju L, et al. Targeting immune suppression with PDE5 inhibition in end-stage multiple myeloma. Cancer Immunol Res 2014;2:725–731.
  • Ghosh N, Rudraraju L, Ye X, et al. Administration of an oral PDE5 inhibitor, tadalafil in conjunction with a lenalidomide containing regimen in patients with multiple myeloma. Blood 2013;122(Suppl. 1): Abstract 1959.
  • De Santo C. Nitroaspirin corrects immune dysfunction in tumor-bearing hosts and promotes tumor eradication by cancer vaccination. Proc Natl Acad Sci USA 2005;102:4185–4190.
  • Mundy-Bosse BL, Lesinski GB, Jaime-Ramirez AC, et al. Myeloid-derived suppressor cell inhibition of the IFN response in tumor-bearing mice. Cancer Res 2011;71:5101–5110.
  • Nagaraj S, Gupta K, Pisarev V, et al. Altered recognition of antigen is a mechanism of CD8 + T cell tolerance in cancer. Nat Med 2007;13:828–835.
  • Molon B, Ugel S, Del Pozzo F, et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J Exp Med 2011; 208:1949–1962.
  • Fiorucci S, Santucci L, Cirino G, et al. IL-1 beta converting enzyme is a target for nitric oxide-releasing aspirin: new insights in the antiinflammatory mechanism of nitric oxide-releasing nonsteroidal antiinflammatory drugs. J Immunol 2000;165:5245–5254.
  • Thimmulappa R, Fuchs R, Malhotra D, et al. Preclinical evaluation of targeting the Nrf2 pathway by triterpenoids (CDDO-Im and CDDO-Me) for protection from LPS-induced inflammatory response and reactive oxygen species in human peripheral blood mononuclear cells and neutrophils. Antioxid Redox Signal 2007;9:1963–1970.
  • Li B, Abdalrahman A, Lai Y, et al. Dihydro-CDDO-trifluoroethyl amide suppresses inflammatory responses in macrophages via activation of Nrf2. Biochem Biophys Res Commun 2014;444:555–561.
  • Ling X, Konopleva M, Zeng Z, et al. The novel triterpenoid C-28 methyl ester of 2-cyano-3, 12-dioxoolen-1, 9-dien-28-oic acid inhibits metastatic murine breast tumor growth through inactivation of STAT3 signaling. Cancer Res 2007;67:4210–4218.
  • Wang YY, Zhe H, Zhao R. Preclinical evidences toward the use of triterpenoid CDDO-Me for solid cancer prevention and treatment. Mol Cancer 2014;13:30.
  • Place AE, Suh N, Williams CR, et al. The novel synthetic triterpenoid, CDDO-imidazolide, inhibits inflammatory response and tumor growth in vivo. Clin Cancer Res 2003;9:2798–2806.
  • Nagaraj S, Youn J, Weber H, et al. Anti-inflammatory triterpenoid blocks immune suppressive function of MDSCs and improves immune response in cancer. Clin Cancer Res 2010;16:1812–1823.
  • Ma X, Kundu N, Rifat S, et al. Prostaglandin E receptor EP4 antagonism inhibits breast cancer metastasis. Cancer Res 2006;66: 2923–2927.
  • Chan G, Boyle JO, Yang EK, et al. Cyclooxygenase-2 expression is up-regulated in squamous cell carcinoma of the head and neck. Cancer Res 1999;59:991–994.
  • Talmadge J, Hood K, Zobel L, et al. Chemoprevention by cyclooxygenase-2 inhibition reduces immature myeloid suppressor cell expansion. Int Immunopharmacol 2007;7:140–151.
  • Veltman J, Lambers M, van Nimwegen M, et al. COX-2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid-derived suppressor cells in mesothelioma. Celecoxib influences MDSC function. BMC Cancer 2010;10:464.
  • Sokal JE, Baccarani M, Russo D, et al. Staging and prognosis in chronic myelogenous leukemia. Semin Hematol 1988;25:49–61.
  • Gabrilove JL, Jakubowski A. Hematopoietic growth factors: biology and clinical application. J Natl Cancer Inst Monogr 1990:73–77.
  • Chen X, Liu S, Wang L, et al. Clinical significance of B7-H1 (PD-L1) expression in human acute leukemia. Cancer Biol Ther 2008;7:622–627.
  • Gupta V, Tallman MS, Weisdorf DJ. Allogeneic hematopoietic cell transplantation for adults with acute myeloid leukemia: myths, controversies, and unknowns. Blood 2011;117:2307–2318.
  • Jitschin R, Braun M, Buttner M, et al. CLL-cells induce IDOhi CD14 + HLA-DRlo myeloid-derived suppressor cells that inhibit T-cell responses and promote TRegs. Blood 2014;124:750–760.
  • Zhang L, Chen X, Liu X, et al. CD40 ligation reverses T cell tolerance in acute myeloid leukemia. J Clin Invest 2013;123:1999–2010.
  • Mussai F, De Santo C, Abu-Dayyeh I, et al. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood 2013;122:749–758.
  • Christiansson L, Soderlund S, Svensson E, et al. Increased level of myeloid-derived suppressor cells, programmed death receptor ligand 1/programmed death receptor 1, and soluble CD25 in Sokal high risk chronic myeloid leukemia. PLoS One 2013;8:e55818.
  • Jiang HJ, Fu R, Wang HQ, et al. Increased circulating of myeloid-derived suppressor cells in myelodysplastic syndrome. Chin Med J (Engl) 2013;126:2582–2584.
  • Chen X, Eksioglu EA, Zhou J, et al. Induction of myelodysplasia by myeloid-derived suppressor cells. J Clin Invest 2013;123:4595–4611.
  • Sato N, Caux C, Kitamura T, et al. Expression and factor-dependent modulation of the interleukin-3 receptor subunits on human hematopoietic cells. Blood 1993;82:752–761.
  • Huang S, Chen Z, Yu JF, et al. Correlation between IL-3 receptor expression and growth potential of human CD34 + hematopoietic cells from different tissues. Stem Cells 1999;17:265–272.
  • Testa U, Fossati C, Samoggia P, et al. Expression of growth factor receptors in unilineage differentiation culture of purified hematopoietic progenitors. Blood 1996;88:3391–3406.
  • Robinson SP, Patterson S, English N, et al. Human peripheral blood contains two distinct lineages of dendritic cells. Eur J Immunol 1999;29:2769–2778.
  • Cella M, Jarrossay D, Facchetti F, et al. Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon. Nat Med 1999;5:919–923.
  • Testa U, Riccioni R, Diverio D, et al. Interleukin-3 receptor in acute leukemia. Leukemia 2004;18:219–226.
  • Testa U, Pelosi E, Frankel A. CD 123 is a membrane biomarker and a therapeutic target in hematologic malignancies. Biomark Res 2014;2:4.
  • Jin L, Lee EM, Ramshaw HS, et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell 2009;5:31–42.
  • Kusmartsev S, Nefedova Y, Yoder D, et al. 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.
  • Kusmartsev S. Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J Leukoc Biol 2003;74:186–196.
  • Sallmyr A, Fan J, Datta K, et al. Internal tandem duplication of FLT3 (FLT3/ITD) induces increased ROS production, DNA damage, and misrepair: implications for poor prognosis in AML. Blood 2008;111:3173–3182.
  • Zhu QS, Xia L, Mills GB, et al. G-CSF induced reactive oxygen species involves Lyn-PI3-kinase-Akt and contributes to myeloid cell growth. Blood 2006;107:1847–1856.
  • Maraldi T, Prata C, Fiorentini D, et al. Induction of apoptosis in a human leukemic cell line via reactive oxygen species modulation by antioxidants. Free Radic Biol Med 2009;46:244–252.
  • Zhou FL, Zhang WG, Wei YC, et al. Involvement of oxidative stress in the relapse of acute myeloid leukemia. J Biol Chem 2010; 285:15010–15015.
  • Zhou F, Shen Q, Claret FX. Novel roles of reactive oxygen species in the pathogenesis of acute myeloid leukemia. J Leukoc Biol 2013;94:423–429.
  • Sonoda Y, Kimura T, Sakabe H, et al. Human FLT3 ligand acts on myeloid as well as multipotential progenitors derived from purified CD34 + blood progenitors expressing different levels of c-kit protein. Eur J Haematol 1997;58:257–264.
  • Solheim J, Reber A, Ashour A, et al. Spleen but not tumor infiltration by dendritic and T cells is increased by intravenous adenovirus-Flt3 ligand injection. Cancer Gene Ther 2007;14:364–371.
  • Rosborough BR, Mathews LR, Matta BM, et al. Cutting edge: Flt3 ligand mediates STAT3-independent expansion but STAT3-dependent activation of myeloid-derived suppressor cells. J Immunol 2014;192:3470–3473.
  • Drexler HG, Quentmeier H. FLT3: receptor and ligand. Growth Factors 2004;22:71–73.
  • Gilliland DG, Griffin JD. Role of FLT3 in leukemia. Curr Opin Hematol 2002;9:274–281.
  • Wang A, Braun SE, Sonpavde G, et al. Antileukemic activity of Flt3 ligand in murine leukemia. Cancer Res 2000;60:1895–1900.
  • Drexler HG. Expression of FLT3 receptor and response to FLT3 ligand by leukemic cells. Leukemia 1996;10:588–599.
  • Dehmel U, Zaborski M, Meierhoff G, et al. Effects of FLT3 ligand on human leukemia cells. I. Proliferative response of myeloid leukemia cells. Leukemia 1996;10:261–270.
  • Piacibello W, Fubini L, Sanavio F, et al. Effects of human FLT3 ligand on myeloid leukemia cell growth: heterogeneity in response and synergy with other hematopoietic growth factors. Blood 1995; 86:4105–4114.
  • Eder M, Hemmati P, Kalina U, et al. Effects of Flt3 ligand and interleukin-7 on in vitro growth of acute lymphoblastic leukemia cells. Exp Hematol 1996;24:371–377.
  • Almand B, Clark JI, Nikitina E, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol 2001;166:678–689.
  • Gabrilovich DI, Velders MP, Sotomayor EM, et al. Mechanism of immune dysfunction in cancer mediated by immature Gr-1 + myeloid cells. J Immunol 2001;166:5398–5406.
  • Mirza N. All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Cancer Res 2006;66:9299–9307.
  • Gervais A, Leveque J, Bouet-Toussaint F, et al. Dendritic cells are defective in breast cancer patients: a potential role for polyamine in this immunodeficiency. Breast Cancer Res 2005;7:R326–R335.
  • van Bockstaele DR, Lenjou M, Snoeck HW, et al. Direct effects of 13-cis and all-trans retinoic acid on normal bone marrow (BM) progenitors: comparative study on BM mononuclear cells and on isolated CD34 + BM cells. Ann Hematol 1993;66:61–66.
  • Van Bockstaele DR, Lenjou M, Snoeck HW, et al. Effect of retinoic acid (RA) on myeloid progenitors in normal bone marrow (BM). Leukemia 1994;8:214–215.
  • Castaigne S, Chomienne C, Daniel MT, et al. All-trans retinoic acid as a differentiation therapy for acute promyelocytic leukemia. I. Clinical results. Blood 1990;76:1704–1709.
  • Warrell RP Jr, Frankel SR, Miller WH Jr, et al. Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid). N Engl J Med 1991;324:1385–1393.
  • Breitman TR, Collins SJ, Keene BR. Terminal differentiation of human promyelocytic leukemic cells in primary culture in response to retinoic acid. Blood 1981;57:1000–1004.
  • Oranger A, Carbone C, Izzo M, et al. Cellular mechanisms of multiple myeloma bone disease. Clin Dev Immunol 2013;2013: 289458.
  • Escors D. Tumour immunogenicity, antigen presentation and immunological barriers in cancer immunotherapy. New J Sci 2014;2014.
  • Parrinello N, LaCava P, Tibullo D, et al. Myeloid-derived suppressor cells in patients with multiple myeloma and monoclnal gammopathy of undetermined significance. Haematologica 2010; 95(Suppl. 2): Abstract 0930.
  • Ramachandran IR, Martner A, Pisklakova A, et al. Myeloid-derived suppressor cells regulate growth of multiple myeloma by inhibiting T cells in bone marrow. J Immunol 2013;190:3815–3823.
  • Gorgun GT, Whitehill G, Anderson JL, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood 2013;121:2975–2987.
  • Favaloro J, Liyadipitiya T, Brown R, et al. Myeloid derived suppressor cells are numerically, functionally and phenotypically different in patients with multiple myeloma. Leuk Lymphoma 2014 May 12. [Epub ahead of print]
  • Noll JE, Williams SA, Tong CM, et al. Myeloma plasma cells alter the bone marrow microenvironment by stimulating the proliferation of mesenchymal stromal cells. Haematologica 2014;99:163–171.
  • Sawant A, Ponnazhagan S. Myeloid-derived suppressor cells as osteoclast progenitors: a novel target for controlling osteolytic bone metastasis. Cancer Res 2013;73:4606–4610.
  • Danilin S, Merkel AR, Johnson JR, et al. Myeloid-derived suppressor cells expand during breast cancer progression and promote tumor-induced bone destruction. Oncoimmunology 2012;1: 1484–1494.
  • Zhuang J, Zhang J, Lwin ST, et al. Osteoclasts in multiple myeloma are derived from Gr-1 + CD11b+ myeloid-derived suppressor cells. PLoS One 2012;7:e48871.
  • Charles JF, Hsu LY, Niemi EC, et al. Inflammatory arthritis increases mouse osteoclast precursors with myeloid suppressor function. J Clin Invest 2012;122:4592–4605.
  • Sahebi F, Shen Y, Thomas SH, et al. Late relapses following reduced intensity allogeneic transplantation in patients with multiple myeloma: a long-term follow-up study. Br J Haematol 2013;160: 199–206.
  • Semeraro M, Vacchelli E, Eggermont A, et al. Trial watch: lenalidomide-based immunochemotherapy. Oncoimmunology 2013; 2:e26494.
  • Li L, Zhang J, Diao W, et al. MicroRNA-155 and microRNA-21 promote the expansion of functional myeloid-derived suppressor cells. J Immunol 2014;192:1034–1043.
  • Tokioka T, Shimamoto Y, Motoyoshi K, et al. Clinical significance of monocytosis and human monocytic colony-stimulating factor in patients with adult T-cell leukaemia/lymphoma. Haematologia (Budap) 1994;26:1–9.
  • Bari A, Tadmor T, Sacchi S, et al. Monocytosis has adverse prognostic significance and impacts survival in patients with T-cell lymphomas. Leuk Res 2013;37:619–623.
  • Porrata LF, Ristow K, Habermann TM, et al. Absolute monocyte/lymphocyte count prognostic score is independent of immunohistochemically determined cell of origin in predicting survival in diffuse large B-cell lymphoma. Leuk Lymphoma 2012;53: 2159–2165.
  • Wilcox RA, Ristow K, Habermann TM, et al. The absolute monocyte and lymphocyte prognostic score predicts survival and identifies high-risk patients in diffuse large-B-cell lymphoma. Leukemia 2011;25:1502–1509.
  • Wilcox RA, Ristow K, Habermann TM, et al. The absolute monocyte count is associated with overall survival in patients newly diagnosed with follicular lymphoma. Leuk Lymphoma 2012;53: 575–580.
  • Lin Y, Gustafson MP, Bulur PA, et al. Immunosuppressive CD14 + HLA-DR(low)/- monocytes in B-cell non-Hodgkin lymphoma. Blood 2011;117:872–881.
  • Porrata LF, Ristow K, Habermann TM, et al. Peripheral blood lymphocyte/monocyte ratio at diagnosis and survival in nodular lymphocyte-predominant Hodgkin lymphoma. Br J Haematol 2012; 157:321–330.
  • Takasaki Y, Iwanaga M, Tsukasaki K, et al. Impact of visceral involvements and blood cell count abnormalities on survival in adult T-cell leukemia/lymphoma (ATLL). Leuk Res 2007;31:751–757.
  • Armitage JO. The aggressive peripheral T-cell lymphomas: 2012 update on diagnosis, risk stratification, and management. Am J Hematol 2012;87:511–519.
  • Wilcox RA, Wada DA, Ziesmer SC, et al. Monocytes promote tumor cell survival in T-cell lymphoproliferative disorders and are impaired in their ability to differentiate into mature dendritic cells. Blood 2009;114:2936–2944.
  • Farinha P, Masoudi H, Skinnider BF, et al. Analysis of multiple biomarkers shows that lymphoma-associated macrophage (LAM) content is an independent predictor of survival in follicular lymphoma (FL). Blood 2005;106:2169–2174.
  • Canioni D, Salles G, Mounier N, et al. High numbers of tumor-associated macrophages have an adverse prognostic value that can be circumvented by rituximab in patients with follicular lymphoma enrolled onto the GELA-GOELAMS FL-2000 trial. J Clin Oncol 2008;26:440–446.
  • Andjelic B, Mihaljevic B, Todorovic M, et al. The number of lymphoma-associated macrophages in tumor tissue is an independent prognostic factor in patients with follicular lymphoma. Appl Immunohistochem Mol Morphol 2012;20:41–46.
  • Dave SS, Wright G, Tan B, et al. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med 2004;351:2159–2169.
  • Ruan J, Hyjek E, Kermani P, et al. Magnitude of stromal hemangiogenesis correlates with histologic subtype of non-Hodgkin's lymphoma. Clin Cancer Res 2006;12:5622–5631.
  • Monestiroli S, Mancuso P, Burlini A, et al. Kinetics and viability of circulating endothelial cells as surrogate angiogenesis marker in an animal model of human lymphoma. Cancer Res 2001; 61:4341–4344.
  • Vacca A, Ribatti D, Ruco L, et al. Angiogenesis extent and macrophage density increase simultaneously with pathological progression in B-cell non-Hodgkin's lymphomas. Br J Cancer 1999;79:965–970.
  • Igreja C, Courinha M, Cachaco AS, et al. Characterization and clinical relevance of circulating and biopsy-derived endothelial progenitor cells in lymphoma patients. Haematologica 2007;92: 469–477.
  • Serafini P, Mgebroff S, Noonan K, et al. Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res 2008;68:5439–5449.
  • Lathers D, Clark J, Achille N, et al. Phase 1B study to improve immune responses in head and neck cancer patients using escalating doses of 25-hydroxyvitamin D3. Cancer Immunol Immunother 2004;53:422–430.
  • Gabitass RF, Annels NE, Crawshaw J, et al. Use of gemcitabine- (Gem) and fluropyrimidine (FP)-based chemotherapy to reduce myeloid-derived suppressor cells (MDSCs) in pancreatic (PC) and esophagogastric cancer (EGC). J Clin Oncol 2011;29(Suppl.): Abstract 2588.
  • Finke JH, Rini B, Ireland J, et al. Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clin Cancer Res 2008;14:6674–6682.
  • Rodriguez P, Ernstoff M, Hernandez C, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res 2009;69: 1553–1560.
  • Obermajer N, Muthuswamy R, Odunsi K, et al. PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res 2011; 71:7463–7470.
  • Wesolowski R, Markowitz J, Carson W. Myeloid derived suppressor cells - a new therapeutic target in the treatment of cancer. J Immunother Cancer 2013;1:10.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.