Skip to main content

Regulation of Tumor Progression and Metastasis by Bone Marrow-Derived Microenvironments

  • Chapter
  • First Online:
Biomarkers of the Tumor Microenvironment

Abstract

Activating mutations in driver oncogenes and loss-of-function mutations in tumor suppressor genes contribute to tumor progression and metastasis. Accordingly, therapies targeting key tumor cell-intrinsic signaling pathways are being used in clinical trials, and some have met FDA approval. However, these treatments benefit only a small proportion of patients harboring key driver mutations, and acquired resistance to these therapies presents a major impediment to effective treatment. More recently, the contribution of the tumor microenvironment (TME) has been an area of active investigation and has begun to provide critical insights into carcinogenesis. The host stromal cells in the TME coevolve with tumors and contribute to carcinogenesis in several ways. Among the host cells, bone marrow (BM)-derived cells constitute a significant fraction and directly contribute to proliferation, invasion, intravasation, extravasation, and outgrowth at the metastatic site. While the tumor-reprogrammed BM cells constitute attractive targets for anticancer therapy, recent studies have also begun to unravel their role as prognostic and predictive molecular markers of the disease.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Institutional subscriptions

References

  1. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–31.

    Article  CAS  PubMed  Google Scholar 

  2. Gao D, Mittal V. The role of bone-marrow-derived cells in tumor growth, metastasis initiation and progression. Trends Mol Med. 2009;15:333–43.

    Article  CAS  PubMed  Google Scholar 

  3. Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9:239–52.

    Article  CAS  PubMed  Google Scholar 

  4. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309–22.

    Article  CAS  PubMed  Google Scholar 

  5. Adams GB, Chabner KT, Alley IR, Olson DP, Szczepiorkowski ZM, Poznansky MC, Kos CH, Pollak MR, Brown EM, Scadden DT. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature. 2006;439:599–603.

    Article  CAS  PubMed  Google Scholar 

  6. Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, Ito K, Koh GY, Suda T. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118:149–61.

    Article  CAS  PubMed  Google Scholar 

  7. Hattori K, Heissig B, Rafii S. The regulation of hematopoietic stem cell and progenitor mobilization by chemokine SDF-1. Leuk Lymphoma. 2003;44:575–82.

    Article  CAS  PubMed  Google Scholar 

  8. Nilsson SK, Johnston HM, Whitty GA, Williams B, Webb RJ, Denhardt DT, Bertoncello I, Bendall LJ, Simmons PJ, Haylock DN. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. 2005;106:1232–9.

    Article  CAS  PubMed  Google Scholar 

  9. Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med. 2006;12:657–64.

    Article  CAS  PubMed  Google Scholar 

  10. Takafuji V, Forgues M, Unsworth E, Goldsmith P, Wang XW. An osteopontin fragment is essential for tumor cell invasion in hepatocellular carcinoma. Oncogene. 2007;26:6361–71.

    Article  CAS  PubMed  Google Scholar 

  11. McQuibban GA, Butler GS, Gong JH, Bendall L, Power C, Clark-Lewis I, Overall CM. Matrix metalloproteinase activity inactivates the CXC chemokine stromal cell-derived factor-1. J Biol Chem. 2001;276:43503–8.

    Article  CAS  PubMed  Google Scholar 

  12. Chantrain CF, Feron O, Marbaix E, DeClerck YA. Bone marrow microenvironment and tumor progression. Cancer Microenviron. 2008;1:23–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Petit I, Szyper-Kravitz M, Nagler A, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol. 2002;3:687–94.

    Article  CAS  PubMed  Google Scholar 

  14. Allavena P, Sica A, Solinas G, Porta C, Mantovani A. The inflammatory micro-environment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol Hematol. 2008;66:1–9.

    Article  PubMed  Google Scholar 

  15. Fujimoto H, Sangai T, Ishii G, Ikehara A, Nagashima T, Miyazaki M, Ochiai A. Stromal MCP-1 in mammary tumors induces tumor-associated macrophage infiltration and contributes to tumor progression. Int J Cancer J Int Cancer. 2009;125:1276–84.

    Article  CAS  Google Scholar 

  16. 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;115:1461–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gao F, Liang B, Reddy S, Farias-Eisner R, Su X. Role of inflammation-associated microenvironment in tumorigenesis and metastasis. Curr Cancer Drug Targets. 2014;14:30–45.

    Article  CAS  PubMed  Google Scholar 

  18. Du R, KV L, Petritsch C, et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell. 2008;13:206–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Steidl C, Lee T, Shah SP, et al. Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N Engl J Med. 2010;362:875–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. DeNardo DG, Brennan DJ, Rexhepaj E, et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011;1:54–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chung F-T, Lee K-Y, Wang C-W, et al. Tumor-associated macrophages correlate with response to epidermal growth factor receptor-tyrosine kinase inhibitors in advanced non-small cell lung cancer. Int J Cancer J Int Cancer. 2012;131:E227–35.

    Article  CAS  Google Scholar 

  23. Ganster RW, Taylor BS, Shao L, Geller DA. Complex regulation of human inducible nitric oxide synthase gene transcription by Stat 1 and NF-κB. Proc Natl Acad Sci U S A. 2001;98:8638–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Krausgruber T, Blazek K, Smallie T, Alzabin S, Lockstone H, Sahgal N, Hussell T, Feldmann M, Udalova IA. IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol. 2011;12:231–8.

    Article  CAS  PubMed  Google Scholar 

  25. Quatromoni JG, Eruslanov E. Tumor-associated macrophages: function, phenotype, and link to prognosis in human lung cancer. Am J Transl Res. 2012;4:376–89.

    PubMed  PubMed Central  Google Scholar 

  26. Redente EF, Dwyer-Nield LD, Merrick DT, Raina K, Agarwal R, Pao W, Rice PL, Shroyer KR, Malkinson AM. Tumor progression stage and anatomical site regulate tumor-associated macrophage and bone marrow-derived monocyte polarization. Am J Pathol. 2010;176:2972–85.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Biswas SK, Gangi L, Paul S, et al. A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-κB and enhanced IRF-3/STAT1 activation). Blood. 2006;107:2112–22.

    Article  CAS  PubMed  Google Scholar 

  28. Joyce JA, Baruch A, Chehade K, Meyer-Morse N, Giraudo E, Tsai F-Y, Greenbaum DC, Hager JH, Bogyo M, Hanahan D. Cathepsin cysteine proteases are effectors of invasive growth and angiogenesis during multistage tumorigenesis. Cancer Cell. 2004;5:443–53.

    Article  CAS  PubMed  Google Scholar 

  29. Gocheva V, Zeng W, Ke D, Klimstra D, Reinheckel T, Peters C, Hanahan D, Joyce JA. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis. Genes Dev. 2006;20:543–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gocheva V, Wang H-W, Gadea BB, Shree T, Hunter KE, Garfall AL, Berman T, Joyce JA. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 2010;24:241–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med. 2001;193:727–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lin EY, Li J-F, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA, Qian H, Xue X, Pollard JW. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006;66:11238–46.

    Article  CAS  PubMed  Google Scholar 

  33. Lin EY, Pollard JW. Tumor-associated macrophages press the angiogenic switch in breast cancer. Cancer Res. 2007;67:5064–6.

    Article  CAS  PubMed  Google Scholar 

  34. Zabuawala T, Taffany DA, Sharma SM, et al. An ets2-driven transcriptional program in tumor-associated macrophages promotes tumor metastasis. Cancer Res. 2010;70:1323–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yeo E-J, Cassetta L, Qian B-Z, et al. Myeloid WNT7b mediates the angiogenic switch and metastasis in breast cancer. Cancer Res. 2014;74:2962–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mazzieri R, Pucci F, Moi D, et al. Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell. 2011;19:512–26.

    Article  CAS  PubMed  Google Scholar 

  37. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, Coussens LM. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 2009;16:91–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chong H, Vodovotz Y, Cox GW, Barcellos-Hoff MH. Immunocytochemical localization of latent transforming growth factor-beta1 activation by stimulated macrophages. J Cell Physiol. 1999;178:275–83.

    Article  CAS  PubMed  Google Scholar 

  39. Thomas DA, Massagué J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8:369–80.

    Article  CAS  PubMed  Google Scholar 

  40. Kuang D-M, Zhao Q, Peng C, Xu J, Zhang J-P, Wu C, Zheng L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206:1327–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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;64:5839–49.

    Article  CAS  PubMed  Google Scholar 

  42. Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, Wang-Gillam A, Goedegebuure SP, Linehan DC, DeNardo DG. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74:5057–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ostrand-Rosenberg S. Immune surveillance: a balance between protumor and antitumor immunity. Curr Opin Genet Dev. 2008;18:11–8.

    Google Scholar 

  44. Gabrilovich DI, Nagaraj S. Myeloid-derived-suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sinha P, Okoro C, Foell D, Freeze HH, Ostrand-Rosenberg S, Srikrishna GO. Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J Immunol Baltim Md. 2008;181:4666–75.

    CAS  Google Scholar 

  46. Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev. 2008;222:162–79.

    Article  CAS  PubMed  Google Scholar 

  47. 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;190:3783–97.

    Article  CAS  PubMed  Google Scholar 

  48. Grohmann U, Fallarino F, Puccetti P. Tolerance, DCs and tryptophan: much ado about IDO. Trends Immunol. 2003;24:242–8.

    Article  CAS  PubMed  Google Scholar 

  49. Yu J, Wang Y, Yan F, Zhang P, Li H, Zhao H, Yan C, Yan F. Ren X (2014) Noncanonical NF-κB activation mediates STAT3-stimulated IDO upregulation in myeloid-derived suppressor cells in breast cancer. J Immunol Baltim Md. 1950;193:2574–86.

    Google Scholar 

  50. Holmgaard RB, Zamarin D, Li Y, Gasmi B, Munn DH, Allison JP, Merghoub T, Wolchok JD. Tumor-expressed IDO recruits and activates MDSCs in a Treg-dependent manner. Cell Rep. 2015;13:412–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Pekarek LA, Starr BA, Toledano AY, Schreiber H. Inhibition of tumor growth by elimination of granulocytes. J Exp Med. 1995;181:435–40.

    Article  CAS  PubMed  Google Scholar 

  52. Shojaei F, Singh M, Thompson JD, Ferrara N. Role of Bv8 in neutrophil-dependent angiogenesis in a transgenic model of cancer progression. Proc Natl Acad Sci U S A. 2008;105:2640–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol Baltim Md. 2008;181:5791–802.

    CAS  Google Scholar 

  54. Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, Fuh G, Gerber H-P, Ferrara N. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25:911–20.

    Article  CAS  PubMed  Google Scholar 

  55. Shojaei F, Wu X, Zhong C, et al. Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature. 2007;450:825–31.

    Article  CAS  PubMed  Google Scholar 

  56. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, Matrisian LM, Carbone DP, Lin PC. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004;6:409–21.

    Article  CAS  PubMed  Google Scholar 

  57. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, Worthen GS, Albelda SM. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16:183–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer. 2013;4:36–44.

    Article  CAS  PubMed  Google Scholar 

  59. Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell. 2015;161:1527–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Lyden D, Hattori K, Dias S, et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med. 2001;7:1194–201.

    Article  CAS  PubMed  Google Scholar 

  61. Peters BA, Diaz LA, Polyak K, et al. Contribution of bone marrow-derived endothelial cells to human tumor vasculature. Nat Med. 2005;11:261–2.

    Article  CAS  PubMed  Google Scholar 

  62. Nolan DJ, Ciarrocchi A, Mellick AS, et al. Bone marrow-derived endothelial progenitor cells are a major determinant of nascent tumor neovascularization. Genes Dev. 2007;21:1546–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gao D, Nolan D, McDonnell K, Vahdat L, Benezra R, Altorki N, Mittal V. Bone marrow-derived endothelial progenitor cells contribute to the angiogenic switch in tumor growth and metastatic progression. Biochim Biophys Acta. 2009;1796:33–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Kerbel RS, Benezra R, Lyden DC, et al. Endothelial progenitor cells are cellular hubs essential for neoangiogenesis of certain aggressive adenocarcinomas and metastatic transition but not adenomas. Proc Natl Acad Sci U S A. 2008;105:E54; author reply E55

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Gao D, Nolan DJ, Mellick AS, Bambino K, McDonnell K, Mittal V. Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science. 2008;319:195–8.

    Article  CAS  PubMed  Google Scholar 

  66. Mellick AS, Plummer PN, Nolan DJ, et al. Using the transcription factor inhibitor of DNA binding 1 to selectively target endothelial progenitor cells offers novel strategies to inhibit tumor angiogenesis and growth. Cancer Res. 2010;70:7273–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Shaked Y, Ciarrocchi A, Franco M, et al. Therapy-induced acute recruitment of circulating endothelial progenitor cells to tumors. Science. 2006;313:1785–7.

    Article  CAS  PubMed  Google Scholar 

  68. Shaked Y, Henke E, Roodhart JML, et al. Rapid chemotherapy-induced acute endothelial progenitor cell mobilization: implications for antiangiogenic drugs as chemosensitizing agents. Cancer Cell. 2008;14:263–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Purhonen S, Palm J, Rossi D, Kaskenpää N, Rajantie I, Ylä-Herttuala S, Alitalo K, Weissman IL, Salven P. Bone marrow-derived circulating endothelial precursors do not contribute to vascular endothelium and are not needed for tumor growth. Proc Natl Acad Sci U S A. 2008;105:6620–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Burchfield JS, Dimmeler S. Role of paracrine factors in stem and progenitor cell mediated cardiac repair and tissue fibrosis. Fibrogenesis Tissue Repair. 2008;1:4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Stockmann C, Doedens A, Weidemann A, Zhang N, Takeda N, Greenberg JI, Cheresh DA, Johnson RS. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature. 2008;456:814–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lee S, Chen TT, Barber CL, Jordan MC, Murdock J, Desai S, Ferrara N, Nagy A, Roos KP, Iruela-Arispe ML. Autocrine VEGF signaling is required for vascular homeostasis. Cell. 2007;130:691–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Direkze NC, Forbes SJ, Brittan M, Hunt T, Jeffery R, Preston SL, Poulsom R, Hodivala-Dilke K, Alison MR, Wright NA. Multiple organ engraftment by bone-marrow-derived myofibroblasts and fibroblasts in bone-marrow-transplanted mice. Stem Cells Dayt Ohio. 2003;21:514–20.

    Article  Google Scholar 

  74. Direkze NC, Hodivala-Dilke K, Jeffery R, Hunt T, Poulsom R, Oukrif D, Alison MR, Wright NA. Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts. Cancer Res. 2004;64:8492–5.

    Article  CAS  PubMed  Google Scholar 

  75. Vong S, Kalluri R. The role of stromal myofibroblast and extracellular matrix in tumor angiogenesis. Genes Cancer. 2011;2:1139–45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Rajantie I, Ilmonen M, Alminaite A, Ozerdem U, Alitalo K, Salven P. Adult bone marrow-derived cells recruited during angiogenesis comprise precursors for periendothelial vascular mural cells. Blood. 2004;104:2084–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Song S, Ewald AJ, Stallcup W, Werb Z, Bergers G. PDGFRbeta+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat Cell Biol. 2005;7:870. –879

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Jodele S, Chantrain CF, Blavier L, Lutzko C, Crooks GM, Shimada H, Coussens LM, Declerck YA. The contribution of bone marrow-derived cells to the tumor vasculature in neuroblastoma is matrix metalloproteinase-9 dependent. Cancer Res. 2005;65:3200–8.

    Google Scholar 

  79. Lamagna C, Bergers G. The bone marrow constitutes a reservoir of pericyte progenitors. J Leukoc Biol. 2006;80:677–81.

    Article  CAS  PubMed  Google Scholar 

  80. Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER, Graf T, Pollard JW, Segall J, Condeelis J. A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res. 2004;64:7022–9.

    Article  CAS  PubMed  Google Scholar 

  81. Goswami S, Sahai E, Wyckoff JB, Cammer M, Cox D, Pixley FJ, Stanley ER, Segall JE, Condeelis JS. Macrophages promote the invasion of breast carcinoma cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res. 2005;65:5278–83.

    Article  CAS  PubMed  Google Scholar 

  82. Ishihara D, Dovas A, Hernandez L, Pozzuto M, Wyckoff J, Segall JE, Condeelis JS, Bresnick AR, Cox D. Wiskott-Aldrich syndrome protein regulates leukocyte-dependent breast cancer metastasis. Cell Rep. 2013;4:429–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Hernandez L, Smirnova T, Kedrin D, et al. The EGF/CSF-1 paracrine invasion loop can be triggered by heregulin beta1 and CXCL12. Cancer Res. 2009;69:3221–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Gertler F, Condeelis J. Metastasis: tumor cells becoming Menacing. Trends Cell Biol. 2011;21:81–90.

    Article  CAS  PubMed  Google Scholar 

  85. Roussos ET, Balsamo M, Alford SK, et al. Mena invasive (MenaINV) promotes multicellular streaming motility and transendothelial migration in a mouse model of breast cancer. J Cell Sci. 2011;124:2120–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Harney AS, Arwert EN, Entenberg D, Wang Y, Guo P, Qian B-Z, Oktay MH, Pollard JW, Jones JG, Condeelis JS. Real-time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage-derived VEGFA. Cancer Discov. 2015;5:932–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Wyckoff JB, Wang Y, Lin EY, Li J, Goswami S, Stanley ER, Segall JE, Pollard JW, Condeelis J. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007;67:2649–56.

    Article  CAS  PubMed  Google Scholar 

  88. Roh-Johnson M, Bravo-Cordero JJ, Patsialou A, Sharma VP, Guo P, Liu H, Hodgson L, Condeelis J. Macrophage contact induces RhoA GTPase signaling to trigger tumor cell intravasation. Oncogene. 2014;33:4203–12.

    Article  CAS  PubMed  Google Scholar 

  89. Liu C-Y, J-Y X, Shi X-Y, Huang W, Ruan T-Y, Xie P, Ding J-L. M2-polarized tumor-associated macrophages promoted epithelial–mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Lab Investig. 2013;93:844–54.

    Article  CAS  PubMed  Google Scholar 

  90. Bekes EM, Schweighofer B, Kupriyanova TA, Zajac E, Ardi VC, Quigley JP, Deryugina EI. Tumor-recruited neutrophils and neutrophil TIMP-free MMP-9 regulate coordinately the levels of tumor angiogenesis and efficiency of malignant cell intravasation. Am J Pathol. 2011;179:1455–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhou S-L, Dai Z, Zhou Z-J, et al. CXCL5 contributes to tumor metastasis and recurrence of intrahepatic cholangiocarcinoma by recruiting infiltrative intratumoral neutrophils. Carcinogenesis. 2014;35:597–605.

    Article  CAS  PubMed  Google Scholar 

  92. Sharma B, Nannuru KC, Varney ML, Singh RK. Host Cxcr2-dependent regulation of mammary tumor growth and metastasis. Clin Exp Metastasis. 2015;32:65–72.

    Article  CAS  PubMed  Google Scholar 

  93. Yang L, Huang J, Ren X, et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13:23–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Kitamura T, Kometani K, Hashida H, et al. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nat Genet. 2007;39:467–75.

    Article  CAS  PubMed  Google Scholar 

  95. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R, Weinberg RA. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449:557–63.

    Article  CAS  PubMed  Google Scholar 

  96. Maheswaran S, Haber DA. Circulating tumor cells: a window into cancer biology and metastasis. Curr Opin Genet Dev. 2010;20:96–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Reymond N, d’Água BB, Ridley AJ. Crossing the endothelial barrier during metastasis. Nat Rev Cancer. 2013;13:858–70.

    Article  CAS  PubMed  Google Scholar 

  98. Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, Jirousková M, Degen JL. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood. 2005;105:178–85.

    Article  CAS  PubMed  Google Scholar 

  99. Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, Hu Z, Barney KA, Degen JL. Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood. 2007;110:133–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Camerer E, Qazi AA, Duong DN, Cornelissen I, Advincula R, Coughlin SR. Platelets, protease-activated receptors, and fibrinogen in hematogenous metastasis. Blood. 2004;104:397–401.

    Article  CAS  PubMed  Google Scholar 

  101. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011;20:576–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Ferjančič Š, Gil-Bernabé AM, Hill SA, Allen PD, Richardson P, Sparey T, Savory E, McGuffog J, Muschel RJ. VCAM-1 and VAP-1 recruit myeloid cells that promote pulmonary metastasis in mice. Blood. 2013;121:3289–97.

    Article  PubMed  CAS  Google Scholar 

  103. Chen Q, Zhang XH-F, Massagué J. Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell. 2011;20:538–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Huh SJ, Liang S, Sharma A, Dong C, Robertson GP. Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development. Cancer Res. 2010;70:6071–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Spicer JD, McDonald B, Cools-Lartigue JJ, Chow SC, Giannias B, Kubes P, Ferri LE. Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells. Cancer Res. 2012;72:3919–27.

    Article  CAS  PubMed  Google Scholar 

  106. Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias B, Bourdeau F, Kubes P, Ferri L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 2013; doi:10.1172/JCI67484.

  107. Coupland LA, Chong BH, Parish CR. Platelets and P-selectin control tumor cell metastasis in an organ-specific manner and independently of NK cells. Cancer Res. 2012;72:4662–71.

    Article  CAS  PubMed  Google Scholar 

  108. Irimura T, Nakamori S, Matsushita Y, Taniuchi Y, Todoroki N, Tsuji T, Izumi Y, Kawamura Y, Hoff SD, Cleary KR. Colorectal cancer metastasis determined by carbohydrate-mediated cell adhesion: role of sialyl-LeX antigens. Semin Cancer Biol. 1993;4:319–24.

    CAS  PubMed  Google Scholar 

  109. O I OL, Kieber-Emmons T, Blaszczyk-Thurin M. Role of SA-Le(a) and E-selectin in metastasis assessed with peptide antagonist. Peptides. 2002;23:999–1010.

    Article  PubMed  Google Scholar 

  110. Zipin A, Israeli-Amit M, Meshel T, et al. Tumor-microenvironment interactions: the fucose-generating FX enzyme controls adhesive properties of colorectal cancer cells. Cancer Res. 2004;64:6571–8.

    Article  CAS  PubMed  Google Scholar 

  111. Qian B, Deng Y, Im JH, Muschel RJ, Zou Y, Li J, Lang RA, Pollard JW. A distinct macrophage population mediates metastatic breast cancer cell extravasation, establishment and growth. PLoS One. 2009;4:e6562.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Qian B-Z, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, Kaiser EA, Snyder LA, Pollard JW. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475:222–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Zhao L, Lim SY, Gordon-Weeks AN, Tapmeier TT, Im JH, Cao Y, Beech J, Allen D, Smart S, Muschel RJ. Recruitment of a myeloid cell subset (CD11b/Gr1 mid) via CCL2/CCR2 promotes the development of colorectal cancer liver metastasis. Hepatol Baltim Md. 2013;57:829–39.

    Article  CAS  Google Scholar 

  114. Viguier M, Lemaître F, Verola O, Cho M-S, Gorochov G, Dubertret L, Bachelez H, Kourilsky P. Ferradini L (2004) Foxp3 expressing CD4+CD25(high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. J Immunol Baltim Md. 1950;173:1444–53.

    Google Scholar 

  115. Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9.

    Article  CAS  PubMed  Google Scholar 

  116. Fidler IJ. The pathogenesis of cancer metastasis: the “seed and soil” hypothesis revisited. Nat Rev Cancer. 2003;3:453–8.

    Article  CAS  PubMed  Google Scholar 

  117. Psaila B, Lyden D. The metastatic niche: adapting the foreign soil. Nat Rev Cancer. 2009;9:285–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Hiratsuka S, Watanabe A, Aburatani H, Maru Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat Cell Biol. 2006;8:1369–75.

    Article  CAS  PubMed  Google Scholar 

  120. Hiratsuka S, Watanabe A, Sakurai Y, Akashi-Takamura S, Ishibashi S, Miyake K, Shibuya M, Akira S, Aburatani H, Maru Y. The S100A8–serum amyloid A3–TLR4 paracrine cascade establishes a pre-metastatic phase. Nat Cell Biol. 2008;10:1349–55.

    Article  CAS  PubMed  Google Scholar 

  121. Hiratsuka S, Ishibashi S, Tomita T, Watanabe A, Akashi-Takamura S, Murakami M, Kijima H, Miyake K, Aburatani H, Maru Y. Primary tumours modulate innate immune signalling to create pre-metastatic vascular hyperpermeability foci. Nat Commun. 2013;4:1853.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Hiratsuka S, Nakamura K, Iwai S, Murakami M, Itoh T, Kijima H, Shipley JM, Senior RM, Shibuya M. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell. 2002;2:289–300.

    Article  CAS  PubMed  Google Scholar 

  123. Bergers G, Brekken R, McMahon G, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol. 2000;2:737–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. 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;109:625–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Sceneay J, Chow MT, Chen A, et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 2012;72:3906–11.

    Article  CAS  PubMed  Google Scholar 

  126. Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, Le Q-T, Giaccia AJ. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009;15:35–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Cox TR, Bird D, Baker A-M, Barker HE, Ho MW-Y, Lang G, Erler JT. LOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasis. Cancer Res. 2013;73:1721–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Cox TR, Erler JT. Molecular pathways: connecting fibrosis and solid tumor metastasis. Clin Cancer Res. 2014;20:3637–43.

    Article  CAS  PubMed  Google Scholar 

  129. Cox TR, Rumney RMH, Schoof EM, et al. The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase. Nature. 2015;522:106–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Said N, Sanchez-Carbayo M, Smith SC, Theodorescu D. RhoGDI2 suppresses lung metastasis in mice by reducing tumor versican expression and macrophage infiltration. J Clin Invest. 2012;122:1503–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. van Deventer HW, Palmieri DA, Wu QP, McCook EC. Serody JS (2013) Circulating fibrocytes prepare the lung for cancer metastasis by recruiting Ly-6C+ monocytes via CCL2. J Immunol Baltim Md. 1950;190:4861–7.

    Google Scholar 

  132. Gil-Bernabé AM, Ferjancic S, Tlalka M, et al. Recruitment of monocytes/macrophages by tissue factor-mediated coagulation is essential for metastatic cell survival and premetastatic niche establishment in mice. Blood. 2012;119:3164–75.

    Article  PubMed  CAS  Google Scholar 

  133. Catena R, Bhattacharya N, Rayes TE, et al. Bone marrow-derived Gr1+ cells can generate a metastasis-resistant microenvironment via induced secretion of thrombospondin-1. Cancer Discov. 2013;3:578–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Kowanetz M, Wu X, Lee J, et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc Natl Acad Sci U S A. 2010;107:21248–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Granot Z, Henke E, Comen EA, King TA, Norton L, Benezra R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 2011;20:300–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Deng J, Liu Y, Lee H, et al. S1PR1-STAT3 signaling is crucial for myeloid cell colonization at future metastatic sites. Cancer Cell. 2012;21:642–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Monteiro AC, Leal AC, Gonçalves-Silva T, Mercadante ACT, Kestelman F, Chaves SB, Azevedo RB, Monteiro JP, Bonomo A. T cells induce pre-metastatic osteolytic disease and help bone metastases establishment in a mouse model of metastatic breast cancer. PLoS One. 2013;8:e68171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Olkhanud PB, Baatar D, Bodogai M, Hakim F, Gress R, Anderson RL, Deng J, Xu M, Briest S, Biragyn A. Breast cancer lung metastasis requires expression of chemokine receptor CCR4 and regulatory T cells. Cancer Res. 2009;69:5996–6004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16:717–27.

    Article  CAS  PubMed  Google Scholar 

  140. Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA, Massagué J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3:537–49.

    Article  CAS  PubMed  Google Scholar 

  142. Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9:274–84.

    Article  CAS  PubMed  Google Scholar 

  143. Obenauf AC, Massagué J. Surviving at a distance: organ specific metastasis. Trends Cancer. 2015;1:76–91.

    Article  PubMed Central  Google Scholar 

  144. Müller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410:50–6.

    Article  PubMed  Google Scholar 

  145. Li YM, Pan Y, Wei Y, et al. Upregulation of CXCR4 is essential for HER2-mediated tumor metastasis. Cancer Cell. 2004;6:459–69.

    Article  CAS  PubMed  Google Scholar 

  146. Marchesi F, Piemonti L, Fedele G, et al. The chemokine receptor CX3CR1 is involved in the neural tropism and malignant behavior of pancreatic ductal adenocarcinoma. Cancer Res. 2008;68:9060–9.

    Article  CAS  PubMed  Google Scholar 

  147. Wang H, Fu W, Im JH, et al. Tumor cell alpha3beta1 integrin and vascular laminin-5 mediate pulmonary arrest and metastasis. J Cell Biol. 2004;164:935–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Kim S, Takahashi H, Lin W-W, Descargues P, Grivennikov S, Kim Y, Luo J-L, Karin M. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009;457:102–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer. 2002;2:584–93.

    Article  CAS  PubMed  Google Scholar 

  150. Jones DH, Nakashima T, Sanchez OH, et al. Regulation of cancer cell migration and bone metastasis by RANKL. Nature. 2006;440:692–6.

    Article  CAS  PubMed  Google Scholar 

  151. Sohara Y, Shimada H, DeClerck YA. Mechanisms of bone invasion and metastasis in human neuroblastoma. Cancer Lett. 2005;228:203–9.

    Article  CAS  PubMed  Google Scholar 

  152. Ara T, Song L, Shimada H, Keshelava N, Russell HV, Metelitsa LS, Groshen SG, Seeger RC, DeClerck YA. Interleukin-6 in the bone marrow microenvironment promotes the growth and survival of neuroblastoma cells. Cancer Res. 2009;69:329–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Hoshino A, Costa-Silva B, Shen T-L, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015; doi:10.1038/nature15756.

  154. El Rayes T, Catena R, Lee S, et al. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc Natl Acad Sci U S A. 2015;112:16000–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Wculek SK, Malanchi I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature. 2015;528:413–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Gao D, Joshi N, Choi H, et al. Myeloid progenitor cells in the premetastatic lung promote metastases by inducing mesenchymal to epithelial transition. Cancer Res. 2012;72:1384–94.

    Article  CAS  PubMed  Google Scholar 

  157. Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr H-A, Delaloye J-F, Huelsken J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2012;481:85–9.

    Article  CAS  Google Scholar 

  158. Wang Z, Ouyang G. Periostin: a bridge between cancer stem cells and their metastatic niche. Cell Stem Cell. 2012;10:111–2.

    Article  CAS  PubMed  Google Scholar 

  159. Coutu DL, JH W, Monette A, Rivard G-É, Blostein MD, Galipeau J. Periostin, a member of a novel family of vitamin K-dependent proteins, is expressed by mesenchymal stromal cells. J Biol Chem. 2008;283:17991–8001.

    Article  CAS  PubMed  Google Scholar 

  160. Ghajar CM, Peinado H, Mori H, et al. The perivascular niche regulates breast tumour dormancy. Nat Cell Biol. 2013;15:807–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Sanford DE, Belt BA, Panni RZ, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res Off J Am Assoc Cancer Res. 2013;19:3404–15.

    Article  CAS  Google Scholar 

  162. Tomita M, Shimizu T, Ayabe T, Yonei A, Onitsuka T. Preoperative neutrophil to lymphocyte ratio as a prognostic predictor after curative resection for non-small cell lung cancer. Anticancer Res. 2011;31:2995–8.

    PubMed  Google Scholar 

  163. Robinson BD, Sica GL, Liu Y-F, Rohan TE, Gertler FB, Condeelis JS, Jones JG. Tumor microenvironment of metastasis in human breast carcinoma: a potential prognostic marker linked to hematogenous dissemination. Clin Cancer Res Off J Am Assoc Cancer Res. 2009;15:2433–41.

    Article  CAS  Google Scholar 

  164. Rohan TE, Xue X, Lin H-M, et al. Tumor microenvironment of metastasis and risk of distant metastasis of breast cancer. J Natl Cancer Inst. 2014; doi:10.1093/jnci/dju136.

  165. Welsh TJ, Green RH, Richardson D, Waller DA, O’Byrne KJ, Bradding P. Macrophage and mast-cell invasion of tumor cell islets confers a marked survival advantage in non-small-cell lung cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2005;23:8959–67.

    Article  Google Scholar 

  166. Kim D-W, Min HS, Lee K-H, et al. High tumour islet macrophage infiltration correlates with improved patient survival but not with EGFR mutations, gene copy number or protein expression in resected non-small cell lung cancer. Br J Cancer. 2008;98:1118–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Dai F, Liu L, Che G, Yu N, Pu Q, Zhang S, Ma J, Ma L, You Z. The number and microlocalization of tumor-associated immune cells are associated with patient’s survival time in non-small cell lung cancer. BMC Cancer. 2010;10:220.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Ohri CM, Shikotra A, Green RH, Waller DA, Bradding P. Macrophages within NSCLC tumour islets are predominantly of a cytotoxic M1 phenotype associated with extended survival. Eur Respir J Off J Eur Soc Clin Respir Physiol. 2009;33:118–26.

    CAS  Google Scholar 

  169. Takanami I, Takeuchi K, Kodaira S. Tumor-associated macrophage infiltration in pulmonary adenocarcinoma: association with angiogenesis and poor prognosis. Oncology. 1999;57:138–42.

    Article  CAS  PubMed  Google Scholar 

  170. Chen JJW, Yao P-L, Yuan A, Hong T-M, Shun C-T, Kuo M-L, Lee Y-C, Yang P-C. Up-regulation of tumor interleukin-8 expression by infiltrating macrophages: its correlation with tumor angiogenesis and patient survival in non-small cell lung cancer. Clin Cancer Res Off J Am Assoc. Cancer Res. 2003;9:729–37.

    CAS  Google Scholar 

  171. Ohtaki Y, Ishii G, Nagai K, Ashimine S, Kuwata T, Hishida T, Nishimura M, Yoshida J, Takeyoshi I, Ochiai A. Stromal macrophage expressing CD204 is associated with tumor aggressiveness in lung adenocarcinoma. J Thorac Oncol Off Publ Int Assoc Study Lung Cancer. 2010;5:1507–15.

    Google Scholar 

  172. Zhang B, Yao G, Zhang Y, Gao J, Yang B, Rao Z, Gao J. M2-polarized tumor-associated macrophages are associated with poor prognoses resulting from accelerated lymphangiogenesis in lung adenocarcinoma. Clin São Paulo Braz. 2011;66:1879–86.

    Article  Google Scholar 

  173. Wang R, Lu M, Zhang J, Chen S, Luo X, Qin Y, Chen H. Increased IL-10 mRNA expression in tumor-associated macrophage correlated with late stage of lung cancer. J Exp Clin Cancer Res CR. 2011;30:62.

    Article  PubMed  CAS  Google Scholar 

  174. Zeni E, Mazzetti L, Miotto D, et al. Macrophage expression of interleukin-10 is a prognostic factor in nonsmall cell lung cancer. Eur Respir J Off J Eur Soc Clin Respir Physiol. 2007;30:627–32.

    CAS  Google Scholar 

  175. Wang R, Zhang J, Chen S, Lu M, Luo X, Yao S, Liu S, Qin Y, Chen H. Tumor-associated macrophages provide a suitable microenvironment for non-small lung cancer invasion and progression. Lung Cancer Amst Neth. 2011;74:188–96.

    Article  Google Scholar 

  176. Nagaraj S, Youn J-I, Weber H, et al. Anti-inflammatory triterpenoid blocks immune suppressive function of MDSCs and improves immune response in cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2010;16:1812–23.

    Article  CAS  Google Scholar 

  177. Huang A, Zhang B, Wang B, Zhang F, Fan K-X, Guo Y-J. Increased CD14(+)HLA-DR (−/low) myeloid-derived suppressor cells correlate with extrathoracic metastasis and poor response to chemotherapy in non-small cell lung cancer patients. Cancer Immunol Immunother CII. 2013;62:1439–51.

    Article  CAS  PubMed  Google Scholar 

  178. Zhang B, Wang Z, Wu L, Zhang M, Li W, Ding J, Zhu J, Wei H, Zhao K. Circulating and tumor-infiltrating myeloid-derived suppressor cells in patients with colorectal carcinoma. PLoS One. 2013;8:e57114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Meyer C, Cagnon L, Costa-Nunes CM, Baumgaertner P, Montandon N, Leyvraz L, Michielin O, Romano E, Speiser DE. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother CII. 2014;63:247–57.

    Article  CAS  PubMed  Google Scholar 

  180. Raychaudhuri B, Rayman P, Ireland J, Ko J, Rini B, Borden EC, Garcia J, Vogelbaum MA, Finke J. Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma. Neuro-Oncol. 2011;13:591–9.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Najjar YG, Finke JH. Clinical perspectives on targeting of myeloid derived suppressor cells in the treatment of cancer. Front Oncol. 2013; doi:10.3389/fonc.2013.00049.

  182. Gabrilovich DI, Velders MP, Sotomayor EM. Kast WM (2001) Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol Baltim Md. 1950;166:5398–406.

    Google Scholar 

  183. Mirza N, Fishman M, Fricke I, Dunn M, Neuger AM, Frost TJ, Lush RM, Antonia S, Gabrilovich DI. All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Cancer Res. 2006;66:9299–307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. 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 Off J Am Assoc Cancer Res. 2009;15:2148–57.

    Article  CAS  Google Scholar 

  185. Ko JS, Rayman P, Ireland J, Swaidani S, Li G, Bunting KD, Rini B, Finke JH, Cohen PA. Direct and differential suppression of myeloid-derived suppressor cell subsets by sunitinib is compartmentally constrained. Cancer Res. 2010;70:3526–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Friberg M, Jennings R, Alsarraj M, Dessureault S, Cantor A, Extermann M, Mellor AL, Munn DH, Antonia SJ. Indoleamine 2,3-dioxygenase contributes to tumor cell evasion of T cell-mediated rejection. Int J Cancer. 2002;101:151–5.

    Article  CAS  PubMed  Google Scholar 

  187. Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E, Prendergast GC. Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med. 2005;11:312–9.

    Article  CAS  PubMed  Google Scholar 

  188. Gu T, Rowswell-Turner RB, Kilinc MO, Egilmez NK. Central role of IFNγ–indoleamine 2,3-dioxygenase axis in regulation of interleukin-12-mediated antitumor immunity. Cancer Res. 2010;70:129–38.

    Article  CAS  PubMed  Google Scholar 

  189. Holmgaard RB, Zamarin D, Munn DH, Wolchok JD, Allison JP. Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4. J Exp Med. 2013;210:1389–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Allinen M, Beroukhim R, Cai L, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004;6:17–32.

    Article  CAS  PubMed  Google Scholar 

  191. Durrans A, Gao D, Gupta R, et al. Identification of reprogrammed myeloid cell transcriptomes in NSCLC. PLoS One. 2015;10:e0129123.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  192. Choi H, Sheng J, Gao D, et al. Transcriptome analysis of individual stromal cell populations identifies stroma-tumor crosstalk in mouse lung cancer model. Cell Rep. 2015;10:1187–201.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Tina El Rayes or Vivek Mittal .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2017 Springer International Publishing Switzerland

About this chapter

Cite this chapter

El Rayes, T., Gao, D., Altorki, N.K., Cox, T.R., Erler, J.T., Mittal, V. (2017). Regulation of Tumor Progression and Metastasis by Bone Marrow-Derived Microenvironments. In: Akslen, L., Watnick, R. (eds) Biomarkers of the Tumor Microenvironment. Springer, Cham. https://doi.org/10.1007/978-3-319-39147-2_13

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-39147-2_13

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-39145-8

  • Online ISBN: 978-3-319-39147-2

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics