Skip to main content

Advertisement

Log in

Mammary Gland Involution as an Immunotherapeutic Target for Postpartum Breast Cancer

  • Published:
Journal of Mammary Gland Biology and Neoplasia Aims and scope Submit manuscript

Abstract

Postpartum mammary gland involution has been identified as tumor-promotional and is proposed to contribute to the increased rates of metastasis and poor survival observed in postpartum breast cancer patients. In rodent models, the involuting mammary gland microenvironment is sufficient to induce enhanced tumor cell growth, local invasion, and metastasis. Postpartum involution shares many attributes with wound healing, including upregulation of genes involved in immune responsiveness and infiltration of tissue by immune cells. In rodent models, treatment with non-steroidal anti-inflammatory drugs (NSAIDs) ameliorates the tumor-promotional effects of involution, consistent with the immune milieu of the involuting gland contributing to tumor promotion. Currently, immunotherapy is being investigated as a means of breast cancer treatment with the purpose of identifying ways to enhance anti-tumor immune responses. Here we review evidence for postpartum mammary gland involution being a uniquely defined ‘hot-spot’ of pro-tumorigenic immune cell infiltration, and propose that immunotherapy should be explored for prevention and treatment of breast cancers that arise in this environment.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig 1
Fig 2
Fig 3
Fig 4

Similar content being viewed by others

Abbreviations

ATP:

adenosine triphosphate

arg-1:

arginase 1

AMP:

adenosine monophosphate

BMI:

body mass index

CCL:

chemokine (C-C motif) ligand

CD:

cluster of differentiation

CK:

cytokeratin

COX-2:

cyclooxygenase-2

CSF-1:

colony stimulating factor-1

CSF-1R:

colony stimulating factor-1 receptor

CTLA-4:

cytotoxic T-lymphocyte antigen 4

CXCL:

chemoattractant chemokine (C-X-C motif) ligand

ECM:

extracellular matrix

EGF:

epidermal growth factor

FDA:

Food and Drug Administration

FGF:

fibroblast growth factor

GM-CSF:

granulocyte-macrophage colony-stimulating factor

ICE:

interleukin-1β converting enzyme

IFNγ:

interferon gamma

IL:

interleukin

iNOS:

inducible nitric oxide synthase

Inv:

Involution

Lac:

lactation

LBP:

lipopolysaccharide binding protein

LRP1:

low density lipoprotein-related protein 1

LPC:

lysophosphatidylcholine

LPS:

lipopolysaccharide

MCP-1:

monocyte chemoattractant protein 12

MHC:

major histocompatibility complex

MMPs:

matrix metalloproteinases

MMTV:

mouse mammary tumor virus

MSC:

myeloid suppressor cell

NK:

natural killer

NOD:

non-obese diabetic

NSAIDs:

non-steroidal anti-inflammatory drugs

PD-L1:

programmed death ligand 1

PD-1:

programmed cell death protein 1

PGE2 :

prostaglandin E2

Preg:

Pregnant

PyMT:

polyoma virus middle T antigen

Reg:

Regressed

SCID:

severe combined immunodeficiency

STAT3:

signal transducer and activator of transcription 3

TGF-β:

transforming growth factor beta

TNFα:

tumor necrosis factor alpha

Treg:

regulatory T cell

uPA:

urokinase-type plasminogen activator

UTP:

uridine-5’-triphosphate

VEGF:

vascular endothelial growth factor

Vir:

Virgin

References

  1. Sasmono RT, Oceandy D, Pollard JW, Tong W, Pavli P, Wainwright BJ, et al. A macrophage colony-stimulating factor receptor-green fluorescent protein transgene is expressed throughout the mononuclear phagocyte system of the mouse. Blood. 2003;101(3):1155–63. doi:10.1182/blood-2002-02-0569.

    CAS  PubMed  Google Scholar 

  2. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445–55. doi:10.1038/nature12034.

    CAS  PubMed Central  PubMed  Google Scholar 

  3. Gouon-Evans V, Rothenberg ME, Pollard JW. Postnatal mammary gland development requires macrophages and eosinophils. Development. 2000;127(11):2269–82.

    CAS  PubMed  Google Scholar 

  4. Van Nguyen A, Pollard JW. Colony stimulating factor-1 is required to recruit macrophages into the mammary gland to facilitate mammary ductal outgrowth. Dev Biol. 2002;247(1):11–25. doi:10.1006/dbio.2002.0669.

    PubMed  Google Scholar 

  5. Chua AC, Hodson LJ, Moldenhauer LM, Robertson SA, Ingman WV. Dual roles for macrophages in ovarian cycle-associated development and remodelling of the mammary gland epithelium. Development. 2010;137(24):4229–38. doi:10.1242/dev.059261.

    CAS  PubMed  Google Scholar 

  6. Hodson LJ, Chua AC, Evdokiou A, Robertson SA, Ingman WV. Macrophage phenotype in the mammary gland fluctuates over the course of the estrous cycle and is regulated by ovarian steroid hormones. Biol Reprod. 2013;89(3):6. doi:10.1095/biolreprod.113.109561.

    Google Scholar 

  7. Pollard JW, Hennighausen L. Colony stimulating factor 1 is required for mammary gland development during pregnancy. Proc Natl Acad Sci U S A. 1994;91(20):9312–6.

    CAS  PubMed Central  PubMed  Google Scholar 

  8. O’Brien J, Martinson H, Durand-Rougely C, Schedin P. Macrophages are crucial for epithelial cell death and adipocyte repopulation during mammary gland involution. Development. 2012;139(2):269–75. doi:10.1242/dev.071696.

    PubMed  Google Scholar 

  9. Lilla JN, Joshi RV, Craik CS, Werb Z. Active plasma kallikrein localizes to mast cells and regulates epithelial cell apoptosis, adipocyte differentiation, and stromal remodeling during mammary gland involution. J Biol Chem. 2009;284(20):13792–803. doi:10.1074/jbc.M900508200.

    CAS  PubMed Central  PubMed  Google Scholar 

  10. Lambe M, Hsieh C, Trichopoulos D, Ekbom A, Pavia M, Adami HO. Transient increase in the risk of breast cancer after giving birth. N Engl J Med. 1994;331(1):5–9. doi:10.1056/NEJM199407073310102.

    CAS  PubMed  Google Scholar 

  11. Liu Q, Wuu J, Lambe M, Hsieh SF, Ekbom A, Hsieh CC. Transient increase in breast cancer risk after giving birth: postpartum period with the highest risk (Sweden). Cancer Causes Control. 2002;13(4):299–305.

    PubMed  Google Scholar 

  12. Schedin P. Pregnancy-associated breast cancer and metastasis. Nat Rev Cancer. 2006;6(4):281–91. doi:10.1038/nrc1839.

    CAS  PubMed  Google Scholar 

  13. Albrektsen G, Heuch I, Hansen S, Kvale G. Breast cancer risk by age at birth, time since birth and time intervals between births: exploring interaction effects. Br J Cancer. 2005;92(1):167–75. doi:10.1038/sj.bjc.6602302.

    CAS  PubMed Central  PubMed  Google Scholar 

  14. Chie WC, Hsieh C, Newcomb PA, Longnecker MP, Mittendorf R, Greenberg ER, et al. Age at any full-term pregnancy and breast cancer risk. Am J Epidemiol. 2000;151(7):715–22.

    CAS  PubMed  Google Scholar 

  15. Albrektsen G, Heuch I, Kvale G. The short-term and long-term effect of a pregnancy on breast cancer risk: a prospective study of 802,457 parous Norwegian women. Br J Cancer. 1995;72(2):480–4.

    CAS  PubMed Central  PubMed  Google Scholar 

  16. Leon DA, Carpenter LM, Broeders MJ, Gunnarskog J, Murphy MF. Breast cancer in Swedish women before age 50: evidence of a dual effect of completed pregnancy. Cancer Causes Control. 1995;6(4):283–91.

    CAS  PubMed  Google Scholar 

  17. Janerich DT, Hoff MB. Evidence for a crossover in breast cancer risk factors. Am J Epidemiol. 1982;116(5):737–42.

    CAS  PubMed  Google Scholar 

  18. Lyons TR, Schedin PJ, Borges VF. Pregnancy and breast cancer: when they collide. J Mammary Gland Biol Neoplasia. 2009;14(2):87–98. doi:10.1007/s10911-009-9119-7.

    PubMed Central  PubMed  Google Scholar 

  19. Stensheim H, Moller B, van Dijk T, Fossa SD. Cause-specific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J Clin Oncol. 2009;27(1):45–51. doi:10.1200/JCO.2008.17.4110.

    PubMed  Google Scholar 

  20. Johansson AL, Andersson TM, Hsieh CC, Cnattingius S, Lambe M. Increased mortality in women with breast cancer detected during pregnancy and different periods postpartum. Cancer Epidemiol Biomarkers Prev. 2011;20(9):1865–72. doi:10.1158/1055-9965.EPI-11-0515.

    PubMed  Google Scholar 

  21. Callihan EB, Gao D, Jindal S, Lyons TR, Manthey E, Edgerton S, et al. Postpartum diagnosis demonstrates a high risk for metastasis and merits an expanded definition of pregnancy-associated breast cancer. Breast Cancer Res Treat. 2013;138(2):549–59. doi:10.1007/s10549-013-2437-x.

    PubMed Central  PubMed  Google Scholar 

  22. Bonnier P, Romain S, Dilhuydy JM, Bonichon F, Julien JP, Charpin C, et al. Influence of pregnancy on the outcome of breast cancer: a case–control study. Societe Francaise de senologie et de pathologie mammaire study group. Int J Cancer. 1997;72(5):720–7. doi:10.1002/(SICI)1097-0215(19970904)72:5<720::AID-IJC3>3.0.CO;2-U.

    CAS  PubMed  Google Scholar 

  23. Daling JR, Malone KE, Doody DR, Anderson BO, Porter PL. The relation of reproductive factors to mortality from breast cancer. Cancer Epidemiol Biomarkers Prev. 2002;11(3):235–41.

    PubMed  Google Scholar 

  24. Olson SH, Zauber AG, Tang J, Harlap S. Relation of time since last birth and parity to survival of young women with breast cancer. Epidemiology. 1998;9(6):669–71.

  25. O’Brien J, Schedin P. Macrophages in breast cancer: do involution macrophages account for the poor prognosis of pregnancy-associated breast cancer? J Mammary Gland Biol Neoplasia. 2009;14(2):145–57. doi:10.1007/s10911-009-9118-8.

  26. Jindal S, Gao D, Bell P, Albrektsen G, Edgerton S, Ambrosone C, et al. Postpartum breast involution reveals regression of secretory lobules mediated by tissue-remodeling. Breast Cancer Res. 2014;16(2):R31.

    PubMed Central  PubMed  Google Scholar 

  27. Lund LR, Romer J, Thomasset N, Solberg H, Pyke C, Bissell MJ, et al. Two distinct phases of apoptosis in mammary gland involution: proteinase-independent and -dependent pathways. Development. 1996;122(1):181–93.

    CAS  PubMed Central  PubMed  Google Scholar 

  28. McDaniel SM, Rumer KK, Biroc SL, Metz RP, Singh M, Porter W, et al. Remodeling of the mammary microenvironment after lactation promotes breast tumor cell metastasis. Am J Pathol. 2006;168(2):608–20. doi:10.2353/ajpath.2006.050677.

    CAS  PubMed Central  PubMed  Google Scholar 

  29. Stein T, Morris J, Davies C, Weber-Hall S, Duffy M-A, Heath V, et al. Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3. Breast Cancer Res. 2004;6(2):R75–91.

    CAS  PubMed Central  PubMed  Google Scholar 

  30. Walker NI, Bennett RE, Kerr JF. Cell death by apoptosis during involution of the lactating breast in mice and rats. Am J Anat. 1989;185(1):19–32. doi:10.1002/aja.1001850104.

    CAS  PubMed  Google Scholar 

  31. Li M, Liu X, Robinson G, Bar-Peled U, Wagner KU, Young WS, et al. Mammary-derived signals activate programmed cell death during the first stage of mammary gland involution. Proc Natl Acad Sci U S A. 1997;94(7):3425–30.

    CAS  PubMed Central  PubMed  Google Scholar 

  32. Marti A, Feng Z, Altermatt HJ, Jaggi R. Milk accumulation triggers apoptosis of mammary epithelial cells. Eur J Cell Biol. 1997;73(2):158–65.

    CAS  PubMed  Google Scholar 

  33. Kreuzaler PA, Staniszewska AD, Li W, Omidvar N, Kedjouar B, Turkson J, et al. Stat3 controls lysosomal-mediated cell death in vivo. Nat Cell Biol. 2011;13(3):303–9. doi:10.1038/ncb2171.

    CAS  PubMed  Google Scholar 

  34. Dickson SR, Warburton MJ. Enhanced synthesis of gelatinase and stromelysin by myoepithelial cells during involution of the rat mammary gland. J Histochem Cytochem. 1992;40(5):697–703.

    CAS  PubMed  Google Scholar 

  35. Lefebvre O, Wolf C, Limacher JM, Hutin P, Wendling C, LeMeur M, et al. The breast cancer-associated stromelysin-3 gene is expressed during mouse mammary gland apoptosis. J Cell Biol. 1992;119(4):997–1002.

    CAS  PubMed  Google Scholar 

  36. Strange R, Li F, Saurer S, Burkhardt A, Friis RR. Apoptotic cell death and tissue remodelling during mouse mammary gland involution. Development. 1992;115(1):49–58.

    CAS  PubMed  Google Scholar 

  37. Talhouk RS, Bissell MJ, Werb Z. Coordinated expression of extracellular matrix-degrading proteinases and their inhibitors regulates mammary epithelial function during involution. J Cell Biol. 1992;118(5):1271–82.

    CAS  PubMed  Google Scholar 

  38. Clarkson R, Wayland M, Lee J, Freeman T, Watson C. Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression. Breast Cancer Res. 2004;6(2):R92–109.

    CAS  PubMed Central  PubMed  Google Scholar 

  39. O’Brien J, Lyons T, Monks J, Lucia MS, Wilson RS, Hines L, et al. Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. Am J Pathol. 2010;176(3):1241–55. doi:10.2353/ajpath.2010.090735.

    PubMed Central  PubMed  Google Scholar 

  40. Schedin P, Mitrenga T, McDaniel S, Kaeck M. Mammary ECM composition and function are altered by reproductive state. Mol Carcinog. 2004;41(4):207–20. doi:10.1002/mc.20058.

    CAS  PubMed  Google Scholar 

  41. Schedin P, O’Brien J, Rudolph M, Stein T, Borges V. Microenvironment of the involuting mammary gland mediates mammary cancer progression. J Mammary Gland Biol Neoplasia. 2007;12(1):71–82. doi:10.1007/s10911-007-9039-3.

    PubMed  Google Scholar 

  42. Schedin P, Strange R, Mitrenga T, Wolfe P, Kaeck M. Fibronectin fragments induce MMP activity in mouse mammary epithelial cells: evidence for a role in mammary tissue remodeling. J Cell Sci. 2000;113(Pt 5):795–806.

    CAS  PubMed  Google Scholar 

  43. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860–7. doi:10.1038/nature01322.

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Fornetti J, Jindal S, Middleton KA, Borges VF, Schedin P. Physiological COX-2 expression in breast epithelium associates with COX-2 levels in ductal carcinoma in situ and invasive breast cancer in young women. Am J Pathol. 2014;184(4):1219–29. doi:10.1016/j.ajpath.2013.12.026.

    CAS  PubMed  Google Scholar 

  45. Gupta PB, Proia D, Cingoz O, Weremowicz J, Naber SP, Weinberg RA, et al. Systemic stromal effects of estrogen promote the growth of estrogen receptor–negative cancers. Cancer Res. 2007;67(5):2062–71. doi:10.1158/0008-5472.can-06-3895.

    CAS  PubMed  Google Scholar 

  46. Fornetti J, Martinson H, Borges V, Schedin P. Emerging targets for the prevention of pregnancy-associated breast cancer. Cell Cycle. 2012;11(4):639–40. doi:10.4161/cc.11.4.19358.

    CAS  PubMed Central  PubMed  Google Scholar 

  47. Stein T, Salomonis N, Nuyten DS, van de Vijver MJ, Gusterson BA. A mouse mammary gland involution mRNA signature identifies biological pathways potentially associated with breast cancer metastasis. J Mammary Gland Biol Neoplasia. 2009;14(2):99–116. doi:10.1007/s10911-009-9120-1.

    PubMed  Google Scholar 

  48. Monks J, Smith-Steinhart C, Kruk ER, Fadok VA, Henson PM. Epithelial cells remove apoptotic epithelial cells during post-lactation involution of the mouse mammary gland. Biol Reprod. 2008;78(4):586–94. doi:10.1095/biolreprod.107.065045.

    CAS  PubMed  Google Scholar 

  49. Hughes K, Wickenden JA, Allen JE, Watson CJ. Conditional deletion of Stat3 in mammary epithelium impairs the acute phase response and modulates immune cell numbers during post-lactational regression. J Pathol. 2012;227(1):106–17. doi:10.1002/path.3961.

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Ramirez RA, Lee A, Schedin P, Russell JS, Masso-Welch PA. Alterations in mast cell frequency and relationship to angiogenesis in the rat mammary gland during windows of physiologic tissue remodeling. Dev Dyn. 2012;241(5):890–900. doi:10.1002/dvdy.23778.

    CAS  PubMed Central  PubMed  Google Scholar 

  51. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–86.

    CAS  PubMed  Google Scholar 

  52. Sica A, Schioppa T, Mantovani A, Allavena P. Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer. 2006;42(6):717–27. doi:10.1016/j.ejca.2006.01.003.

    CAS  PubMed  Google Scholar 

  53. Sica A, Larghi P, Mancino A, Rubino L, Porta C, Totaro MG, et al. Macrophage polarization in tumour progression. Semin Cancer Biol. 2008;18(5):349–55.

    CAS  PubMed  Google Scholar 

  54. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(12):958–69.

    CAS  PubMed Central  PubMed  Google Scholar 

  55. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23(11):549–55.

  56. Sohn BH, Moon HB, Kim TY, Kang HS, Bae YS, Lee KK, et al. Interleukin-10 up-regulates tumour-necrosis-factor-alpha-related apoptosis-inducing ligand (TRAIL) gene expression in mammary epithelial cells at the involution stage. Biochem J. 2001;360(Pt 1):31–8.

    CAS  PubMed Central  PubMed  Google Scholar 

  57. Chang SH, Liu CH, Conway R, Han DK, Nithipatikom K, Trifan OC, et al. Role of prostaglandin E2-dependent angiogenic switch in cyclooxygenase 2-induced breast cancer progression. Proc Natl Acad Sci U S A. 2004;101(2):591–6. doi:10.1073/pnas.2535911100.

    CAS  PubMed Central  PubMed  Google Scholar 

  58. Hu M, Peluffo G, Chen H, Gelman R, Schnitt S, Polyak K. Role of COX-2 in epithelial-stromal cell interactions and progression of ductal carcinoma in situ of the breast. Proc Natl Acad Sci U S A. 2009;106((9):3372–7. doi:10.1073/pnas.0813306106.

    Google Scholar 

  59. Karavitis J, Hix LM, Shi YH, Schultz RF, Khazaie K, Zhang M. Regulation of COX2 expression in mouse mammary tumor cells controls bone metastasis and PGE2-induction of regulatory T cell migration. PLoS One. 2012;7(9):e46342. doi:10.1371/journal.pone.0046342.

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Larkins T, Nowell M, Singh S, Sanford G. Inhibition of cyclooxygenase-2 decreases breast cancer cell motility, invasion and matrix metalloproteinase expression. BMC Cancer. 2006;6(1):181.

    PubMed Central  PubMed  Google Scholar 

  61. Liu CH, Chang S-H, Narko K, Trifan OC, Wu M-T, Smith E, et al. Overexpression of cyclooxygenase-2 is sufficient to induce tumorigenesis in transgenic mice. J Biol Chem. 2001;276(21):18563–9. doi:10.1074/jbc.M010787200.

    CAS  PubMed  Google Scholar 

  62. Lyons TR, O’Brien J, Borges VF, Conklin MW, Keely PJ, Eliceiri KW et al. Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2. Nat Med. 2011;17(9):1109–15. doi:http://www.nature.com/nm/journal/v17/n9/abs/nm.2416.html#supplementary-information.

  63. Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67(9):4507–13. doi:10.1158/0008-5472.CAN-06-4174.

    CAS  PubMed  Google Scholar 

  64. Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009;124(11):2621–33. doi:10.1002/ijc.24249.

    CAS  PubMed Central  PubMed  Google Scholar 

  65. Chen EP, Smyth EM. COX-2 and PGE2-dependent immunomodulation in breast cancer. Prostaglandins Other Lipid Mediat. 2011;96(1–4):14–20. doi:10.1016/j.prostaglandins.2011.08.005.

    CAS  PubMed Central  PubMed  Google Scholar 

  66. O’Brien J, Hansen K, Barkan D, Green J, Schedin P. Non-steroidal anti-inflammatory drugs target the pro-tumorigenic extracellular matrix of the postpartum mammary gland. Int J Dev Biol. 2011;55(7–9):745–55. doi:10.1387/ijdb.113379jo.

    PubMed  Google Scholar 

  67. Sandahl M, Hunter DM, Strunk KE, Earp HS, Cook RS. Epithelial cell-directed efferocytosis in the post-partum mammary gland is necessary for tissue homeostasis and future lactation. BMC Dev Biol. 2010;10:122. doi:10.1186/1471-213X-10-122.

    CAS  PubMed Central  PubMed  Google Scholar 

  68. Fadok VA, Bratton DL, Konowal A, Freed OW. Macrophages That Have Ingested Apoptotic Cells In Vitro Inhibit Proinflammatory Cytokine Production Through Autocrine/Paracrine Mechanisms Involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998;101.

  69. Chung EY, Liu J, Homma Y, Zhang Y, Brendolan A, Saggese M, et al. Interleukin-10 expression in macrophages during phagocytosis of apoptotic cells is mediated by homeodomain proteins Pbx1 and prep-1. Immunity. 2007;27(6):952–64. doi:10.1016/j.immuni.2007.11.014.

    CAS  PubMed Central  PubMed  Google Scholar 

  70. Brecht K, Weigert A, Hu J, Popp R, Fisslthaler B, Korff T, et al. Macrophages programmed by apoptotic cells promote angiogenesis via prostaglandin E2. FASEB J. 2011;25(7):2408–17. doi:10.1096/fj.10-179473.

    CAS  PubMed  Google Scholar 

  71. Monks J, Rosner D, Geske FJ, Lehman L, Hanson L, Neville MC, et al. Epithelial cells as phagocytes: apoptotic epithelial cells are engulfed by mammary alveolar epithelial cells and repress inflammatory mediator release. Cell Death Differ. 2005;12(2):107–14. doi:10.1038/sj.cdd.4401517.

    CAS  PubMed  Google Scholar 

  72. Chapman RS, Lourenco P, Tonner E, Flint D, Selbert S, Takeda K, et al. The role of Stat3 in apoptosis and mammary gland involution. Conditional deletion of Stat3. Adv Exp Med Biol. 2000;480:129–38.

    CAS  PubMed  Google Scholar 

  73. Golpon HA, Fadok VA, Taraseviciene-Stewart L, Scerbavicius R, Sauer C, Welte T, et al. Life after corpse engulfment: phagocytosis of apoptotic cells leads to VEGF secretion and cell growth. FASEB J. 2004;18(14):1716–8. doi:10.1096/fj.04-1853fje.

    CAS  PubMed  Google Scholar 

  74. Gregory CD, Pound JD. Cell death in the neighbourhood: direct microenvironmental effects of apoptosis in normal and neoplastic tissues. J Pathol. 2011;223(2):177–94. doi:10.1002/path.2792.

    CAS  PubMed  Google Scholar 

  75. Somersan S. Tethering and tickling: a new role for the phosphatidylserine receptor. J Cell Biol. 2001;155(4):501–4. doi:10.1083/jcb.200110066.

    CAS  PubMed Central  PubMed  Google Scholar 

  76. Lauber K, Bohn E, Kröber SM, Xiao Y-j, Blumenthal SG, Lindemann RK, et al. Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell. 2003;113(6):717–30. doi:10.1016/s0092-8674(03)00422-7.

    CAS  PubMed  Google Scholar 

  77. Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461(7261):282–6. doi:10.1038/nature08296.

    CAS  PubMed Central  PubMed  Google Scholar 

  78. Yamaguchi H, Maruyama T, Urade Y, Nagata S. Immunosuppression via adenosine receptor activation by adenosine monophosphate released from apoptotic cells. eLife. 2014. doi:10.7554/eLife.02172.001 10.7554/eLife.02172.002.

    Google Scholar 

  79. Chen W, Frank ME, Jin W, Wahl SM. TGF-beta Released by Apoptotic T Cells Contributes to an Immunosuppressive Milieu. Immunity. 2001;14.

  80. Li F, Huang Q, Chen J, Peng Y, Roop DR, Bedford JS, et al. Apoptotic cells activate the “phoenix rising” pathway to promote wound healing and tissue regeneration. Sci Signal. 2010;3(110):ra13. doi:10.1126/scisignal.2000634.

    PubMed Central  PubMed  Google Scholar 

  81. Yañez R, Oviedo A, Aldea M, Bueren JA, Lamana ML. Prostaglandin E2 plays a key role in the immunosuppressive properties of adipose and bone marrow tissue-derived mesenchymal stromal cells. Exp Cell Res. 2010;316(19):3109–23. doi:10.1016/j.yexcr.2010.08.008.

    PubMed  Google Scholar 

  82. O’Brien JH, Vanderlinden LA, Schedin PJ, Hansen KC. Rat mammary extracellular matrix composition and response to ibuprofen treatment during postpartum involution by differential GeLC-MS/MS analysis. J Proteome Res. 2012;11(10):4894–905. doi:10.1021/pr3003744.

    PubMed  Google Scholar 

  83. Morwood SR, Nicholson LB. Modulation of the immune response by extracellular matrix proteins. Arch Immunol Ther Exp (Warsz). 2006;54(6):367–74. doi:10.1007/s00005-006-0043-x.

    CAS  Google Scholar 

  84. Sorokin L. The impact of the extracellular matrix on inflammation. Nat Rev Immunol. 2010;0((0)):712–23. doi:10.1038/nri2852.

    Google Scholar 

  85. Laskin DL, Soltys RA, Berg RA, Riley DJ. Activation of alveolar macrophages by native and synthetic collagen-like polypeptides. Am J Respir Cell Mol Biol. 1994;10(1):58–64. doi:10.1165/ajrcmb.10.1.8292381.

    CAS  PubMed  Google Scholar 

  86. Mydel P, Shipley JM, Adair-Kirk TL, Kelley DG, Broekelmann TJ, Mecham RP, et al. Neutrophil elastase cleaves laminin-332 (laminin-5) generating peptides that are chemotactic for neutrophils. J Biol Chem. 2008;283(15):9513–22. doi:10.1074/jbc.M706239200.

    CAS  PubMed Central  PubMed  Google Scholar 

  87. Kaplan G. In vitro differentiation of human monocytes. Monocytes cultured on glass are cytotoxic to tumor cells but monocytes cultured on collagen are not. J Exp Med. 1983;157(6):2061–72.

    CAS  PubMed  Google Scholar 

  88. Hauzenberger D, Olivier P, Gundersen D, Ruegg C. Tenascin-C inhibits beta1 integrin-dependent T lymphocyte adhesion to fibronectin through the binding of its fnIII 1–5 repeats to fibronectin. Eur J Immunol. 1999;29(5):1435–47. doi:10.1002/(SICI)1521-4141(199905)29:05<1435::AID-IMMU1435>3.0.CO;2-N.

    CAS  PubMed  Google Scholar 

  89. Hemesath TJ, Marton LS, Stefansson K. Inhibition of T cell activation by the extracellular matrix protein tenascin. J Immunol. 1994;152(11):5199–207.

    CAS  PubMed  Google Scholar 

  90. Hibino S, Kato K, Kudoh S, Yagita H, Okumura K. Tenascin suppresses CD3-mediated T cell activation. Biochem Biophys Res Commun. 1998;250(1):119–24. doi:10.1006/bbrc.1998.9258.

    CAS  PubMed  Google Scholar 

  91. Puente Navazo MD, Valmori D, Ruegg C. The alternatively spliced domain TnFnIII A1A2 of the extracellular matrix protein tenascin-C suppresses activation-induced T lymphocyte proliferation and cytokine production. J Immunol. 2001;167(11):6431–40.

    CAS  PubMed  Google Scholar 

  92. Ruegg CR, Chiquet-Ehrismann R, Alkan SS. Tenascin, an extracellular matrix protein, exerts immunomodulatory activities. Proc Natl Acad Sci U S A. 1989;86(19):7437–41.

    CAS  PubMed Central  PubMed  Google Scholar 

  93. Maller O, Martinson H, Schedin P. Extracellular matrix composition reveals complex and dynamic stromal-epithelial interactions in the mammary gland. J Mammary Gland Biol Neoplasia. 2010;15(3):301–18. doi:10.1007/s10911-010-9189-6.

    PubMed  Google Scholar 

  94. Korpos E, Wu C, Sorokin L. Multiple roles of the extracellular matrix in inflammation. Curr Pharm Des. 2009;15(12):1349–57.

    CAS  PubMed  Google Scholar 

  95. Vaday GG, Lider O. Extracellular matrix moieties, cytokines, and enzymes: dynamic effects on immune cell behavior and inflammation. J Leukoc Biol. 2000;67(2):149–59.

    CAS  PubMed  Google Scholar 

  96. Lortat-Jacob H, Garrone P, Banchereau J, Grimaud JA. Human interleukin 4 is a glycosaminoglycan-binding protein. Cytokine. 1997;9(2):101–5. doi:10.1006/cyto.1996.0142.

    CAS  PubMed  Google Scholar 

  97. Schedin P, Mitrenga T, Kaeck M. Estrous cycle regulation of mammary epithelial cell proliferation, differentiation, and death in the Sprague–Dawley Rat: a model for investigating the role of estrous cycling in mammary carcinogenesis. J Mammary Gland Biol Neoplasia. 2000;5(2):211–25. doi:10.1023/a:1026447506666.

    CAS  PubMed  Google Scholar 

  98. Hinz B. Formation and function of the myofibroblast during tissue repair. J Investig Dermatol. 2007;127(3):526–37. doi:10.1038/sj.jid.5700613.

    CAS  PubMed  Google Scholar 

  99. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta activation. J Cell Sci. 2003;116(Pt 2):217–24.

    CAS  PubMed  Google Scholar 

  100. Flanders KC, Wakefield LM. Transforming growth factor-(beta)s and mammary gland involution; functional roles and implications for cancer progression. J Mammary Gland Biol Neoplasia. 2009;14(2):131–44. doi:10.1007/s10911-009-9122-z.

    PubMed Central  PubMed  Google Scholar 

  101. Ahmadzadeh M, Rosenberg SA. TGF-beta 1 attenuates the acquisition and expression of effector function by tumor antigen-specific human memory CD8 T cells. J Immunol. 2005;174(9):5215–23.

    CAS  PubMed Central  PubMed  Google Scholar 

  102. Kitamura M. Identification of an inhibitor targeting macrophage production of monocyte chemoattractant protein-1 as TGF-beta 1. J Immunol. 1997;159(3):1404–11.

    CAS  PubMed  Google Scholar 

  103. Chen JJ, Sun Y, Nabel GJ. Regulation of the proinflammatory effects of Fas ligand (CD95L). Science. 1998;282(5394):1714–7.

    CAS  PubMed  Google Scholar 

  104. Ghiringhelli F, Menard C, Terme M, Flament C, Taieb J, Chaput N, et al. CD4 + CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner. J Exp Med. 2005;202(8):1075–85. doi:10.1084/jem.20051511.

    CAS  PubMed Central  PubMed  Google Scholar 

  105. Geissmann F, Revy P, Regnault A, Lepelletier Y, Dy M, Brousse N, et al. TGF-beta 1 prevents the noncognate maturation of human dendritic Langerhans cells. J Immunol. 1999;162(8):4567–75.

    CAS  PubMed  Google Scholar 

  106. Wahl SM, Chen W. Transforming growth factor-beta-induced regulatory T cells referee inflammatory and autoimmune diseases. Arthritis Res Ther. 2005;7(2):62–8. doi:10.1186/ar1504.

    CAS  PubMed Central  PubMed  Google Scholar 

  107. O’Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell. 1997;88(2):277–85.

    PubMed  Google Scholar 

  108. Hamano Y, Zeisberg M, Sugimoto H, Lively JC, Maeshima Y, Yang C, et al. Physiological levels of tumstatin, a fragment of collagen IV alpha3 chain, are generated by MMP-9 proteolysis and suppress angiogenesis via alphaV beta3 integrin. Cancer Cell. 2003;3(6):589–601.

    CAS  PubMed Central  PubMed  Google Scholar 

  109. Adair-Kirk TL, Senior RM. Fragments of extracellular matrix as mediators of inflammation. Int J Biochem Cell Biol. 2008;40(6–7):1101–10. doi:10.1016/j.biocel.2007.12.005.

    CAS  PubMed Central  PubMed  Google Scholar 

  110. Brown EJ. The role of extracellular matrix proteins in the control of phagocytosis. J Leukoc Biol. 1986;39(5):579–91.

    CAS  PubMed  Google Scholar 

  111. Yang KD, Augustine NH, Shaio MF, Bohnsack JF, Hill HR. Effects of fibronectin on actin organization and respiratory burst activity in neutrophils, monocytes, and macrophages. J Cell Physiol. 1994;158(2):347–53. doi:10.1002/jcp.1041580217.

    CAS  PubMed  Google Scholar 

  112. Beezhold DH, Personius C. Fibronectin fragments stimulate tumor necrosis factor secretion by human monocytes. J Leukoc Biol. 1992;51(1):59–64.

    CAS  PubMed  Google Scholar 

  113. Marom B, Rahat MA, Lahat N, Weiss-Cerem L, Kinarty A, Bitterman H. Native and fragmented fibronectin oppositely modulate monocyte secretion of MMP-9. J Leukoc Biol. 2007;81(6):1466–76. doi:10.1189/jlb.0506328.

    CAS  PubMed  Google Scholar 

  114. Adair-Kirk TL, Atkinson JJ, Broekelmann TJ, Doi M, Tryggvason K, Miner JH, et al. A site on laminin alpha 5, AQARSAASKVKVSMKF, induces inflammatory cell production of matrix metalloproteinase-9 and chemotaxis. J Immunol. 2003;171(1):398–406.

    CAS  PubMed  Google Scholar 

  115. Adair-Kirk TL, Atkinson JJ, Kelley DG, Arch RH, Miner JH, Senior RM. A chemotactic peptide from laminin alpha 5 functions as a regulator of inflammatory immune responses via TNF alpha-mediated signaling. J Immunol. 2005;174(3):1621–9.

    CAS  PubMed  Google Scholar 

  116. McCready J, Arendt LM, Glover E, Iyer V, Briendel JL, Lyle SR, et al. Pregnancy-associated breast cancers are driven by differences in adipose stromal cells present during lactation. Breast Cancer Res. 2014;16(1):R2. doi:10.1186/bcr3594.

    PubMed Central  PubMed  Google Scholar 

  117. Kamei N, Tobe K, Suzuki R, Ohsugi M, Watanabe T, Kubota N, et al. Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. J Biol Chem. 2006;281(36):26602–14. doi:10.1074/jbc.M601284200.

    CAS  PubMed  Google Scholar 

  118. Kanda H, Tateya S, Tamori Y, Kotani K. Hiasa K-i, Kitazawa R et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116(6):1494–505.

    CAS  PubMed Central  PubMed  Google Scholar 

  119. Acedo SC, Gambero S, Cunha FG, Lorand-Metze I, Gambero A. Participation of leptin in the determination of the macrophage phenotype: an additional role in adipocyte and macrophage crosstalk. In Vitro Cell Dev Biol Anim. 2013;49(6):473–8. doi:10.1007/s11626-013-9629-x.

    CAS  PubMed  Google Scholar 

  120. Klein-Wieringa IR, Andersen SN, Kwekkeboom JC, Giera M, de Lange-Brokaar BJ, van Osch GJ, et al. Adipocytes modulate the phenotype of human macrophages through secreted lipids. J Immunol. 2013;191(3):1356–63. doi:10.4049/jimmunol.1203074.

    CAS  PubMed  Google Scholar 

  121. Gruen ML, Hao M, Piston DW, Hasty AH. Leptin requires canonical migratory signaling pathways for induction of monocyte and macrophage chemotaxis. Am J Physiol Cell Physiol. 2007;293(5):C1481–8. doi:10.1152/ajpcell.00062.2007.

    CAS  PubMed  Google Scholar 

  122. Lin Y, Li Q. Expression and function of leptin and its receptor in mouse mammary gland. Sci China C Life Sci. 2007;50(5):669–75. doi:10.1007/s11427-007-0077-2.

    CAS  PubMed  Google Scholar 

  123. Routley CE, Ashcroft GS. Effect of estrogen and progesterone on macrophage activation during wound healing. Wound Repair Regen. 2009;17(1):42–50. doi:10.1111/j.1524-475X.2008.00440.x.

    PubMed  Google Scholar 

  124. Calippe B, Douin-Echinard V, Delpy L, Laffargue M, Lelu K, Krust A, et al. 17Beta-estradiol promotes TLR4-triggered proinflammatory mediator production through direct estrogen receptor alpha signaling in macrophages in vivo. J Immunol. 2010;185(2):1169–76. doi:10.4049/jimmunol.0902383.

    CAS  PubMed  Google Scholar 

  125. Jensen F, Woudwyk M, Teles A, Woidacki K, Taran F, Costa S, et al. Estradiol and progesterone regulate the migration of mast cells from the periphery to the uterus and induce their maturation and degranulation. PLoS One. 2010;5(12):e14409. doi:10.1371/journal.pone.0014409.

    CAS  PubMed Central  PubMed  Google Scholar 

  126. Zaitsu M, Narita S, Lambert KC, Grady JJ, Estes DM, Curran EM, et al. Estradiol activates mast cells via a non-genomic estrogen receptor-alpha and calcium influx. Mol Immunol. 2007;44(8):1977–85. doi:10.1016/j.molimm.2006.09.030.

    CAS  PubMed Central  PubMed  Google Scholar 

  127. Lelu K, Laffont S, Delpy L, Paulet PE, Perinat T, Tschanz SA, et al. Estrogen receptor alpha signaling in T lymphocytes is required for estradiol-mediated inhibition of Th1 and Th17 cell differentiation and protection against experimental autoimmune encephalomyelitis. J Immunol. 2011;187(5):2386–93. doi:10.4049/jimmunol.1101578.

    CAS  PubMed  Google Scholar 

  128. Pernis AB. Estrogen and CD4+ T cells. Curr Opin Rheumatol. 2007;19(5):414–20. doi:10.1097/BOR.0b013e328277ef2a.

    CAS  PubMed  Google Scholar 

  129. Tai P, Wang J, Jin H, Song X, Yan J, Kang Y, et al. Induction of regulatory T cells by physiological level estrogen. J Cell Physiol. 2008;214(2):456–64. doi:10.1002/jcp.21221.

    CAS  PubMed  Google Scholar 

  130. Fu Y, Li L, Liu X, Ma C, Zhang J, Jiao Y, et al. Estrogen promotes B cell activation in vitro through down-regulating CD80 molecule expression. Gynecol Endocrinol. 2011;27(8):593–6. doi:10.3109/09513590.2010.507281.

    CAS  PubMed  Google Scholar 

  131. Grimaldi CM, Cleary J, Dagtas AS, Moussai D, Diamond B. Estrogen alters thresholds for B cell apoptosis and activation. J Clin Invest. 2002;109(12):1625–33. doi:10.1172/JCI14873.

    CAS  PubMed Central  PubMed  Google Scholar 

  132. Grimaldi CM, Jeganathan V, Diamond B. Hormonal regulation of B cell development: 17 beta-estradiol impairs negative selection of high-affinity DNA-reactive B cells at more than one developmental checkpoint. J Immunol. 2006;176(5):2703–10.

    CAS  PubMed  Google Scholar 

  133. Tsutsui S, Yasuda K, Suzuki K, Tahara K, Higashi H, Era S. Macrophage infiltration and its prognostic implications in breast cancer: the relationship with VEGF expression and microvessel density. Oncol Rep. 2005;14(2):425–31.

    CAS  PubMed  Google Scholar 

  134. Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL. Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res. 1996;56(20):4625–9.

    CAS  PubMed  Google Scholar 

  135. Goede V, Brogelli L, Ziche M, Augustin HG. Induction of inflammatory angiogenesis by monocyte chemoattractant protein-1. Int J Cancer. 1999;82(5):765–70. doi:10.1002/(sici)1097-0215(19990827)82:5<765::aid-ijc23>3.0.co;2-f.

    CAS  PubMed  Google Scholar 

  136. Lee AH, Happerfield LC, Bobrow LG, Millis RR. Angiogenesis and inflammation in invasive carcinoma of the breast. J Clin Pathol. 1997;50(8):669–73. doi:10.1136/jcp.50.8.669.

    CAS  PubMed Central  PubMed  Google Scholar 

  137. Volodko N, Reiner A, Rudas M, Jakesz R. Tumour-associated macrophages in breast cancer and their prognostic correlations. Breast. 1998;7(2):99–105. doi:10.1016/S0960-9776(98)90065-0.

    Google Scholar 

  138. McDermott RS, Deneux L, Mosseri V, Vedrenne J, Clough K, Fourquet A, et al. Circulating macrophage colony stimulating factor as a marker of tumour progression. Eur Cytokine Netw. 2002;13(1):121–7.

    CAS  PubMed  Google Scholar 

  139. Ueno T, Toi M, Saji H, Muta M, Bando H, Kuroi K, et al. Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer. Clin Cancer Res. 2000;6(8):3282–9.

    CAS  PubMed  Google Scholar 

  140. Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. 2001. p. 727–40.

  141. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, et al. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 2009;16(2):91–102. doi:10.1016/j.ccr.2009.06.018.

    CAS  PubMed Central  PubMed  Google Scholar 

  142. DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011;1(1):54–67. doi:10.1158/2159-8274.cd-10-0028.

    CAS  PubMed Central  PubMed  Google Scholar 

  143. Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2009;58(1):49–59. doi:10.1007/s00262-008-0523-4.

    CAS  PubMed Central  PubMed  Google Scholar 

  144. Montero AJ, Diaz-Montero CM, Deutsch YE, Hurley J, Koniaris LG, Rumboldt T, 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(1):215–23. doi:10.1007/s10549-011-1889-0.

    CAS  PubMed Central  PubMed  Google Scholar 

  145. Ohara M, Yamaguchi Y, Matsuura K, Murakami S, Arihiro K, Okada M. Possible involvement of regulatory T cells in tumor onset and progression in primary breast cancer. Cancer Immunol Immunother. 2009;58(3):441–7.

    CAS  PubMed  Google Scholar 

  146. Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, et al. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol. 2006;24(34):5373–80.

    PubMed  Google Scholar 

  147. Yan M, Jene N, Byrne D, Millar EK, O’Toole SA, McNeil CM, et al. Recruitment of regulatory T cells is correlated with hypoxia-induced CXCR4 expression, and is associated with poor prognosis in basal-like breast cancers. Breast Cancer Res. 2011;13(2):R47. doi:10.1186/bcr2869.

    CAS  PubMed Central  PubMed  Google Scholar 

  148. Rodriguez PC, Zea AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC. Regulation of T cell receptor CD3zeta chain expression by L-arginine. J Biol Chem. 2002;277(24):21123–9. doi:10.1074/jbc.M110675200.

    CAS  PubMed  Google Scholar 

  149. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70(19):7465–75. doi:10.1158/0008-5472.CAN-10-1439.

    CAS  PubMed Central  PubMed  Google Scholar 

  150. Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science. 2013;339(6117):286–91. doi:10.1126/science.1232227.

    CAS  PubMed Central  PubMed  Google Scholar 

  151. Fend L, Accart N, Kintz J, Cochin S, Reymann C, Le Pogam F, et al. Therapeutic effects of anti-CD115 monoclonal antibody in mouse cancer models through dual inhibition of tumor-associated macrophages and osteoclasts. PLoS One. 2013;8(9):e73310. doi:10.1371/journal.pone.0073310.

    CAS  PubMed Central  PubMed  Google Scholar 

  152. Strachan DC, Ruffell B, Oei Y, Bissell MJ, Coussens LM, Pryer N, et al. CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8 T cells. Oncolmmunology. 2013;2(12):e26968. doi:10.4161/onci.26968.

    Google Scholar 

  153. Fridlender ZG, Buchlis G, Kapoor V, Cheng G, Sun J, Singhal S, et al. CCL2 blockade augments cancer immunotherapy. Cancer Res. 2010;70(1):109–18. doi:10.1158/0008-5472.can-09-2326.

    CAS  PubMed Central  PubMed  Google Scholar 

  154. Eubank TD, Roberts RD, Khan M, Curry JM, Nuovo GJ, Kuppusamy P, et al. Granulocyte macrophage colony-stimulating factor inhibits breast cancer growth and metastasis by invoking an anti-angiogenic program in tumor-educated macrophages. Cancer Res. 2009;69(5):2133–40. doi:10.1158/0008.

    CAS  PubMed Central  PubMed  Google Scholar 

  155. Zhang X, Tian W, Cai X, Wang X, Dang W, Tang H, et al. Hydrazinocurcumin Encapsuled nanoparticles “re-educate” tumor-associated macrophages and exhibit anti-tumor effects on breast cancer following STAT3 suppression. PLoS One. 2013;8(6):e65896. doi:10.1371/journal.pone.0065896.

    CAS  PubMed Central  PubMed  Google Scholar 

  156. Faure E, Heisterkamp N, Groffen J, Kaartinen V. Differential expression of TGF-beta isoforms during postlactational mammary gland involution. Cell Tissue Res. 2000;300(1):89–95.

    CAS  PubMed  Google Scholar 

  157. Nguyen AV, Pollard JW. Transforming growth factor beta3 induces cell death during the first stage of mammary gland involution. Development. 2000;127(14):3107–18.

    CAS  PubMed  Google Scholar 

  158. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64. doi:10.1038/nrc3239.

    CAS  PubMed  Google Scholar 

  159. Aliperti LA, Predina JD, Vachani A, Singhal S. Local and systemic recurrence is the Achilles heel of cancer surgery. Ann Surg Oncol. 2011;18(3):603–7. doi:10.1245/s10434-010-1442-0.

    PubMed  Google Scholar 

  160. Emens LA. Breast cancer immunobiology driving immunotherapy: vaccines and immune checkpoint blockade. Expert Rev Anticancer Ther. 2012;12(12):1597–611. doi:10.1586/era.12.147.

    CAS  PubMed Central  PubMed  Google Scholar 

  161. Silanikove N. Natural and abrupt involution of the mammary gland affects differently the metabolic and health consequences of weaning. Life Sci. 2014;102(1):10–5. doi:10.1016/j.lfs.2014.02.034.

    CAS  PubMed  Google Scholar 

  162. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820–7. doi:10.1038/nature04186.

    CAS  PubMed Central  PubMed  Google Scholar 

  163. Faupel-Badger JM, Arcaro KF, Balkam JJ, Eliassen AH, Hassiotou F, Lebrilla CB, et al. Postpartum remodeling, lactation, and breast cancer risk: summary of a national cancer institute-sponsored workshop. J Natl Cancer Inst. 2013;105(3):166–74. doi:10.1093/jnci/djs505.

    PubMed Central  PubMed  Google Scholar 

  164. Bernier MO, Plu-Bureau G, Bossard N, Ayzac L, Thalabard JC. Breastfeeding and risk of breast cancer: a metaanalysis of published studies. Hum Reprod Update. 2000;6(4):374–86.

    CAS  PubMed  Google Scholar 

  165. Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet. 2002;360(9328):187–95. doi:10.1016/s0140-6736(02)09454-0.

  166. Michels KB, Willett WC, Rosner BA, Manson JE, Hunter DJ, Colditz GA, et al. Prospective assessment of breastfeeding and breast cancer incidence among 89,887 women. Lancet. 1996;347(8999):431–6.

    CAS  PubMed  Google Scholar 

  167. Jordan I, Hebestreit A, Swai B, Krawinkel MB. Breast cancer risk among women with long-standing lactation and reproductive parameters at low risk level: a case–control study in Northern Tanzania. Breast Cancer Res Treat. 2013;142(1):133–41. doi:10.1007/s10549-010-1255-7.

    PubMed  Google Scholar 

  168. Millikan RC, Newman B, Tse CK, Moorman PG, Conway K, Dressler LG, et al. Epidemiology of basal-like breast cancer. Breast Cancer Res Treat. 2008;109(1):123–39. doi:10.1007/s10549-007-9632-6.

    PubMed Central  PubMed  Google Scholar 

  169. Palmer JR, Boggs DA, Wise LA, Ambrosone CB, Adams-Campbell LL, Rosenberg L. Parity and lactation in relation to estrogen receptor negative breast cancer in African American women. Cancer Epidemiol Biomarkers Prev. 2011;20(9):1883–91. doi:10.1158/1055-9965.EPI-11-0465.

    CAS  PubMed Central  PubMed  Google Scholar 

  170. Li CI, Beaber EF, Tang MT, Porter PL, Daling JR, Malone KE. Reproductive factors and risk of estrogen receptor positive, triple-negative, and HER2-neu overexpressing breast cancer among women 20–44 years of age. Breast Cancer Res Treat. 2013;137(2):579–87. doi:10.1007/s10549-012-2365-1.

    CAS  PubMed Central  PubMed  Google Scholar 

  171. Stuebe AM, Willett WC, Xue F, Michels KB. Lactation and incidence of premenopausal breast cancer: a longitudinal study. Arch Intern Med. 2009;169(15):1364–71. doi:10.1001/archinternmed.2009.231.

    PubMed Central  PubMed  Google Scholar 

  172. Ishida T, Yokoe T, Kasumi F, Sakamoto G, Makita M, Tominaga T, et al. Clinicopathologic characteristics and prognosis of breast cancer patients associated with pregnancy and lactation: analysis of case–control study in Japan. Jpn J Cancer Res. 1992;83(11):1143–9.

    CAS  PubMed  Google Scholar 

  173. Martinez ME, Wertheim BC, Natarajan L, Schwab R, Bondy M, Daneri-Navarro A, et al. Reproductive factors, heterogeneity, and breast tumor subtypes in women of Mexican descent. Cancer Epidemiol Biomark Prev. 2013;22(10):1853–61. doi:10.1158/1055-9965.epi-13-0560.

    Google Scholar 

  174. Gustbee E, Anesten C, Markkula A, Simonsson M, Rose C, Ingvar C, et al. Excessive milk production during breast-feeding prior to breast cancer diagnosis is associated with increased risk for early events. Springerplus. 2013;2(1):298. doi:10.1186/2193-1801-2-298.

    PubMed Central  PubMed  Google Scholar 

  175. Sotgia F, Casimiro MC, Bonuccelli G, Liu M, Whitaker-Menezes D, Er O, et al. Loss of caveolin-3 induces a lactogenic microenvironment that is protective against mammary tumor formation. Am J pathol. 2009;174(2):613–29. doi:10.2353/ajpath.2009.080653.

    CAS  PubMed Central  PubMed  Google Scholar 

  176. Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER, et al. A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res. 2004;64(19):7022–9. doi:10.1158/0008-5472.CAN-04-1449.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Pepper Schedin.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fornetti, J., Martinson, H.A., Betts, C.B. et al. Mammary Gland Involution as an Immunotherapeutic Target for Postpartum Breast Cancer. J Mammary Gland Biol Neoplasia 19, 213–228 (2014). https://doi.org/10.1007/s10911-014-9322-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10911-014-9322-z

Keywords

Navigation