Skip to main content

Advertisement

Log in

Role of the CXCR4/CXCL12 signaling axis in breast cancer metastasis to the brain

  • Review
  • Published:
Clinical & Experimental Metastasis Aims and scope Submit manuscript

Abstract

Breast cancer is the most common malignancy and second leading cause of cancer death in women. Ninety percent of mortality in breast cancer is often associated with metastatic progression or relapse in patients. Critical stages in the development of aggressive breast cancer include the growth of primary tumors and their ability to spread to foreign organs and form metastases, as well as the establishment of an independent blood supply within the new tumors. Hence, it is imperative to characterize the key molecules that regulate the metastasis of human breast cancer cells. The expression of CXCR4/CXCL12 in breast tumors has been correlated with a poor prognosis, increased metastasis, resistance to conventional therapeutic agents and a poor outcome in the pathogenesis of breast cancer. However, effective anti-CXCR4 therapy remains a challenge. Here, we will review the putative involvement of the CXCR4/CXCL12 signaling axis in breast cancer metastasis to the brain. Characterization of signaling events important for breast cancer cell growth and their metastasis to the brain should provide insights into breast cancer therapies and improved, successful treatments for breast cancer.

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

Similar content being viewed by others

Abbreviations

Akt:

Protein kinase B

BBB:

Blood brain barrier

Ca2+ :

Calcium ion

CHK:

Csk homologous kinase

CNS:

Central nervous system

Cox2:

Cyclooxygenase-2 (PTGS2)

CXCL12:

Chemokine (C-X-C motif) ligand 12

CXCR4:

CXC chemokine receptor 4

EAE:

Experimental autoimmune encephalomyelitis

EGFR:

Epidermal growth factor receptor

ErbB2:

Epidermal growth factor receptor 2

ERK1/2:

Extracellular signal-regulated kinases 1 and 2

FAK:

Focal adhesion kinase

FKHRL1:

Forkhead transcription factor like 1

HBMEC:

Human brain microvascular endothelial cells

HER2/Neu:

Epidermal growth factor receptor 2

IL:

Interleukins

KDR/Flt-1:

Kinase insert domain receptor

MAPK:

Mitogen-activated protein (MAP) kinases

MMP:

Matrix metalloproteinases

NF-κB:

Nuclear factor-kappa B

NGF:

Nerve growth factor

NP-1:

Neuropilin-1

PI-3K:

Phosphoinositide 3-kinase

PLC:

Phosphoinositide phospholipase C

RNAi:

RNA interference

SDF-1:

Stromal cell-derived factor-1

TGF-β:

Transforming growth factor beta

VEGF:

Vascular endothelial growth factor

VEGFR1:

Vascular endothelial growth factor receptor 1

YY1B:

Yin Yang 1b transcription factor

References

  1. Internet publication www.womancando.org

  2. Karnoub AE, Weinberg RA (2006–2007) Chemokine networks and breast cancer metastasis. Breast Dis 26:75–85

    CAS  Google Scholar 

  3. Minn AJ, Gupta GP, Padua D, Bos P, Nguyen DX, Nuyten D et al (2007) Lung metastasis genes couple breast tumor size and metastatic spread. Proc Natl Acad Sci USA 104:6740–6745. doi:10.1073/pnas.0701138104

    Article  CAS  PubMed  Google Scholar 

  4. Dupont VN, Gentien D, Oberkampf M, De Rycke Y, Blin N (2007) A gene expression signature associated with metastatic cells in effusions of breast carcinoma patients. Int J Cancer 121:1036–1046. doi:10.1002/ijc.22775

    Article  CAS  PubMed  Google Scholar 

  5. Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY, Nadal C et al (2007) Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature 446:765–770. doi:10.1038/nature05760

    Article  CAS  PubMed  Google Scholar 

  6. Gumireddy K, Sun F, Klein-Szanto AJ, Gibbins JM, Gimotty PA, Saunders AJ et al (2007) In vivo selection for metastasis promoting genes in the mouse. Proc Natl Acad Sci USA 104:6696–6701. doi:10.1073/pnas.0701145104

    Article  CAS  PubMed  Google Scholar 

  7. Chang EL, Lo S (2003) Diagnosis and management of central nervous system metastases from breast cancer. Oncologist 8:398–410. doi:10.1634/theoncologist.8-5-398

    Article  PubMed  Google Scholar 

  8. Boogerd W (1996) Central nervous system metastasis in breast cancer. Radiother Oncol 40:5–22. doi:10.1016/0167-8140(96)01766-5

    Article  CAS  PubMed  Google Scholar 

  9. Boogerd W, Vos VW, Hart AA, Baris G (1993) Brain metastases in breast cancer; natural history, prognostic factors and outcome. J Neurooncol 15:165–174. doi:10.1007/BF01053937

    Article  CAS  PubMed  Google Scholar 

  10. Stewart JF, King RJ, Sexton SA, Millis RR, Rubens RD, Hayward JL (1981) Oestrogen receptors, sites of metastatic disease and survival in recurrent breast cancer. Eur J Cancer 17:449–453

    CAS  PubMed  Google Scholar 

  11. Sparrow GE, Rubens RD (1981) Brain metastases from breast cancer: clinical course, prognosis and influence of treatment. Clin Oncol 7:291–301

    CAS  PubMed  Google Scholar 

  12. Nicolson GL, Menter DG, Herrmann J, Cavanaugh P, Jia L, Hamada J et al (1994) Tumor metastasis to brain: role of endothelial cells, neurotrophins, and paracrine growth factors. Crit Rev Oncog 5:451–471

    CAS  PubMed  Google Scholar 

  13. Wiseman BS, Werb Z (2002) Stromal effects on mammary gland development and breast cancer. Science 296:1046–1049. doi:10.1126/science.1067431

    Article  CAS  PubMed  Google Scholar 

  14. Hesselgesser J, Halks-Miller M, DelVecchio V, Peiper SC, Hoxie J, Kolson DL et al (1997) CD4-independent association between HIV-1 gp120 and CXCR4: functional chemokine receptors are expressed in human neurons. Curr Biol 7:112–121. doi:10.1016/S0960-9822(06)00055-8

    Article  CAS  PubMed  Google Scholar 

  15. Ohtani Y, Minami M, Kawaguchi N, Nishiyori A, Yamamoto J, Takami S et al (1998) Expression of stromal cell-derived factor-1 and CXCR4 chemokine receptor mRNAs in cultured rat glial and neuronal cells. Neurosci Lett 249:163–166. doi:10.1016/S0304-3940(98)00425-X

    Article  CAS  PubMed  Google Scholar 

  16. Tanabe S, Heesen M, Yoshizawa I, Berman MA, Luo Y, Bleul CC et al (1997) Functional expression of the CXC-chemokine receptor-4/fusin on mouse microglial cells and astrocytes. J Immunol (Baltimore, MD.: 1950) 159:905–911

    CAS  Google Scholar 

  17. Volin MV, Joseph L, Shockley MS, Davies PF (1998) Chemokine receptor CXCR4 expression in endothelium. Biochem Biophys Res Commun 242:46–53. doi:10.1006/bbrc.1997.7890

    Article  CAS  PubMed  Google Scholar 

  18. Feil C, Augustin HG (1998) Endothelial cells differentially express functional CXC-chemokine receptor-4 (CXCR-4/fusin) under the control of autocrine activity and exogenous cytokines. Biochem Biophys Res Commun 247:38–45. doi:10.1006/bbrc.1998.8499

    Article  CAS  PubMed  Google Scholar 

  19. Dollé L, El Yazidi-Belkoura I, Adriaenssens E, Nurcombe V, Hondermarck H (2003) Nerve growth factor overexpression and autocrine loop in breast cancer cells. Oncogene 22:5592–5601. doi:10.1038/sj.onc.1206805

    Article  PubMed  Google Scholar 

  20. Dollé L, Adriaenssens E, El Yazidi-Belkoura I, Le Bourhis X, Nurcombe V, Hondermarck H (2004) Nerve growth factor receptors and signaling in breast cancer. Curr Cancer Drug Targets 4:463–470. doi:10.2174/1568009043332853

    Article  PubMed  Google Scholar 

  21. Adriaenssens E, Vanhecke E, Saule P, Mougel A, Page A, Romon R et al (2008) Nerve growth factor is a potential therapeutic target in breast cancer. Cancer Res 68:346–351. doi:10.1158/0008-5472.CAN-07-1183

    Article  CAS  PubMed  Google Scholar 

  22. Prince JM, Klinowska TC, Marshman E, Lowe ET, Mayer U, Miner J et al (2002) Cell-matrix interactions during development and apoptosis of the mouse mammary gland in vivo. Dev Dyn 223:497–516. doi:10.1002/dvdy.10070

    Article  CAS  PubMed  Google Scholar 

  23. El Yazidi-Belkoura I, Adriaenssens E, Dollé L, Descamps S, Hondermarck H (2003) Tumor necrosis factor receptor-associated death domain protein is involved in the neurotrophin receptor-mediated antiapoptotic activity of nerve growth factor in breast cancer cells. J Biol Chem 278:16952–16956. doi:10.1074/jbc.M300631200

    Article  CAS  PubMed  Google Scholar 

  24. Dollé L, Oliveira MJ, Bruyneel E, Hondermarck H, Bracke M (2005) Nerve growth factor mediates its pro-invasive effect in parallel with the release of a soluble E-cadherin fragment from breast cancer MCF-7/AZ cells. J Dairy Res 72:20–26. doi:10.1017/S0022029905001160

    Article  PubMed  Google Scholar 

  25. Dollé JP, Rezvan A, Allen FD, Lazarovici P, Lelkes PI (2005) Nerve growth factor-induced migration of endothelial cells. J Pharmacol Exp Ther 315:1220–1227. doi:10.1124/jpet.105.093252

    Article  PubMed  Google Scholar 

  26. Lai R, Dang C, Malkin M, Abrey L (2004) The risk of central nervous system metastases after trastuzumab therapy in patients with breast carcinoma. Cancer 101:810–816. doi:10.1002/cncr.20418

    Article  CAS  PubMed  Google Scholar 

  27. Weil RJ, Palmieri DC, Bronder JL, Stark AM, Steeg PS (2005) Breast cancer metastasis to the central nervous system. Am J Pathol 167:913–920

    CAS  PubMed  Google Scholar 

  28. Bendell JC, Domchek SM, Burstein HJ, Harris L, Younger J, Kuter I et al (2003) Central nervous system metastases in women who receive trastuzumab-based therapy for metastatic breast carcinoma. Cancer 97:2972–2977. doi:10.1002/cncr.11436

    Article  PubMed  Google Scholar 

  29. Palmieri D, Bronder JL, Herring JM, Yoneda T, Weil RJ, Stark AM et al (2007) Her-2 overexpression increases the metastatic outgrowth of breast cancer cells in the brain. Cancer Res 67:4190–4198. doi:10.1158/0008-5472.CAN-06-3316

    Article  CAS  PubMed  Google Scholar 

  30. Pestalozzi BC, Zahrieh D, Price KN, Holmberg SB, Lindtner J, Collins J, International Breast Cancer Study Group (IBCSG) et al (2006) Identifying breast cancer patients at risk for Central Nervous System (CNS) metastases in trials of the International Breast Cancer Study Group (IBCSG). Ann Oncol 17:935–944. doi:10.1093/annonc/mdl064

    Article  CAS  PubMed  Google Scholar 

  31. Cheng X, Hung MC (2007) Breast cancer brain metastases. Cancer Metastasis Rev 26:635–643. doi:10.1007/s10555-007-9083-x

    Article  PubMed  Google Scholar 

  32. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A et al (2001) Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 344:783–792. doi:10.1056/NEJM200103153441101

    Article  CAS  PubMed  Google Scholar 

  33. Clayton AJ, Danson S, Jolly S, Ryder WD, Burt PA, Stewart AL et al (2004) Incidence of cerebral metastases in patients treated with trastuzumab for metastatic breast cancer. Br J Cancer 91:639–643

    CAS  PubMed  Google Scholar 

  34. Miller KD, Weathers T, Haney LG, Timmerman R, Dickler M, Shen J et al (2003) Occult central nervous system involvement in patients with metastatic breast cancer: prevalence, predictive factors and impact on overall survival. Ann Oncol 14:1072–1077. doi:10.1093/annonc/mdg300

    Article  CAS  PubMed  Google Scholar 

  35. Fuchs IB, Loebbecke M, Buhler H, Stoltenburg-Didinger G, Heine B, Lichtenegger W et al (2002) HER2 in brain metastases: issues of concordance, survival, and treatment. J Clin Oncol 20:4130–4133. doi:10.1200/JCO.2002.04.016

    Article  PubMed  Google Scholar 

  36. Grossi PM, Ochiai H, Archer GE, McLendon RE, Zalutsky MR, Friedman AH et al (2003) Efficacy of intracerebral microinfusion of trastuzumab in an athymic rat model of intracerebral metastatic breast cancer. Clin Cancer Res 9:5514–5520

    CAS  PubMed  Google Scholar 

  37. Montemurro F, Valabrega G, Aglietta M (2007) Lapatinib: a dual inhibitor of EGFR and HER2 tyrosine kinase activity. Expert Opin Biol Ther 7:257–268. doi:10.1517/14712598.7.2.257

    Article  CAS  PubMed  Google Scholar 

  38. Dewji MR (2004) Early phase I data on an irreversible pan-erb inhibitor: CI-1033. What did we learn? J Chemother (Florence, Italy) 16(Suppl 4):44–48

    CAS  Google Scholar 

  39. Lin NU, Bellon JR, Winer EP (2004) CNS metastases in breast cancer. J Clin Oncol 22:3608–3617. doi:10.1200/JCO.2004.01.175

    Article  PubMed  Google Scholar 

  40. Olesen SP (1986) Rapid increase in blood-brain barrier permeability during severe hypoxia and metabolic inhibition. Brain Res 368:24–29. doi:10.1016/0006-8993(86)91038-3

    Article  CAS  PubMed  Google Scholar 

  41. Tanno H, Nockels RP, Pitts LH, Noble LJ (1992) Breakdown of the blood-brain barrier after fluid percussion brain injury in the rat: part 2: effect of hypoxia on permeability to plasma proteins. J Neurotrauma 9:335–347

    Article  CAS  PubMed  Google Scholar 

  42. Ferrara N, Davis-Smyth T (1997) The biology of vascular endothelial growth factor. Endocr Rev 18:4–25. doi:10.1210/er.18.1.4

    Article  CAS  PubMed  Google Scholar 

  43. Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z (1999) Vascular endothelial growth factor (VEGF) and its receptors. FASEB J 13:9–22

    CAS  PubMed  Google Scholar 

  44. Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, Ferrara N (1993) Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362:841–844

    Article  CAS  PubMed  Google Scholar 

  45. Brekken RA, Overholser JP, Stastny VA, Waltenberger J, Minna JD, Thorpe PE (2000) Selective inhibition of vascular endothelial growth factor (VEGF) receptor 2 (KDR/Flk-1) activity by a monoclonal anti-VEGF antibody blocks tumor growth in mice. Cancer Res 60:5117–5124

    CAS  PubMed  Google Scholar 

  46. Senger DR, Perruzzi CA, Feder J, Dvorak HF (1986) A highly conserved vascular permeability factor secreted by a variety of human and rodent tumor cell lines. Cancer Res 46:5629–5632

    CAS  PubMed  Google Scholar 

  47. Brown LF, Berse B, Jackman RW, Tognazzi K, Guidi AJ, Dvorak HF et al (1995) Expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in breast cancer. Hum Pathol 26:86–91. doi:10.1016/0046-8177(95)90119-1

    Article  CAS  PubMed  Google Scholar 

  48. Berkman RA, Merrill MJ, Reinhold WC, Monacci WT, Saxena A, Clark WC et al (1993) Expression of the vascular permeability factor/vascular endothelial growth factor gene in central nervous system neoplasms. J Clin Invest 91:153–159. doi:10.1172/JCI116165

    Article  CAS  PubMed  Google Scholar 

  49. Lee TH, Avraham HK, Jiang S, Avraham S (2003) Vascular endothelial growth factor modulates the transendothelial migration of MDA-MB-231 breast cancer cells through regulation of brain microvascular endothelial cell permeability. J Biol Chem 278:5277–5284. doi:10.1074/jbc.M210063200

    Article  CAS  PubMed  Google Scholar 

  50. Kim LS, Huang S, Lu W, Lev DC, Price JE (2004) Vascular endothelial growth factor expression promotes the growth of breast cancer brain metastases in nude mice. Clin Exp Metastasis 21:107–118. doi:10.1023/B:CLIN.0000024761.00373.55

    Article  CAS  PubMed  Google Scholar 

  51. Stockhammer G, Poewe W, Burgstaller S, Deisenhammer F, Muigg A, Kiechl S et al (2000) Vascular endothelial growth factor in CSF: a biological marker for carcinomatous meningitis. Neurology 54:1670–1676

    CAS  PubMed  Google Scholar 

  52. Rollins BJ (1997) Chemokines. Blood 90:909–928

    CAS  PubMed  Google Scholar 

  53. Premack BA, Schall TJ (1996) Chemokine receptors: gateways to inflammation and infection. Nat Med 2:1174–1178. doi:10.1038/nm1196-1174

    Article  CAS  PubMed  Google Scholar 

  54. Bokoch GM (1995) Chemoattractant signaling and leukocyte activation. Blood 86:1649–1660

    CAS  PubMed  Google Scholar 

  55. Gerard C, Gerard NP (1994) The pro-inflammatory seven-transmembrane segment receptors of the leukocyte. Curr Opin Immunol 6:140–145. doi:10.1016/0952-7915(94)90045-0

    Article  CAS  PubMed  Google Scholar 

  56. Taub DD, Oppenheim JJ (1994) Chemokines, inflammation and the immune system. Ther Immunol 1:229–246

    CAS  PubMed  Google Scholar 

  57. Luster AD (1998) Chemokines-chemotactic cytokines that mediate inflammation. N Engl J Med 338:436–445. doi:10.1056/NEJM199802123380706

    Article  CAS  PubMed  Google Scholar 

  58. Loetscher P, Moser B, Baggiolini M (2000) Chemokines and their receptors in lymphocyte traffic and HIV infection. Adv Immunol 74:127–180. doi:10.1016/S0065-2776(08)60910-4

    Article  CAS  PubMed  Google Scholar 

  59. Miller MD, Krangel MS (1992) Biology and biochemistry of the chemokines: a family of chemotactic and inflammatory cytokines. Crit Rev Immunol 12:17–46

    CAS  PubMed  Google Scholar 

  60. Appleton I (1994) Wound repair: the role of cytokines and vasoactive mediators. J R Soc Med 87:500–502

    CAS  PubMed  Google Scholar 

  61. Gupta SK, Lysko PG, Pillarisetti K, Ohlstein E, Stadel JM (1998) Chemokine receptors in human endothelial cells. Functional expression of CXCR4 and its transcriptional regulation by inflammatory cytokines. J Biol Chem 273:4282–4287. doi:10.1074/jbc.273.7.4282

    Article  CAS  PubMed  Google Scholar 

  62. Zlotnik A, Yoshie O (2000) Chemokines: a new classification system and their role in immunity. Immunity 12:121–127. doi:10.1016/S1074-7613(00)80165-X

    Article  CAS  PubMed  Google Scholar 

  63. Lau EK, Allen S, Hsu AR, Handel TM (2004) Chemokine-receptor interactions: GPCRs, glycosaminoglycans and viral chemokine binding proteins. Adv Protein Chem 68:351–391. doi:10.1016/S0065-3233(04)68010-7

    Article  CAS  PubMed  Google Scholar 

  64. Negus RP (1996) The chemokines: cytokines that direct leukocyte migration. J R Soc Med 89:312–314

    CAS  PubMed  Google Scholar 

  65. Clore GM, Gronenborn AM (1995) Three-dimensional structures of alpha and beta chemokines. FASEB J 9:57–62

    CAS  PubMed  Google Scholar 

  66. Baggiolini M, Dewald B, Moser B (1997) Human chemokines: an update. Annu Rev Immunol 15:675–705. doi:10.1146/annurev.immunol.15.1.675

    Article  CAS  PubMed  Google Scholar 

  67. Balkwill F (2004) Cancer and the chemokine network. Nat Rev Cancer 4:540–550. doi:10.1038/nrc1388

    Article  CAS  PubMed  Google Scholar 

  68. Shirozu M, Nakano T, Inazawa J, Tashiro K, Tada H, Shinohara T et al (1995) Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics 28:495–500. doi:10.1006/geno.1995.1180

    Article  CAS  PubMed  Google Scholar 

  69. Hamada T, Tashiro K, Tada H, Inazawa J, Shirozu M, Shibahara K et al (1996) Isolation and characterization of a novel secretory protein, stromal cell-derived factor-2 (SDF-2) using the signal sequence trap method. Gene 176:211–214. doi:10.1016/0378-1119(96)00251-X

    Article  CAS  PubMed  Google Scholar 

  70. Bleul CC, Farzan M, Choe H, Parolin C, Clark-Lewis I, Sodroski J et al (1996) The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry. Nature 382:829–833. doi:10.1038/382829a0

    Article  CAS  PubMed  Google Scholar 

  71. Feng Y, Broder CC, Kennedy PE, Berger EA (1996) HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 272:872–877. doi:10.1126/science.272.5263.872

    Article  CAS  PubMed  Google Scholar 

  72. Federsppiel B, Melhado IG, Duncan AM, Delaney A, Schappert K, Clark-Lewis I et al (1993) Molecular cloning of the cDNA and chromosomal localization of the gene for a putative seven-transmembrane segment (7-TMS) receptor isolated from human spleen. Genomics 16:707–712. doi:10.1006/geno.1993.1251

    Article  CAS  PubMed  Google Scholar 

  73. Baggiolini M (1998) Chemokines and leukocyte traffic. Nature 392:565–568. doi:10.1038/33340

    Article  CAS  PubMed  Google Scholar 

  74. Nagasawa T, Kikutani H, Kishimoto T (1994) Molecular cloning and structure of a pre-B-cell growth-stimulating factor. Proc Natl Acad Sci USA 91:2305–2309. doi:10.1073/pnas.91.6.2305

    Article  CAS  PubMed  Google Scholar 

  75. Nomura H, Nielsen BW, Matsushima K (1993) Molecular cloning of cDNAs encoding a LD78 receptor and putative leukocyte chemotactic peptide receptors. Int Immunol 5:1239–1249. doi:10.1093/intimm/5.10.1239

    Article  CAS  PubMed  Google Scholar 

  76. Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA (1996) A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1). J Exp Med 184:1101–1109. doi:10.1084/jem.184.3.1101

    Article  CAS  PubMed  Google Scholar 

  77. Ma Q, Jones D, Borghesani PR, Segal RA, Nagasawa T, Kishimoto T et al (1998) Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad Sci USA 95:9448–9453. doi:10.1073/pnas.95.16.9448

    Article  CAS  PubMed  Google Scholar 

  78. Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR (1998) Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature 393:595–599. doi:10.1038/31269

    Article  CAS  PubMed  Google Scholar 

  79. Lataillade JJ, Clay D, Bourin P, Hérodin F, Dupuy C, Jasmin C et al (2002) Stromal cell-derived factor 1 regulates primitive hematopoiesis by suppressing apoptosis and by promoting G(0)/G(1) transition in CD34(+) cells: evidence for an autocrine/paracrine mechanism. Blood 99:1117–1129. doi:10.1182/blood.V99.4.1117

    Article  CAS  PubMed  Google Scholar 

  80. Müller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME et al (2001) Involvement of chemokine receptors in breast cancer metastasis. Nature 410:50–56. doi:10.1038/35065016

    Article  PubMed  Google Scholar 

  81. Kato M, Kitayama J, Kazama S, Nagawa H (2003) Expression pattern of CXC chemokine receptor-4 is correlated with lymph node metastasis in human invasive ductal carcinoma. Breast Cancer Res 5:R144–R150. doi:10.1186/bcr627

    Article  CAS  PubMed  Google Scholar 

  82. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R et al (2005) Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121:335–348. doi:10.1016/j.cell.2005.02.034

    Article  CAS  PubMed  Google Scholar 

  83. Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H et al (2004) Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 6:17–32. doi:10.1016/j.ccr.2004.06.010

    Article  CAS  PubMed  Google Scholar 

  84. Rubin JB, Kung AL, Klein RS, Chan JA, Sun Y, Schmidt K et al (2003) A small-molecule antagonist of CXCR4 inhibits intracranial growth of primary brain tumors. Proc Natl Acad Sci USA 100:13513–13518. doi:10.1073/pnas.2235846100

    Article  CAS  PubMed  Google Scholar 

  85. Zhou Y, Larsen PH, Hao C, Yong VW (2002) CXCR4 is a major chemokine receptor on glioma cells and mediates their survival. J Biol Chem 277:49481–49487. doi:10.1074/jbc.M206222200

    Article  CAS  PubMed  Google Scholar 

  86. Geminder H, Sagi-Assif O, Goldberg L, Meshel T, Rechavi G, Witz IP et al (2001) A possible role for CXCR4 and its ligand, the CXC chemokine stromal cell-derived factor-1, in the development of bone marrow metastases in neuroblastoma. J Immunol (Baltimore, MD.: 1950) 167:4747–4757

    CAS  Google Scholar 

  87. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C et al (2003) A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3:537–549. doi:10.1016/S1535-6108(03)00132-6

    Article  CAS  PubMed  Google Scholar 

  88. Chen Y, Stamatoyannopoulos G, Song CZ (2003) Down-regulation of CXCR4 by inducible small interfering RNA inhibits breast cancer cell invasion in vitro. Cancer Res 63:4801–4804

    CAS  PubMed  Google Scholar 

  89. Helbig G, Christopherson KW 2nd, Bhat-Nakshatri P, Kumar S, Kishimoto H, Miller KD et al (2003) NF-κB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem 278:21631–21638. doi:10.1074/jbc.M300609200

    Article  CAS  PubMed  Google Scholar 

  90. Ries LAG, Eisner MP, Kosary CL, Hankey BF, Miller BA, Clegg L (eds) (2001) SEER cancer statistics review. Natl Cancer Inst Bethesda, MD

    Google Scholar 

  91. Tamamura H, Hori A, Kanzaki N, Hiramatsu K, Mizumoto M, Nakashima H, Yamamoto N, Otaka A, Fujii N (2003) NT140 analogs as CXCR4 antagonists identified as anti-metastatic agents in the treatment of breast cancer. FEBS Lett 550:79–83. doi:10.1016/S0014-5793(03)00824-X

    Article  CAS  PubMed  Google Scholar 

  92. Fischer OM, Hart S, Gschwind A, Ullrich A (2003) EGFR signal transactivation in cancer cells. Biochem Soc Trans 31:1203–1208

    Article  CAS  PubMed  Google Scholar 

  93. Luttrell DK, Luttrell LM (2004) Not so strange bedfellows: G-protein-coupled receptors and Src family kinases. Oncogene 23:7969–7978. doi:10.1038/sj.onc.1208162

    Article  CAS  PubMed  Google Scholar 

  94. Porcile C, Bajetto A, Barbero S, Pirani P, Schettini G (2004) CXCR4 activation induces epidermal growth factor receptor transactivation in an ovarian cancer cell line. Ann N Y Acad Sci 1030:162–169. doi:10.1196/annals.1329.021

    Article  CAS  PubMed  Google Scholar 

  95. Venkatakrishnan G, Salgia R, Groopman JE (2000) Chemokine receptors CXCR-1/2 activate mitogen-activated protein kinase via the epidermal growth factor receptor in ovarian cancer cells. J Biol Chem 275(10):6868–6875. doi:10.1074/jbc.275.10.6868

    Article  CAS  PubMed  Google Scholar 

  96. Li YM, Pan Y, Wei Y, Cheng X, Zhou BP, Tan M et al (2004) Upregulation of CXCR4 is essential for HER2-mediated tumor metastasis. Cancer Cell 6:459–469. doi:10.1016/j.ccr.2004.09.027

    Article  CAS  PubMed  Google Scholar 

  97. Cabioglu N, Summy J, Miller C, Parikh NU, Sahin AA, Tuzlali S et al (2005) CXCL-12/stromal cell-derived factor-1 transactivates HER2-neu in breast cancer cells by a novel pathway involving Src kinase activation. Cancer Res 65:6493–6497. doi:10.1158/0008-5472.CAN-04-1303

    Article  CAS  PubMed  Google Scholar 

  98. Lin J, Freeman MR (2003) Transactivation of ErbB1 and ErbB2 receptors by angiotensin II in normal human prostate stromal cells. Prostate 54:1–7. doi:10.1002/pros.10160

    Article  PubMed  Google Scholar 

  99. Gschwind A, Prenzel N, Ullrich A (2002) Lysophosphatidic acid-induced squamous cell carcinoma cell proliferation and motility involves epidermal growth factor receptor signal transactivation. Cancer Res 62:6329–6336

    CAS  PubMed  Google Scholar 

  100. Chinni SR, Yamamoto H, Dong Z, Sabbota A, Bonfil RD, Cher ML (2008) CXCL12/CXCR4 transactivates HER2 in lipid rafts of prostate cancer cells and promotes growth of metastatic deposits in bone. Mol Cancer Res 6:446–457. doi:10.1158/1541-7786.MCR-07-0117

    Article  CAS  PubMed  Google Scholar 

  101. Kishimoto H, Wang Z, Bhat-Nakshatri P, Chang D, Clarke R, Nakshatri H (2005) The p160 family coactivators regulate breast cancer cell proliferation and invasion through autocrine/paracrine activity of SDF-1alpha/CXCL12. Carcinogenesis 26:1706–1715. doi:10.1093/carcin/bgi137

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Limitations of space preclude extensive citation of the literature; we apologize to those whose work is not mentioned herein. Research in this lab is supported, in part, by National Institutes of Health Grants HL80699 (SA), CA096805 (HA), HL00791708 (CVH), Army Breast Cancer Concept Award (SA), the Susan G. Komen Fellowship (SA).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hava Karsenty Avraham.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hinton, C.V., Avraham, S. & Avraham, H.K. Role of the CXCR4/CXCL12 signaling axis in breast cancer metastasis to the brain. Clin Exp Metastasis 27, 97–105 (2010). https://doi.org/10.1007/s10585-008-9210-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10585-008-9210-2

Keywords

Navigation