Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jun 1.
Published in final edited form as: Clin Lymphoma Myeloma Leuk. 2011 Apr 30;11(Suppl 1):S65–S69. doi: 10.1016/j.clml.2011.03.022

Targeting the bone marrow in Waldenstrom Macroglobulinemia

Irene M Ghobrial 1, Yong Zhang 1, Yang Liu 1, Hai Ngo 1, Feda Azab 1, Antonio Sacco 1, Abdelkareem Azab 1, Patricia Maiso 1, Brittany Morgan 1, Phong Quang 1, Ghayas Issa 1, Xavier Leleu 1, Aldo Roccaro 1
PMCID: PMC3212729  NIHMSID: NIHMS330953  PMID: 22035751

Abstract

Waldesntrom Macroglobulinemia (WM) is a low-grade B cell lymphoma characterized by widespread involvement of the bone marrow with lymphoplasmacytic cells. In about 20% of patients, the malignant clone also involves the lymph nodes and induces hepatosplenomegaly. The mechanisms by which the tumor cells home to the bone marrow and preferentially reside in the marrow niches are not fully elucidated. In this review, we examine the role of the bone marrow microenvironment in the regulation of cell growth, survival and cell dissemination in WM. We also summarize specific regulators of niche-dependent tumor proliferation in WM. These include chemokines, adhesion molecules, Src/PI3K/Akt/mTOR signaling, NF-kB activation, and miRNA regulation in WM. Targeting these pathways in clinical trials could lead to significant responses in this rare disease.

Keywords: Waldenstrom, cell trafficking, homing, niche, bone marrow, adhesion

Introduction

Waldesntrom Macroglobulinemia is a low-grade B cell lymphoma characterized by widespread involvement of the bone marrow with lymphoplasmacytic cells. In about 20% of patients, the malignant clone also involves the lymph nodes and induces hepatosplenomegaly1-3. The mechanisms by which the tumor cells home to the bone marrow and preferentially reside in the marrow niches are not fully elucidated. However, many studies have shown that the microenvironment plays a crucial role in tumor cell proliferation, survival, and drug-resistance in WM.

The bone marrow niche is a term that was first coined by Schofield who indicated that the bone marrow microenvironment structures are essential for the long-term maintenance of a stable hematopoetici stem cell pool4. The BM microenvironment consists of hematopoietic and non-hematopoietic cells, as well as an extracellular and liquid compartment organized in a complex architecture of sub-microenvironments or “niches”5. At least 2 distinct niches have been identified in BM: the osteoblastic niche and the vascular niche6,7. The stroma of the bone marrow is composed of cells of various lineages, including osteoblasts, endothelial cells, fibroblasts, and adipocytes, but the relative contributions of each lineage have remained elusive.

Genetic and epigenetic events in the tumor clone of WM lead to its oncogenesis. WM is thought to arise from B cells in the germinal center that were arrested between the stages of somatic hypermutation and isotype class switching8,9. Deletion of the long arm of chromosome 6 (6q-) is the most frequent cytogenetic abnormality in WM. In 102 WM patients, the incidence of 6q21 deletion was found to be 7% by conventional cytogenetics and 34% when analyzed by fluorescent in situ hybridization (FISH). Patients with deletion of 6q had worse prognostic features that correlated with poor prognostic features of WM. In addition, patients with smoldering WM who displayed the abnormality showed a trend to an earlier requirement of therapy10. Other cytogenetic abnormalities include trisomy 4, trisomy 5, monsomy 8 and deletion 20q11-13. However, these events may not be sufficient, and a permissive microenvironment has been suggested to be required for frank malignancy to emerge14. Examples of microenvironmental contributions to neoplasia include a necessary mast cell contribution to Nf1-induced neurofibromas15 and mesenchymal cell alteration of epithelial tumor growth kinetics16. Changes in a tissue microenvironment have also been suggested to precede and promote the initiation of genetic events by creating a ‘pre-malignant’ state characterized by disruption of quiescence-inducing signals or increases in proliferative signaling17. This has been validated experimentally with altered TGF-β signaling in tissue fibroblasts18 and with myeloid progenitor expansion after RAR-γ deletion in bone marrow or Rb deficiency in haematopoietic and microenvironmental cells19. In addition, the relevance of niche-mediated oncogenesis has become evident with studies showing that the microenvironment plays an integral role in the development of myeloproliferative syndromes14.

Although no such evidence of niche-mediated oncogenesis is present in WM, several studies have shown that the interaction of WM cells with their microenvironment leads to tumor proliferation, survival and drug-resistance. In this review, we describe studies that demonstrate the critical role of the bone marrow niche in regulating tumor progression in WM.

Cell trafficking in WM

WM represents a spectrum from early asymptomatic monoclonal gammopathy of undetermined significance (IgM-MGUS), where there are small numbers of lymphoplasmacytic cells in the bone marrow (<10%), to active WM with anemia, hyperviscosity and widespread disease20,21. In a similar plasma cell dyscrasia such as Multiple Myeloma, studies have demonstrated the presence of a small number of circulating plasma cells in over 70% of patients with MM22. The number of circulating cells in the peripheral blood increased with progression of the disease, and was an independent unfavorable prognostic marker22. This data implies that progression of these plasma cell dyscrasias occurs through the continuous trafficking of the malignant cells to new sites of the BM.

The CXCR4/SDF-1 axis and migration in WM

Migration of cells through the blood to the bone marrow niches requires active navigation through the process of homing. Homing is thought to be a coordinated, multistep process, which involves signaling by stromal derived factor (SDF-1), activation of lymphocyte function–associated antigen 1 (LFA-1), VLA-4/5, and activation of MMP2/923-25. SDF-1 is chemokine that has been extensively studied in the regulation of homing and migration of hematopoietic cells and lymphocytes. The receptor of SDF-1 is CXCR4. Recently, a new receptor was discovered that is activated by SDF-1, namely CXCR7 (RDC-1)26. SDF-1 is primarily produced by stromal cells. The major biological effects of SDF-1 are related to the ability of this chemokine to induce (a) motility, (b) chemotactic responses, (c) adhesion, and (d) secretion of MMPs and angiopoietic factors (e.g., VEGF)27-29. SDF-1 has been reported to induce firm adhesion and migration by inducing activation of integrins LFA-1, VLA-4, and VLA-5 on stem cells27,29-31. CXCR4 is expressed on the surface of normal cells such as hematopoeitic stem cells and T and B lymphocytes, as well as malignant cells such as breast cancer cells and lymphoid malignancies32-36. CXCR4 knockout mice die neonatally due to multiple developmental defects, with lymphopoiesis and myelopoiesis dramatically defective in these fetuses37. Plasma cells in chimeric mice reconstituted with CXCR4-deficient fetal liver cells were found in elevated numbers in the blood, and failed to accumulate in the BM, indicating an important role of CXCR4 in the homing of malignant plasma cells38. The most important pathways involved in signaling that regulate migration or adhesion include activation of the MAPK p42/44–ERK-1, PI3K–AKT, PKC, NF-kB pathways, focal adhesion components such as focal adhesion kinase (FAK), paxillin, Nck, Crk, Crk-L, protein kinase C (PKC), phospholipase C (PLC), and calcium flux29,39-41,42.

We previously showed that WM cells express high levels of chemokine and adhesion receptors, including CXCR4 and VLA-443. We showed that CXCR4 was essential for the migration and trans-endothelial migration of WM cells under static and dynamic shear flow conditions, with significant inhibition of migration using CXCR4 knockdown or the CXCR4 inhibitor plerixafor43. Similarly, CXCR4 or VLA-4 inhibition led to significant inhibition of adhesion to fibronectin, stromal cells, and endothelial cells43. Decreased adhesion of WM cells to stromal cells by plerixafor led to increased sensitivity of these cells to cytotoxicity by bortezomib. To further investigate the mechanisms of CXCR4-dependent adhesion, we showed that CXCR4 and VLA-4 directly interact in response to SDF-1. We further investigated downstream signaling pathways regulating migration and adhesion in WM, including the PI3K and ERK pathways and found that these pathways are activated in response to CXCR4 activation43.

Another study was recently published that examined the distribution and the clinical influence of the SDF-1 (CXCL12) (-801GA) polymorphism in a series of 114 WM patients44. CXCL12 (-801AA) genotype was more frequent in WM patients compared with control subjects (p = 0.01). On the other hand, CXCL12 (-801GG) patients had a shorter median survival after initiation of first line therapy than remaining patients that did not have CXCL12 (-801GG), (p = 0.01). In conclusion, the CXCL12 (-801GA) polymorphism may either be associated with a high incidence of WM or influence clinical outcome.

CXCR4 is not the only receptor for CXCL12 (SDF-1). The more recently identified CXCR7 receptor has been identified in other hematological malignancies, specifically multiple myeloma. Its role in WM is currently being evaluated. These studies indicate that CXCR4 inhibitors may be used in the therapy of patients with WM. The preclinical data showing that inhibition of CXCR4 with plerixafor leads to sensitization of tumor cells to therapy with bortezomib indicate that this strategy may be used in clinical trials. The use of plerixafor in chemosensitization therapy in clinical trials of AML and multiple myeloma are ongoing and the results are promising.

Adhesion receptors and WM

Proteomic analysis of WM cells compared to normal CD19+ cells has shown that adhesion –related proteins are upregulated in WM. These included Integrin b3 (CD61), MDC9 (ADAM), JAM-1, Mena, Maspin, LAR, annexin II, p62 lck ligand, pp120 src substrate, CLA-1 (CD36), RPTPb, nexilin,contactin, and tensin.

Previous studies presented by our group and others have shown that VLA-4 and LFA-1 are highly expressed in WM and that adhesion to fibronectin, endothelial cells and stromal cells are regulated by VLA-4. We also showed that VLA-4 interacts with CXCR4, indicating the complexity and interactions between adhesion and migration.

Src tyrosine kinase is known to regulate cell adhesion and migration45. A recent study has shown that Src was overexpressed in WM45. Inhibition of Src by AZD0530 (Astra Zeneca Inc) was shown to inhibit the basal activation of Src in BCWM1, as well as to inhibit the SDF1-induced activation of Src and other cytoskeletal-related proteins and polymerization of actin, indicating an important role of Src in the signaling cascade downstream of CXCR4 leading to an increase of migration and adhesion of WM45. Inhibition of CXCR4 by plerixafor or inhibition of Src results in similar effects of inhibition of the chemotaxis. Therefore, Src inhibition may be a good target for the development of therapeutic agents in WM.

The PI3K/mTOR pathway

Tumorigenesis results from synergistic interactions of a complex of signal transduction processes, including multiple onco-proteins and tumor suppressors such as Ras, Myc, PKB/Akt, Her-2/Neu, p53 and PTEN46-48. Overexpression of Akt plays an important role in the initiation and progression of malignancies. The PI3K pathway is important in enhancing cell survival by stimulating cell proliferation and inhibiting apoptosis49-53. Regulators of this pathway include PTEN, HSP90, P18, P15, P19, P16 and P2154,55.

There is no evidence that there are activating mutations in the PI3K/Akt/mTOR pathway in WM. Therefore, activation of this pathway may be due to external stimulation though the bone marrow microenvironment such as stimulation through IGF-1 or SDF-1. Previous studies have shown activation of Akt and mTOR in WM. Inhibition of Akt by perifosine (Keryx Inc)56, mTOR by RAD001 (Novartis Inc) or the PI3K/mTOR inhibitor (NVP-BEZ235)57 have shown activity on WM cells even in the presence of bone marrow stromal cells.

Based on the preclinical studies, a phase II trial of single agent perifosine was conducted in patients with relapsed or refractory WM58. The study showed that of the 37 patients, 4 achieved partial response (11%), 9 minimal response (24%), and 20 showed stable disease (54%). The median progression-free survival was 12.6 months. Gene expression profiling of samples obtained from patients before and after therapy with perifosine showed that there were 162 genes significantly changed in expression in response to perifosine. These included reduced expression of several genes involved in the adhesion and migration processes, as well as regulators of the NF-kB pathway.

RAD001 (Everolimus, Novartis Inc) has shown preclinical activity in WM. In additon, it inhibited adhesion of WM cells to stromal cells and enhanced activity of rituximab and bortezomib (unpublished data from our lab). A Phase II trial of single agent RAD001 in patients with relapsed WM was conducted in 50 patients59. The response rate was 42% (all PR) with 28% MR for an overall clinical benefit rate of 70%. The estimated PFS at 6 and 12 months is 75% (95%CI: 64-89%) and 62% (95%CI: 48-80%)59. Prior studies of mTOR inhibitors have shown that these agents can induce tumor mobilization of the malignant cells from the lymph node compartment to th e peripheral blood compartment, such as in Mantle cell lymphoma. Similar observations were seen with PI3K delta inhibitors in chronic lymphocytic leukemia (CLL), indicating that inhibitors of this pathway can induce mobilization of the tumor clone. Mobilization of tumor cells to the peripheral blood can make them more sensitive to therapeutic agents that are usually more active on tumor cells that are not adherent to stroma. We did not examine peripheral mobilization of WM cells in the RAD001 study, but future studies of mTOR inhibitors and other inhibitors of this pathway will examine the effect of these agents on mobilization of the tumor cells to the peripheral blood and their sensitization to therapeutic agents while they are in circulation.

In a recent study using the PI3K/mTOR inhibitor NVP-BEZ235, Roccaro et al showed that bone marrow stromal cells (BMSCs) triggered increase of 52% in proliferation of WM cells, which was inhibited by NVP- BEZ235 in a dose-dependent manner57. Adherence of BCWM.1 cells to BMSCs induced phosphorylation of Akt and mTOR; conversely, NVP-BEZ235 abrogated BMSC adhesion- induced phosphorylation of Akt and mTOR in WM cells, indicating that NVP-BEZ235 exerts its antitumor activity even when WM cells were in contact with BM milieu. Similarly, NVP-BEZ235 targeted rictor and raptor in WM cells, even when cultured in presence of BMSCs, which induced up-regulation of raptor, but not rictor. NVP-BEZ235 induced significant inhibition of adhesion to either fibronectin or BMSCs. NVP-BEZ235 also inhibited migration of WM cells in response to SDF-1. NVP-BEZ235 inhibited phosphorylation of focal adhesion kinase, paxillin, and cofilin; important proteins that act as key regulators of adhesion and cell migration . Finally, NVP-BEZ235 resulted in a significant inhibition of homing of WM cells to the BM in vivo.

The NF-kB pathway

The NF-kB pathway in malignant transformation, progression and resistance: NF-kB comprises a family of transcription factors that regulate the transcription of hundreds of genes involved in inflammation, innate immunity, cell growth and apoptosis 60. Inhibition of the NFkB pathway by proteasome inhibitors such as bortezomib and other agents such as NPI-0052m carfilzomib and Onyx0912 can inhibit WM cells even in the presence of bone marrow stromal cells61,62. Bortezomib is known to inhibit adhesion of plasma cells in MM. Studies using bortezomib alone or in combination with rituximab have shown significant activity with over 80% response rate even in relapsed or newly diagnosed WM63-65. Future studies using carfilzomib are planned in WM.

Epigenetics in the bone marrow of WM

miRNA 155 in WM. miRNA-155 plays a pivotal role in B-cell malignancies including diffuse large B-cell lymphomas, primary mediastinal B-cell lymphomas, and Hodgkin lymphomas66. We found that miRNA-155 is increased in WM tumor cells compared to normal CD19+ cells66. Therefore we studied the functional role of miRNA-155 in WM by examining proliferation, adhesion, and migration in miR-155 LNA knockdown WM cells compared with controls (control probe–transfected BCWM.1 cells or nontransfected BCWM.1 cells). Our studies have shown that knocking down miRNA-155 in WM cells reduced the adhesion and migration in vitro; and significantly decreased proliferation even in the presence of bone marrow stromal cells66. In vivo studies confirmed significant inhibition of homing and proliferation of miRNA-155 knockdown WM cells; decreased IgM secretion; and a significant survival benefit in mice. We further confirmed that specific knockdown of miRNA-155 inhibits specific genes regulating cell cycle as well as regulators of adhesion and migration. Together, these studies confirm that miRNA-155 is a critical regulator of proliferation, cell cycle regulation, migration and adhesion66. Further studies to use miRNA 155 in the treatment of patients with WM are ongoing.

In summary, we show that the bone marrow microenvironment is a critical regulator of cell growth, survival and cell dissemination in WM. Targeting this niche-dependent tumor growth is important in the design of future clinical trials in this disease. Specific targets that can be used for clinical trials include chemokine receptors, adhesion molecules, downstream signaling targets such as Src, PI3K/Akt/mTOR, or specific epigenetic changes such as miRNA regulation

Table of novel agents targeting migration and adhesion.

Pathway or Target Therapeutic agent Clinical or preclinical testing Reference
CXCR4 Plerixafor Preclinical testing 43
VLA-4 and LFA-1 VLA-4 antibodies Preclinical testing 43
Src AZD0530 Preclinical testing 45
PI3K/Akt/mTOR pathway Perifosine, RAD001, BEZ-235 ○ Phase II trial of perifosine showed an ORR of 35% including MR in relapsed or refractory WM.
○ Phase II trial of RAD001 showed an ORR of 70% including MR in relapsed or refractory WM.
○ NVP-BEZ-235 completed preclinical testing.
56, 57, 58, 59,
NF-kB Bortezomib, NPI-0052, carfilzomib and Onyx 0912 ○ Bortezomib single agent or in combination with rituximab has completed phase II clinical trials in WM with an ORR of about 80-90% including MR in newly diagnosed or relapsed WM.
○ NPI-0052, Carfilzomib and Onyx-0912 completed preclinical testing.
61-65
miRNA Anti-miR-155 LNA Preclinical testing 66

Acknowledgments

Supported in part by the International Waldenstrom Macroglobulinemia Foundation (IWMF), Kirsch Lab for Waldenstrom and Heje Fellowship for WM.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Treon SP. How I treat Waldenstrom's macroglobulinemia. Blood. 2009 doi: 10.1182/blood-2009-05-174359. [DOI] [PubMed] [Google Scholar]
  • 2.Vijay A, Gertz MA. Waldenstrom Macroglobulinemia. Blood. 2007 doi: 10.1182/blood-2006-11-055012. [DOI] [PubMed] [Google Scholar]
  • 3.Ghobrial IM, Witzig TE. Waldenstrom macroglobulinemia. Curr Treat Options Oncol. 2004;5:239–247. doi: 10.1007/s11864-004-0015-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Scadden DT. Circadian rhythms: stem cells traffic in time. Nature. 2008;452:416–417. doi: 10.1038/452416a. [DOI] [PubMed] [Google Scholar]
  • 5.Roodman GD. Role of the bone marrow microenvironment in multiple myeloma. J Bone Miner Res. 2002;17:1921–1925. doi: 10.1359/jbmr.2002.17.11.1921. [DOI] [PubMed] [Google Scholar]
  • 6.Sipkins DA, Wei X, Wu JW, et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature. 2005;435:969–973. doi: 10.1038/nature03703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yin T, Li L. The stem cell niches in bone. J Clin Invest. 2006;116:1195–1201. doi: 10.1172/JCI28568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kriangkum J, Taylor BJ, Treon SP, Mant MJ, Belch AR, Pilarski LM. Clonotypic IgM V/D/J sequence analysis in Waldenstrom macroglobulinemia suggests an unusual B-cell origin and an expansion of polyclonal B cells in peripheral blood. Blood. 2004;104:2134–2142. doi: 10.1182/blood-2003-11-4024. [DOI] [PubMed] [Google Scholar]
  • 9.Sahota S, Forconi F, Ottensmeier CH, Provan D, Oscier DG, Hamblin T, et al. Typical Waldenström macroglobulinemia is derived from a B-cell arrested after cessation of somatic mutation but prior to isotype switch events. Blood. 2002;100:1505–1507. [PubMed] [Google Scholar]
  • 10.Schop RF, Kuehl WM, Van Wier SA, et al. Waldenstrom macroglobulinemia neoplastic cells lack immunoglobulin heavy chain locus translocations but have frequent 6q deletions. Blood. 2002;100:2996–3001. doi: 10.1182/blood.V100.8.2996. [DOI] [PubMed] [Google Scholar]
  • 11.Schop RF, Jalal SM, Van Wier SA, et al. Deletions of 17p13.1 and 13q14 are uncommon in Waldenstrom macroglobulinemia clonal cells and mostly seen at the time of disease progression. Cancer Genet Cytogenet. 2002;132:55–60. doi: 10.1016/s0165-4608(01)00526-x. [DOI] [PubMed] [Google Scholar]
  • 12.Terre C, Nguyen-Khac F, Barin C, Mozziconacci MJ, Eclache V, Leonard C, et al. Trisomy 4, a new chromosomal abnormality in Waldenstro□m's macroglobulinemia: a study of 39 cases. Leukemia. 2006;20:1634–1636. doi: 10.1038/sj.leu.2404314. [DOI] [PubMed] [Google Scholar]
  • 13.Liu Y, Miyazawa K, Sashida G, Kodama A, Ohyashiki K. Deletion (20q) as the sole abnormality in Waldenstro□m macroglobulinemia suggests distinct pathogenesis of 20q11 anomaly. Cancer Genet Cytogenet. 2006;169:69–72. doi: 10.1016/j.cancergencyto.2006.03.013. [DOI] [PubMed] [Google Scholar]
  • 14.Raaijmakers MH, Mukherjee S, Guo S, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 464:852–857. doi: 10.1038/nature08851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yang FC, Ingram DA, Chen S, et al. Nf1-dependent tumors require a microenvironment containing Nf1+/-- and c-kit-dependent bone marrow. Cell. 2008;135:437–448. doi: 10.1016/j.cell.2008.08.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Trimboli AJ, Cantemir-Stone CZ, Li F, et al. Pten in stromal fibroblasts suppresses mammary epithelial tumours. Nature. 2009;461:1084–1091. doi: 10.1038/nature08486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sneddon JB, Werb Z. Location, location, location: the cancer stem cell niche. Cell Stem Cell. 2007;1:607–611. doi: 10.1016/j.stem.2007.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bhowmick NA, Chytil A, Plieth D, et al. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303:848–851. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
  • 19.Walkley CR, Shea JM, Sims NA, Purton LE, Orkin SH. Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment. Cell. 2007;129:1081–1095. doi: 10.1016/j.cell.2007.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kyle RA, Rajkumar SV. Monoclonal gammopathy of undetermined significance. Clin Lymphoma Myeloma. 2005;6:102–114. doi: 10.3816/CLM.2005.n.036. [DOI] [PubMed] [Google Scholar]
  • 21.Kyle RA, Therneau TM, Rajkumar SV, Larson DR, Plevak MF, Melton LJ., 3rd Long-term follow-up of 241 patients with monoclonal gammopathy of undetermined significance: the original Mayo Clinic series 25 years later. Mayo Clin Proc. 2004;79:859–866. doi: 10.4065/79.7.859. [DOI] [PubMed] [Google Scholar]
  • 22.Nowakowski GS, Witzig TE, Dingli D, et al. Circulating plasma cells detected by flow cytometry as a predictor of survival in 302 patients with newly diagnosed multiple myeloma. Blood. 2005;106:2276–2279. doi: 10.1182/blood-2005-05-1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lapidot T, Dar A, Kollet O. How do stem cells find their way home? Blood. 2005;106:1901–1910. doi: 10.1182/blood-2005-04-1417. [DOI] [PubMed] [Google Scholar]
  • 24.Avecilla ST, Hattori K, Heissig B, et al. Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med. 2004;10:64–71. doi: 10.1038/nm973. [DOI] [PubMed] [Google Scholar]
  • 25.Kollet O, Shivtiel S, Chen YQ, et al. HGF, SDF-1, and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liver. J Clin Invest. 2003;112:160–169. doi: 10.1172/JCI17902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Balabanian K, Lagane B, Infantino S, et al. The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem. 2005;280:35760–35766. doi: 10.1074/jbc.M508234200. [DOI] [PubMed] [Google Scholar]
  • 27.Peled A, Kollet O, Ponomaryov T, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood. 2000;95:3289–3296. [PubMed] [Google Scholar]
  • 28.Lapidot T, Kollet O. The essential roles of the chemokine SDF-1 and its receptor CXCR4 in human stem cell homing and repopulation of transplanted immune-deficient NOD/SCID and NOD/SCID/B2m(null) mice. Leukemia. 2002;16:1992–2003. doi: 10.1038/sj.leu.2402684. [DOI] [PubMed] [Google Scholar]
  • 29.Kucia M, Reca R, Miekus K, et al. Trafficking of normal stem cells and metastasis of cancer stem cells involve similar mechanisms: pivotal role of the SDF-1-CXCR4 axis. Stem Cells. 2005;23:879–894. doi: 10.1634/stemcells.2004-0342. [DOI] [PubMed] [Google Scholar]
  • 30.Peled A, Grabovsky V, Habler L, et al. The chemokine SDF-1 stimulates integrin-mediated arrest of CD34(+) cells on vascular endothelium under shear flow. J Clin Invest. 1999;104:1199–1211. doi: 10.1172/JCI7615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wysoczynski M, Reca R, Ratajczak J, et al. Incorporation of CXCR4 into membrane lipid rafts primes homing-related responses of hematopoietic stem/progenitor cells to an SDF-1 gradient. Blood. 2005;105:40–48. doi: 10.1182/blood-2004-04-1430. [DOI] [PubMed] [Google Scholar]
  • 32.Juarez J, Bendall L. SDF-1 and CXCR4 in normal and malignant hematopoiesis. Histol Histopathol. 2004;19:299–309. doi: 10.14670/HH-19.299. [DOI] [PubMed] [Google Scholar]
  • 33.Murphy PM. Chemokines and the molecular basis of cancer metastasis. N Engl J Med. 2001;345:833–835. doi: 10.1056/NEJM200109133451113. [DOI] [PubMed] [Google Scholar]
  • 34.Nakayama T, Hieshima K, Izawa D, Tatsumi Y, Kanamaru A, Yoshie O. Cutting edge: profile of chemokine receptor expression on human plasma cells accounts for their efficient recruitment to target tissues. J Immunol. 2003;170:1136–1140. doi: 10.4049/jimmunol.170.3.1136. [DOI] [PubMed] [Google Scholar]
  • 35.Ghobrial IM, Bone ND, Stenson MJ, et al. Expression of the chemokine receptors CXCR4 and CCR7 and disease progression in B-cell chronic lymphocytic leukemia/ small lymphocytic lymphoma. Mayo Clin Proc. 2004;79:318–325. doi: 10.4065/79.3.318. [DOI] [PubMed] [Google Scholar]
  • 36.Smith MC, Luker KE, Garbow JR, et al. CXCR4 regulates growth of both primary and metastatic breast cancer. Cancer Res. 2004;64:8604–8612. doi: 10.1158/0008-5472.CAN-04-1844. [DOI] [PubMed] [Google Scholar]
  • 37.Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature. 1998;393:595–599. doi: 10.1038/31269. [DOI] [PubMed] [Google Scholar]
  • 38.Erickson LD, Lin LL, Duan B, Morel L, Noelle RJ. A genetic lesion that arrests plasma cell homing to the bone marrow. Proc Natl Acad Sci U S A. 2003;100:12905–12910. doi: 10.1073/pnas.2131686100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kucia M, Jankowski K, Reca R, et al. CXCR4-SDF-1 signalling, locomotion, chemotaxis and adhesion. J Mol Histol. 2004;35:233–245. doi: 10.1023/b:hijo.0000032355.66152.b8. [DOI] [PubMed] [Google Scholar]
  • 40.Ganju RK, Brubaker SA, Meyer J, et al. The alpha-chemokine, stromal cell-derived factor-1alpha, binds to the transmembrane G-protein-coupled CXCR-4 receptor and activates multiple signal transduction pathways. J Biol Chem. 1998;273:23169–23175. doi: 10.1074/jbc.273.36.23169. [DOI] [PubMed] [Google Scholar]
  • 41.Zhang XF, Wang JF, Matczak E, Proper JA, Groopman JE. Janus kinase 2 is involved in stromal cell-derived factor-1alpha-induced tyrosine phosphorylation of focal adhesion proteins and migration of hematopoietic progenitor cells. Blood. 2001;97:3342–3348. doi: 10.1182/blood.v97.11.3342. [DOI] [PubMed] [Google Scholar]
  • 42.Petit I, Goichberg P, Spiegel A, et al. Atypical PKC-zeta regulates SDF-1-mediated migration and development of human CD34+ progenitor cells. J Clin Invest. 2005;115:168–176. doi: 10.1172/JCI21773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ngo HT, Leleu X, Lee J, et al. SDF-1/CXCR4 and VLA-4 interaction regulates homing in Waldenstrom macroglobulinemia. Blood. 2008;112:150–158. doi: 10.1182/blood-2007-12-129395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Poulain S, Ertault M, Leleu X, et al. SDF1/CXCL12 (-801GA) polymorphism is a prognostic factor after treatment initiation in Waldenstrom macroglobulinemia. Leuk Res. 2009;33:1204–1207. doi: 10.1016/j.leukres.2009.03.031. [DOI] [PubMed] [Google Scholar]
  • 45.Ngo HT, Azab AK, Farag M, et al. Src tyrosine kinase regulates adhesion and chemotaxis in Waldenstrom macroglobulinemia. Clin Cancer Res. 2009;15:6035–6041. doi: 10.1158/1078-0432.CCR-09-0718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rosen N, She QB. AKT and cancer--is it all mTOR? Cancer Cell. 2006;10:254–256. doi: 10.1016/j.ccr.2006.10.001. [DOI] [PubMed] [Google Scholar]
  • 47.Yao D, Alexander CL, Quinn JA, Porter MJ, Wu H, Greenhalgh DA. PTEN loss promotes rasHa-mediated papillomatogenesis via dual up-regulation of AKT activity and cell cycle deregulation but malignant conversion proceeds via PTEN-associated pathways. Cancer Res. 2006;66:1302–1312. doi: 10.1158/0008-5472.CAN-05-2341. [DOI] [PubMed] [Google Scholar]
  • 48.Li L, Ross AH. Why is PTEN an important tumor suppressor? J Cell Biochem. 2007;102:1368–1374. doi: 10.1002/jcb.21593. [DOI] [PubMed] [Google Scholar]
  • 49.Cantrell DA. Phosphoinositide 3-kinase signalling pathways. J Cell Sci. 2001;114:1439–1445. doi: 10.1242/jcs.114.8.1439. [DOI] [PubMed] [Google Scholar]
  • 50.Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C, Gonzalez-Baron M. PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 2004;30:193–204. doi: 10.1016/j.ctrv.2003.07.007. [DOI] [PubMed] [Google Scholar]
  • 51.Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;4:988–1004. doi: 10.1038/nrd1902. [DOI] [PubMed] [Google Scholar]
  • 52.Pene F, Claessens YE, Muller O, et al. Role of the phosphatidylinositol 3-kinase/Akt and mTOR/P70S6-kinase pathways in the proliferation and apoptosis in multiple myeloma. Oncogene. 2002;21:6587–6597. doi: 10.1038/sj.onc.1205923. [DOI] [PubMed] [Google Scholar]
  • 53.Dancey JE. Molecular targeting: PI3 kinase pathway. Ann Oncol. 2004;15(Suppl 4):iv233–239. doi: 10.1093/annonc/mdh932. [DOI] [PubMed] [Google Scholar]
  • 54.Choi Y, Zhang J, Murga C, et al. PTEN, but not SHIP and SHIP2, suppresses the PI3K/Akt pathway and induces growth inhibition and apoptosis of myeloma cells. Oncogene. 2002;21:5289–5300. doi: 10.1038/sj.onc.1205650. [DOI] [PubMed] [Google Scholar]
  • 55.Shi Y, Gera J, Hu L, et al. Enhanced sensitivity of multiple myeloma cells containing PTEN mutations to CCI-779. Cancer Res. 2002;62:5027–5034. [PubMed] [Google Scholar]
  • 56.Leleu X, Jia X, Runnels J, et al. The Akt pathway regulates survival and homing in Waldenstrom macroglobulinemia. Blood. 2007;110:4417–4426. doi: 10.1182/blood-2007-05-092098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Roccaro AM, Sacco A, Husu EN, et al. Dual targeting of the PI3K/Akt/mTOR pathway as an antitumor strategy in Waldenstrom macroglobulinemia. Blood. 115:559–569. doi: 10.1182/blood-2009-07-235747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ghobrial IM, Roccaro A, Hong F, et al. Clinical and translational studies of a phase II trial of the novel oral Akt inhibitor perifosine in relapsed or relapsed/refractory Waldenstrom's macroglobulinemia. Clin Cancer Res. 16:1033–1041. doi: 10.1158/1078-0432.CCR-09-1837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ghobrial IC, S. Sam A, et al. Phase II Trial of the mTOR Inhibitor RAD001 in Relapsed and/or Refractory Waldenstrom Macroglobulinemia: The Dana Farber Cancer Institute Experience Blood. 2008. Abstract.
  • 60.Hayden MSGS. Signaling to NF-kappaB. Genes and Development. 2004;18:2195–2224. doi: 10.1101/gad.1228704. [DOI] [PubMed] [Google Scholar]
  • 61.Roccaro AM, Leleu X, Sacco A, et al. Dual targeting of the proteasome regulates survival and homing in Waldenstrom's Macroglobulinemia. Blood. 2008 doi: 10.1182/blood-2007-11-120972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Roccaro AM, Sacco A, Aujay M, et al. Selective inhibition of chymotrypsin-like activity of the immunoproteasome and constitutive proteasome in Waldenstrom macroglobulinemia. Blood. doi: 10.1182/blood-2009-09-243402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Treon SP, Ioakimidis L, Soumerai JD, et al. Primary Therapy of Waldenstrom Macroglobulinemia With Bortezomib, Dexamethasone, and Rituximab: WMCTG Clinical Trial 05-180. J Clin Oncol. 2009 doi: 10.1200/JCO.2008.20.4677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ghobrial IM, Xie W, Padmanabhan S, et al. Phase II trial of weekly bortezomib in combination with rituximab in untreated patients with Waldenstrom Macroglobulinemia. Am J Hematol. 85:670–674. doi: 10.1002/ajh.21788. [DOI] [PubMed] [Google Scholar]
  • 65.Ghobrial IM, Hong F, Padmanabhan S, et al. Phase II trial of weekly bortezomib in combination with rituximab in relapsed or relapsed and refractory Waldenstrom macroglobulinemia. J Clin Oncol. 28:1422–1428. doi: 10.1200/JCO.2009.25.3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Roccaro AM, Sacco A, Chen C, et al. microRNA expression in the biology, prognosis and therapy of Waldenstrom macroglobulinemia. Blood. 2008 doi: 10.1182/blood-2008-09-178228. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES