Skip to main content
BoneKEy Reports logoLink to BoneKEy Reports
. 2014 Dec 3;3:600. doi: 10.1038/bonekey.2014.95

Immune regulation of bone metastasis

Aude-Hélène Capietto 1, Roberta Faccio 1,a
PMCID: PMC4260446  PMID: 25512853

Abstract

Metastases to bone occur in about 70% of patients with metastatic prostate and breast cancers. Unfortunately, bone metastases result in significant morbidity and mortality and treatment options are limited. Thus, significant effort has focused on understanding the mechanisms that drive tumor dissemination to bone. Bone metastases are typically characterized by a self-perpetuating ‘vicious' cycle wherein tumor cells and bone-resorbing cells (osteoclasts) are locked in a cycle that leads to osteoclast-driven bone destruction and the release of bone-stored factors that in turn stimulate tumor cell proliferation and survival. To break this ‘vicious' cycle, potent antiresorptive agents such as zoledronic acid (ZOL) have been used. However, in the clinical setting, ZOL failed to improve the overall survival of cancer patients even though it inhibited osteoclast resorptive activity. Thus, other cells in addition to osteoclasts are likely involved in modulating tumor growth in the bone. The immune system has the ability to eliminate tumor cells. Nevertheless, tumor cells can acquire the ability to escape immune control. Our recent observations indicated that a decline in the ability of the immune cells to recognize and kill the tumor drives tumor dissemination to bone even when osteoclasts are inhibited by potent antiresorptive agents. This review focuses on the antitumor and protumor effects of various immune cell populations involved in the bone metastatic process. We also discuss strategies to enhance antitumor immune responses and bypass cancer immune resistance.

Introduction

Cancer immunosurveillance occurs when the immune system identifies danger signals such as tumor-specific antigens or stress ligands on transformed cells that have escaped cell-intrinsic tumor suppressor mechanisms and eliminates them before they can establish malignancy.1 Unfortunately, antitumor immune responses are not always efficient in eliminating incipient tumors, thus allowing the transformed cells to escape immune control. Many mechanisms are involved in the escape phase including intrinsic cancer cell alterations and tumor-induced immunosuppression.1 The result of the escape phase is the tumor outgrowth and dissemination to distant sites.

The skeleton is the predominant metastatic site for many cancers, including breast, prostate and lung cancers.2,3,4,5 Tumor invasion into bone is associated with marked skeletal-related events (SREs) such as fractures, bone pain, hypercalcemia and spinal cord compression.6 The current model for the pathophysiology of bone metastasis centers on the interaction between tumor cells and osteoclasts (OCs) and is known as the ‘bone tumor vicious cycle'. Tumor cells secrete a plethora of factors and cytokines that can directly activate the OC or increase their maturation by stimulating osteoblast-mediated production of receptor activator of nuclear factor-κB ligand (RANKL). Once mature OCs start to resorb the bone, they release bone-stored factors such as tumor growth factor-β (TGF-β) that further stimulate tumor cell recruitment and proliferation.7 Thus, potent antiresorptive agents such as zoledronic acid (ZOL) and the anti-RANKL monoclonal antibody (Ab) such as Denosumab have become common treatments to minimize the risk of fractures in patients with bone metastasis.4,8,9 Despite the central role that the OC has in creating a hospitable niche for tumor colonization and growth in the bone microenvironment, the antitumor effects of ZOL are controversial. The AZURE trial did not support the use of ZOL as adjuvant therapy in breast cancer.10 Further analysis indicated that ZOL adjuvant therapy improved the disease-free survival in postmenopausal breast cancer patients but not in premenopausal women. These findings were in contrast to another study, the ABCSG-12 trial, showing improved disease-free survival in premenopausal early-stage breast cancer patients receiving ZOL adjuvant therapy.11 Thus, there are likely other cells/factors modulating the ZOL antitumor effects in addition to targeting the OCs. Emerging evidence suggests that immune populations have a critical role in controlling local tumor growth within the bone microenvironment.12,13,14 This review aims to discuss the recent findings showing the antitumor and protumor effects of immune cells in skeletal metastases and their interplay with ZOL.

CD4+ and CD8+ T cells

During the antitumor immune response, the capture of tumor antigens by dendritic cells (DCs) induces their maturation and migration to tumor-draining lymph nodes where DCs cross-present these antigens to T cells leading to their activation. Tumor-specific cytotoxic CD8+ T cells (CTLs) participate in the killing of antigen-positive tumor cells,15,16 and activated CD4+ T cells are shown to further facilitate the development of CTLs. Thus, the presence of activated T cells at tumor sites or in circulation generally has a good prognostic value. Although the percentage of T cells in the bone marrow is usually very low, activated CD4+ and CD8+ T cells have been observed in the bone marrow of untreated patients with breast cancer,17 thus suggesting that they may have a protective role in the bone metastatic dissemination. However, the authors also observed increased memory T cells in breast cancer patients with disseminated tumor cells (DTCs) in the bone marrow compared with those without detectable DTCs. As the presence of DTCs is associated with an increased risk of bone metastasis,18,19 clinical data correlating patient immune profiles with progression of tumors in the bone need to be further evaluated.

In recent years, a few laboratories have started investigating the role of T cells in tumor growth in the bone using various animal models of bone metastases. Our laboratory demonstrated that both CD4+ and CD8+ T-cell populations exert antitumor effects in the context of bone metastases.13 Depletion of either cell type, alone or in combination, significantly increases the growth and metastatic dissemination of melanoma tumors in bone. Upon activation, T cells produce interferon-γ (IFNγ), a cytokine that has a critical role in antitumor immune responses. Human T-cell leukemia virus type 1 (HTLV-1) Tax transgenic mice develop spontaneous bone tumors. In this tumor model, genetic deletion of IFNγ increases the number of bone tumors.20 This result indicates that IFNγ, mainly produced by HTLV-1-specific T cells (and possibly natural killer cells), has protective effects on Tax(+) tumor cell growth in the bone.

Activation of T-cell responses via injection of ipilimumab, an Ab that blocks the inhibitory signal CTLA4 expressed on T cells, suppresses tumor growth in the bone following intracardiac injection of B16 melanoma cells.13 Ipilimumab injection was approved by the Food and Drug Administration (FDA) in 2011 for the treatment of unresectable or metastatic melanoma. However, its protective effects in patients with bone metastases have only been indirectly evaluated in a recent phase III clinical trial. This trial included 799 metastatic castration-resistant prostate cancer (mCRPC) patients with progressive disease and at least one bone metastasis at the time of inclusion. Unfortunately, no significant differences between the ipilimumab group and the placebo group were found in terms of overall survival.21 Nevertheless, an improved 3-month progression-free survival and a marked reduction in the prostate-specific antigen were observed in the ipilimumab group.21 Even though α-CTLA4 Ab has been shown to stimulate T-cell responses in preclinical or early clinical studies in metastatic prostate or breast cancers,22,23,24,25 the immune activation in this trial needs to be evaluated to understand why it was not fully successful. Nevertheless, this first finding suggests that patients with bone metastases might benefit from T-cell-activating therapies.

Another small study examined the tumor-protective effects of a T-cell adoptive transfer in breast carcinoma patients, some of which presented skeletal metastases at the time of inclusion.26 In this pilot study, memory T cells isolated from the patient bone marrow were reactivated ex vivo with autologous DC pulsed with lysate from the MCF-7 breast cancer cells as a source of tumor antigens and adoptively transferred back into the patients. Regrettably, patients with bone metastases at the time of inclusion were not responsive to the treatment. As a high proportion of immunosuppressive regulatory T cells (Tregs) was found in the T-cell cultures of the non-responding patients, depletion of Tregs before the adoptive transfer should be considered.

Bisphosphonates, such as ZOL, are commonly used in patients with skeletal metastases to protect from SREs because of their OC inhibitory actions. Interestingly, bisphosphonates have also shown the ability to activate γδ T cells. γδ T cells, similar to CD4+ and CD8+ T cells, can exert antitumor effects. This T-cell population is found at tumor sites in vivo and can induce tumor cell cytolysis in vitro.27 Thus, a phase I clinical trial in mCRPC patients with bone metastases has recently tested the immunomodulatory effects of ZOL via stimulation of γδ T cells. Patients were divided into two groups, one receiving ZOL plus interleukin-2 (IL-2) (to further activate T-cell proliferation) and another receiving ZOL alone.28 A favorable clinical response, or at least disease stabilization, was observed in six out of nine patients treated with the combined therapy, and the results correlated with increased peripheral γδ T cells. As patients treated with ZOL alone did not show γδ T-cell expansion, this study supports the need for IL-2 to maintain γδ T-cell activation in vivo.

The efficacy of ZOL in reducing the incidence of bone metastases in early-stage breast cancer patients has been controversial. Meta-analysis studies by Valachis et al.29 did not show significant differences for the disease-free survival outcome or incidence of bone metastases. He et al.,30 found improved bone metastasis-free survival in the ZOL adjuvant therapy group compared with placebo or delayed ZOL administration. The reasons for these differential effects are unknown, and the immune status of these patients has not been evaluated. We have recently shown that, in animals with bone metastases, T-cell deficiency reduces the antitumor effects of ZOL compared with immunocompetent mice.13 Interestingly, the antiresorptive therapy is still able to suppress tumor-induced bone loss, but ZOL loses its efficacy in constraining tumor growth in the bone. This observation has important clinical implications, as it suggests that reduced T-cell numbers or impaired T-cell activation might be the cause for the failure of ZOL to reduce tumor burden and increase survival in breast cancer patients. More studies to evaluate the role of T cells in bone metastatic dissemination using clinically relevant models of bone metastases, such as prostate and breast cancers, need to be performed.

Regulatory T cells and T helper type 17

Among CD4+ T cells, Tregs are known to be potent immune suppressors that protect tissues from autoimmune reactions by suppressing self-reactive cells. Unfortunately, activated Tregs have been observed in cancer patients, including with invasive breast carcinoma, and their presence predicts worse relapse-free survival and decreased overall survival.31,32 The mechanisms underlying immunosuppression include production of TGF-β, IL-35 and IL-10, and also direct T-cell cytolytic effects through the release of perforin and granzyme (reviewed in Linehan and Goedegebuure33). Tregs are also responsible for impaired γδ T-cell proliferation.34 Several studies in animal models of myeloma, leukemia, sarcoma and lung metastases have shown the antitumor effects of Treg depletion using anti-CD25 Ab.35,36 However, opposite results in which Treg depletion showed no beneficial effects were also reported.37,38,39 As suggested by the authors, different animal models and/or cell lines may explain the controversial effects of targeting Tregs in cancer.

In addition to their immunosuppressive capabilities, infiltrating Tregs have also been recently shown to be a major source of RANKL,40 the critical cytokine required for OC differentiation. As RANKL has been reported to regulate cancer cell mobility and bone metastasis,41 it is possible that RANKL+ Tregs could favor skeletal tumor dissemination.

Clinical studies have correlated the beneficial effects of antitumor approaches with a reduction in the Treg population. Low doses of cyclophosphamide, a DNA alkylating agent, have been shown to selectively deplete Tregs and restore T- and natural killer cell effector functions in end-stage cancer patients.42 In another study conducted in metastatic breast cancer, including in patients with skeletal metastases, the low-dose regimen of the alkylating agent induced a 40% reduction in circulating Tregs.43 Despite having only transitory effects on Treg depletion, patients receiving cyclophosphamide showed increased tumor-reactive T cells in blood, and increased overall survival by ∼3 months.43 Despite these exciting results, cyclophosphamide administration has been associated with increased circulating myeloid-derived suppressor cells (MDSCs) in animal models and cancer patients44 that might counterbalance the positive effects of Treg depletion. Thus, further studies are required to fully characterize the antitumor effects of this agent and in particular of Treg depletion.

T helper type 17 (Th17) cells are another subset of CD4+ T cells that might be more harmful than beneficial in the context of bone metastases. These IL-17 producing T lymphocytes were shown to induce osteoclastogenesis and bone damage through RANKL production in the context of inflammatory arthritis.45 To study the link between arthritis-mediated inflammation and secondary metastases from breast cancer, Roy et al.46 injected arthritis-inducing type II collagen in MMTV-PyVMT mice that develop spontaneous metastases to the lung and bone. Interestingly, the treatment with anti-IL-17 Ab in those arthritic mice reduced the percentage of secondary metastases compared with controls.46 Monteiro et al.47 further investigated the role of tumor-specific Th17 cells on OC activation in the context of bone metastases. Tumor-specific Th17 cells promote OC activation and induce osteolytic bone disease via the production of RANKL, and the effects in the bone microenvironment are observed before arrival of tumor cells. Furthermore, tumor-specific RANKL+ Th17 cell adoptive transfer into mice orthotopically injected with 4T1 breast cancer cells increases tumor colonization to bone. Surprisingly, the growth of the tumor at the primary and metastatic sites was not affected by Th17 adoptive transfer.47 Interestingly, although high IL-17 levels are observed in vivo, its blockade does not abolish the pro-osteoclastogenic activity of tumor-specific RANKL+ Th17 cells. Nevertheless, another study indicated that human bone marrow-derived stem cells produce IL-17 and favor skeletal metastatic dissemination of breast cancer cells overexpressing IL-17 receptor (IL-17R).48 Altogether, these data suggest that Th17 cells favor tumor growth in the bone, a process that is likely to be mediated by IL-17 and RANKL production.

Dendritic cells

DCs are important regulators of T-cell activation by virtue of their antigen-presenting capacities (APCs). Thus, DC-based vaccines have been used in various cancers to induce tumor-specific T-cell responses. Sipuleucel-T was the first therapeutic vaccine approved by the FDA to treat mCRPC. Sipuleucel-T consists of the injection of ex vivo processed APCs, including DC, that express a key tumor antigen to stimulate patient's T-cell responses. In a phase III clinical trial, men receiving Sipuleucel-T experienced an overall reduced risk of death and improved overall survival of ∼20 months compared with placebo.49 However, the beneficial antitumor effects of Sipuleucel-T in mCRPC patients older than 65 years remain controversial.50 Results from the ongoing clinical trial combining ipilimumab and Sipuleucel-T will help define new strategies to improve antitumor immunotherapy efficacy.

Unfortunately, like CTLs, DCs can be the target of immunosuppressive effects used by the tumor. Data suggest that tumor-infiltrating DCs remain in an immature state, which suppresses their ability to properly activate T cells.51 Tumor-infiltrating DCs have also been reported to suppress CD8+ T cells through the production of TGF-β, nitric oxide, IL-10, VEGF (vascular endothelial growth factor) and arginase I.52,53,54 Furthermore, the accumulation of DCs with immunosuppressive properties at a tumor site promotes the recruitment of other immunosuppressive populations such as Tregs and MDSCs, thus supporting tumor progression and metastasis.55

In a recent study, Sawant et al.56 reported elevated numbers of plasmacytoid DC (pDCs) within the bone of mice inoculated with 4T1 mammary cancer cells. Depletion of pDCs by PDCA-1 showed reduced lung and bone metastases. Interestingly, accumulation of Tregs and the monocytic fraction of MDSCs within the bone was also reduced by PDCA-1 Ab. As Tregs and MDSCs have been shown to have a critical role in late stages of tumor development, it would be interesting to test the therapeutic effects of the PDCA-1 Ab on established tumors.

Myeloid-derived suppressor cells

MDSCs are a heterogeneous cell population of immature myeloid cells derived from the bone marrow, which potently suppress T-cell-mediated antitumor responses. MDSCs are identified by the coexpression of αm integrin (CD11b) and Gr-1 in mice and CD11b and CD33 in humans.57,58,59,60,61,62,63 Stimulated by tumor, MDSCs leave the bone marrow and are found in high numbers in circulation, spleen and tumor sites.64 Up to a 10-fold increase in MDSC numbers is also detected in the blood of cancer patients.65 Recent evidence indicates a correlation between MDSC numbers, stage of malignancy and poor prognosis, with the highest numbers observed in advanced cancers with worst prognostic outcomes. Mechanistically, MDSCs exert their proneoplastic effects through the release of small soluble oxidizers, impairment of T-cell/antigen recognition and depletion of essential amino acids from the local extracellular environment, all ultimately leading to T-cell suppression.61,62,66,67 In addition, it has been suggested that MDSCs can induce the expansion of Tregs.68 Furthermore, MDSCs can favor tumor proliferation through the overproduction of cytokines and angiogenic factors.69 In a recent phase I study with advanced stage of small-cell lung cancer patients, the addition of all-trans retinoic acid (ATRA) induced a greater immune response to DC-expressing p53 vaccine. As ATRA has been shown to induce MDSC apoptosis, this result suggests that MDSC depletion may benefit patients with metastatic disease. Nevertheless, further studies are required to validate these combined therapies prospectively in patients with bone metastases.

The role of MDSCs in bone metastases has only become apparent in recent years. We found that tumor-bearing PLCγ2−/− mice have an increased percentage of MDSCs in the spleen and bone marrow.12 Interestingly, despite the blockade of OC differentiation and resorption, tumor growth in the bone of PLCγ2−/− mice was significantly higher than that in the WT mice because of the inhibition of antitumor T-cell responses.13 The effects of PLCγ2 in the T-cell population were not intrinsic, as PLCγ2 is not required for T-cell activity. Thus, modulation of T-cell responses in this context is likely due to aberrant expansion/activation of MDSCs. This study indicates that the immune phenotype of PLCγ2−/− animals overrides the requirement for active OC in promoting tumor growth in the bone.13 These data support the assumption that immunosuppressive myeloid populations modulate tumor growth in the bone, independent of the OC status, by affecting T-cell responses.

However, a role for MDSCs in promoting bone metastases through OCs has also been proposed. Sawant et al.,70 using an immunocompetent model of breast cancer bone metastases, showed that MDSCs isolated from the tumor–bone microenvironment differentiate into resorbing OCs in vitro. Remarkably, OC differentiation does not occur if MDSCs are isolated from tumor-free mice or tumor-bearing animals without bone metastases.70 Similarly, in the context of multiple myeloma (MM), Zhuang et al.71 discovered that MDSCs from tumor-bearing mice have increased osteoclastogenic potential. Importantly, coinjection of tumor-challenged MDSCs together with MM cells leads to increased tumor burden and osteolytic lesions, an effect that is inhibited by administration of ZOL. Despite the evidence that MDSCs can become OCs in vitro or can induce OC activation in vivo when adoptively transferred into tumor-bearing mice, the study with PLCγ2−/− mice, which have intrinsic OC defects, suggests that MDSCs can enhance tumor growth in the bone independent of their ability to differentiate into OCs.12

Macrophages

Macrophages are mature tissue-resident myeloid cells72 that originate from circulating bone marrow-derived monocytic precursors, suggested to be a subset of MDSCs.57,66,73 In recent years, macrophages have been divided into two major subsets (proinflammatory M1 and anti-inflammatory M2) with many other subsets in between. M1 macrophages detect endogenous danger signals present in the debris of necrotic cells through Toll-like receptors 2, 5 and 6, intracellular pattern recognition receptors and the IL-1R. Once activated, M1 macrophages produce high levels of proinflammatory cytokines such as IL-1, IL-6, IL-12, IL-23 and IFN-γ,72,74 and participate in the elimination of tumor cells.75 Distinct from M1, M2 macrophages, also called tumor-associated macrophages (TAMs), have a negative prognostic value for the overall survival in patients with breast, gastric, ovarian and thyroid cancers.76 Tumor-derived factors such as IL-4, IL-10, IL-13, TGF-β and prostaglandin 2 promulgate an anti-inflammatory macrophage polarization associated with tumor progression. M2 macrophages are characterized by high expression of IL-1Rα (IL-1 decoy receptor), mannose receptors, scavenger receptors and elevated CCL17 and CCL22 secretion. Activated M2 macrophages also produce high levels of IL-10 and TGF-β, which alter the activation of CD4+ and CD8+ T cells.77 Furthermore, their ability to express VEGF, matrix metalloproteinase 9 (MMP9) and other proangiogenic factors has been associated with their positive role in metastatic dissemination.78,79 Thus, depleting agents targeting the infiltrating macrophage population are under intensive investigation for their antitumor effects.

Recent studies using clodronate-coated liposomes, which target phagocytic cells including macrophages, showed reduced incidence of bone metastases, as well as a number of metastatic lesions in the hindlimbs of nude mice injected with human lung cancer cells.80 In this study, macrophages and OCs were reduced at the tumor sites. The anti-mouse CD115 mAb (CSF1R antagonist) reduces the recruitment of TAMs to the primary sites in the MMTV-PyMT mammary tumor model81 and decreases the osteolytic bone lesions in nude mice injected intracardiacally with breast tumor MDA-MB-231 cells.82 Altogether, these studies suggest a role for macrophages in supporting tumor growth in bone. Nevertheless, so far there is no direct evidence suggesting that the antitumor effects of targeting TAMs in the context of bone metastases are not simply because of the inhibition of OC differentiation.

Neutrophils

Polymorphonuclear neutrophils (PMNs) are the predominant leukocyte subset in human peripheral blood. They are released from the bone marrow to the blood as mature cells. PMNs have been extensively studied for their proinflammatory role in host defense against microorganisms. Based on similarities in inflammatory reactions between cancer and infection, PMNs have recently emerged as new infiltrating myeloid cells in the tumor microenvironment. Indeed, PMNs represent a significant proportion of the inflammatory cell infiltrate in human cancers, leading to the term tumor-associated neutrophils or TANs.83,84,85 Some in vitro studies reported that PMNs may display antitumor effects by inducing tumor cell lysis via Ab-dependent cell-mediated cytotoxicity.86 However, more recent studies in humans suggest the involvement of TANs in tumor progression rather than in antitumor responses.83,84,85 Several groups proposed the neutrophil-to-lymphocyte ratio as a useful prognostic biomarker for predicting overall survival in metastatic colorectal cancer87,88 and advanced gastric cancer.89 In addition, neutrophils are able to release VEGF and MMP9, two factors involved in the metastatic process.90 Considering the abundance of neutrophils in bone marrow, their involvement in favoring tumor growth in bone requires further investigations.

Conclusion

Unfortunately, there is no curative treatment for bone metastasis. Tumor cells that reach the bone environment are usually resistant to current antitumor therapeutic approaches. The only options for these patients are palliative treatments to reduce pain and prevent additional bone destruction.

The presence of CD8+ tumor-infiltrating lymphocytes in primary breast tumors correlates with reduced metastatic invasion and increased overall survival.91 In this review, we describe several strategies to stimulate therapeutic antitumor cytotoxic T-cell responses. We also discuss that immunosuppressive cells are major obstacles in the development of active immunotherapy for cancer patients. Unfortunately, because of the limited number of orthotopic bone metastatic tumor models available in immunocompetent mice, the role of immunosurveillance in bone metastasis is understudied. There are still major holes in the relevance of specific immune cell types regulating metastasis to bone, especially in breast or prostate cancer. Additional studies are needed to further elucidate the key immunosuppressive mechanisms at play and to identify possible biomarkers that would predict responsiveness to therapy. Completion of this task could lead to the design of appropriate clinical trials based on the identification of immunosuppressive/promoting phenotypes in a particular cancer type.

Acknowledgments

We thank the Washington University Muskuloskeletal Research Center and the Washington University Bright Institute and Molecular Imaging Center (P50 CA94056ADD). This work was supported by a National Institutes of Health (NIH) grant (R01 AR52921 to RF), Shriners Hospital Fund and Bright Institute Pilot Research grant (to RF).

Footnotes

The authors declare no conflict of interest.

References

  1. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 2011;331:1565–1570. [DOI] [PubMed] [Google Scholar]
  2. Hess KR, Varadhachary GR, Taylor SH, Wei W, Raber MN, Lenzi R et al. Metastatic patterns in adenocarcinoma. Cancer 2006;106:1624–1633. [DOI] [PubMed] [Google Scholar]
  3. Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer 2002;2:584–593. [DOI] [PubMed] [Google Scholar]
  4. Roodman GD. Mechanisms of bone metastasis. Discov Med 2004;4:144–148. [PubMed] [Google Scholar]
  5. Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal attraction. Nat Rev Cancer 2011;11:411–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Clemons M, Gelmon KA, Pritchard KI, Paterson AH. Bone-targeted agents and skeletal-related events in breast cancer patients with bone metastases: the state of the art. Curr Oncol 2012;19:259–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Kakonen SM, Selander KS, Chirgwin JM, Yin JJ, Burns S, Rankin WA et al. Transforming growth factor-beta stimulates parathyroid hormone-related protein and osteolytic metastases via Smad and mitogen-activated protein kinase signaling pathways. J Biol Chem 2002;277:24571–24578. [DOI] [PubMed] [Google Scholar]
  8. Clines GA, Guise TA. Molecular mechanisms and treatment of bone metastasis. Expert Rev Mol Med 2008;10:e7. [DOI] [PubMed] [Google Scholar]
  9. Guise TA. Molecular mechanisms of osteolytic bone metastases. Cancer 2000;88(Suppl): 2892–2898. [DOI] [PubMed] [Google Scholar]
  10. Coleman R, Cameron D, Dodwell D, Bell R, Wilson C, Rathbone E et al. Adjuvant zoledronic acid in patients with early breast cancer: final efficacy analysis of the AZURE (BIG 01/04) randomised open-label phase 3 trial. Lancet Oncol 2014;15:997–1006. [DOI] [PubMed] [Google Scholar]
  11. Gnant M, Mlineritsch B, Stoeger H, Luschin-Ebengreuth G, Heck D, Menzel C et al. Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 62-month follow-up from the ABCSG-12 randomised trial. Lancet Oncol 2011;12:631–641. [DOI] [PubMed] [Google Scholar]
  12. Capietto AH, Kim S, Sanford DE, Linehan DC, Hikida M, Kumosaki T et al. Down-regulation of PLCgamma2-beta-catenin pathway promotes activation and expansion of myeloid-derived suppressor cells in cancer. J Exp Med 2013;210:2257–2271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Zhang K, Kim S, Cremasco V, Hirbe AC, Collins L, Piwnica-Worms D et al. CD8+ T cells regulate bone tumor burden independent of osteoclast resorption. Cancer Res 2011;71:4799–4808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bidwell BN, Slaney CY, Withana NP, Forster S, Cao Y, Loi S et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat Med 2012;18:1224–1231. [DOI] [PubMed] [Google Scholar]
  15. Sallusto F, Lanzavecchia A. Understanding dendritic cell and T-lymphocyte traffic through the analysis of chemokine receptor expression. Immunol Rev 2000;177:134–140. [DOI] [PubMed] [Google Scholar]
  16. Huang AY, Golumbek P, Ahmadzadeh M, Jaffee E, Pardoll D, Levitsky H. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science 1994;264:961–965. [DOI] [PubMed] [Google Scholar]
  17. Feuerer M, Rocha M, Bai L, Umansky V, Solomayer EF, Bastert G et al. Enrichment of memory T cells and other profound immunological changes in the bone marrow from untreated breast cancer patients. Int J Cancer 2001;92:96–105. [PubMed] [Google Scholar]
  18. Pantel K, Brakenhoff RH, Brandt B. Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat Rev Cancer 2008;8:329–340. [DOI] [PubMed] [Google Scholar]
  19. Siddappa CM, Watson MA, Pillai SG, Trinkaus K, Fleming T, Aft R. Detection of disseminated tumor cells in the bone marrow of breast cancer patients using multiplex gene expression measurements identifies new therapeutic targets in patients at high risk for the development of metastatic disease. Breast Cancer Res Treat 2013;137:45–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Xu Z, Hurchla MA, Deng H, Uluckan O, Bu F, Berdy A et al. Interferon-gamma targets cancer cells and osteoclasts to prevent tumor-associated bone loss and bone metastases. J Biol Chem 2009;284:4658–4666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJ et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Kwon ED, Foster BA, Hurwitz AA, Madias C, Allison JP, Greenberg NM et al. Elimination of residual metastatic prostate cancer after surgery and adjunctive cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade immunotherapy. Proc Natl Acad Sci USA 1999;96:15074–15079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. van den Eertwegh AJ, Versluis J, van den Berg HP, Santegoets SJ, van Moorselaar RJ, van der Sluis TM et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012;13:509–517. [DOI] [PubMed] [Google Scholar]
  24. Allard B, Pommey S, Smyth MJ, Stagg J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res 2013;19:5626–5635. [DOI] [PubMed] [Google Scholar]
  25. Vonderheide RH, LoRusso PM, Khalil M, Gartner EM, Khaira D, Soulieres D et al. Tremelimumab in combination with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin Cancer Res 2010;16:3485–3494. [DOI] [PubMed] [Google Scholar]
  26. Schuetz F, Ehlert K, Ge Y, Schneeweiss A, Rom J, Inzkirweli N et al. Treatment of advanced metastasized breast cancer with bone marrow-derived tumour-reactive memory T cells: a pilot clinical study. Cancer Immunol Immunother 2009;58:887–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Capietto AH, Martinet L, Fournie JJ. Stimulated gammadelta T cells increase the in vivo efficacy of trastuzumab in HER-2+ breast cancer. J Immunol 2011;187:1031–1038. [DOI] [PubMed] [Google Scholar]
  28. Dieli F, Vermijlen D, Fulfaro F, Caccamo N, Meraviglia S, Cicero G et al. Targeting human {gamma}delta} T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res 2007;67:7450–7457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Valachis A, Polyzos NP, Coleman RE, Gnant M, Eidtmann H, Brufsky AM et al. Adjuvant therapy with zoledronic acid in patients with breast cancer: a systematic review and meta-analysis. Oncologist 2013;18:353–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. He M, Fan W, Zhang X. Adjuvant zoledronic acid therapy for patients with early stage breast cancer: an updated systematic review and meta-analysis. J Hematol Oncol 2013;6:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. 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;24:5373–5380. [DOI] [PubMed] [Google Scholar]
  32. Wolf AM, Wolf D, Steurer M, Gastl G, Gunsilius E, Grubeck-Loebenstein B. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res 2003;9:606–612. [PubMed] [Google Scholar]
  33. Linehan DC, Goedegebuure PS. CD25+ CD4+ regulatory T-cells in cancer. Immunol Res 2005;32:155–168. [DOI] [PubMed] [Google Scholar]
  34. Kunzmann V, Kimmel B, Herrmann T, Einsele H, Wilhelm M. Inhibition of phosphoantigen-mediated gammadelta T-cell proliferation by CD4+ CD25+ FoxP3+ regulatory T cells. Immunology 2009;126:256–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Olkhanud PB, Baatar D, Bodogai M, Hakim F, Gress R, Anderson RL et al. Breast cancer lung metastasis requires expression of chemokine receptor CCR4 and regulatory T cells. Cancer Res 2009;69:5996–6004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Onizuka S, Tawara I, Shimizu J, Sakaguchi S, Fujita T, Nakayama E. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor alpha) monoclonal antibody. Cancer Res 1999;59:3128–3133. [PubMed] [Google Scholar]
  37. Hong H, Gu Y, Zhang H, Simon AK, Chen X, Wu C et al. Depletion of CD4+CD25+ regulatory T cells enhances natural killer T cell-mediated anti-tumour immunity in a murine mammary breast cancer model. Clin Exp Immunol 2010;159:93–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Ostrand-Rosenberg S, Clements VK, Terabe M, Park JM, Berzofsky JA, Dissanayake SK. Resistance to metastatic disease in STAT6-deficient mice requires hemopoietic and nonhemopoietic cells and is IFN-gamma dependent. J Immunol 2002;169:5796–5804. [DOI] [PubMed] [Google Scholar]
  39. Terabe M, Swann J, Ambrosino E, Sinha P, Takaku S, Hayakawa Y et al. A nonclassical non-Valpha14Jalpha18 CD1d-restricted (type II) NKT cell is sufficient for down-regulation of tumor immunosurveillance. J Exp Med 2005;202:1627–1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, Hoffman RM et al. Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL–RANK signalling. Nature 2011;470:548–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Jones DH, Nakashima T, Sanchez OH, Kozieradzki I, Komarova SV, Sarosi I et al. Regulation of cancer cell migration and bone metastasis by RANKL. Nature 2006;440:692–696. [DOI] [PubMed] [Google Scholar]
  42. Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother 2007;56:641–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ge Y, Domschke C, Stoiber N, Schott S, Heil J, Rom J et al. Metronomic cyclophosphamide treatment in metastasized breast cancer patients: immunological effects and clinical outcome. Cancer Immunol Immunother 2012;61:353–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. 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;58:49–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Okamoto K, Takayanagi H. Regulation of bone by the adaptive immune system in arthritis. Arthritis Res Ther 2011;13:219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Roy LD, Ghosh S, Pathangey LB, Tinder TL, Gruber HE, Mukherjee P. Collagen induced arthritis increases secondary metastasis in MMTV-PyV MT mouse model of mammary cancer. BMC Cancer 2011;11:365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Monteiro AC, Leal AC, Goncalves-Silva T, Mercadante AC, Kestelman F, Chaves SB et al. T cells induce pre-metastatic osteolytic disease and help bone metastases establishment in a mouse model of metastatic breast cancer. PLoS One 2013;8:e68171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Goldstein RH, Reagan MR, Anderson K, Kaplan DL, Rosenblatt M. Human bone marrow-derived MSCs can home to orthotopic breast cancer tumors and promote bone metastasis. Cancer Res 2010;70:10044–10050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–422. [DOI] [PubMed] [Google Scholar]
  50. Huber ML, Haynes L, Parker C, Iversen P. Interdisciplinary critique of sipuleucel-T as immunotherapy in castration-resistant prostate cancer. J Natl Cancer Inst 2012;104:273–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gabrilovich DI, Corak J, Ciernik IF, Kavanaugh D, Carbone DP. Decreased antigen presentation by dendritic cells in patients with breast cancer. Clin Cancer Res 1997;3:483–490. [PubMed] [Google Scholar]
  52. Dumitriu IE, Dunbar DR, Howie SE, Sethi T, Gregory CD. Human dendritic cells produce TGF-beta 1 under the influence of lung carcinoma cells and prime the differentiation of CD4+CD25+Foxp3+ regulatory T cells. J Immunol 2009;182:2795–2807. [DOI] [PubMed] [Google Scholar]
  53. Norian LA, Rodriguez PC, O'Mara LA, Zabaleta J, Ochoa AC, Cella M et al. Tumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolism. Cancer Res 2009;69:3086–3094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Liu Q, Zhang C, Sun A, Zheng Y, Wang L, Cao X. Tumor-educated CD11bhighIalow regulatory dendritic cells suppress T cell response through arginase I. J Immunol 2009;182:6207–6216. [DOI] [PubMed] [Google Scholar]
  55. Shurin MR, Naiditch H, Zhong H, Shurin GV. Regulatory dendritic cells: new targets for cancer immunotherapy. Cancer Biol Ther 2011;11:988–992. [DOI] [PubMed] [Google Scholar]
  56. Sawant A, Hensel JA, Chanda D, Harris BA, Siegal GP, Maheshwari A et al. Depletion of plasmacytoid dendritic cells inhibits tumor growth and prevents bone metastasis of breast cancer cells. J Immunol 2012;189:4258–4265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Bronte V, Apolloni E, Cabrelle A, Ronca R, Serafini P, Zamboni P et al. Identification of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progenitor capable of activating or suppressing CD8(+) T cells. Blood 2000;96:3838–3846. [PMC free article] [PubMed] [Google Scholar]
  58. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol 2001;166:5398–5406. [DOI] [PubMed] [Google Scholar]
  59. Gallina G, Dolcetti L, Serafini P, De Santo C, Marigo I, Colombo MP et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest 2006;116:2777–2790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kusmartsev S, Nagaraj S, Gabrilovich DI. Tumor-associated CD8+ T cell tolerance induced by bone marrow-derived immature myeloid cells. J Immunol 2005;175:4583–4592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol 2004;172:989–999. [DOI] [PubMed] [Google Scholar]
  62. Liu Y, Van Ginderachter JA, Brys L, De Baetselier P, Raes G, Geldhof AB. Nitric oxide-independent CTL suppression during tumor progression: association with arginase-producing (M2) myeloid cells. J Immunol 2003;170:5064–5074. [DOI] [PubMed] [Google Scholar]
  63. Zea AH, Rodriguez PC, Atkins MB, Hernandez C, Signoretti S, Zabaleta J et al. Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res 2005;65:3044–3048. [DOI] [PubMed] [Google Scholar]
  64. Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L et al. Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity 2010;32:790–802. [DOI] [PubMed] [Google Scholar]
  65. Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol 2001;166:678–689. [DOI] [PubMed] [Google Scholar]
  66. Kusmartsev S, Gabrilovich DI. Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J Leukocyte Biol 2003;74:186–196. [DOI] [PubMed] [Google Scholar]
  67. Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E, Serafini P et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol 2002;168:689–695. [DOI] [PubMed] [Google Scholar]
  68. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009;9:162–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kusmartsev S, Gabrilovich DI. Role of immature myeloid cells in mechanisms of immune evasion in cancer. Cancer Immunol Immunother 2006;55:237–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Sawant A, Deshane J, Jules J, Lee CM, Harris BA, Feng X et al. Myeloid-derived suppressor cells function as novel osteoclast progenitors enhancing bone loss in breast cancer. Cancer Res 2013;73:672–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Zhuang J, Zhang J, Lwin ST, Edwards JR, Edwards CM, Mundy GR et al. Osteoclasts in multiple myeloma are derived from Gr-1+CD11b+myeloid-derived suppressor cells. PLoS ONE 2012;7:e48871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008;8:958–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Li Q, Pan PY, Gu P, Xu D, Chen SH. Role of immature myeloid Gr-1+ cells in the development of antitumor immunity. Cancer Res 2004;64:1130–1139. [DOI] [PubMed] [Google Scholar]
  74. Mantovani A, Sica A. Macrophages, innate immunity and cancer: balance, tolerance, and diversity. Curr Opin Immunol 2010;22:231–237. [DOI] [PubMed] [Google Scholar]
  75. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012;12:253–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Zhang QW, Liu L, Gong CY, Shi HS, Zeng YH, Wang XZ et al. Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS ONE 2012;7:e50946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 2010;11:889–896. [DOI] [PubMed] [Google Scholar]
  78. Coffelt SB, Hughes R, Lewis CE. Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta 2009;1796:11–18. [DOI] [PubMed] [Google Scholar]
  79. Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res 2006;66:11238–11246. [DOI] [PubMed] [Google Scholar]
  80. Hiraoka K, Zenmyo M, Watari K, Iguchi H, Fotovati A, Kimura YN et al. Inhibition of bone and muscle metastases of lung cancer cells by a decrease in the number of monocytes/macrophages. Cancer Sci 2008;99:1595–1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. 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;1:54–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. 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:e73310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Bellocq A, Antoine M, Flahault A, Philippe C, Crestani B, Bernaudin JF et al. Neutrophil alveolitis in bronchioloalveolar carcinoma: induction by tumor-derived interleukin-8 and relation to clinical outcome. Am J Pathol 1998;152:83–92. [PMC free article] [PubMed] [Google Scholar]
  84. Jensen HK, Donskov F, Marcussen N, Nordsmark M, Lundbeck F, von der Maase H. Presence of intratumoral neutrophils is an independent prognostic factor in localized renal cell carcinoma. J Clin Oncol 2009;27:4709–4717. [DOI] [PubMed] [Google Scholar]
  85. Wislez M, Rabbe N, Marchal J, Milleron B, Crestani B, Mayaud C et al. Hepatocyte growth factor production by neutrophils infiltrating bronchioloalveolar subtype pulmonary adenocarcinoma: role in tumor progression and death. Cancer Res 2003;63:1405–1412. [PubMed] [Google Scholar]
  86. Di Carlo E, Forni G, Lollini P, Colombo MP, Modesti A, Musiani P. The intriguing role of polymorphonuclear neutrophils in antitumor reactions. Blood 2001;97:339–345. [DOI] [PubMed] [Google Scholar]
  87. Chua W, Charles KA, Baracos VE, Clarke SJ. Neutrophil/lymphocyte ratio predicts chemotherapy outcomes in patients with advanced colorectal cancer. Br J Cancer 2011;104:1288–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Kishi Y, Kopetz S, Chun YS, Palavecino M, Abdalla EK, Vauthey JN. Blood neutrophil-to-lymphocyte ratio predicts survival in patients with colorectal liver metastases treated with systemic chemotherapy. Ann Surg Oncol 2009;16:614–622. [DOI] [PubMed] [Google Scholar]
  89. Lee S, Oh SY, Kim SH, Lee JH, Kim MC, Kim KH et al. Prognostic significance of neutrophil lymphocyte ratio and platelet lymphocyte ratio in advanced gastric cancer patients treated with FOLFOX chemotherapy. BMC Cancer 2013;13:350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. van Kempen LC, Coussens LM. MMP9 potentiates pulmonary metastasis formation. Cancer Cell 2002;2:251–252. [DOI] [PubMed] [Google Scholar]
  91. Pages F, Galon J, Dieu-Nosjean MC, Tartour E, Sautes-Fridman C, Fridman WH. Immune infiltration in human tumors: a prognostic factor that should not be ignored. Oncogene 2010;29:1093–1102. [DOI] [PubMed] [Google Scholar]

Articles from BoneKEy Reports are provided here courtesy of Nature Publishing Group

RESOURCES