Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Feb 1.
Published in final edited form as: Pharmacol Ther. 2013 Oct 15;141(2):222–233. doi: 10.1016/j.pharmthera.2013.10.006

Targeting tumor-stromal interactions in bone metastasis

Mark Esposito 1, Yibin Kang 1
PMCID: PMC3947254  NIHMSID: NIHMS532250  PMID: 24140083

Abstract

Bone metastasis is a frequent occurrence in late stage solid tumors, including breast cancers, prostate or lung. However, the causes for this proclivity have only recently been elucidated. Significant progress has been made in the past decade toward understanding the molecular underpinnings of bone metastasis, and much of this research reveals a crucial role of the host stroma in each step of the metastatic cascade. Tumor-stromal interactions are crucial in engineering a pre-metastatic niche, accommodating metastatic seeding, and establishing the vicious cycle of bone metastasis. Current treatments in bone metastasis focus on latter steps of the metastatic cascade, with most treatments targeting the process of bone remodeling; however, emerging research identifies many other candidates as promising targets. Host stromal cells including platelets and endothelial cells are important in the early steps of metastatic homing, attachment and extravasation while a variety of immune cells, parenchymal cells and mesenchymal cells of the bone marrow are important in the establishment of overt, immune-suppressed metastatic lesions. Many participants during these steps have been identified and functionally validated. Significant contributors include integrins, (αvβ3, α2β1, α4β1), TGFβ family members, bone resident proteins (BSP, OPG, SPARC, OPN), RANKL, and PTHrP. In this review, we will discuss the contribution of host stromal cells to pre-metastatic niche conditioning, seeding, dormancy, bone-remodeling, immune regulation, and chemotherapeutic shielding in bone metastasis. Research exploring these interactions between bone metastases and stromal cells has yielded many therapeutic targets, and we will discuss both the current and future therapeutic avenues in treating bone metastasis.

Keywords: Bone metastasis, metastatic niche, tumor dormancy, osteoclast inhibitors, tumor-stromal interactions, Immune surveillance

1. Introduction

Metastasis to the bone is one of the most common and devastating complications in patients with advanced cancers of the breast, prostate or lung. Also manifest in other cancers (thyroid, renal cell, colon, esophageal or rectum), bone metastasis is a pathological process notable for the ability of tumor cells to exploit endogenous stromal environments and coerce other host cell types into cooperation. Despite gross morphological differences between the bone and the soft tissues from which bone metastases originate, the underlying molecular interactions between disseminated tumor cells (DTCs) and bone tissues make bone a particularly attractive niche for the growth of metastatic lesions. Lending credence to this idea, many of the genes associated with breast cancer metastasis to bone are surface interaction proteins or secreted growth factors, demonstrating that mechanisms extrinsic to the tumor cells are paramount to metastatic progression (Kang, et al., 2003). From instructing the pre-metastatic niche to establishing a vicious cycle of bone remodeling and tumor growth, tumor-stromal interactions are crucial to metastatic expansion in bone. Although this tumor-stromal relationship endows resistance to many conventional therapeutic approaches, exploiting the crosstalk between tumor cells and the bone stromal compartment may provide an effective means to thwart cancer metastasis to bone.

Myriad evidence has emerged within the last decade which indicates the need to target early tumor-stromal interactions (pre-metastatic niche conditioning, seeding and dormancy) to best treat bone metastasis. Current treatments target overt, established metastases and the symptoms associated with increased bone remodeling (Ell & Kang, 2012; Roodman, 2004; Weilbaecher, Guise, & McCauley, 2011). Meanwhile, recent research implicates multiple novel therapeutic opportunities within the priming of the pre-metastatic niche, metastatic seeding, micrometastatic dormancy and immune surveillance (Catena, et al., 2010; Sipkins, et al., 2005) (Table 1). Knowledge regarding the later steps of metastasis, such as bone-remodeling and the establishment of an immune-suppressive environment, has also advanced in recent years and future treatments may strive to transform overt metastasis into a chronic, treatable condition. Furthermore, select patients are receptive to current immune-modulatory therapies, yet the factors that govern a positive response are unknown and require further research to elucidate.

Table 1.

Current and potential therapeutics targeting tumor-stromal interactions in bone metastasis

Therapeutic Target Mechanism Agent Reference
FDA-approved therapies
Prenylation Osteolysis inhibitor Zoledronic Acid Rosen, et al., 2001
RANKL Osteoclastogenesis inhibitor Denosumab Fizazi, et al., 2009
CTLA4 Cytotoxic T cell activator Ipilimumab Hodi, et al., 2010
Cytotoxic cells NK cell activator Interleukin 2 Rosenberg, et al., 2004
Potential therapies
EGFR Osteoclastogenesis inhibitor Gefitinib Normanno, et al., 2005
PD1 Immunosuppression antagonist CT-011 Dulos, et al., 2012
CD137 Immunosuppression antagonist BMS-663513 Simeone & Ascierto, 2005
Gal3 CTC Attachment ligand mimic Lactulose-L-Leucine Heimburg, et al., 2006
c-MET Pre-metastatic conditioning Inhibitor Tivantinib Previdi, et al., 2011
CXCR4 Homing and dormancy inhibitor AMD3100 Shiozawa, et al., 2011
CCL2 Homing and growth antagonist Carlumab Loberg, et al., 2008
ET1 Osteogenesis antagonist Atrasentan Yin, et al., 2003
Metastatic cells Suppressive pathway activator Interferon 7 Bidwell, et al., 2012
PSA+ cells PSA based vaccine PROSTVAC-VF Kantoff, et al., 2010
VWF Platelet shielding inhibitor ARC1779 Karpatkin, et al., 1988
β3 integrin Seeding and growth antagonist RGD-mimetic/ mAb Matsuura, et al., 1996
Cathepsin B Osteolysis inhibitor CA-074 Withana, et al., 2012
c-FMS Osteoclastogenesis inhibitor Ki-20227 Ohno, et al., 2006
SRC Proliferation inhibior Dasatinib (Zhang, et al., 2009)
Cathepsin K Osteolysis inhibitor Odanacatib Le Gall, et al., 2007
TGFβ Osteoclastogenesis inhibitor Ki26894, SD-208, LY2109761 (Ehata, et al., 2007;Korpal, et al., 2009; Mohammad, et al., 2011)

2. Educating the bone: forming the pre-metastatic niche

The discovery that primary tumor cells are able to instruct the adaptation of foreign sites for future colonization represents a paradigmatic shift in cancer research. Rather than a stochastic process through which a certain proportion of CTCs are able to colonize sites of distant metastasis, the ability of the primary tumor to influence future routes of metastasis both supports Stephen Paget’s well established seed and soil hypothesis (Paget, 1989), and presents new opportunities for therapeutic intervention. Considerable evidence points to the formation of perivascular pre-metastatic niches in the lung by bone-derived cells such as Toll-like receptor 4+ (TLR4+) myeloid cells or vascular endothelial growth factor receptor-1+ (VEGFR+) hematopoetic progenitor cells (Hiratsuka, et al., 2008; Kaplan, et al., 2005). Similar mechanisms are observed in liver metastasis as cytokine secretions from metastatic cells rapidly upregulate intercellular adhesion molecules in the liver and allow for enhanced adherence (Khatib, et al., 1999). Evidence also exists for the presence of pre-metastatic conditioning in bone marrow (Kelly, et al., 2005; Peinado, et al., 2012), however the results of many studies suggest that the bone already possesses many features ideal for fostering metastatic colonization.

Peinado and colleagues recently demonstrated the ability of tumor-derived exosomes, or small lipid coated vesicles containing tumor-derived proteins, to educate bone marrow derived cells, thus facilitating metastasis to the bone or lung in a melanoma model (Peinado, et al., 2012). This conditioning was attributed to increased Met signaling in bone marrow stromal cells. Likewise, inhibition of the hepatocyte growth factor (HGF) receptor (c-MET) interaction has shown promise in weakening the migratory phenotype of breast cancer metastatic cells, and in vivo administration of Tivantinib is able to significantly delay bone metastatic progression (Previdi, Abbadessa, Dalo, France, & Broggini, 2011). Changes in bone marrow structural components has also been observed (Figure 1), as Heparanase (HPSE) secreted by primary tumors increases bone degradation in the absence of metastatic lesions (Kelly, et al., 2005).

Figure 1. Primary tumor-derived factors predispose the bone stroma for colonization.

Figure 1

Tumor secretion of enzymes, growth factors and cytokines alters the distant bone environment to accommodate metastatic seeding. Tumor-derived exosomes enhance MET signaling in bone marrow-derived cells while unknown factors inhibit osteoblast differentiation by upregulating DKK1 plasma levels. Proteases such as HPSE alter the bone stroma by digesting extracellular matrix proteins.

Understanding the role of mesenchymal stem cells (MSCs) in tumor-stromal interaction has become an important field of study (Koh & Kang, 2012). Patients with advanced lung or breast cancer, but without bone metastasis, exhibit changes in MSC plasticity which predisposes the bone toward enhanced osteolysis (Fernandez Vallone, et al., 2013). This predisposition was accompanied by altered serum levels of Dickkopf 1 (DKK1), an inhibitor of osteoblast differentiation, reflecting perturbations in bone marrow homeostasis prior to metastatic seeding (Fernandez Vallone, et al., 2013).

Contrary to the hypothesis of pre-metastatic conditioning, the existence of sites permissive for tumor engraftment in healthy mice has also been established. Work by Sipkins et al utilized in vivo imaging to show that both leukemic cells and hematopoetic stem cells (HSCs) home to discrete bone marrow sites expressing stromal-derived factor-1 (SDF1/ CXCL12) and E-selectin (SELE) (Sipkins, et al., 2005). Cell homing and attachment was reduced by 80% upon ablation of SDF1 and 20% upon loss of SELE. Implicated elsewhere in metastasis, cell adhesion molecules VCAM1, ICAM1 and PECAM-1 were not directly associated with metastatic seeding. This work demonstrates that bone marrow may not be conditioned as extensively as in pulmonary metastasis, and rather that the same mechanisms that govern HSC homing in healthy individuals are co-opted by tumor cells.

3. Metastatic seeding: survival in circulation, homing, and attaching to bone parenchyma

Metastatic cells are especially vulnerable during transit from the primary tumor to distant metastatic sites. Selective pressures placed on metastatic cells during seeding and extravasation results in an high attrition rate—only an estimated 0.2% of experimentally introduced circulating tumor cells (CTC) successfully accomplish distant colonization (Chambers, Groom, & MacDonald, 2002). During the traverse from primary tumor to bone marrow, circulating tumor cells (CTCs) must both evade immune surveillance and breach the normal vascular endothelium. This process is accomplished by co-opting circulating platelets and leukocytes, avoiding recognition by immune cells, and deploying immune-like strategies to attach and extravasate into foreign sites (Figure 2).

Figure 2. Survival, seeding and arrest are accomplished by co-opting immune cells and mimicking immune-based strategies of intravasation.

Figure 2

Circulating tumor cell survival is enhanced by platelet secretion of TGFβ and formation of platelet aggregates. In a multistep cascade, metastatic cells home to the bone upon exposure to bone endogenous proteins such as SDF1, SPARC and CCL2. Initial attachment to the endothelium is then mediated by lectin-family proteins, leading to integrin clustering. Engagement of integrins to bone resident proteins enhances both migration and survival, allowing for trans-endothelial extravasation and colonization.

Survival

Survival in circulation is attributed to a combination of cell intrinsic programs, such as decoupling of the anoikis pathway (Demers, et al., 2009), and interactions with circulating stromal cells, including platelets and natural killer (NK) cells. The role of platelet adherence in metastasis was recognized early (Gasic, Gasic, & Stewart, 1968). Multiple genetic deficiency models have identified crucial molecular mediators of CTC interaction with platelets; these interactions are largely dependent on integrin, membrane glycoprotein complex Gp11b-Gp111a, and platelet selectin (SELP) engagement to ligands on metastatic cells. Bone metastasis in β3 integrin-null mice exhibited a 95% decrease in bone tumor burden (Bakewell, et al., 2003), and antibody blocking of the β3 ligand Fibronectin (FN) demonstrated similar reductions. Antibodies to mouse von Willebrand factor (VWF), the ligand for the Gp11b-Gp111a complex, demonstrate a 50–75% reduction in metastatic tumor burden that can be reconstituted through infusion with human platelets (Karpatkin, Pearlstein, Ambrogio, & Coller, 1988). Selp-deficient mice also show attenuated metastatic progression, a result which the authors attribute to reduced platelet shielding (Y. J. Kim, Borsig, Varki, & Varki, 1998).

Surface shielding of CTCs from NK cell-mediated lysis is thought to be the major functional role for platelets during metastasis, however recent studies have begun to challenge this notion. While metastatic cells sensitive to NK cell-mediated lysis are more efficient in seeding when bound to platelets (Nieswandt, Hafner, Echtenacher, & Mannel, 1999; Palumbo, et al., 2007; Palumbo, et al., 2005), models of breast cancer and melanoma show platelets enhance metastasis independently of NK cells (Coupland, Chong, & Parish, 2012). P2Y2 receptor is an endothelial receptor activated by platelet-derived ATP which in turn facilitates trans-endothelial migration of CTCs (Schumacher, Strilic, Sivaraj, Wettschureck, & Offermanns, 2013). Perhaps most significantly, platelets in contact with CTCs during transit are sources of both TGFβ1 and activators of the NF-κB pathway. These factors are responsible for a transition to a mesenchymal morphology and enhanced metastasis (Labelle, Begum, & Hynes, 2011).

Homing

Multiple cytokines are responsible for increased chemotaxis and homing to the bone marrow (Figure 1). The most strongly associated protein receptor, CXC chemokine receptor 4 (CXCR4), provokes actin polymerization and pseudopodia formation resulting in migration upon exposure to bone endothelial cell secretions (A. Muller, et al., 2001). CXCR4 is normally expressed on HSCs, and this expression is important for guiding marrow engraftment of HSCs to organs with high levels of the CXCR4 ligand, SDF1 (Brenner, et al., 2004). In a functional screen for bone metastatic variants, CXCR4 expression was strongly selected for in bone metastatic cells (Kang, et al., 2003), and synthetic peptide inhibition of CXCR4 binding is able to reduce breast cancer metastatic burden (Liang, et al., 2004). Another ligand in the C-C class of chemokines, Chemokine C-C motif ligand 2 (CCL2), is a chemoattractant for myeloma cell homing and plays an instructive role in trans-endothelial migration to the bone marrow via the CCR2 receptor (Craig & Loberg, 2006). Supporting the role for CCL2 in bone metastasis (X. Lu & Kang, 2009a), CCR2 expression in prostate cancer correlates with progression (Yi Lu, et al., 2007), and systemic administration of CCR2-neutralizing antibodies lessens metastatic burden after intracardiac injection of prostate metastatic cells (Loberg, et al., 2007).

In addition to the influence of chemokines, secreted proteins and lipids normally expressed in bone have demonstrated a strong ability to induce bone metastatic cell chemotaxis. For example, the glycoprotein osteonectin (SPARC) acts as a chemoattractant for both breast and prostate cancer cells (Jacob, Webber, Benayahu, & Kleinman, 1999). Preliminary evidence has established a possible role for adipocytes in metastatic homing to the bone. Prostate cells preferentially home to lipid rich regions of the bone stroma; this effect was attributed to Omega-6 exposure and could be inhibited by increased Omega-3 concentrations (Brown, Hart, Gazi, Bagley, & Clarke, 2006). Furthermore, adipose tissue enhances both primary tumor growth and metastasis in an estrogen dependent manner (Elliott, Tam, Dexter, & Chen, 1992). Much work remains in elucidating how the bone marrow stroma affects tumor homing. For example, bone marrow-derived cells home to soft tissues following chemotherapy or radiation (Epperly, Guo, Shields, Zhang, & Greenberger, 2004; Ortiz, et al., 2003), how this affects bone metastasis is yet unexplored.

Attachment and escape to bone parenchyma

Numerous molecular mechanisms have been elucidated which endow bone metastatic CTCs with the ability to attach to and invade bone tissue (Figure 2). Broad classes of stromal interactions, such as integrin- and lectin-mediated attachment or protease-dependent invasion have been characterized. Whereas multiple trials with Matrix Metalloprotease (MMP) inhibitors have met with failure (Coussens, 2002), many known inhibitors of surface adhesion molecules have not yet been tested in clinical trials.

With the ability to induce mitogenic intracellular signaling via outside-in activation, integrins are broadly implicated in bone metastasis. Three major integrins have demonstrated instructive roles in bone metastatic seeding: integrins αvβ3, α2β1, and α4β1 (Schneider, Amend, & Weilbaecher, 2011). These integrins are expressed by bone metastatic cells and bind extracellular matrix proteins normally expressed by bone-associated cells. Following binding, pleiotropic intracellular signaling programs induce migration, invasion and proliferation.

The αvβ3 integrin functions in breast cancer bone metastasis through binding either osteopontin (OPN), bone sialoprotein (BSP), or CD44 (Hayashi, et al., 2007; Senger, Perruzzi, & Papadopoulos, 1989; Takayama, et al., 2005; Zohar, et al., 2000), and targeted αv integrin-inhibitory peptide mimics are effective in attenuating both early metastatic seeding and proliferation (Shannon, et al., 2004). OPN is a major extracellular component of multiple bone tissue cells, and engagement by αvβ3 results in MEK-induced upregulation of matrix metalloproteinase 9 (MMP9) (Chen, et al., 2009). Additionally, αvβ3 binding to serum OPN induces the migration of B16 melanoma cells, and forced expression is able to redirect a spontaneous lung metastasis model to the bone marrow (Hayashi, et al., 2007; Sloan, et al., 2006). BSP is a distinct αvβ3 ligand normally expressed in bone tissue which is upregulated in bone metastatic lesions compared to metastases in other organs (Waltregny, et al., 2000). BSP, MMP2 and αvβ3 may form a tripartite complex responsible for increased invasiveness of thyroid carcinoma cells (Karadag, Ogbureke, Fedarko, & Fisher, 2004) which can potentially be abrogated by overexpression of tissue inhibitor of MMP2 (TIMP2) (Yoneda, et al., 1997).

The α2β1 and α4β1 integrins bind Collagen 1 (COL1) and VCAM1 in bone metastasis. COL1 is the major structural component of the bone matrix; when bound by α2β1, subsequent RhoC activation primes cell morphology for invasion (Hall, 2006; Hall, et al., 2008). VCAM1 constitutively expressed by bone endothelial cells binds α4β1 (Miyake, et al., 1991), and forced expression of α4β1 is sufficient to endow Chinese hamster ovary (CHO) cells with bone metastatic ability. Preclinical evidence suggests that antibody blocking of this interaction is able to suppress the establishment of osteolytic myeloma (Matsuura, et al., 1996).

Whereas integrins mediate mitogeneic and migratory signaling, the initial attachment of metastatic cells to bone endothelial cells is largely attributed to the lectin class of protein adhesion molecules (Barthel, et al., 2013; Lehr & Pienta, 1998). Glycosylated ligands present on CTCs such as PSGL1 and CD44 engage endothelial selectin (SELE), and mediate initial cell attachment to and subsequent rolling along bone endothelial cells in both HSCs and CTCs (Barthel, et al., 2013; Dimitroff, Lee, Rafii, Fuhlbrigge, & Sackstein, 2001; Katayama, et al., 2003). This initial attachment facilitates integrin clustering and subsequent trans-endothelial migration. Two other selectins, SELP and leukocyte selectin (SELL), are also shown to engage metastatic ligands and mediate CTC-endothelial cell attachment through the action of platelets or circulating leukocytes (Borsig, Wong, Hynes, Varki, & Varki, 2002). Galectin 3 (GAL3), an additional carbohydrate binding protein, is able to engage in heterotypic interactions between CTCs and bone endothelium (Heimburg, et al., 2006; Lehr & Pienta, 1998; Pienta, et al., 1995). Therapeutics targeting lectin family proteins are able to diminish metastasis in both animal models and human patients. The GAL3 ligand mimic lactulose-L-leucine has shown therapeutic promise in models of breast, prostate and lung metastasis to bone (Glinskii, et al., 2012; Heimburg, et al., 2006), while the mechanism of cimetidine-induced survival in colorectal patients was attributed to decreased SELE expression (K. Kobayashi, Matsumoto, Morishima, Kawabe, & Okamoto, 2000).

4. Metastatic niches and metastatic dormancy

The existence of dormant cells in bone metastasis has long been hypothesized; however, research on the molecular regulation of metastatic dormancy only started to gain momentum in recent years (Aguirre-Ghiso, 2007). Existing knowledge regarding dormancy indicates that metastatic quiescence is largely dependent on the stromal environment and specifically on the niche where the DTCs reside. How stromal cells affect cell dormancy and the extent to which either the HSC niche or perivascular niche regulates cell proliferation is a promising new avenue for the development of novel therapeutics.

Early work on cancer depicted metastasis as the final step in primary tumor progression, however, clinical evidence has established that bone metastasis can be an early event in the progression of cancer. Using cytokeratin staining of bone marrow aspirates, studies have shown that DTCs are present in patients with locally restricted prostate or breast cancer (Banys, et al., 2011; Braun, et al., 2005; Hüsemann, et al., 2008; Sänger, et al., 2011; Solakoglu, et al., 2002). The link between DTCs and metastasis is not straightforward as detection rates of bone marrow DTCs in patients with localized disease (55%) do not reflect the overall relapse rates (12–30%) (Melchior, et al., 1997), indicating that exit from dormancy cannot be achieved by all DTCs within the bone marrow. Reflecting the complexity of connection between DTC and metastasis, substantial proportion of CTCs lose epithelial traits such as cytokeratins, via the epithelial to mesenchymal transition (Kang & Pantel, 2013; Yu, et al., 2013), and the presence of epithelial DTCs in bone marrow may not accurately depict the most invasive population (Gradilone, et al., 2011).

Programs mediating dormancy

Current evidence suggests that metastatic dormancy is the result of both a permissive endogenous bone marrow environment as well as a specific reaction to the presence of invading metastatic cells (Figure 3). For example, bone endothelial cells normally secrete basic fibroblast growth factor (FGF2) which induces breast cancer cell dormancy. FGF2-stimulated cells are able to maintain a dormant state upon α5β1 attachment to fibronectin and will otherwise undergo apoptosis (Korah, Boots, & Wieder, 2004). Bone endothelial cells also express Duffy antigen receptor for chemokines (DARC) which causes growth arrest upon binding to KAI1 expressed by metastatic cells (Bandyopadhyay, et al., 2006). Alternately, the identification of several metastasis suppressor genes has demonstrated that a stromal reaction to tumor cells induces dormancy. Kisspeptin-1 (KiSS1) is a G-protein-coupled receptor ligand which is secreted by tumor cells and processed by stromal cells which functions to suppress metastasis (Lee, et al., 1996; Nash, et al., 2007). Furthermore, the serine protease inhibitor Maspin is responsible for attenuating uPA activity on the surrounding stroma (Cher, et al., 2003). Taken together, these data indicate that perturbations in the normal bone environment may induce dormancy.

Figure 3. Metastatic dormancy is dependent on bone marrow stromal elements.

Figure 3

Two distinct niches are host to invading tumor cells in the bone marrow: the hematopoetic stem niche or the perivascular niche. In either niche, the bone microenvironment invokes tumor dormancy through growth factor secretion, activation of metastasis suppressor proteins, or binding to anti-proliferative signals. Metastatic cells respond and proliferate by secreting proteases or growth factors to accommodate growth and niche remodeling.

Regardless of the programs which induce dormancy, survival and exit from dormancy is achieved through a complex series of steps such as adaptation of dormant cells, changes in systemic endocrine levels by stress, or the effects of distant established tumors (Ben-Eliyahu, Shakhar, Page, Stefanski, & Shakhar, 2000; McAllister, et al., 2008). For example, distant metastatic cells are able to mobilize bone marrow endothelial cells through OPN secretion. Mobilized cells migrate into dormant lesions and lead to proliferation (McAllister, et al., 2008). Alternately, formation of a complex between urokinase type plasminogen activator (uPAR) and the dormancy-sustaining α5β1-Fn interaction may be responsible for an exit from dormancy (Aguirre Ghiso, Kovalski, & Ossowski, 1999). The dormancy state of DTCs is further influenced by the balance of ERK1/2 and p38α/β signaling activity (Sosa, Avivar-Valderas, Bragado, Wen, & Aguirre-Ghiso, 2011). In bone metastasis, early osteoclast activity is potentially important, and engagement of α4β1 to VCAM1 expressed on osteoclasts provokes growth of indolent breast cancer metastases (Xin Lu, et al., 2011). The αvβ3 integrin is also important in tumor proliferation and angiogenesis (Nemeth, et al., 2003). As a potential ligand of αvβ3 integrin, periostin is associated with breast cancer metastasis (Sasaki, et al., 2003), and enhances endothelial cell vascularization as well as tumor intrinsic survival by binding αvβ3 (Bao, et al., 2004).

Members of the TGFβ family are known to have opposing roles in tumor growth. Bone morphogenic protein 7 (BMP7) secreted from endothelial cells effects a reversible cell cycle block in prostate cancer stem cells through p38 induction. Constant exposure of BMP7 leads to senescence and death (A. Kobayashi, et al., 2011). Alternatively, upon induction of angiogenesis, TGFβ1 from sprouting vascular tips is linked to an exit from dormancy and proliferation (Ghajar, et al., 2013). Administration of a TGFβ1 receptor inhibitor or genetic ablation of the TGFβ signaling pathway reduces proliferation of bone metastasis by inhibiting the production of pro-bone metastatic genes, including Interleukin-11 (IL11), Jagged1 and Parathyroid hormone-related protein (PTHrP) (Ehata, et al., 2007; Korpal, et al., 2009; Mohammad, et al., 2011; Sethi, Dai, Winter, & Kang, 2011).

Tumor cell niches

Two distinct stromal environments have been depicted as potential niches to invading metastatic cells—the hematopoetic stem cell (osteoblastic) niche and the perivascular niche. Both niches are theorized to impact metastatic survival and proliferation, albeit through distinct mechanisms. The HSC niche shares many of the same programs characterized in bone metastases, and direct competition has been observed between HSCs and bone metastatic cells (Shiozawa, et al., 2011). Recently, Shiozawa and colleagues demonstrated that metastatic cells and HSCs utilize SDF1/CXCR4 signaling for ingress and egress to the endosteal compartment. Remarkably, antagonism with either the CXCR4 inhibitor AMD3100 or recombinant GM-CSF is able to mobilize both HSCs and DTCs from bone marrow (Shiozawa, et al., 2011). Interestingly, the HSC niche, which is composed of MSCs and osteoblasts, is stimulated by many of the same signals which cause bone metastatic cells to proliferate (Adams, et al., 2007; Calvi, et al., 2003; Méndez-Ferrer, et al., 2010). The similarities between HSCs and metastatic cell behavior in the niche have profound implications on strategies for therapeutic intervention—namely, the same agents which affect HSCs may also have therapeutic values in controlling bone metastasis (Shiozawa, et al., 2011). While this increases the arsenal of drugs to treat bone metastasis, targeting the niche may also exacerbate HSC death and hematopenia. Compounding this issue, active TGFβ signaling by myeloma cells reduces the plasticity of hematopoetic stem progenitor cells and MSCs, resulting in diminished ability for hematopoesis (Bruns, et al., 2012).

Interestingly, breast or prostate cancer patients with locally restricted disease who coincidentally received radiation to lumbar or thoracic vertebrae experienced a significant reduction in subsequent metastasis to these regions (Bagshaw, Kaplan, Valdagni, & Cox, 1992; Hercbergs, Werner, & Brenner, 1985). The cause for the phenomenon remains unidentified, yet may be linked to changes in HSC niche composition.

Angiopoietin-2 (ANG2) is a potential regulator of the HSC niche and metastatic dormancy. Expression of ANG2 is upregulated in metastatic prostate and breast tissue (Imanishi, et al., 2007; Lind, et al., 2005), but expression has not been tied to metastatic dormancy and the HSC niche. ANG2 and ANG1 directly compete for binding to the TIE2 receptor (Maisonpierre, et al., 1997). Upon binding, TIE2 signaling regulates HSC quiescence (Arai, et al., 2004), sensitizes endothelial cells to inflammation (Fiedler, et al., 2006), and disrupts angiogenesis (Maisonpierre, et al., 1997). Similar pathways are instrumental in metastasis, and recently developed TIE2 inhibitors (rebastinib) may be effective in attenuating bone metastasis (Lamontanara, Gencer, Kuzyk, & Hantschel, 2013).

The perivascular niche has been characterized as an alternate site for bone metastatic colonization. This niche bears similarity to the sites of metastatic seeding in pulmonary, brain, or hepatic metastasis: endothelial progenitor cells, pericytes and other cell types are recruited to form a supportive stroma around the tumor cells. High resolution imaging of DTCs directly associated with bone marrow microvasculature after intracardiac injection provides direct evidence for tumor cells residing in the perivascular compartment. Furthermore, engineered culture conditions mimicking bone microvasculature were able to support metastatic cell attachment and induce dormancy (Ghajar, et al., 2013). Only cells in direct contact with bone endothelial cells were negative for Ki67 staining, and the metastasis suppressor Thrombospondin-1 (TSP1) was shown to be causal in this dormancy by inhibiting angiogenic programs (Ghajar, et al., 2013). Many protein dynamics which have been implicated in DTC progression independent of niche specification are likely important in the perivascular niche, for example periostin and VEGF are strong candidates for master regulators of this niche.

5. The osteolytic/osteogenic axis in bone metastasis

The bone is best known for two of the most vital roles in normal physiology: structural support and hematopoesis. The ability of bone metastases to subvert both processes is the primary cause of morbidity and mortality in cancer patients with bone metastasis. The bone is in a constant state of dynamic remodeling by a balance between osteolytic and osteogenic programs. Metastatic cancer cells often successfully exploit the normal bone homeostatic process and tip the equilibrium toward either bone lysis or bone growth to facilitate the formation of bone metastases (Ell & Kang, 2012; Mundy, 2002; Weilbaecher, et al., 2011).

Osteolytic lesions

The paracrine regulatory program of osteoclastogenesis controlled by metastatic cells is the most researched and targeted tumor-stromal interaction in bone metastasis. Multiple cytokines and growth factors have shown pleiotropic effects in stimulating hyperactive osteoclast activity and promoting osteolytic metastasis (Ell & Kang, 2012; Weilbaecher, et al., 2011).

The most prominent cytokine inducer of osteoclastogenesis is receptor activator of NF-κB ligand (RANKL) (Mundy, 2002; Weilbaecher, et al., 2011). RANKL binding to its receptor (RANK) expressed on both osteoclast precursors and metastatic cells activates NF-κB signaling (Eghbali-Fatourechi, et al., 2003), leading to both the transcription of key osteoclastogenic factors (Park, et al., 2007) and enhanced breast cancer cell migration selectively to bone marrow (Jones, et al., 2006). Similar to other TNF family members, RANKL is bound to the extracellular matrix, and is activated and released upon cleavage by tumor-secreted MMP7 (Lynch, et al., 2005). RANKL activity is balanced in normal bone homeostasis by osteoprotegerin (OPG), a decoy receptor for RANKL secreted by osteoblasts. Experimental introduction of OPG results in a pronounced decrease in osteolytic tumor growth (Morony, et al., 2001). Multiple other cytokines or ligands have also shown importance in osteoclast activation; TNFα increases the preosteoclast pool in the bone marrow and synergistically complements RANKL-induced osteoclast activation (Lam, et al., 2000). Tumor-derived Jagged1 can directly stimulate osteoclast differentiation by activating Notch signaling (Sethi, et al., 2011). Both Granulocyte macrophage-colony stimulating factor (GM-CSF/CSF2) and Macrophage-colony stimulating factor (M-CSF/CSF1) are also shown to be necessary for the maturation of preosteoclasts into osteoclasts (Lacey, et al., 1998; Park, et al., 2007). M-CSF receptor (c-FMS) antagonists have shown a significant ability to suppress osteoclast differentiation and osteolytic bone destruction (Ohno, et al., 2006).

Following osteoclast maturation, the “vicious cycle” of lytic bone metastasis takes place whereby degradation of the bone matrix by osteoclasts releases various growth factors such as IGFs, TGFβ, EGFs and FGFs, spurring tumor cell growth and further promoting osteoclast activation (Hauschka, Mavrakos, Iafrati, Doleman, & Klagsbrun, 1986; Mundy, 2002).

Dynamic in vivo imaging coupled with genetic and pharmacological inhibition of the TGFβ pathway showed that TGFβ signaling is most important in the early establishment of breast cancer metastatic lesions (Korpal, et al., 2009), and a milestone study by Yin and colleagues demonstrated that blockade of TGFβ signaling in tumor cells inhibits PTHrP secretion and reduces osteolysis (Yin, et al., 1999). In addition to inducing RANKL production from osteoblasts to increase osteoclastogenesis, the pro-metastatic function of PTHrP has been attributed to other important functions. PTHrP increases the expression of CCL2 which functions to promote osteoclast activation and bone metastasis in both breast cancer prostate cancers (X. Lu & Kang, 2009b; Y. Lu, et al., 2008; Y. Lu, et al., 2007). PTHrP is also responsible for activating the transcription of the Wnt inhibitor DKK1. DKK1 is highly expressed in myeloma, a purely lytic cancer type, and prevents the maturation of osteoblasts (Tian, et al., 2003). Paradoxically, patients with high PTHrP levels in primary tumors experience increased survival, suggesting the potential differential role of PTHrP in different tumor microenvironments (Henderson, et al., 2006).

Other factors affecting osteoclastogenesis have also emerged. For example, the proteases MMP1 and ADAMTS1 were found to release EGF ligands from the metastatic cell matrix and inhibit osteoblast differentiation from MSCs, decoupling the ability to regulate osteoclast activation (X. Lu, et al., 2009). Many other inducers of RANKL have also been discovered, including Macrophage inflammatory protein 1α (MIP1α) (Oba, et al., 2005; Tsubaki, et al., 2007), and IL8 (Bendre, et al., 2003). Tumor associated fibroblasts are also sources of RANKL, and secretion of RANKL is able to induce osteoclast differentiation from tumor associated macrophages (Lau, et al., 2006).

Osteoblastic metastasis

Parallel to the RANKL/RANK/OPG system in osteolysis, the gene regulatory networks governed by the transcription factor RUNX2 are the most heavily implicated in osteoblastic bone metastasis. Normally expressed in mesenchymal progenitor cells during osteoblast differentiation, RUNX2 activates bone matrix protein transcription such as BSP and osteocalcin (Ducy, Zhang, Geoffroy, Ridall, & Karsenty, 1997). RUNX2 is also expressed in prostate cancer cells (Akech, et al., 2010), and is theorized to induce osteomimicry in these cells through expression of pro-metastatic, bone-specific matrix proteins (Koeneman, Yeung, & Chung, 1999) such as BSP, MMP9 and SDF1 (Baniwal, et al., 2010; Barnes, et al., 2003). Evidence for osteomimicry in advanced bone metastasis is observed in both osteolytic and osteoblastic lesions. Expression of bone specific markers likely has numerous effects in increasing invasion, angiogenesis, survival and any other numbers of functions necessary to endogenous bone cells. Proteins such as osteoblastic cadherin (CDH11) are also implicated in this role (Chu, et al., 2008).

Both Endothelin-1 (ET1), a secreted osteoblast mitogen, and Prostate specific antigen (PSA), a serine protease, have been very well established in the pathology of osteoblastic metastasis (Nelson, et al., 1995). ET1 is secreted from both breast and prostate cancers; Yin and colleagues demonstrated a causal role in increasing osteoblastic tumor burden through binding the ETA receptor of osteoblasts, and orally available antagonists were able to reduce tumor burden (Yin, et al., 2003). A Phase III clinical trial demonstrated that ET1 antagonist Atrasentan did not decrease the risk of disease progression in metastatic prostate cancer despite reduced levels of PSA and bone alkaline phosphatase (BAP) (Carducci, et al., 2007). PSA is shown to activate growth factors such as PTHrP (Cramer, Chen, & Peehl, 1996; Iwamura, Hellman, Cockett, Lilja, & Gershagen, 1996), and clinical trials of PSA vaccines show strong promise in improving overall survival (Kantoff, et al., 2010).

Osteoclast and osteoblast activity are believed to be coupled in bone metastasis, with most bone lesions exhibiting varying degrees of both osteolytic and osteoblastic activities. It is perhaps not surprising that several factors are implicated in both osteolytic and osteoblastic metastasis, perhaps through activating different downstream genes in different cancer types. For example, TGFβ activates the expression of osteoclastogenic genes, such as Jagged1, PTHrP and IL-11 (Kang, et al., 2003; Sethi, et al., 2011; Yin, et al., 1999), as well as osteoblastic genes such as ET1.

6. Current therapies targeting bone remodeling

Therapeutics targeting the bone resorptive cycles of myeloma and breast cancer are a palliative rather than curative approach toward treatment of bone metastasis as these therapies slow osteoclast activity and thereby extend patient lifespan. Anti-resorptive therapies similarly extend time to relapse in bone when tested in patients with osteoblastic metastases, demonstrating that the cross communication between osteoclasts and osteoblasts is important to progression in both subtypes of bone metastasis (Figure 4). Of the osteoclast inhibitors, two have shown efficacy through Phase III clinical trials: bisphosphonates, and Denosumab (anti-RANKL antibody). Additional pharmacological agents have demonstrated promise in preclinical experiments but have yet to be tested in clinical trials.

Figure 4. The vicious cycle of osteolytic metastasis offers many targets for therapeutic intervention.

Figure 4

Tumor cell induction of RANKL/ GM-CSF/ M-CSF results in fusion of osteoclast precursor cells into multinucleated osteoclasts. Enhanced osteoclast-mediated lysis of the bone matrix releases cytokines such as TGFβ, which spur tumor cell growth and stimulate the expression of pro-metastatic factors such as Jagged1 and PTHrP from tumor cells to further induce osteoclast differentiation. Exposure to TNFα increases the pre-osteoclast pool, while osteoblasts are also important mediators of the tumor-osteoclast crosstalk.

The activity of bisphosphonates in diminishing bone resorption has long been known (van Holten-Verzantvoort, et al., 1987), and has become a standard of care in the treatment of osteolytic bone metastasis. Bisphosphonates, such as zoledronic acid, pamidronate or ibandronate, specifically bind to the bone matrix and are internalized by osteoclasts upon resorption. Once internalized, these drugs inhibit various metabolic processes such as prenylation, and lead to apoptosis (Mundy, 2002). Multiple bisphosphonates have been developed, and of these, zoledronic acid shows the highest efficacy in a Phase III clinical trial versus pamindronate for breast cancer and myeloma (Rosen, et al., 2001).

While treatment with bisphosphonates is considered the standard of care for bone metastasis, the mechanisms of bisphosphonate therapy are not fully understood. For example, the impact of ovarian activity in osteolytic bone metastasis for breast cancer patients was recognized, as treatment with zolendroate does not increase lifespan in pre-menopausal women (Gnant, et al., 2009) unless they are placed under ovarian suppression. Mechanisms for this response have not been identified (Steinman, Brufsky, & Oesterreich, 2012). Furthermore, limited studies have shown cytotoxic effects of bisphosphonates (Hiraga, Williams, Mundy, & Yoneda, 2001), however, the concentrations required for this effect may not replicate the local bone environment.

Compared to bisphosphonates, Denosumab and recombinant OPG demonstrate higher clinical efficacy by targeting RANKL. Both have shown similar promise in clinical trials, however only Denosumab has advanced through Phase III. Administration of Denosumab showed significant efficacy by inhibiting osteolysis in patients with prostate or breast cancer bone metastasis, based on the analysis of urinary N-telopeptide levels as a marker for bone turnover (Fizazi, Bosserman, Gao, Skacel, & Markus, 2013; Fizazi, et al., 2009). A single dosing system of recombinant OPG (AMGN-0007) in a Phase I trial demonstrated a similar sustained reduction in bone lysis (Body, et al., 2003).

A second cohort of lesser known anti-osteoclastogenic factors are also emerging for clinical trial. EGF signaling is able to induce RANKL and M-CSF secretion, and treatment with the EGFR kinase inhibitor Gefinitib suppresses osteoclast activity (Normanno, et al., 2005) and inhibits bone metastasis (X. Lu, et al., 2009). Similarly, small molecule antagonists of c-FMS can attenuate growth by 89% (Murray, et al., 2003; Ohno, et al., 2006). The vitamin K analogue from traditional medicine, plumbagin, inhibits NF-κB signaling through RANK and inhibits osteolytic bone metastasis (Sung, Oyajobi, & Aggarwal, 2011). Both Cathepsins B and K are functional in osteolytic bone metastasis, and specific inhibitors are available which show therapeutic promise in bone metastasis (Le Gall, et al., 2007; Withana, et al., 2012). Unexpectedly, the use of Inhibitor of apoptosis protein (IAP) antagonists has shown an activating effect on osteoclasts and preliminary trials showed accelerated metastatic growth in patients treated with IAP inhibitors (Yang, et al., 2012).

7. Immune regulation and immune suppression

The immune system plays a critical role in each step of tumor progression—from the original transformation to macrometastatic progression, immune suppression and/or immune evasion is required for the survival and expansion of tumors. In a successful metastatic progression, the immune system is often reconfigured to become a malignant accomplice able to assist in multiple functions. Several programs, such as NK cell- or effector T cell-mediated lysis, are antagonistic to progression. However the majority of immune cells, such as regulatory T cells (Treg), dendritic cells (DC), MDSCs, and macrophages, can be converted to a tumor-promoting phenotype. Therapeutic strategies, such as DC vaccination or interferon therapy, which give the immune system stimulus, are currently under investigation and show clinical promise. The role of the immune system in the progression of primary tumors and dissemination has been extensively reviewed elsewhere (Shiao, Ganesan, Rugo, & Coussens, 2011; Smith & Kang, 2013). In this section, we will focus on immune targets which have shown direct impact in bone metastasis or in clinical studies involving cancers which show a predisposition toward bone metastasis.

Tumor-antagonistic functions

Both the innate and adaptive immune responses are attributed with tumor-antagonistic roles early in metastatic progression. These responses are often attenuated as the tumor develops the ability to suppress T cell activation or shield itself from NK cell activity.

Traditional antigen presenting machinery through the MHC class I and immune surveillance is sometimes active in delaying metastatic progression. A spontaneous mouse melanoma model which constitutively expresses the MHC antigen AAD has been shown to disseminate early in tumor progression but the development of lung metastasis was kept in check by immune surveillance by CD8+ T cells (Eyles, et al., 2010). A similar mechanism is seen in bone metastasis as lesions which rapidly proliferate in athymic mice are dormant when introduced into immune competent mice, and proliferation is restored upon CD8-specific depletion (M. Muller, et al., 1998). While platelets generally exhibit multiple pro-metastatic functions, bone marrow megakaryocytes, which are platelet precursor cells, are antagonistic to growth of metastatic lesions. This is attributed to the ability of megakaryocytes to inhibit osteoclast maturation from monocytes (Beeton, Bord, Ireland, & Compston, 2006).

Immune suppression

The establishment of immune suppression in both primary tumors and metastases is well recognized – while multiple cell-intrinsic programs exist which interfere with the antigen presenting pathway, crosstalk between tumor cells and the immune system is responsible for establishing a state of chronic inflammation and immune dysregulation (Smith & Kang, 2013). Much evidence indicates that tumor-antagonistic cells are present in the bone microenvironment, however, pervasive programs of immune suppression keep these cells at bay. Immune-modulatory therapies aimed at reactivating host immune response have demonstrated clinical success (Lode, et al., 1998; Quesada, Swanson, Trindade, & Gutterman, 1983).

Myeloid derived suppressor cells (MDSC), plasmacytoid dendritic cells (pDC), and Treg cells are strongly implicated in bone metastasis. It is generally accepted that infiltrating macrophages, pDCs, Treg cells, and MDSCs induce a sustained Th2 response, suppress CD8 or NK cell activity and exacerbate bone remodeling stresses (A. Sawant, et al., 2012; Anandi Sawant & Ponnazhagan, 2013; Sisirak, et al., 2013). Several major signaling hubs are identified in this network—notably, GM-CSF and IL3 are necessary for immune-suppressive activity (Salgia, et al., 2003; Young, Wright, & Young, 1991). On the other hand, Bidwell et al demonstrated that a loss of Interferon-7 (IRF7) pathways was responsible for mammary tumor progression to bone metastasis, and treatment with IRF7 was able to restore host CD8+ T cell or NK cell response (Bidwell, et al., 2012). The STAT6 pathway is important in CD8 T cell suppression, however this functions independently of Treg cells (Ostrand-Rosenberg, Grusby, & Clements, 2000).

Successful metastatic lesions thrive in an environment replete with tumor-promoting myeloid cells and osteoclasts that secrete inflammatory cytokines. For example, the ECM molecule versican engages TLR2 and TLR6 to stimulate macrophage secretion of TNFα, which in turn stimulates tumor growth (S. Kim, et al., 2009). The creation of this environment can foster bone invasion, as bone-derived MSCs are able to infiltrate primary breast carcinoma and promote bone metastasis through IL17B secretion (Goldstein, Reagan, Anderson, Kaplan, & Rosenblatt, 2010). Similarly, Lysophosphatidic acid released from activated platelets in the bone microenvironment stimulates IL6 and IL8 secretion, enhancing tumor proliferation through increased osteolysis (Boucharaba, et al., 2004).

Paradoxically, tumor-associated antigen (TAA)-specific memory T cells selectively increase in the bone marrow of advanced breast cancer patients with bone marrow DTCs (Feuerer, et al., 2001). The bone marrow is also a significant reservoir of Treg cells (Zou, et al., 2004), indicating that activity of these TAA-specific memory T cells is potentially suppressed by Treg cells. Metastasis associated Treg cells also specifically increase in prostate bone metastasis microenvironments, and inhibit bone resorption by exhibiting a broad immune-suppressive profile (Zhao, et al., 2012). The creation of an inflammatory microenvironment with an abundance of proliferative and angiogenic signals is conducive for both primary and metastatic cell growth. Indeed, even treatment with aspirin shows significant decreases in tumor osteolytic activity (Powles, Dowsett, Easty, Easty, & Neville, 1976).

While the establishment of immune suppression is a hallmark of most metastatic tumors, few immune-modulatory therapies have demonstrated clinical success. This type of therapy is theorized as a molecular reboot, the programs which maintain immune-suppression are temporarily disrupted allowing for either host response or response by allogenic cells. Both metastatic renal cell carcinoma and melanoma seem especially susceptible to immune-modulatory therapies and several therapies have resulted in complete relapse. In chemoresistant metastatic renal cell cancer or melanoma, treatment with interleukin-2 (IL2) provoked an anti-metastatic response in 20% of patients and is used in clinical practice currently (Rosenberg, et al., 1994). IL2 therapy was also proved effective exclusively through NK cell activity in a model of neuroblastoma bone metastasis (Lode, et al., 1998). Interferon α treatment is marginally effective in renal cell metastasis with 30% of patients showing a response (Quesada, et al., 1983). A separate trial with chemoresistant renal cell metastasis demonstrated that non-myeloablative immune suppression followed by stem cell transplantation from allogenic donors resulted in metastatic regression in multiple sites, including bone (Childs, et al., 2000). Similar responses have been anecdotally reported in advanced breast cancer patients (Eibl, et al., 1996). Taken together, these trials demonstrate promise for a specific cohort of patients, however, considerable work remains to identify patient populations by likelihood of clinical response.

Blockade of the specific molecules promoting immune suppression in advanced tumors has similarly received attention. CD-28 activates T cells when bound to CD-80 expressed on antigen presenting cells, cytotoxic T-lymphocyte associated antigen-4 (CTLA4) competes with CD-28 for CD80/86 binding and inhibits T cell activation (O’Day, Hamid, & Urba, 2007). Phase III clinical trials with Ipilimumab (Anti-CTLA4) in metastatic melanoma patients demonstrated a median lifespan of 10 months compared to 6 months for placebo (Hodi, et al., 2010). This lifespan extension is attributed to CD8+ T Cell activity at sites of metastasis (Demaria, et al., 2005). A similar mechanism is targeted with anti- Programmed Death-1 (PD1) or anti-CD137 antibodies, which shift T cell populations from a tumor-promoting Th2 population to a tumor-antagonistic Th1/ Th17 population (Dulos, et al., 2012; Simeone & Ascierto, 2012). Furthermore, dendritic cell vaccination in patients with metastasis demonstrates reactivation of CD4/CD8 T cell-mediated metastatic clearance (Gong, Chen, Kashiwaba, & Kufe, 1997). Further studies show that the machinery for antigen mediated clearance is intact in at least 7 of 17 patients (Kugler, et al., 2000).

8. Stromal-endowed therapeutic resistance

Bone stromal cells not only participate in the pathogenesis of bone lesions, but also make them refractory to traditional chemotherapies. Chemotherapy has no effect on dormant DTCs in the bone marrow (Braun, et al., 2000), and many groundbreaking studies have attributed this phenomenon to various stromal cell programs mediating cancer cell survival. Acharyya et al found that a chemoresistance cycle was instated when chemotherapy elicits TNFα secretion from the stroma – this then increases CXCL1/2 secretion from tumor cells which in turn attracts S100A8/9 secretion from myeloid cells to ensure survival (Acharyya, et al., 2012). Alternately, cellular contacts with components of the HSC niche or osteoclasts have also been shown to enhance tumor cell survival during adjuvant therapy (Abe, 2004). Inhibition of these tumor-stromal programs sensitizes DTCs to conventional therapeutic approaches. The use of AMD3100 to antagonize CXCR4 makes prostate or leukemic cells in the bone marrow more receptive to chemotherapy (Domanska, et al., 2012; Nervi, et al., 2009), while use of CXCR2 antagonists is similarly able to enervate the survival phenotype (Acharyya, et al., 2012).

9. Conclusions and future directions

The understanding of bone metastasis has progressed considerably in recent years, with particular emphasis on the contribution of the host to metastatic progression. Bone metastases often acquire a unique set of molecular traits which are able to temper the bone microenvironment to facilitate the growth of metastatic tumors in bone. These traits are only part of the pathogenesis of bone metastasis—contributions of associated stromal cells are active participants in every step of progression. While these interactions are potential therapeutic targets, the similarities between bone metastatic cells and the stem cells of the bone marrow may make the treatment of engrafted metastatic lesions difficult using conventional therapeutic approaches. New molecular targeting strategies should combine with immune modulation in order to achieve effective eradication of metastatic lesions with minimal side effects.

Current anti-resorptive treatments used in bone metastasis do not constitute a curative approach toward bone metastasis. Much progress needs to be made in the clinical setting to bring our improved basic understanding of bone metastasis into clinical practice, yet many hurdles exist in developing novel anti-metastasis drugs for clinical trial. Primarily, the endpoints used in treating primary tumors, such as shrinkage in tumor size, need to be adjusted for evaluating the therapeutic efficacy of targeting the metastatic process (Steeg, 2012). The clinical course of bone metastasis is highly variable, and early stage processes such as tumor seeding, dormancy, and local niche remodeling need to be assessed, mostly likely in an adjuvant setting. This also necessitates new surrogate markers for each stage of metastatic progression. While the use of urinary bone turnover markers provides an invaluable surrogate for osteolytic bone metastasis (Kamiya, et al., 2012), minimally invasive methods for the detection of CTCs or dormant metastatic cells must be developed.

Acknowledgments

We thank members of our laboratories for helpful discussions and critical reading of the manuscript. We also apologize to the many investigators whose important studies could not be cited directly here owing to space limitations. Research in the authors’ laboratory was supported by grants from the Brewster Foundation, the Champalimaud Foundation, Department of Defense, Komen for the Cure, and the National Institutes of Health to Y.K.

Abbreviations

CTC

circulating tumor cell

DC

dendritic cell

DTC

disseminated tumor cell

HSC

hematopoetic stem cell

MSC

mesenchymal stem cell

NK

natural killer

Treg

regulatory T cell

Footnotes

Conflict of Interest: The authors declare that there are no conflicts of interest.

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. Abe M. Osteoclasts enhance myeloma cell growth and survival via cell-cell contact: a vicious cycle between bone destruction and myeloma expansion. Blood. 2004;104:2484–2491. doi: 10.1182/blood-2003-11-3839. [DOI] [PubMed] [Google Scholar]
  2. Acharyya S, Oskarsson T, Vanharanta S, Malladi S, Kim J, Morris Patrick G, Manova-Todorova K, Leversha M, Hogg N, Seshan Venkatraman E, Norton L, Brogi E, Massagué J. A CXCL1 Paracrine Network Links Cancer Chemoresistance and Metastasis. Cell. 2012;150:165–178. doi: 10.1016/j.cell.2012.04.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Adams GB, Martin RP, Alley IR, Chabner KT, Cohen KS, Calvi LM, Kronenberg HM, Scadden DT. Therapeutic targeting of a stem cell niche. Nature Biotechnology. 2007;25:238–243. doi: 10.1038/nbt1281. [DOI] [PubMed] [Google Scholar]
  4. Aguirre-Ghiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer. 2007;7:834–846. doi: 10.1038/nrc2256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Aguirre Ghiso JA, Kovalski K, Ossowski L. Tumor dormancy induced by downregulation of urokinase receptor in human carcinoma involves integrin and MAPK signaling. J Cell Biol. 1999;147:89–104. doi: 10.1083/jcb.147.1.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Akech J, Wixted JJ, Bedard K, van der Deen M, Hussain S, Guise TA, van Wijnen AJ, Stein JL, Languino LR, Altieri DC, Pratap J, Keller E, Stein GS, Lian JB. Runx2 association with progression of prostate cancer in patients: mechanisms mediating bone osteolysis and osteoblastic metastatic lesions. Oncogene. 2010;29:811–821. doi: 10.1038/onc.2009.389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, Ito K, Koh GY, Suda T. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118:149–161. doi: 10.1016/j.cell.2004.07.004. [DOI] [PubMed] [Google Scholar]
  8. Bagshaw MA, Kaplan ID, Valdagni R, Cox RS. Radiation treatment of prostate bone metastases and the biological considerations. Adv Exp Med Biol. 1992;324:255–268. doi: 10.1007/978-1-4615-3398-6_28. [DOI] [PubMed] [Google Scholar]
  9. Bakewell SJ, Nestor P, Prasad S, Tomasson MH, Dowland N, Mehrotra M, Scarborough R, Kanter J, Abe K, Phillips D, Weilbaecher KN. Platelet and osteoclast beta3 integrins are critical for bone metastasis. Proc Natl Acad Sci U S A. 2003;100:14205–14210. doi: 10.1073/pnas.2234372100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bandyopadhyay S, Zhan R, Chaudhuri A, Watabe M, Pai SK, Hirota S, Hosobe S, Tsukada T, Miura K, Takano Y, Saito K, Pauza ME, Hayashi S, Wang Y, Mohinta S, Mashimo T, Iiizumi M, Furuta E, Watabe K. Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression. Nature Medicine. 2006;12:933–938. doi: 10.1038/nm1444. [DOI] [PubMed] [Google Scholar]
  11. Baniwal SK, Khalid O, Gabet Y, Shah RR, Purcell DJ, Mav D, Kohn-Gabet AE, Shi Y, Coetzee GA, Frenkel B. Runx2 transcriptome of prostate cancer cells: insights into invasiveness and bone metastasis. Mol Cancer. 2010;9:258. doi: 10.1186/1476-4598-9-258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Banys M, Gruber I, Krawczyk N, Becker S, Kurth R, Wallwiener D, Jakubowska J, Hoffmann J, Rothmund R, Staebler A, Fehm T. Hematogenous and lymphatic tumor cell dissemination may be detected in patients diagnosed with ductal carcinoma in situ of the breast. Breast Cancer Research and Treatment. 2011;131:801–808. doi: 10.1007/s10549-011-1478-2. [DOI] [PubMed] [Google Scholar]
  13. Bao S, Ouyang G, Bai X, Huang Z, Ma C, Liu M, Shao R, Anderson RM, Rich JN, Wang XF. Periostin potently promotes metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. Cancer Cell. 2004;5:329–339. doi: 10.1016/s1535-6108(04)00081-9. [DOI] [PubMed] [Google Scholar]
  14. Barnes GL, Javed A, Waller SM, Kamal MH, Hebert KE, Hassan MQ, Bellahcene A, Van Wijnen AJ, Young MF, Lian JB, Stein GS, Gerstenfeld LC. Osteoblast-related transcription factors Runx2 (Cbfa1/AML3) and MSX2 mediate the expression of bone sialoprotein in human metastatic breast cancer cells. Cancer Res. 2003;63:2631–2637. [PubMed] [Google Scholar]
  15. Barthel SR, Hays DL, Yazawa EM, Opperman M, Walley KC, Nimrichter L, Burdick MM, Gillard BM, Moser MT, Pantel K, Foster BA, Pienta KJ, Dimitroff CJ. Definition of molecular determinants of prostate cancer cell bone extravasation. Cancer Res. 2013;73:942–952. doi: 10.1158/0008-5472.CAN-12-3264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Beeton CA, Bord S, Ireland D, Compston JE. Osteoclast formation and bone resorption are inhibited by megakaryocytes. Bone. 2006;39:985–990. doi: 10.1016/j.bone.2006.06.004. [DOI] [PubMed] [Google Scholar]
  17. Ben-Eliyahu S, Shakhar G, Page GG, Stefanski V, Shakhar K. Suppression of NK cell activity and of resistance to metastasis by stress: a role for adrenal catecholamines and beta-adrenoceptors. Neuroimmunomodulation. 2000;8:154–164. doi: 10.1159/000054276. [DOI] [PubMed] [Google Scholar]
  18. Bendre MS, Montague DC, Peery T, Akel NS, Gaddy D, Suva LJ. Interleukin-8 stimulation of osteoclastogenesis and bone resorption is a mechanism for the increased osteolysis of metastatic bone disease. Bone. 2003;33:28–37. doi: 10.1016/s8756-3282(03)00086-3. [DOI] [PubMed] [Google Scholar]
  19. Bidwell BN, Slaney CY, Withana NP, Forster S, Cao Y, Loi S, Andrews D, Mikeska T, Mangan NE, Samarajiwa SA, de Weerd NA, Gould J, Argani P, Möller A, Smyth MJ, Anderson RL, Hertzog PJ, Parker BS. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nature Medicine. 2012;18:1224–1231. doi: 10.1038/nm.2830. [DOI] [PubMed] [Google Scholar]
  20. Body JJ, Greipp P, Coleman RE, Facon T, Geurs F, Fermand JP, Harousseau JL, Lipton A, Mariette X, Williams CD, Nakanishi A, Holloway D, Martin SW, Dunstan CR, Bekker PJ. A phase I study of AMGN-0007, a recombinant osteoprotegerin construct, in patients with multiple myeloma or breast carcinoma related bone metastases. Cancer. 2003;97:887–892. doi: 10.1002/cncr.11138. [DOI] [PubMed] [Google Scholar]
  21. Borsig L, Wong R, Hynes RO, Varki NM, Varki A. Synergistic effects of L- and P-selectin in facilitating tumor metastasis can involve non-mucin ligands and implicate leukocytes as enhancers of metastasis. Proc Natl Acad Sci U S A. 2002;99:2193–2198. doi: 10.1073/pnas.261704098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Boucharaba A, Serre CM, Gres S, Saulnier-Blache JS, Bordet JC, Guglielmi J, Clezardin P, Peyruchaud O. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J Clin Invest. 2004;114:1714–1725. doi: 10.1172/JCI22123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Braun S, Kentenich C, Janni W, Hepp F, de Waal J, Willgeroth F, Sommer H, Pantel K. Lack of effect of adjuvant chemotherapy on the elimination of single dormant tumor cells in bone marrow of high-risk breast cancer patients. J Clin Oncol. 2000;18:80–86. doi: 10.1200/JCO.2000.18.1.80. [DOI] [PubMed] [Google Scholar]
  24. Braun S, Vogl FD, Naume B, Janni W, Osborne MP, Coombes RC, Schlimok G, Diel IJ, Gerber B, Gebauer G, Pierga JY, Marth C, Oruzio D, Wiedswang G, Solomayer EF, Kundt G, Strobl B, Fehm T, Wong GY, Bliss J, Vincent-Salomon A, Pantel K. A pooled analysis of bone marrow micrometastasis in breast cancer. N Engl J Med. 2005;353:793–802. doi: 10.1056/NEJMoa050434. [DOI] [PubMed] [Google Scholar]
  25. Brenner S, Whiting-Theobald N, Kawai T, Linton GF, Rudikoff AG, Choi U, Ryser MF, Murphy PM, Sechler JMG, Malech HL. CXCR4-Transgene Expression Significantly Improves Marrow Engraftment of Cultured Hematopoietic Stem Cells. Stem Cells. 2004;22:1128–1133. doi: 10.1634/stemcells.2003-0196. [DOI] [PubMed] [Google Scholar]
  26. Brown MD, Hart CA, Gazi E, Bagley S, Clarke NW. Promotion of prostatic metastatic migration towards human bone marrow stoma by Omega 6 and its inhibition by Omega 3 PUFAs. Br J Cancer. 2006;94:842–853. doi: 10.1038/sj.bjc.6603030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Bruns I, Cadeddu RP, Brueckmann I, Frobel J, Geyh S, Bust S, Fischer JC, Roels F, Wilk CM, Schildberg FA, Hunerliturkoglu AN, Zilkens C, Jager M, Steidl U, Zohren F, Fenk R, Kobbe G, Brors B, Czibere A, Schroeder T, Trumpp A, Haas R. Multiple myeloma-related deregulation of bone marrow-derived CD34(+) hematopoietic stem and progenitor cells. Blood. 2012;120:2620–2630. doi: 10.1182/blood-2011-04-347484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841–846. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
  29. Carducci MA, Saad F, Abrahamsson PA, Dearnaley DP, Schulman CC, North SA, Sleep DJ, Isaacson JD, Nelson JB. A phase 3 randomized controlled trial of the efficacy and safety of atrasentan in men with metastatic hormone-refractory prostate cancer. Cancer. 2007;110:1959–1966. doi: 10.1002/cncr.22996. [DOI] [PubMed] [Google Scholar]
  30. Catena R, Luis-Ravelo D, Anton I, Zandueta C, Salazar-Colocho P, Larzabal L, Calvo A, Lecanda F. PDGFR Signaling Blockade in Marrow Stroma Impairs Lung Cancer Bone Metastasis. Cancer Research. 2010;71:164–174. doi: 10.1158/0008-5472.CAN-10-1708. [DOI] [PubMed] [Google Scholar]
  31. Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2:563–572. doi: 10.1038/nrc865. [DOI] [PubMed] [Google Scholar]
  32. Chen YJ, Wei YY, Chen HT, Fong YC, Hsu CJ, Tsai CH, Hsu HC, Liu SH, Tang CH. Osteopontin increases migration and MMP-9 up-regulation via αvβ3 integrin, FAK, ERK, and NF-κB-dependent pathway in human chondrosarcoma cells. Journal of Cellular Physiology. 2009;221:98–108. doi: 10.1002/jcp.21835. [DOI] [PubMed] [Google Scholar]
  33. Cher ML, Biliran HR, Jr, Bhagat S, Meng Y, Che M, Lockett J, Abrams J, Fridman R, Zachareas M, Sheng S. Maspin expression inhibits osteolysis, tumor growth, and angiogenesis in a model of prostate cancer bone metastasis. Proc Natl Acad Sci U S A. 2003;100:7847–7852. doi: 10.1073/pnas.1331360100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Childs R, Chernoff A, Contentin N, Bahceci E, Schrump D, Leitman S, Read EJ, Tisdale J, Dunbar C, Linehan WM, Young NS, Barrett AJ. Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation. N Engl J Med. 2000;343:750–758. doi: 10.1056/NEJM200009143431101. [DOI] [PubMed] [Google Scholar]
  35. Chu K, Cheng CJ, Ye X, Lee YC, Zurita AJ, Chen DT, Yu-Lee LY, Zhang S, Yeh ET, Hu MCT, Logothetis CJ, Lin SH. Cadherin-11 Promotes the Metastasis of Prostate Cancer Cells to Bone. Molecular Cancer Research. 2008;6:1259–1267. doi: 10.1158/1541-7786.MCR-08-0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Coupland LA, Chong BH, Parish CR. Platelets and P-selectin control tumor cell metastasis in an organ-specific manner and independently of NK cells. Cancer Res. 2012;72:4662–4671. doi: 10.1158/0008-5472.CAN-11-4010. [DOI] [PubMed] [Google Scholar]
  37. Coussens LM. Matrix Metalloproteinase Inhibitors and Cancer--Trials and Tribulations. Science. 2002;295:2387–2392. doi: 10.1126/science.1067100. [DOI] [PubMed] [Google Scholar]
  38. Craig MJ, Loberg RD. CCL2 (Monocyte Chemoattractant Protein-1) in cancer bone metastases. Cancer Metastasis Rev. 2006;25:611–619. doi: 10.1007/s10555-006-9027-x. [DOI] [PubMed] [Google Scholar]
  39. Cramer SD, Chen Z, Peehl DM. Prostate specific antigen cleaves parathyroid hormone-related protein in the PTH-like domain: inactivation of PTHrP-stimulated cAMP accumulation in mouse osteoblasts. J Urol. 1996;156:526–531. doi: 10.1097/00005392-199608000-00076. [DOI] [PubMed] [Google Scholar]
  40. Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, Formenti SC. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005;11:728–734. [PubMed] [Google Scholar]
  41. Demers MJ, Thibodeau S, Noël D, Fujita N, Tsuruo T, Gauthier R, Arguin M, Vachon PH. Intestinal epithelial cancer cell anoikis resistance: EGFR-mediated sustained activation of Src overrides Fak-dependent signaling to MEK/Erk and/or PI3-K/Akt-1. Journal of Cellular Biochemistry. 2009;107:639–654. doi: 10.1002/jcb.22131. [DOI] [PubMed] [Google Scholar]
  42. Dimitroff CJ, Lee JY, Rafii S, Fuhlbrigge RC, Sackstein R. CD44 is a major E-selectin ligand on human hematopoietic progenitor cells. J Cell Biol. 2001;153:1277–1286. doi: 10.1083/jcb.153.6.1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Domanska UM, Timmer-Bosscha H, Nagengast WB, Oude Munnink TH, Kruizinga RC, Ananias HJ, Kliphuis NM, Huls G, De Vries EG, de Jong IJ, Walenkamp AM. CXCR4 inhibition with AMD3100 sensitizes prostate cancer to docetaxel chemotherapy. Neoplasia. 2012;14:709–718. doi: 10.1593/neo.12324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell. 1997;89:747–754. doi: 10.1016/s0092-8674(00)80257-3. [DOI] [PubMed] [Google Scholar]
  45. Dulos J, Carven GJ, van Boxtel SJ, Evers S, Driessen-Engels LJ, Hobo W, Gorecka MA, de Haan AF, Mulders P, Punt CJ, Jacobs JF, Schalken JA, Oosterwijk E, van Eenennaam H, Boots AM. PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer. J Immunother. 2012;35:169–178. doi: 10.1097/CJI.0b013e318247a4e7. [DOI] [PubMed] [Google Scholar]
  46. Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, Lacey DL, Riggs BL. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. Journal of Clinical Investigation. 2003;111:1221–1230. doi: 10.1172/JCI17215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Ehata S, Hanyu A, Fujime M, Katsuno Y, Fukunaga E, Goto K, Ishikawa Y, Nomura K, Yokoo H, Shimizu T, Ogata E, Miyazono K, Shimizu K, Imamura T. Ki26894, a novel transforming growth factor-beta type I receptor kinase inhibitor, inhibits in vitro invasion and in vivo bone metastasis of a human breast cancer cell line. Cancer Sci. 2007;98:127–133. doi: 10.1111/j.1349-7006.2006.00357.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Eibl B, Schwaighofer H, Nachbaur D, Marth C, Gachter A, Knapp R, Bock G, Gassner C, Schiller L, Petersen F, Niederwieser D. Evidence for a graft-versus-tumor effect in a patient treated with marrow ablative chemotherapy and allogeneic bone marrow transplantation for breast cancer. Blood. 1996;88:1501–1508. [PubMed] [Google Scholar]
  49. Ell B, Kang Y. SnapShot: Bone Metastasis. Cell. 2012;151:690–690. e691. doi: 10.1016/j.cell.2012.10.005. [DOI] [PubMed] [Google Scholar]
  50. Elliott BE, Tam SP, Dexter D, Chen ZQ. Capacity of adipose tissue to promote growth and metastasis of a murine mammary carcinoma: effect of estrogen and progesterone. Int J Cancer. 1992;51:416–424. doi: 10.1002/ijc.2910510314. [DOI] [PubMed] [Google Scholar]
  51. Epperly MW, Guo H, Shields D, Zhang X, Greenberger JS. Correlation of ionizing irradiation-induced late pulmonary fibrosis with long-term bone marrow culture fibroblast progenitor cell biology in mice homozygous deletion recombinant negative for endothelial cell adhesion molecules. In Vivo. 2004;18:1–14. [PubMed] [Google Scholar]
  52. Eyles J, Puaux AL, Wang X, Toh B, Prakash C, Hong M, Tan TG, Zheng L, Ong LC, Jin Y, Kato M, Prevost-Blondel A, Chow P, Yang H, Abastado JP. Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J Clin Invest. 2010;120:2030–2039. doi: 10.1172/JCI42002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Fernandez Vallone VB, Hofer EL, Choi H, Bordenave RH, Batagelj E, Feldman L, La Russa V, Caramutti D, Dimase F, Labovsky V, Martinez LM, Chasseing NA. Behaviour of mesenchymal stem cells from bone marrow of untreated advanced breast and lung cancer patients without bone osteolytic metastasis. Clin Exp Metastasis. 2013;30:317–332. doi: 10.1007/s10585-012-9539-4. [DOI] [PubMed] [Google Scholar]
  54. Feuerer M, Rocha M, Bai L, Umansky V, Solomayer EF, Bastert G, Diel IJ, Schirrmacher V. 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]
  55. Fiedler U, Reiss Y, Scharpfenecker M, Grunow V, Koidl S, Thurston G, Gale NW, Witzenrath M, Rosseau S, Suttorp N, Sobke A, Herrmann M, Preissner KT, Vajkoczy P, Augustin HG. Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat Med. 2006;12:235–239. doi: 10.1038/nm1351. [DOI] [PubMed] [Google Scholar]
  56. Fizazi K, Bosserman L, Gao G, Skacel T, Markus R. Denosumab treatment of prostate cancer with bone metastases and increased urine N-telopeptide levels after therapy with intravenous bisphosphonates: results of a randomized phase II trial. J Urol. 2013;189:S51–57. doi: 10.1016/j.juro.2012.11.022. discussion S57–58. [DOI] [PubMed] [Google Scholar]
  57. Fizazi K, Lipton A, Mariette X, Body JJ, Rahim Y, Gralow JR, Gao G, Wu L, Sohn W, Jun S. Randomized phase II trial of denosumab in patients with bone metastases from prostate cancer, breast cancer, or other neoplasms after intravenous bisphosphonates. J Clin Oncol. 2009;27:1564–1571. doi: 10.1200/JCO.2008.19.2146. [DOI] [PubMed] [Google Scholar]
  58. Gasic GJ, Gasic TB, Stewart CC. Antimetastatic effects associated with platelet reduction. Proc Natl Acad Sci U S A. 1968;61:46–52. doi: 10.1073/pnas.61.1.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Ghajar CM, Peinado H, Mori H, Matei IR, Evason KJ, Brazier H, Almeida D, Koller A, Hajjar KA, Stainier DYR, Chen EI, Lyden D, Bissell MJ. The perivascular niche regulates breast tumour dormancy. Nature Cell Biology. 2013;15:807–817. doi: 10.1038/ncb2767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Glinskii OV, Sud S, Mossine VV, Mawhinney TP, Anthony DC, Glinsky GV, Pienta KJ, Glinsky VV. Inhibition of prostate cancer bone metastasis by synthetic TF antigen mimic/galectin-3 inhibitor lactulose-L-leucine. Neoplasia. 2012;14:65–73. doi: 10.1593/neo.111544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Gnant M, Mlineritsch B, Schippinger W, Luschin-Ebengreuth G, Postlberger S, Menzel C, Jakesz R, Seifert M, Hubalek M, Bjelic-Radisic V, Samonigg H, Tausch C, Eidtmann H, Steger G, Kwasny W, Dubsky P, Fridrik M, Fitzal F, Stierer M, Rucklinger E, Greil R, Marth C. Endocrine therapy plus zoledronic acid in premenopausal breast cancer. N Engl J Med. 2009;360:679–691. doi: 10.1056/NEJMoa0806285. [DOI] [PubMed] [Google Scholar]
  62. 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: 10.1158/0008-5472.CAN-10-1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Gong J, Chen D, Kashiwaba M, Kufe D. Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells. Nat Med. 1997;3:558–561. doi: 10.1038/nm0597-558. [DOI] [PubMed] [Google Scholar]
  64. Gradilone A, Raimondi C, Nicolazzo C, Petracca A, Gandini O, Vincenzi B, Naso G, Agliano AM, Cortesi E, Gazzaniga P. Circulating tumour cells lacking cytokeratin in breast cancer: the importance of being mesenchymal. J Cell Mol Med. 2011;15:1066–1070. doi: 10.1111/j.1582-4934.2011.01285.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hall CL. Type I Collagen Receptor (2 1) Signaling Promotes the Growth of Human Prostate Cancer Cells within the Bone. Cancer Research. 2006;66:8648–8654. doi: 10.1158/0008-5472.CAN-06-1544. [DOI] [PubMed] [Google Scholar]
  66. Hall CL, Dubyk CW, Riesenberger TA, Shein D, Keller ET, van Golen KL. Type I collagen receptor (alpha2beta1) signaling promotes prostate cancer invasion through RhoC GTPase. Neoplasia. 2008;10:797–803. doi: 10.1593/neo.08380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hauschka PV, Mavrakos AE, Iafrati MD, Doleman SE, Klagsbrun M. Growth factors in bone matrix. Isolation of multiple types by affinity chromatography on heparin-Sepharose. J Biol Chem. 1986;261:12665–12674. [PubMed] [Google Scholar]
  68. Hayashi C, Rittling S, Hayata T, Amagasa T, Denhardt D, Ezura Y, Nakashima K, Noda M. Serum osteopontin, an enhancer of tumor metastasis to bone, promotes B16 melanoma cell migration. Journal of Cellular Biochemistry. 2007;101:979–986. doi: 10.1002/jcb.21298. [DOI] [PubMed] [Google Scholar]
  69. Heimburg J, Yan J, Morey S, Glinskii OV, Huxley VH, Wild L, Klick R, Roy R, Glinsky VV, Rittenhouse-Olson K. Inhibition of spontaneous breast cancer metastasis by anti-Thomsen-Friedenreich antigen monoclonal antibody JAA-F11. Neoplasia. 2006;8:939–948. doi: 10.1593/neo.06493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Henderson MA, Danks JA, Slavin JL, Byrnes GB, Choong PF, Spillane JB, Hopper JL, Martin TJ. Parathyroid hormone-related protein localization in breast cancers predict improved prognosis. Cancer Res. 2006;66:2250–2256. doi: 10.1158/0008-5472.CAN-05-2814. [DOI] [PubMed] [Google Scholar]
  71. Hercbergs A, Werner A, Brenner HJ. Reduced thoracic vertebrae metastases following post mastectomy parasternal irradiation. Int J Radiat Oncol Biol Phys. 1985;11:773–776. doi: 10.1016/0360-3016(85)90310-4. [DOI] [PubMed] [Google Scholar]
  72. Hiraga T, Williams PJ, Mundy GR, Yoneda T. The bisphosphonate ibandronate promotes apoptosis in MDA-MB-231 human breast cancer cells in bone metastases. Cancer Res. 2001;61:4418–4424. [PubMed] [Google Scholar]
  73. Hiratsuka S, Watanabe A, Sakurai Y, Akashi-Takamura S, Ishibashi S, Miyake K, Shibuya M, Akira S, Aburatani H, Maru Y. The S100A8–serum amyloid A3–TLR4 paracrine cascade establishes a pre-metastatic phase. Nature Cell Biology. 2008;10:1349–1355. doi: 10.1038/ncb1794. [DOI] [PubMed] [Google Scholar]
  74. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Hüsemann Y, Geigl JB, Schubert F, Musiani P, Meyer M, Burghart E, Forni G, Eils R, Fehm T, Riethmüller G, Klein CA. Systemic Spread Is an Early Step in Breast Cancer. Cancer Cell. 2008;13:58–68. doi: 10.1016/j.ccr.2007.12.003. [DOI] [PubMed] [Google Scholar]
  76. Imanishi Y, Hu B, Jarzynka MJ, Guo P, Elishaev E, Bar-Joseph I, Cheng SY. Angiopoietin-2 stimulates breast cancer metastasis through the alpha(5)beta(1) integrin-mediated pathway. Cancer Res. 2007;67:4254–4263. doi: 10.1158/0008-5472.CAN-06-4100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Iwamura M, Hellman J, Cockett AT, Lilja H, Gershagen S. Alteration of the hormonal bioactivity of parathyroid hormone-related protein (PTHrP) as a result of limited proteolysis by prostate-specific antigen. Urology. 1996;48:317–325. doi: 10.1016/S0090-4295(96)00182-3. [DOI] [PubMed] [Google Scholar]
  78. Jacob K, Webber M, Benayahu D, Kleinman HK. Osteonectin promotes prostate cancer cell migration and invasion: a possible mechanism for metastasis to bone. Cancer Res. 1999;59:4453–4457. [PubMed] [Google Scholar]
  79. Jones DH, Nakashima T, Sanchez OH, Kozieradzki I, Komarova SV, Sarosi I, Morony S, Rubin E, Sarao R, Hojilla CV, Komnenovic V, Kong YY, Schreiber M, Dixon SJ, Sims SM, Khokha R, Wada T, Penninger JM. Regulation of cancer cell migration and bone metastasis by RANKL. Nature. 2006;440:692–696. doi: 10.1038/nature04524. [DOI] [PubMed] [Google Scholar]
  80. Kamiya N, Suzuki H, Endo T, Yano M, Naoi M, Nishimi D, Kawamura K, Imamoto T, Ichikawa T. Clinical usefulness of bone markers in prostate cancer with bone metastasis. International Journal of Urology. 2012;19:968–979. doi: 10.1111/j.1442-2042.2012.03098.x. [DOI] [PubMed] [Google Scholar]
  81. Kang Y, Pantel K. Tumor cell dissemination: emerging biological insights from animal models and cancer patients. Cancer Cell. 2013;23:573–581. doi: 10.1016/j.ccr.2013.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon-Cardo C, Guise TA, Massague J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3:537–549. doi: 10.1016/s1535-6108(03)00132-6. [DOI] [PubMed] [Google Scholar]
  83. Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, Dahut WL, Arlen PM, Gulley JL, Godfrey WR. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28:1099–1105. doi: 10.1200/JCO.2009.25.0597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jensen KK, Rafii S, Lyden D. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–827. doi: 10.1038/nature04186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Karadag A, Ogbureke KU, Fedarko NS, Fisher LW. Bone sialoprotein, matrix metalloproteinase 2, and alpha(v)beta3 integrin in osteotropic cancer cell invasion. J Natl Cancer Inst. 2004;96:956–965. doi: 10.1093/jnci/djh169. [DOI] [PubMed] [Google Scholar]
  86. Karpatkin S, Pearlstein E, Ambrogio C, Coller BS. Role of adhesive proteins in platelet tumor interaction in vitro and metastasis formation in vivo. J Clin Invest. 1988;81:1012–1019. doi: 10.1172/JCI113411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Katayama Y, Hidalgo A, Furie BC, Vestweber D, Furie B, Frenette PS. PSGL-1 participates in E-selectin-mediated progenitor homing to bone marrow: evidence for cooperation between E-selectin ligands and alpha4 integrin. Blood. 2003;102:2060–2067. doi: 10.1182/blood-2003-04-1212. [DOI] [PubMed] [Google Scholar]
  88. Kelly T, Suva LJ, Huang Y, Macleod V, Miao HQ, Walker RC, Sanderson RD. Expression of heparanase by primary breast tumors promotes bone resorption in the absence of detectable bone metastases. Cancer Res. 2005;65:5778–5784. doi: 10.1158/0008-5472.CAN-05-0749. [DOI] [PubMed] [Google Scholar]
  89. Khatib AM, Kontogiannea M, Fallavollita L, Jamison B, Meterissian S, Brodt P. Rapid induction of cytokine and E-selectin expression in the liver in response to metastatic tumor cells. Cancer Res. 1999;59:1356–1361. [PubMed] [Google Scholar]
  90. Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, Luo JL, Karin M. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009;457:102–106. doi: 10.1038/nature07623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Kim YJ, Borsig L, Varki NM, Varki A. P-selectin deficiency attenuates tumor growth and metastasis. Proc Natl Acad Sci U S A. 1998;95:9325–9330. doi: 10.1073/pnas.95.16.9325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Kobayashi A, Okuda H, Xing F, Pandey PR, Watabe M, Hirota S, Pai SK, Liu W, Fukuda K, Chambers C, Wilber A, Watabe K. Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. Journal of Experimental Medicine. 2011;208:2641–2655. doi: 10.1084/jem.20110840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kobayashi K, Matsumoto S, Morishima T, Kawabe T, Okamoto T. Cimetidine inhibits cancer cell adhesion to endothelial cells and prevents metastasis by blocking E-selectin expression. Cancer Res. 2000;60:3978–3984. [PubMed] [Google Scholar]
  94. Koeneman KS, Yeung F, Chung LW. Osteomimetic properties of prostate cancer cells: a hypothesis supporting the predilection of prostate cancer metastasis and growth in the bone environment. Prostate. 1999;39:246–261. doi: 10.1002/(sici)1097-0045(19990601)39:4<246::aid-pros5>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
  95. Koh BI, Kang Y. The pro-metastatic role of bone marrow-derived cells: a focus on MSCs and regulatory T cells. EMBO reports. 2012;13:412–422. doi: 10.1038/embor.2012.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Korah R, Boots M, Wieder R. Integrin alpha5beta1 promotes survival of growth-arrested breast cancer cells: an in vitro paradigm for breast cancer dormancy in bone marrow. Cancer Res. 2004;64:4514–4522. doi: 10.1158/0008-5472.CAN-03-3853. [DOI] [PubMed] [Google Scholar]
  97. Korpal M, Yan J, Lu X, Xu S, Lerit DA, Kang Y. Imaging transforming growth factor-β signaling dynamics and therapeutic response in breast cancer bone metastasis. Nature Medicine. 2009;15:960–966. doi: 10.1038/nm.1943. [DOI] [PubMed] [Google Scholar]
  98. Kugler A, Stuhler G, Walden P, Zoller G, Zobywalski A, Brossart P, Trefzer U, Ullrich S, Muller CA, Becker V, Gross AJ, Hemmerlein B, Kanz L, Muller GA, Ringert RH. Regression of human metastatic renal cell carcinoma after vaccination with tumor cell-dendritic cell hybrids. Nat Med. 2000;6:332–336. doi: 10.1038/73193. [DOI] [PubMed] [Google Scholar]
  99. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011;20:576–590. doi: 10.1016/j.ccr.2011.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell. 1998;93:165–176. doi: 10.1016/s0092-8674(00)81569-x. [DOI] [PubMed] [Google Scholar]
  101. Lam J, Takeshita S, Barker JE, Kanagawa O, Ross FP, Teitelbaum SL. TNF-alpha induces osteoclastogenesis by direct stimulation of macrophages exposed to permissive levels of RANK ligand. J Clin Invest. 2000;106:1481–1488. doi: 10.1172/JCI11176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Lamontanara AJ, Gencer EB, Kuzyk O, Hantschel O. Mechanisms of resistance to BCR-ABL and other kinase inhibitors. Biochim Biophys Acta. 2013;1834:1449–1459. doi: 10.1016/j.bbapap.2012.12.009. [DOI] [PubMed] [Google Scholar]
  103. Lau YS, Sabokbar A, Giele H, Cerundolo V, Hofstetter W, Athanasou NA. Malignant melanoma and bone resorption. Br J Cancer. 2006;94:1496–1503. doi: 10.1038/sj.bjc.6603103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Le Gall C, Bellahcene A, Bonnelye E, Gasser JA, Castronovo V, Green J, Zimmermann J, Clezardin P. A cathepsin K inhibitor reduces breast cancer induced osteolysis and skeletal tumor burden. Cancer Res. 2007;67:9894–9902. doi: 10.1158/0008-5472.CAN-06-3940. [DOI] [PubMed] [Google Scholar]
  105. Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman BE, Welch DR. KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J Natl Cancer Inst. 1996;88:1731–1737. doi: 10.1093/jnci/88.23.1731. [DOI] [PubMed] [Google Scholar]
  106. Lehr JE, Pienta KJ. Preferential adhesion of prostate cancer cells to a human bone marrow endothelial cell line. J Natl Cancer Inst. 1998;90:118–123. doi: 10.1093/jnci/90.2.118. [DOI] [PubMed] [Google Scholar]
  107. Liang Z, Wu T, Lou H, Yu X, Taichman RS, Lau SK, Nie S, Umbreit J, Shim H. Inhibition of breast cancer metastasis by selective synthetic polypeptide against CXCR4. Cancer Res. 2004;64:4302–4308. doi: 10.1158/0008-5472.CAN-03-3958. [DOI] [PubMed] [Google Scholar]
  108. Lind AJ, Wikstrom P, Granfors T, Egevad L, Stattin P, Bergh A. Angiopoietin 2 expression is related to histological grade, vascular density, metastases, and outcome in prostate cancer. Prostate. 2005;62:394–399. doi: 10.1002/pros.20163. [DOI] [PubMed] [Google Scholar]
  109. Loberg RD, Ying C, Craig M, Day LL, Sargent E, Neeley C, Wojno K, Snyder LA, Yan L, Pienta KJ. Targeting CCL2 with Systemic Delivery of Neutralizing Antibodies Induces Prostate Cancer Tumor Regression In vivo. Cancer Research. 2007;67:9417–9424. doi: 10.1158/0008-5472.CAN-07-1286. [DOI] [PubMed] [Google Scholar]
  110. Lode HN, Xiang R, Dreier T, Varki NM, Gillies SD, Reisfeld RA. Natural killer cell-mediated eradication of neuroblastoma metastases to bone marrow by targeted interleukin-2 therapy. Blood. 1998;91:1706–1715. [PubMed] [Google Scholar]
  111. Lu X, Kang Y. Chemokine (C-C motif) ligand 2 engages CCR2+ stromal cells of monocytic origin to promote breast cancer metastasis to lung and bone. J Biol Chem. 2009a;284:29087–29096. doi: 10.1074/jbc.M109.035899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Lu X, Kang Y. Chemokine (C-C Motif) Ligand 2 Engages CCR2+ Stromal Cells of Monocytic Origin to Promote Breast Cancer Metastasis to Lung and Bone. Journal of Biological Chemistry. 2009b;284:29087–29096. doi: 10.1074/jbc.M109.035899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Lu X, Mu E, Wei Y, Riethdorf S, Yang Q, Yuan M, Yan J, Hua Y, Tiede Benjamin J, Lu X, Haffty Bruce G, Pantel K, Massagué J, Kang Y. VCAM-1 Promotes Osteolytic Expansion of Indolent Bone Micrometastasis of Breast Cancer by Engaging α4β1-Positive Osteoclast Progenitors. Cancer Cell. 2011;20:701–714. doi: 10.1016/j.ccr.2011.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Lu X, Wang Q, Hu G, Van Poznak C, Fleisher M, Reiss M, Massague J, Kang Y. ADAMTS1 and MMP1 proteolytically engage EGF-like ligands in an osteolytic signaling cascade for bone metastasis. Genes & Development. 2009;23:1882–1894. doi: 10.1101/gad.1824809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Lu Y, Cai Z, Xiao G, Liu Y, Keller ET, Yao Z, Zhang J. CCR2 expression correlates with prostate cancer progression. Journal of Cellular Biochemistry. 2007;101:676–685. doi: 10.1002/jcb.21220. [DOI] [PubMed] [Google Scholar]
  116. Lu Y, Chen Q, Corey E, Xie W, Fan J, Mizokami A, Zhang J. Activation of MCP-1/CCR2 axis promotes prostate cancer growth in bone. Clinical & Experimental Metastasis. 2008;26:161–169. doi: 10.1007/s10585-008-9226-7. [DOI] [PubMed] [Google Scholar]
  117. Lu Y, Xiao G, Galson DL, Nishio Y, Mizokami A, Keller ET, Yao Z, Zhang J. PTHrP-induced MCP-1 production by human bone marrow endothelial cells and osteoblasts promotes osteoclast differentiation and prostate cancer cell proliferation and invasion in vitro. Int J Cancer. 2007;121:724–733. doi: 10.1002/ijc.22704. [DOI] [PubMed] [Google Scholar]
  118. Lynch CC, Hikosaka A, Acuff HB, Martin MD, Kawai N, Singh RK, Vargo-Gogola TC, Begtrup JL, Peterson TE, Fingleton B, Shirai T, Matrisian LM, Futakuchi M. MMP-7 promotes prostate cancer-induced osteolysis via the solubilization of RANKL. Cancer Cell. 2005;7:485–496. doi: 10.1016/j.ccr.2005.04.013. [DOI] [PubMed] [Google Scholar]
  119. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN, Yancopoulos GD. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science. 1997;277:55–60. doi: 10.1126/science.277.5322.55. [DOI] [PubMed] [Google Scholar]
  120. Matsuura N, Puzon-McLaughlin W, Irie A, Morikawa Y, Kakudo K, Takada Y. Induction of experimental bone metastasis in mice by transfection of integrin alpha 4 beta 1 into tumor cells. Am J Pathol. 1996;148:55–61. [PMC free article] [PubMed] [Google Scholar]
  121. McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince TA, Reinhardt F, Harris LN, Hylander BL, Repasky EA, Weinberg RA. Systemic Endocrine Instigation of Indolent Tumor Growth Requires Osteopontin. Cell. 2008;133:994–1005. doi: 10.1016/j.cell.2008.04.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Melchior SW, Corey E, Ellis WJ, Ross AA, Layton TJ, Oswin MM, Lange PH, Vessella RL. Early tumor cell dissemination in patients with clinically localized carcinoma of the prostate. Clin Cancer Res. 1997;3:249–256. [PubMed] [Google Scholar]
  123. Méndez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, MacArthur BD, Lira SA, Scadden DT, Ma’ayan A, Enikolopov GN, Frenette PS. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829–834. doi: 10.1038/nature09262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Miyake K, Medina K, Ishihara K, Kimoto M, Auerbach R, Kincade PW. A VCAM-like adhesion molecule on murine bone marrow stromal cells mediates binding of lymphocyte precursors in culture. J Cell Biol. 1991;114:557–565. doi: 10.1083/jcb.114.3.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Mohammad KS, Javelaud D, Fournier PG, Niewolna M, McKenna CR, Peng XH, Duong V, Dunn LK, Mauviel A, Guise TA. TGF-beta-RI kinase inhibitor SD-208 reduces the development and progression of melanoma bone metastases. Cancer Res. 2011;71:175–184. doi: 10.1158/0008-5472.CAN-10-2651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Morony S, Capparelli C, Sarosi I, Lacey DL, Dunstan CR, Kostenuik PJ. Osteoprotegerin inhibits osteolysis and decreases skeletal tumor burden in syngeneic and nude mouse models of experimental bone metastasis. Cancer Res. 2001;61:4432–4436. [PubMed] [Google Scholar]
  127. Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410:50–56. doi: 10.1038/35065016. [DOI] [PubMed] [Google Scholar]
  128. Muller M, Gounari F, Prifti S, Hacker HJ, Schirrmacher V, Khazaie K. EblacZ tumor dormancy in bone marrow and lymph nodes: active control of proliferating tumor cells by CD8+ immune T cells. Cancer Res. 1998;58:5439–5446. [PubMed] [Google Scholar]
  129. Mundy GR. Metastasis: Metastasis to bone: causes, consequences and therapeutic opportunities. Nature Reviews Cancer. 2002;2:584–593. doi: 10.1038/nrc867. [DOI] [PubMed] [Google Scholar]
  130. Murray LJ, Abrams TJ, Long KR, Ngai TJ, Olson LM, Hong W, Keast PK, Brassard JA, O’Farrell AM, Cherrington JM, Pryer NK. SU11248 inhibits tumor growth and CSF-1R-dependent osteolysis in an experimental breast cancer bone metastasis model. Clin Exp Metastasis. 2003;20:757–766. doi: 10.1023/b:clin.0000006873.65590.68. [DOI] [PubMed] [Google Scholar]
  131. Nash KT, Phadke PA, Navenot JM, Hurst DR, Accavitti-Loper MA, Sztul E, Vaidya KS, Frost AR, Kappes JC, Peiper SC, Welch DR. Requirement of KISS1 Secretion for Multiple Organ Metastasis Suppression and Maintenance of Tumor Dormancy. JNCI Journal of the National Cancer Institute. 2007;99:309–321. doi: 10.1093/jnci/djk053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Nelson JB, Hedican SP, George DJ, Reddi AH, Piantadosi S, Eisenberger MA, Simons JW. Identification of endothelin-1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nat Med. 1995;1:944–949. doi: 10.1038/nm0995-944. [DOI] [PubMed] [Google Scholar]
  133. Nemeth JA, Cher ML, Zhou Z, Mullins C, Bhagat S, Trikha M. Inhibition of alpha(v)beta3 integrin reduces angiogenesis, bone turnover, and tumor cell proliferation in experimental prostate cancer bone metastases. Clin Exp Metastasis. 2003;20:413–420. doi: 10.1023/a:1025461507027. [DOI] [PubMed] [Google Scholar]
  134. Nervi B, Ramirez P, Rettig MP, Uy GL, Holt MS, Ritchey JK, Prior JL, Piwnica-Worms D, Bridger G, Ley TJ, DiPersio JF. Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100. Blood. 2009;113:6206–6214. doi: 10.1182/blood-2008-06-162123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Nieswandt B, Hafner M, Echtenacher B, Mannel DN. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 1999;59:1295–1300. [PubMed] [Google Scholar]
  136. Normanno N, De Luca A, Aldinucci D, Maiello MR, Mancino M, D’Antonio A, De Filippi R, Pinto A. Gefitinib inhibits the ability of human bone marrow stromal cells to induce osteoclast differentiation: implications for the pathogenesis and treatment of bone metastasis. Endocr Relat Cancer. 2005;12:471–482. doi: 10.1677/erc.1.00956. [DOI] [PubMed] [Google Scholar]
  137. O’Day SJ, Hamid O, Urba WJ. Targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4): a novel strategy for the treatment of melanoma and other malignancies. Cancer. 2007;110:2614–2627. doi: 10.1002/cncr.23086. [DOI] [PubMed] [Google Scholar]
  138. Oba Y, Lee JW, Ehrlich LA, Chung HY, Jelinek DF, Callander NS, Horuk R, Choi SJ, Roodman GD. MIP-1alpha utilizes both CCR1 and CCR5 to induce osteoclast formation and increase adhesion of myeloma cells to marrow stromal cells. Exp Hematol. 2005;33:272–278. doi: 10.1016/j.exphem.2004.11.015. [DOI] [PubMed] [Google Scholar]
  139. Ohno H, Kubo K, Murooka H, Kobayashi Y, Nishitoba T, Shibuya M, Yoneda T, Isoe T. A c-fms tyrosine kinase inhibitor, Ki20227, suppresses osteoclast differentiation and osteolytic bone destruction in a bone metastasis model. Mol Cancer Ther. 2006;5:2634–2643. doi: 10.1158/1535-7163.MCT-05-0313. [DOI] [PubMed] [Google Scholar]
  140. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, Phinney DG. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A. 2003;100:8407–8411. doi: 10.1073/pnas.1432929100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Ostrand-Rosenberg S, Grusby MJ, Clements VK. Cutting edge: STAT6-deficient mice have enhanced tumor immunity to primary and metastatic mammary carcinoma. J Immunol. 2000;165:6015–6019. doi: 10.4049/jimmunol.165.11.6015. [DOI] [PubMed] [Google Scholar]
  142. Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 1989;8:98–101. [PubMed] [Google Scholar]
  143. Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, Hu Z, Barney KA, Degen JL. Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood. 2007;110:133–141. doi: 10.1182/blood-2007-01-065995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, Jirouskova M, Degen JL. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood. 2005;105:178–185. doi: 10.1182/blood-2004-06-2272. [DOI] [PubMed] [Google Scholar]
  145. Park BK, Zhang H, Zeng Q, Dai J, Keller ET, Giordano T, Gu K, Shah V, Pei L, Zarbo RJ, McCauley L, Shi S, Chen S, Wang CY. NF-kappaB in breast cancer cells promotes osteolytic bone metastasis by inducing osteoclastogenesis via GM-CSF. Nat Med. 2007;13:62–69. doi: 10.1038/nm1519. [DOI] [PubMed] [Google Scholar]
  146. Peinado H, Ale kovi M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, García-Santos G, Ghajar CM, Nitadori-Hoshino A, Hoffman C, Badal K, Garcia BA, Callahan MK, Yuan J, Martins VR, Skog J, Kaplan RN, Brady MS, Wolchok JD, Chapman PB, Kang Y, Bromberg J, Lyden D. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nature Medicine. 2012;18:883–891. doi: 10.1038/nm.2753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Pienta KJ, Naik H, Akhtar A, Yamazaki K, Replogle TS, Lehr J, Donat TL, Tait L, Hogan V, Raz A. Inhibition of spontaneous metastasis in a rat prostate cancer model by oral administration of modified citrus pectin. J Natl Cancer Inst. 1995;87:348–353. doi: 10.1093/jnci/87.5.348. [DOI] [PubMed] [Google Scholar]
  148. Powles TJ, Dowsett M, Easty GC, Easty DM, Neville AM. Breast-cancer osteolysis, bone metastases, and anti-osteolytic effect of aspirin. Lancet. 1976;1:608–610. doi: 10.1016/s0140-6736(76)90416-5. [DOI] [PubMed] [Google Scholar]
  149. Previdi S, Abbadessa G, Dalo F, France DS, Broggini M. Breast Cancer-Derived Bone Metastasis Can Be Effectively Reduced through Specific c-MET Inhibitor Tivantinib (ARQ 197) and shRNA c-MET Knockdown. Molecular Cancer Therapeutics. 2011;11:214–223. doi: 10.1158/1535-7163.MCT-11-0277. [DOI] [PubMed] [Google Scholar]
  150. Quesada JR, Swanson DA, Trindade A, Gutterman JU. Renal cell carcinoma: antitumor effects of leukocyte interferon. Cancer Res. 1983;43:940–947. [PubMed] [Google Scholar]
  151. Roodman GD. Mechanisms of bone metastasis. N Engl J Med. 2004;350:1655–1664. doi: 10.1056/NEJMra030831. [DOI] [PubMed] [Google Scholar]
  152. Rosen LS, Gordon D, Kaminski M, Howell A, Belch A, Mackey J, Apffelstaedt J, Hussein M, Coleman RE, Reitsma DJ, Seaman JJ, Chen BL, Ambros Y. Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial. Cancer J. 2001;7:377–387. [PubMed] [Google Scholar]
  153. Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA. 1994;271:907–913. [PubMed] [Google Scholar]
  154. Salgia R, Lynch T, Skarin A, Lucca J, Lynch C, Jung K, Hodi FS, Jaklitsch M, Mentzer S, Swanson S, Lukanich J, Bueno R, Wain J, Mathisen D, Wright C, Fidias P, Donahue D, Clift S, Hardy S, Neuberg D, Mulligan R, Webb I, Sugarbaker D, Mihm M, Dranoff G. Vaccination with irradiated autologous tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor augments antitumor immunity in some patients with metastatic non-small-cell lung carcinoma. J Clin Oncol. 2003;21:624–630. doi: 10.1200/JCO.2003.03.091. [DOI] [PubMed] [Google Scholar]
  155. Sänger N, Effenberger KE, Riethdorf S, Van Haasteren V, Gauwerky J, Wiegratz I, Strebhardt K, Kaufmann M, Pantel K. Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ. International Journal of Cancer. 2011;129:2522–2526. doi: 10.1002/ijc.25895. [DOI] [PubMed] [Google Scholar]
  156. Sasaki H, Yu CY, Dai M, Tam C, Loda M, Auclair D, Chen LB, Elias A. Elevated serum periostin levels in patients with bone metastases from breast but not lung cancer. Breast Cancer Res Treat. 2003;77:245–252. doi: 10.1023/a:1021899904332. [DOI] [PubMed] [Google Scholar]
  157. Sawant A, Hensel JA, Chanda D, Harris BA, Siegal GP, Maheshwari A, Ponnazhagan S. Depletion of Plasmacytoid Dendritic Cells Inhibits Tumor Growth and Prevents Bone Metastasis of Breast Cancer Cells. The Journal of Immunology. 2012;189:4258–4265. doi: 10.4049/jimmunol.1101855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Sawant A, Ponnazhagan S. Myeloid-derived suppressor cells as a novel target for the control of osteolytic bone disease. OncoImmunology. 2013;2:e24064. doi: 10.4161/onci.24064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Schneider JG, Amend SR, Weilbaecher KN. Integrins and bone metastasis: Integrating tumor cell and stromal cell interactions. Bone. 2011;48:54–65. doi: 10.1016/j.bone.2010.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Schumacher D, Strilic B, Sivaraj KK, Wettschureck N, Offermanns S. Platelet-Derived Nucleotides Promote Tumor-Cell Transendothelial Migration and Metastasis via P2Y2 Receptor. Cancer Cell. 2013;24:130–137. doi: 10.1016/j.ccr.2013.05.008. [DOI] [PubMed] [Google Scholar]
  161. Senger DR, Perruzzi CA, Papadopoulos A. Elevated expression of secreted phosphoprotein I (osteopontin, 2ar) as a consequence of neoplastic transformation. Anticancer Res. 1989;9:1291–1299. [PubMed] [Google Scholar]
  162. Sethi N, Dai X, Winter CG, Kang Y. Tumor-Derived Jagged1 Promotes Osteolytic Bone Metastasis of Breast Cancer by Engaging Notch Signaling in Bone Cells. Cancer Cell. 2011;19:192–205. doi: 10.1016/j.ccr.2010.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Shannon KE, Keene JL, Settle SL, Duffin TD, Nickols MA, Westlin M, Schroeter S, Ruminski PG, Griggs DW. Anti-metastatic properties of RGD-peptidomimetic agents S137 and S247. Clin Exp Metastasis. 2004;21:129–138. doi: 10.1023/b:clin.0000024764.93092.5f. [DOI] [PubMed] [Google Scholar]
  164. Shiao SL, Ganesan AP, Rugo HS, Coussens LM. Immune microenvironments in solid tumors: new targets for therapy. Genes Dev. 2011;25:2559–2572. doi: 10.1101/gad.169029.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Shiozawa Y, Pedersen EA, Havens AM, Jung Y, Mishra A, Joseph J, Kim JK, Patel LR, Ying C, Ziegler AM, Pienta MJ, Song J, Wang J, Loberg RD, Krebsbach PH, Pienta KJ, Taichman RS. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J Clin Invest. 2011;121:1298–1312. doi: 10.1172/JCI43414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Simeone E, Ascierto PA. Immunomodulating antibodies in the treatment of metastatic melanoma: the experience with anti-CTLA-4, anti-CD137, and anti-PD1. J Immunotoxicol. 2012;9:241–247. doi: 10.3109/1547691X.2012.678021. [DOI] [PubMed] [Google Scholar]
  167. Sipkins DA, Wei X, Wu JW, Runnels JM, Cote D, Means TK, Luster AD, Scadden DT, Lin CP. 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]
  168. Sisirak V, Faget J, Vey N, Blay JY, Ménétrier-Caux C, Caux C, Bendriss-Vermare N. Plasmacytoid dendritic cells deficient in IFNα production promote the amplification of FOXP3+ regulatory T cells and are associated with poor prognosis in breast cancer patients. OncoImmunology. 2013;2:e22338. doi: 10.4161/onci.22338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Sloan EK, Pouliot N, Stanley KL, Chia J, Moseley JM, Hards DK, Anderson RL. Tumor-specific expression of alphavbeta3 integrin promotes spontaneous metastasis of breast cancer to bone. Breast Cancer Res. 2006;8:R20. doi: 10.1186/bcr1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Smith HA, Kang Y. The metastasis-promoting roles of tumor-associated immune cells. Journal of Molecular Medicine. 2013;91:411–429. doi: 10.1007/s00109-013-1021-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Solakoglu O, Maierhofer C, Lahr G, Breit E, Scheunemann P, Heumos I, Pichlmeier U, Schlimok G, Oberneder R, Kollermann MW, Kollermann J, Speicher MR, Pantel K. Heterogeneous proliferative potential of occult metastatic cells in bone marrow of patients with solid epithelial tumors. Proc Natl Acad Sci U S A. 2002;99:2246–2251. doi: 10.1073/pnas.042372199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Sosa MS, Avivar-Valderas A, Bragado P, Wen HC, Aguirre-Ghiso JA. ERK1/2 and p38alpha/beta signaling in tumor cell quiescence: opportunities to control dormant residual disease. Clin Cancer Res. 2011;17:5850–5857. doi: 10.1158/1078-0432.CCR-10-2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Steeg PS. Perspective: The right trials. Nature. 2012;485:S58–59. doi: 10.1038/485S58a. [DOI] [PubMed] [Google Scholar]
  174. Steinman RA, Brufsky AM, Oesterreich S. Zoledronic acid effectiveness against breast cancer metastases - a role for estrogen in the microenvironment? Breast Cancer Res. 2012;14:213. doi: 10.1186/bcr3223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Sung B, Oyajobi B, Aggarwal BB. Plumbagin Inhibits Osteoclastogenesis and Reduces Human Breast Cancer-Induced Osteolytic Bone Metastasis in Mice through Suppression of RANKL Signaling. Molecular Cancer Therapeutics. 2011;11:350–359. doi: 10.1158/1535-7163.MCT-11-0731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Takayama S, Ishii S, Ikeda T, Masamura S, Doi M, Kitajima M. The relationship between bone metastasis from human breast cancer and integrin alpha(v)beta3 expression. Anticancer Res. 2005;25:79–83. [PubMed] [Google Scholar]
  177. Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B, Shaughnessy JD., Jr The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med. 2003;349:2483–2494. doi: 10.1056/NEJMoa030847. [DOI] [PubMed] [Google Scholar]
  178. Tsubaki M, Kato C, Manno M, Ogaki M, Satou T, Itoh T, Kusunoki T, Tanimori Y, Fujiwara K, Matsuoka H, Nishida S. Macrophage inflammatory protein-1alpha (MIP-1alpha) enhances a receptor activator of nuclear factor kappaB ligand (RANKL) expression in mouse bone marrow stromal cells and osteoblasts through MAPK and PI3K/Akt pathways. Mol Cell Biochem. 2007;304:53–60. doi: 10.1007/s11010-007-9485-7. [DOI] [PubMed] [Google Scholar]
  179. van Holten-Verzantvoort AT, Bijvoet OL, Cleton FJ, Hermans J, Kroon HM, Harinck HI, Vermey P, Elte JW, Neijt JP, Beex LV, et al. Reduced morbidity from skeletal metastases in breast cancer patients during long-term bisphosphonate (APD) treatment. Lancet. 1987;2:983–985. doi: 10.1016/s0140-6736(87)92555-4. [DOI] [PubMed] [Google Scholar]
  180. Waltregny D, Bellahcene A, de Leval X, Florkin B, Weidle U, Castronovo V. Increased expression of bone sialoprotein in bone metastases compared with visceral metastases in human breast and prostate cancers. J Bone Miner Res. 2000;15:834–843. doi: 10.1359/jbmr.2000.15.5.834. [DOI] [PubMed] [Google Scholar]
  181. Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal attraction. Nature Reviews Cancer. 2011;11:411–425. doi: 10.1038/nrc3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Withana NP, Blum G, Sameni M, Slaney C, Anbalagan A, Olive MB, Bidwell BN, Edgington L, Wang L, Moin K, Sloane BF, Anderson RL, Bogyo MS, Parker BS. Cathepsin B Inhibition Limits Bone Metastasis in Breast Cancer. Cancer Research. 2012;72:1199–1209. doi: 10.1158/0008-5472.CAN-11-2759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Yang C, Davis JL, Zeng R, Vora P, Su X, Collins LI, Vangveravong S, Mach RH, Piwnica-Worms D, Weilbaecher KN, Faccio R, Veis Novack D. Antagonism of Inhibitor of Apoptosis Proteins Increases Bone Metastasis via Unexpected Osteoclast Activation. Cancer Discovery. 2012;3:212–223. doi: 10.1158/2159-8290.CD-12-0271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Yin JJ, Mohammad KS, Kakonen SM, Harris S, Wu-Wong JR, Wessale JL, Padley RJ, Garrett IR, Chirgwin JM, Guise TA. A causal role for endothelin-1 in the pathogenesis of osteoblastic bone metastases. Proc Natl Acad Sci U S A. 2003;100:10954–10959. doi: 10.1073/pnas.1830978100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Yin JJ, Selander K, Chirgwin JM, Dallas M, Grubbs BG, Wieser R, Massague J, Mundy GR, Guise TA. TGF-beta signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J Clin Invest. 1999;103:197–206. doi: 10.1172/JCI3523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Yoneda T, Sasaki A, Dunstan C, Williams PJ, Bauss F, De Clerck YA, Mundy GR. Inhibition of osteolytic bone metastasis of breast cancer by combined treatment with the bisphosphonate ibandronate and tissue inhibitor of the matrix metalloproteinase-2. J Clin Invest. 1997;99:2509–2517. doi: 10.1172/JCI119435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Young MR, Wright MA, Young ME. Antibodies to colony-stimulating factors block Lewis lung carcinoma cell stimulation of immune-suppressive bone marrow cells. Cancer Immunol Immunother. 1991;33:146–152. doi: 10.1007/BF01756134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, Isakoff SJ, Ciciliano JC, Wells MN, Shah AM, Concannon KF, Donaldson MC, Sequist LV, Brachtel E, Sgroi D, Baselga J, Ramaswamy S, Toner M, Haber DA, Maheswaran S. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339:580–584. doi: 10.1126/science.1228522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Zhang XH, Wang Q, Gerald W, Hudis CA, Norton L, Smid M, Foekens JA, Massague J. Latent bone metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell. 2009;16:67–78. doi: 10.1016/j.ccr.2009.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Zhao E, Wang L, Dai J, Kryczek I, Wei S, Vatan L, Altuwaijri S, Sparwasser T, Wang G, Keller ET, Zou W. Regulatory T cells in the bone marrow microenvironment in patients with prostate cancer. OncoImmunology. 2012;1:152–161. doi: 10.4161/onci.1.2.18480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Zohar R, Suzuki N, Suzuki K, Arora P, Glogauer M, McCulloch CA, Sodek J. Intracellular osteopontin is an integral component of the CD44-ERM complex involved in cell migration. J Cell Physiol. 2000;184:118–130. doi: 10.1002/(SICI)1097-4652(200007)184:1<118::AID-JCP13>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  192. Zou L, Barnett B, Safah H, Larussa VF, Evdemon-Hogan M, Mottram P, Wei S, David O, Curiel TJ, Zou W. Bone marrow is a reservoir for CD4+CD25+ regulatory T cells that traffic through CXCL12/CXCR4 signals. Cancer Res. 2004;64:8451–8455. doi: 10.1158/0008-5472.CAN-04-1987. [DOI] [PubMed] [Google Scholar]

RESOURCES