ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Mechanisms of bone metastases

Mechanisms of bone metastases
Literature review current through: Jan 2024.
This topic last updated: Nov 01, 2022.

INTRODUCTION — Bone is the third most frequent site of tumor metastasis; it is estimated that in the United States approximately 350,000 people die each year from bone metastases [1]. Analysis of medical records has demonstrated that 8.4 percent of patients with solid tumors have bone metastases by ten years from the initial diagnosis [2].

The incidence of bone metastases varies based on age, sex, and the primary tumor site [3,4]. Bone is the preferred site for breast and prostate cancer metastasis, with 50 to 70 percent of patients with advanced disease having bone involvement [5,6]. Many other cancers, including lung cancer [7], renal cell cancer, and melanomas, also colonize the skeleton prior to metastasizing to other organs [2]. For example, in autopsy series, 30 to 40 percent of patients with carcinomas of the thyroid, kidney, and bronchus have bone metastases [8]. Although hematologic malignancies do not frequently cause bone disease, multiple myeloma is the most frequent cancer to involve bone, with 70 percent of patients presenting with bone involvement at diagnosis and over 80 percent of patients having bone involvement with advanced disease [9,10]. Importantly, the presence of cancer in bone has been termed "the lethal phenotype" [11] as patients are rarely curable once bone is involved.

Normal bone remodeling is markedly disturbed in bone metastasis, with imbalances in both osteoclast and osteoblast numbers and activity [12]. These disturbances in the bone remodeling process can result in debilitating skeletal-related events (SREs), which are common complications (31 to 47 percent of patients with breast or prostate cancer or myeloma [13]) that have catastrophic sequelae for patients. SREs include severe bone pain, pathologic fractures, spinal cord and nerve compression syndromes, and derangements of calcium and phosphate homeostasis that can result in life-threatening hypercalcemia [14]. Because bone involvement is the main source of cancer-related pain and is the major cause of severe pain for patients with advanced malignancies [15], SREs not only greatly increase morbidity and mortality [16], but also diminish quality of life for patients, which further impacts survival.

In addition to SREs, a significant proportion of patients with bone metastases have systemic muscle weakness that results from cancer-related bone destruction [17], which increases the risk of falls, which can result in fractures, and negatively impacts performance status, survival, and quality of life. Advances in our understanding of the mechanisms responsible for bone metastasis have provided the basis for development of mechanistic-based therapies to treat these patients and potentially reverse or prevent the severe sequelae of cancer in bone. Several of these therapies are in clinical trials. (See "Overview of cancer pain syndromes", section on 'Multifocal bone pain' and "Clinical presentation and evaluation of complete and impending pathologic fractures in patients with metastatic bone disease, multiple myeloma, and lymphoma" and "Clinical features and diagnosis of neoplastic epidural spinal cord compression" and "Hypercalcemia of malignancy: Mechanisms".)

This topic will review the mechanisms by which bone metastases develop and affect normal bone. Control of normal bone remodeling is discussed separately. (See "Normal skeletal development and regulation of bone formation and resorption".)

OSTEOLYTIC VERSUS OSTEOBLASTIC BONE METASTASES — Normal bone constantly undergoes a remodeling process that involves the resorption of bone by osteoclasts and the deposition of new bone by osteoblasts. The bone remodeling process, including osteoclast and osteoblast activity, is regulated by the osteocyte [18].

Bone metastases are generally classified as osteolytic or osteoblastic based upon a radiologic appearance that demonstrates predominant bone destruction or deposition of new bone, respectively. However, this distinction is not absolute, and many patients have a mixed picture of both osteolytic and osteoblastic metastases (table 1). Both types of bone metastases are characterized by dysregulation of the normal bone remodeling process. (See "Normal skeletal development and regulation of bone formation and resorption", section on 'Bone modeling and remodeling'.)

The spectrum of osteolytic and osteoblastic bone involvement is illustrated by three malignancies that are frequently associated with skeletal metastases:

Multiple myeloma – The classic bone lesions in multiple myeloma are purely osteolytic due to increased bone destruction and suppressed bone formation (image 1) [19]. These lesions are often not detectable on bone scans, which rely upon technetium uptake to identify areas of new bone formation. (See "Multiple myeloma: Clinical features, laboratory manifestations, and diagnosis" and "Epidemiology, clinical presentation, and diagnosis of bone metastasis in adults", section on 'Bone scan'.)

Prostate cancer – Males with bone metastases from prostate cancer predominantly have osteoblastic lesions with increased numbers of irregular bone trabeculae (image 2) [20]. Although these metastases are osteoblastic, bone resorption is also increased [21,22]. Thus, agents that block bone resorption can decrease bone pain and the risk of skeletal-related events in patients with metastatic prostate cancer [23]. (See "Bone metastases in advanced prostate cancer: Management", section on 'Osteoclast inhibitors'.)

Breast cancer – Although bone metastases due to breast cancer are predominantly osteolytic, osteoblastic areas are also usually present, and at least 15 to 20 percent of cases have predominantly osteoblastic lesions [20,24]. In addition, the bone destruction in osteolytic lesions can induce secondary new bone formation.

BONE AS A PREFERENTIAL SITE FOR METASTASIS — Multiple mechanisms have been identified that increase the potential for tumor cells to metastasize to bone. The intrinsic properties of the tumor cells, changes induced by the tumor cells or their products in the bone microenvironment, and the bone microenvironment itself all contribute to the preferential colonization and growth of cancer cells in bone [25]. Stephen Paget in 1889 published the "seed and soil" hypothesis [26], in which he proposed that cancer cells only grow in "congenial" secondary sites, to explain his autopsy findings that bone was a preferred site for breast cancer metastasis. This hypothesis went against the prevailing belief that when cancer cells spread from the primary site, they would grow similarly in any tissue that they "seeded."

Intrinsic tumor cell properties — In preclinical studies of mice injected with human breast cancer cells, breast cancer clones with a high propensity to induce bone metastasis were found to express a specific four-gene signature that increased their bone metastatic potential [27]. These genes included CXCR4 (CXC motif chemokine 4), which binds CXCL12 (the product of the CXC motif chemokine ligand 12 gene) on bone cells to increase bone homing, fibroblast growth factor 5 (an angiogenic factor), interleukin 11 (IL-11; an osteoclast stimulatory factor), and osteopontin. Breast cancer cells that expressed at least three of these four genes had an increased propensity to metastasize to bone. Validation studies are needed to confirm these findings.

The investigators also found that increased expression of both matrix metalloproteinase 1 (MMP1) and A disintegrin-like and metalloproteinase with thrombospondin type 1 motif 1 (ADAMTS1) in breast cancer cells further enhanced their bone metastatic potential. MMP1 and ADAMTS1 cooperated to cleave amphiregulin (AREG), an epidermal growth factor family member, from the breast cancer cell membrane. AREG in turn increased bone resorption by suppressing osteoprotegerin (OPG) expression by osteoblasts [28]. OPG is a decoy receptor for receptor activator of nuclear factor kappa-B ligand (RANKL) that blocks its capacity to induce osteoclast formation and activity and thereby increase bone resorption. Increased bone resorption enhances bone metastasis in breast cancer [29] and prostate cancer [30].

Changes induced by tumor cells or their products in the bone microenvironment — Several changes induced by tumor cells or their products in the bone microenvironment have been implicated in the propensity of tumor cells to metastasize to bone. As examples:

Increased transforming growth factor (TGF)-beta signaling in the tumor cells plays a crucial role in prostate cancer and breast cancer bone metastasis. TGF-beta controls expression of multiple genes that promote bone metastasis, including CXCR4, MMP1, IL-11, Jagged 1 (JAG1), and parathyroid hormone-like hormone, in breast and prostate cancer cells [31,32]. Others have found that TGF-beta also induces expression of an inhibitor of TGF-beta signaling in prostate cancer cells [33]. PMEPA1 (prostate transmembrane protein, androgen induced) expression was higher at the primary tumor site than in bone metastases, and decreased expression of PMEPA1 in the bone metastases was associated with decreased survival and cancer recurrence in patients with breast, lung, and prostate cancer. The decreased expression of PMEPA1 in prostate cancer cells in bone most likely resulted from methylation of the PMEPA1 gene that inhibited PMEPA1 expression. Finally, they showed that increased TGF-beta signaling was required for formation of osteolytic bone metastases by the prostate cancer cells.

Tumor-associated stromal cells in the primary tumor site also can increase the bone metastatic potential of breast cancer cells [34]. One study found that cancer-associated fibroblasts bias the heterogeneous population of breast cancer cells present in the primary tumor toward clones that express SRC (the product of the SRC oncogene), which allows the cancer cells to respond to the CAF-derived factors CXCL12 and insulin-like growth factor 1 (IGF1) [34,35]. Because limiting concentrations of these factors are available at the primary tumor site, cancer cells with high SRC activity are primed for metastasis to the CXCL12-rich bone microenvironment that is present in the bone marrow. SRC activity promotes breast cancer cell growth and survival in bone marrow [36].

Increased expression of adhesive molecules, cytokine receptors, and receptor ligands on the surface of tumor cells also plays an important role in bone metastasis. CXCR4, which binds to CXCL12 and is expressed on the surface of pericytes and bone marrow stromal cells in bone, is a homing receptor for breast cancer, prostate cancer, and multiple myeloma cells to bone [37]. Tumor cells expressing alpha-v beta-3 integrin or E-cadherin can also home to the bone marrow by binding osteopontin, bone sialoprotein, vitronectin, or N-cadherin, respectively [38]. Similarly, multiple myeloma cells express alpha-4 beta-1 integrin to bind vascular cell adhesion molecule 1 on marrow stromal cells [39]. Additionally, breast cancer, melanoma, and prostate cancer cells express RANK (the receptor for RANKL), which promotes their homing to the marrow [40].

Multiple products of tumor cells also prepare the premetastatic niche in bone for tumor cell colonization [41]. These include lysyl oxidase, which increases osteoclast activity; extracellular vesicles (EVs)containing micro-RNAs that induce angiogenesis or increase osteoclast activity [42]; and factors that induce immune suppression in the bone microenvironment by increasing regulatory T cells and myeloid suppressor cells that enhance tumor colonization of bone [43,44].

Tumor cell-derived EVs, including exosomes and microvesicles, contain a variety of macromolecules such as proteins, mRNA, and miRNA. EV release from the primary tumor have been shown to target the bone marrow and establish a premetastatic niche that promotes targeting and growth of cancer cells in the bone microenvironment [45-47].

Cross talk between macrophages and cancer cells in the bone microenvironment promote metastasis progression [48]. Macrophages in the bone marrow and primary tumor site can be activated by tumor cells to change from M1 (antitumorigenic) to M2 (protumorigenic) macrophages [49]. This increase in M2 macrophages in bone contributes to the growth of prostate cancer bone metastases [50]. Further, TGF-beta can induce expression of the notch receptor ligand JAG1 on breast cancer cells to activate notch signaling in osteoblasts and increase production of interleukin 6 (IL-6) [32]. IL-6 enhances tumor growth in bone and increases the size of lytic lesions. In keeping with the importance of this pathway, investigators have developed a therapeutic antibody targeting JAG1 on tumor cells that successfully blocked breast cancer bone metastasis in a preclinical model and sensitized the bone metastases to chemotherapy [51].

Cytokines produced by the primary tumor or the bone microenvironment, such as TGF-beta, also upregulate expression of adhesion molecules on bone marrow stromal cells and osteoblasts, and interact with hypoxia-inducible factor 1 alpha to increase vascular endothelial growth factor and CXCR4 expression in the bone microenvironment, which increases bone metastasis [52].

Physical characteristics of bone — The physical characteristics of bone may also contribute to tumor cell colonization.

The bone extracellular matrix is extremely rigid. Exposure to the high matrix rigidity in bone increases levels of the GLI-Kruppel family member 2 (GLI2) gene transcription factor in breast cancer cells. GLI2 in turn increases parathyroid hormone-related protein promoter activity and TGF-beta signaling, which promote cancer growth in bone [53].

Osteocytes mediate mechanotransduction (ie, the ability to sense and induce cell signaling secondary to mechanical forces). As tumor cells grow, they can induce pressure in the bone microenvironment. The increase in pressure is exacerbated by the rigidness of bone. Intraosseous pressure has been shown to stimulate osteocytes to produce the C-C motif chemokine ligand and MMPs that promote cancer growth and invasion in bone [54].

CONTRIBUTION OF BONE CELLS TO TUMOR CELL DORMANCY, REACTIVATION, AND TUMOR PROGRESSION IN BONE — In the skeleton, the multistep process of metastasis development begins with colonization, when circulating tumor cells enter the bone marrow compartment and engage in specialized microenvironments or niches [55,56]. The second step involves survival and dormancy in that the colonizing tumor cells adapt to their new microenvironment, evade the immune system, and reside in a dormant state for long periods of time [57]. The third step, reactivation and development, requires the ability to escape from the dormant state to actively proliferate and form micrometastases [58]. The final step occurs when cells grow uncontrollably, become independent of the microenvironment, and ultimately modify bone as the metastases flourish.

The molecular mechanisms responsible for both osteolytic and osteoblastic metastases are being identified. The use of gene arrays and proteomics, and the availability of appropriate animal models of bone metastasis have permitted the identification of factors, produced by the tumor cells themselves or by the bone cells or bone microenvironment in response to the tumor, that mediate dormancy and reactivation of tumor cells in bone and the bone destructive process. Advances in understanding these pathways have revealed new potentially beneficial therapeutic approaches to target bone metastases [59,60].

Hematopoietic stem cell niches — When tumor cells colonize bone, they appear to localize to the same specific areas in bone as hematopoietic stem cells (HSCs). This area is the osteoblastic niche, which regulates and supports HSC self-renewal, quiescence, and differentiation [60,61]. In support of this concept, investigators have shown that prostate cancer cells directly compete with HSCs for niche support and can displace HSCs from the osteoblastic niche, releasing HSCs into the circulation [62]. Similarly, when myeloma cells home to bone, they also interact with the osteoblastic niche and are retained in a dormant state [63], which protects the myeloma cells from the effects of chemotherapy. Furthermore, metastatic breast cancer cells localize to the hematopoietic stem cell niche that is heavily populated with osteoblasts [64]. This hematopoietic stem cell mimicry is regulated, in part, by Notch 2 in breast cancer cells [65].

The molecular mechanisms responsible for tumor cell dormancy are not well defined, but they may involve binding of the annexin II receptor on myeloma and prostate cancer cells to annexin II expressed on osteoblastic lineage cells in bone [55]. Annexin II binding to myeloma and prostate cancer cells can regulate tumor cell growth [66,67]. This interaction also increases AXL expression in prostate cancer cells, increasing transforming growth factor (TGF)-beta-2 signaling, which is also associated with tumor cell dormancy [68-70]. Others have shown that dormant breast cancer cells in bone reside on the vasculature of metastatic sites, and that tumor cell dormancy in this case is mediated by endothelial-produced thrombospondin-1 [69] and induction of Notch 2 signaling in the breast cancer cells [65].

Our understanding of how dormant tumor cells escape control of the niche is very limited. Osteoclastic bone resorption can reactivate and release dormant myeloma and breast cancer cells from their niches, analogous to the release of HSCs from their niche [42,71,72]. Other data suggest that reactivation may be under tumor cell control, which may be intrinsic to the colonizing tumor cells or acquired once the cells enter the niche [55,73]. In addition, inflammatory activity appears to promote tumor cell plasticity and dormancy escape [74]. A model depicting the various bone marrow niches, cell types, and cues that might regulate the dormancy of disseminated tumor cells is illustrated in the figure (figure 1) [59].

Osteoclasts — Studies in preclinical models have shown that higher levels of osteoclast activity increase tumor metastasis and growth in bone, while blocking osteoclast bone resorption decreases osteolytic metastasis [75,76]. In clinical studies, blocking bone resorption through the use of osteoclast inhibitors reduces skeletal-related events (SREs) and, at least in multiple myeloma, increases the overall survival of patients with bone metastases. By contrast, while antiresorptive agents decrease SREs and improve quality of life, they appear to have limited impact on overall survival in men with prostate cancer bone metastases [77]. (See "Osteoclast inhibitors for patients with bone metastases from breast, prostate, and other solid tumors" and "Multiple myeloma: The use of osteoclast inhibitors", section on 'Indications'.)

Multiple factors that are induced or produced by tumor cells in the bone microenvironment enhance osteoclastic bone resorption [12]. The resulting increased bone destruction releases growth factors, such as TGF-beta, insulin-like growth factor 1 (IGF1), and others, to increase tumor cell growth. This results in a "vicious cycle" (figure 2) whereby tumor cells in bone induce osteoclastic bone resorption that releases growth factors from the bone matrix to further increase tumor growth.

RANK/RANKL signaling pathway and bone remodeling — Products of tumor cells can activate osteoclastic bone resorption in a number of ways. The receptor activator of nuclear factor kappa-B (RANK)/RANK ligand (RANKL) signaling pathway is a major regulator of both normal and pathologic bone remodeling. Many of the cytokines, chemokines, and hormones produced by tumor cells that induce osteolytic metastasis do so by increasing RANKL expression.

RANKL is a type II homotrimeric (a trimer derived from three identical monomers) transmembrane protein that exists as a membrane-bound or secreted soluble protein that is produced by cleavage of the full-length form on the cell surface [78]. In bone, RANKL is mainly expressed by bone marrow stromal cells, osteoblasts, and osteocytes and is also secreted by activated T lymphocytes [12]. Studies suggest that osteocytes are the major source of RANKL production in bone and produce 10-fold higher levels of RANKL than do osteoblasts [79]. RANKL binds to RANK, a member of the tumor necrosis factor (TNF) receptor superfamily, which is expressed on the surface of osteoclast precursors and mature osteoclasts (figure 3). The binding of RANKL to RANK stimulates a number of signaling cascades that are vital for osteoclast differentiation, survival, and activity [80]. (See "Normal skeletal development and regulation of bone formation and resorption", section on 'Signaling pathways'.)

Osteoprotegerin (OPG), another member of the TNF receptor superfamily, is a soluble decoy receptor for RANKL that is produced by mature osteoblasts and osteocytes and normally blocks the interaction between RANKL and its receptor on osteoclasts [81]. (See 'Osteoblasts' below.)

The RANKL/OPG ratio is crucial for regulating osteoclast formation and activity. Many of the cytokines, chemokines, and hormones produced by tumors cells that induce osteolytic metastasis (eg, parathyroid hormone-related protein [PTHrP], 1,25D3, prostaglandins, interleukin 1 beta [IL1B], macrophage inflammatory protein 1 alpha [MIP-1-alpha], and TNF-alpha) do so by increasing RANKL expression [19].

It is unclear if tumor cells in bone directly produce RANKL or only induce its production by acting on cells (predominantly osteoblasts and osteocytes) in the bone microenvironment. (See 'Osteoblasts' below.)

Several studies report that RANKL is expressed by multiple myeloma cells in bone marrow biopsies and by human and murine multiple myeloma cell lines [82]. Others have shown that multiple myeloma cells from patients expressed RANKL and that RANKL was responsible for the release of TNF-alpha, interleukin 6 (IL-6), and interleukin 8 (IL-8) via an autocrine/paracrine mechanism that increased the survival and growth of malignant cells and exacerbated bone destruction [83]. However, still other investigators have found that certain (syndecan-1 [CD138] positive) multiple myeloma cells and multiple myeloma cell lines do not express RANKL, and that an increased RANKL/OPG ratio occurred only when human multiple myeloma cells were cocultured with bone marrow stromal cells [84]. The increased RANKL/OPG ratio in multiple myeloma patients correlated with poor prognosis and reduced survival [81,85]. Importantly, preclinical models of multiple myeloma bone disease demonstrate that blocking RANKL-induced osteoclast formation, via administration of recombinant OPG or RANK-Fc, significantly decreases osteolytic lesions and tumor growth in mice [86].

The RANK/OPG axis has also been demonstrated to impact progression of prostate cancer bone metastases. Inhibition of RANKL using either OPG [87] or a solubilized form of the RANKL receptor [88] decreases growth of prostate cancer in bone in preclinical models.

Denosumab, a human antibody to RANKL, is highly effective for preventing SREs in patients with bone metastases from prostate cancer, breast cancer, and other solid tumors, and in multiple myeloma. It also prevents treatment-related bone loss in men treated with androgen deprivation therapy for prostate cancer and in women with breast cancer who are treated with aromatase inhibitors. In spite of the efficacy of denosumab for decreasing skeletal-related events, it is accompanied by rare, but serious toxicities such as osteonecrosis of the jaw suggesting that dose optimization and additional therapeutic strategies need consideration. (See "Osteoclast inhibitors for patients with bone metastases from breast, prostate, and other solid tumors" and "Multiple myeloma: The use of osteoclast inhibitors" and "Use of osteoclast inhibitors in early breast cancer" and "Medication-related osteonecrosis of the jaw in patients with cancer" and "Risks of therapy with bone antiresorptive agents in patients with advanced malignancy".)

In addition to inducing RANKL or decreasing OPG, products of tumor cells can activate osteoclastic bone resorption in other ways:

Jagged 1 (JAG1) expressed on breast cancer cells can activate notch signaling in osteoclast precursors to induce osteoclast formation [32].

Metalloproteinases, such as matrix metalloproteinase (MMP) 7, produced by osteoclasts in response to prostate cancer cells can cleave membrane-bound RANKL into a secretable soluble protein that increases bone destruction [89].

Multiple tumor types produce PTHrP including prostate cancer [90,91], multiple myeloma [92], and breast cancer [93]. PTHrP induces osteoclast activity through induction of RANKL.

MMP13 produced by myeloma cells can increase osteoclast precursor fusion to enhance osteolysis. This effect of MMP13 is independent of its proteolytic activity [94].

Osteoblasts — Cells of the osteoblast lineage primarily contribute to bone metastasis through production of RANKL and OPG. Immature osteoblasts/bone marrow stromal cells produce RANKL, while more mature osteoblasts and osteocytes produce OPG. As noted above, multiple tumor-derived factors can induce RANKL expression by immature osteoblasts or block OPG expression by mature osteoblasts or osteocytes. For example, direct interactions between myeloma cells and osteocytes increase RANKL expression and decrease OPG expression by osteocytes [95]. In addition, myeloma cells inhibit osteoblast precursor differentiation, thereby increasing the RANKL/OPG ratio. The RANKL/OPG ratio is crucial for regulating osteoclast formation and activity. (See 'RANK/RANKL signaling pathway and bone remodeling' above.)

As noted above, cells of osteoblast lineage also contribute to maintaining tumor cells in the dormant state within HSC niches. (See 'Hematopoietic stem cell niches' above.)

Advances in understanding the molecular mechanisms underlying these osteoblast functions suggest the potential for new therapeutic approaches to target bone metastases. As an example, it has been reported that hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) alpha, which are present in the tumor microenvironment, induce expression of RANKL and macrophage colony-stimulating factor (MCSF) by mature osteoblasts through transactivation of c-Met, the receptor for HGF, and that this increases osteolysis [96]. These same investigators then showed that a dual kinase inhibitor of c-Met and the VEGF receptor 2 on osteoblasts could significantly reduce the growth of prostate cancer in bone. Thus, targeting these pathways in osteoblasts could represent a new approach to treating prostate cancer metastatic to bone.

Additionally, bone matrix proteins produced by osteoblasts, such as osteopontin, bone sialoprotein, and decorin, affect bone metastasis. High levels of osteopontin increase bone metastasis, while overexpression of decorin inhibits bone metastasis [97,98]. These results suggest that blocking osteopontin-breast cancer cell interaction or overexpressing decorin may provide new ways to treat bone metastases. The bone matrix microstructure itself may regulate bone metastasis. Typically bone matrix is an organized structure, but dysregulated remodeling induced by inflammation or tumors can lead to disorganized crystalline structure. In the case of melanoma and breast cancer, the disorganized crystalline structure is associated with increased homing of cancer cells to bone [99,100].

Tumor cells in bone can also regulate osteoblast differentiation. Tumor cells that cause osteoblastic metastasis produce osteoblast differentiation factors, such as endothelin-1, bone morphogenic proteins (BMPs), insulin-like growth factors, platelet-derived growth factors, and fibroblast growth factors, that drive the development of osteoblastic metastases and increase tumor cell growth [41]. In one study, administration of a selective endothelin-1 receptor antagonist decreased both osteoblastic metastases and tumor burden in an animal model, although it had no effect on tumor growth at orthotopic sites [101].

BMPs, including BMP-6 and BMP-2, also increase osteoblast differentiation in osteoblastic metastasis and appear to act independently of endothelin-1 [102]. As an example, the administration of an anti-BMP antibody inhibited prostate cancer growth in bone in a preclinical model [102]. Both BMP-6 and BMP-2 also increase the local invasiveness of prostate cancer cells in the bone microenvironment. Overproduction of urokinase-type plasminogen activator (uPA) by prostate cancer cells increases in bone metastasis [103], and cells transfected with an antisense DNA to uPA had a threefold decrease in bone metastases compared with empty-vector transfected cells. An anti-urokinase receptor antibody also blocked bone metastasis by prostate cancer cells [104]. These observations suggest that blocking the production of osteoblast-inducing activity by tumors may decrease tumor growth. Thus, as with osteolytic metastasis, a feedback cycle may be present in which tumors induce osteoblast activity and osteoblasts in turn release growth factors that increase tumor growth. (See 'Osteoclasts' above.)

Other soluble factors can modulate osteoblast differentiation and impact bone metastasis. For example, activation of the retinoic acid receptor inhibits the endothelial cell transition to osteoblasts resulting in decreased prostate cancer-induced bone formation [105].

By contrast, tumor cells that induce osteolytic lesions may also inhibit osteoblast differentiation, with the most profound inhibition occurring in myeloma (figure 4). Multiple factors act to suppress osteoblast differentiation in myeloma. These include soluble factors such as interleukin 7 (IL-7) and TNF-alpha, the Wnt signaling antagonists Dickkopf-1 (DKK1) and sclerostin (produced by multiple myeloma cells and osteocytes), as well as adhesive interactions between myeloma cells and bone marrow stromal cells [106]. DKK1 levels are also increased in osteolytic breast cancer bone metastasis and early in the course of prostate cancer bone metastasis [107,108]. However, DKK1 levels decrease in response to the PTHrP produced by prostate cancer cells, allowing formation of the osteoblastic metastases that are characteristic of prostate cancer [107] (see 'Osteolytic versus osteoblastic bone metastases' above). In addition to direct effects on bone, DKK1 may promote bone metastasis through regulation of an antitumor immune response [109].

Importantly, the suppression of bone formation in multiple myeloma persists even when multiple myeloma cells are eradicated and can no longer affect osteoblast differentiation through direct interactions or release of osteoblast inhibitory soluble factors. Epigenetic changes in RUNX2 (runt-related transcription factor 2) and SP7 (also called Osterix), master genes required for osteoblast differentiation, occur in bone marrow stromal cells exposed to myeloma cells [110]. These epigenetic changes play a critical role in the protracted suppression of osteoblast differentiation in multiple myeloma.

The transcriptional repressor growth factor-independent 1 (GFI1) is upregulated in bone marrow stromal cells from myeloma patients and can induce long-term suppression of osteoblast differentiation in multiple myeloma. GFI1 binds to the RUNX2 promoter and recruits chromatin co-repressors to RUNX2 to induce epigenetic changes at the RUNX2 locus that block RUNX2 gene transcription [110]. These repressive chromatin changes persist even after removal of the multiple myeloma cells. Importantly, knockdown of GFI1 or selective pharmacologic inhibition of these co-repressors in bone marrow stromal cells prevented repression of RUNX2 transcription induced by multiple myeloma and restored osteoblast differentiation. These data suggest that treatment of multiple myeloma patients with clinically available co-repressor inhibitors might be beneficial for reversing the profound osteoblast suppression that is associated with multiple myeloma, allowing the repair of osteolytic lesions [110].

Osteocytes — The important contributions of osteocytes to cancer progression in bone are just becoming appreciated. Osteocytes comprise more than 95 percent of all bone cells (compared with 1 to 2 percent for osteoclasts and 3 to 4 percent for osteoblasts) and are central regulators of physiologic bone remodeling [111]. The osteocyte dendritic network allows direct cell-to-cell contact not only between osteocytes but also with cells on the bone surface and in the bone marrow. This network also distributes osteocyte-secreted molecules within the bone/marrow microenvironment and into blood vessels, so that they can enter the general circulation. Osteocytes produce RANKL, OPG, sclerostin, and DKK1, which regulate generation and activity of osteoclasts and osteoblasts. In addition, apoptotic osteocytes target bone remodeling to specific areas in bone [112]. Induction of osteocyte apoptosis is sufficient to increase resorption and bone loss [113], suggesting that osteocyte apoptosis underlies pathologic conditions involving enhanced bone resorption. The mechanisms underlying osteoclast recruitment and differentiation driven by osteocyte apoptosis are not fully understood.

Osteocyte apoptosis is increased in the bone lesions of patients with multiple myeloma, and the number of apoptotic osteocytes positively correlates with the number of osteoclasts in bone [114]. Multiple myeloma cells interact directly with osteocytes, and these interactions activate bidirectional notch signaling between multiple myeloma cells and osteocytes, which increases multiple myeloma cell proliferation and induces osteocyte apoptosis, which is further increased by the TNF-alpha secreted by the multiple myeloma cells [95]. Osteocyte apoptosis induced by multiple myeloma cells in turn increases RANKL expression in osteocytes and stimulates osteoclast recruitment. Inhibiting osteocyte apoptosis prevents these effects. In addition, multiple myeloma cells increase expression of sclerostin in osteocytes, which inhibits osteoblast differentiation. Deletion of the sclerostin gene or neutralization of its product with an anti-sclerostin antibody stimulates bone formation and reduces the bone resorption induced by multiple myeloma cells [115]. Furthermore, myeloma promotes osteocyte production of VEGF alpha and subsequent angiogenesis [116].

There are some potential therapeutic implications of these findings, at least in the setting of multiple myeloma:

Bortezomib, a proteosome inhibitor with antitumor activity in myeloma, decreases osteocyte autophagic/apoptotic death in vitro and the number of dead osteocytes in the bones [117]. Strong immunoexpression of DKK1 is associated with the response to bortezomib [118].

Antibody-based inhibition of DKK1 prevented the development of osteolytic bone disease in a murine model of multiple myeloma [119], and at least one inhibitor (BHQ880) may be beneficial for patients with relapsed or refractory myeloma and prior SREs [120].

Antisclerostin antibody has been shown to decrease myeloma-associated bone loss in a preclinical model [121].

In addition to myeloma, others have also shown a role for osteocytes in prostate cancer bone metastasis by showing that tumor-generated pressure in bone promotes prostate cancer bone metastasis acting through osteocytes [54]. Application of pressure to osteocytes, the main mechanotransducing cells in bone, increased C-C motif chemokine ligand 5 (CCL5) and metalloproteinases in osteocytes, which enhanced prostate cancer growth and invasion. Furthermore, it has been suggested that mechanical stimulation of osteocytes promotes proliferation and migration of breast cancer cells through CXC motif chemokine ligand 12 (CXCL12) [122]. (See 'Physical characteristics of bone' above.)

Finally, in addition to promoting bone metastatic progression, there is evidence that stimulation of Wnt signaling pathway in osteocytes may protect against bone metastasis [123].

Bone marrow adipocytes — Bone marrow adipocytes are one of the most abundant cells in adult bone marrow, and they increase with age, so that by age 65, adipocytes comprise 60 percent of marrow volume [124]. Adipocytes are derived from the same common mesenchymal stromal precursor as osteoblasts, which is induced to differentiate to a particular lineage by differentiation factors in the bone marrow microenvironment [125].

Marrow adipocytes promote bone metastasis and enhance the growth of cancers through the production of adipokines and chemokines that increase tumor cell survival, growth, and osteoclastogenesis [126-128]. The role of adipocytes in the development and progression of bone metastases is supported by the following observations:

Cell-to-cell interactions between cancer cells and adipocytes induce morphologic and phenotypic changes in the adipocytes that decrease their expression of adiponectin, a suppressor of tumor growth [129], decrease their lipid content, and decrease expression of adipocytic genes, while increasing expression of inflammatory cytokines. Inflammatory cytokines and chemokines, such as IL-6, TNF-alpha, CXCL12, and leptin, can enhance multiple myeloma cell growth and migration while preventing apoptosis of myeloma cells [130].

Adipocyte-derived CXC motif chemokine receptor 1 and CXCL12 increase the osteoclast activity that enhances metastatic prostate cancer growth and survival [131].

Secretion of IL1B and leptin by bone marrow adipocytes attracts breast cancer cells to bone [132].

A high-fat diet increases adipocytes in bone marrow with an associated increased caprylic acid, a medium-chain fatty acid, that promoted prostate cancer invasion in a preclinical model [133].

Platelets and megakaryocytes — Platelets also play an important role in bone metastasis. Alpha-2 beta-3 integrin on platelets controls the homing of melanoma cells to bone [134]. Additionally, platelet-derived lipid lysophosphatidic acid induces growth of breast cancer cells in bone and production of the osteoclastogenic factors IL-6, IL-8, and monocyte chemoattractant protein 1 [135]. Consistent with these findings, investigators have shown that antiplatelet therapy decreases breast and melanoma bone metastasis in mice [136].

In contrast to platelets, megakaryocytes (the hematopoietic cells that produce platelets) act to decrease bone metastasis by suppressing osteoclast formation through the production of OPG [137], as well as by increasing bone formation [138]. Increasing megakaryocytes in the marrow through the use of recombinant thrombopoietin has been shown to block prostate cancer bone metastasis [139].

Macrophages modulate bone metastases — The following evidence supports the view that macrophages modulate bone metastases:

Ablation of macrophages inhibits breast cancer growth in bone [140].

Furthermore, it was shown that the macrophage promoted metastasis through interleukin-4 receptor. In prostate cancer CCL5 derived from tumor-associated macrophages promotes metastasis via b-catenin/STAT 3 pathway activation [141].

Bone marrow macrophages have been shown to clear apoptotic cancer cells (a process called efferocytosis) with a resulting increase in production of inflammatory cytokines, that could enhance cancer cell growth in bone [142].

Taken together these studies underscore the importance of future studies delineating the roles of macrophages as mediators and potential therapeutic targets for bone metastasis.

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Bone metastases (The Basics)")

SUMMARY

Morbidity and mortality – Bone is a frequent site of metastasis, and bone metastases cause significant morbidity and increased mortality for patients. (See 'Introduction' above.)

Osteoblastic versus osteolytic metastases – Normal bone constantly undergoes a remodeling process that includes the resorption of bone by osteoclasts and the deposition of new bone by osteoblasts. Bone metastases are generally classified as osteolytic or osteoblastic based upon the radiologic appearance of predominant bone destruction or deposition of new bone. However, this distinction is not absolute, and many patients have a mixed picture of both osteolytic and osteoblastic metastases. Both types of bone metastases are characterized by dysregulation of the normal bone remodeling process. (See 'Osteolytic versus osteoblastic bone metastases' above.)

Bone is a preferential site for metastases – Multiple mechanisms increase the potential for tumor cells to preferentially metastasize to bone, including the intrinsic properties of the tumor cells, changes induced by the tumor cells or their products in the bone microenvironment, and the bone microenvironment itself. (See 'Bone as a preferential site for metastasis' above.)

Interaction of bone cells with metastases and molecular mechanisms – In the skeleton, the multistep process of metastasis development begins with colonization, when circulating tumor cells enter the bone marrow compartment and engage in specialized microenvironments or niches. The second step involves survival and dormancy in that the colonizing tumor cells adapt to their new microenvironment, evade the immune system, and reside in a dormant state, sometimes for long periods of time. The third step, reactivation and development, requires the ability to escape from the dormant state to actively proliferate and form micrometastases. The final step occurs when cells grow uncontrollably, become independent of the microenvironment, and ultimately modify bone as the metastases flourish. (See 'Contribution of bone cells to tumor cell dormancy, reactivation, and tumor progression in bone' above.)

The molecular mechanisms responsible for both osteolytic and osteoblastic metastases are being identified. The use of high throughput genomics, proteomics, and single-cell analytic technology; the availability of appropriate animal models of bone metastasis; and in vitro models have permitted the identification of several factors, produced by the tumor cells themselves or by the bone cells or bone microenvironment in response to the tumor, that mediate dormancy and reactivation of tumor cells in bone and the bone destructive process.

Advances in understanding these pathways have revealed new potentially beneficial therapeutic approaches to target bone metastases in a variety of malignancies, especially multiple myeloma. (See 'Osteocytes' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges David G Roodman, MD, PhD, who contributed to an earlier version of this topic review.

  1. Huang JF, Shen J, Li X, et al. Incidence of patients with bone metastases at diagnosis of solid tumors in adults: a large population-based study. Ann Transl Med 2020; 8:482.
  2. Hernandez RK, Wade SW, Reich A, et al. Incidence of bone metastases in patients with solid tumors: analysis of oncology electronic medical records in the United States. BMC Cancer 2018; 18:44.
  3. Shih JT, Yeh TT, Wang SH, et al. Incidence of bone metastases in patients with organ-specific cancers: A nationwide population-based cohort study. Int J Clin Pract 2021; 75:e13997.
  4. Ryan C, Stoltzfus KC, Horn S, et al. Epidemiology of bone metastases. Bone 2022; 158:115783.
  5. Coleman RE. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res 2006; 12:6243s.
  6. Wedam SB, Beaver JA, Amiri-Kordestani L, et al. US Food and Drug Administration Pooled Analysis to Assess the Impact of Bone-Only Metastatic Breast Cancer on Clinical Trial Outcomes and Radiographic Assessments. J Clin Oncol 2018; 36:1225.
  7. Katakami N, Kunikane H, Takeda K, et al. Prospective study on the incidence of bone metastasis (BM) and skeletal-related events (SREs) in patients (pts) with stage IIIB and IV lung cancer-CSP-HOR 13. J Thorac Oncol 2014; 9:231.
  8. Coleman RE. Skeletal complications of malignancy. Cancer 1997; 80:1588.
  9. Roodman GD. Pathogenesis of myeloma bone disease. J Cell Biochem 2010; 109:283.
  10. van de Donk NWCJ, Pawlyn C, Yong KL. Multiple myeloma. Lancet 2021; 397:410.
  11. Loberg RD, Logothetis CJ, Keller ET, Pienta KJ. Pathogenesis and treatment of prostate cancer bone metastases: targeting the lethal phenotype. J Clin Oncol 2005; 23:8232.
  12. Roodman GD. Mechanisms of bone metastasis. N Engl J Med 2004; 350:1655.
  13. Baek YH, Jeon HL, Oh IS, et al. Incidence of skeletal-related events in patients with breast or prostate cancer-induced bone metastasis or multiple myeloma: A 12-year longitudinal nationwide healthcare database study. Cancer Epidemiol 2019; 61:104.
  14. Terpos E, Berenson J, Raje N, Roodman GD. Management of bone disease in multiple myeloma. Expert Rev Hematol 2014; 7:113.
  15. Portenoy RK. Tolerance to opioid analgesics: clinical aspects. Cancer Surv 1994; 21:49.
  16. Saad F, Lipton A, Cook R, et al. Pathologic fractures correlate with reduced survival in patients with malignant bone disease. Cancer 2007; 110:1860.
  17. Waning DL, Mohammad KS, Reiken S, et al. Excess TGF-β mediates muscle weakness associated with bone metastases in mice. Nat Med 2015; 21:1262.
  18. Robling AG, Bonewald LF. The Osteocyte: New Insights. Annu Rev Physiol 2020; 82:485.
  19. Marino S, Roodman GD. Multiple Myeloma and Bone: The Fatal Interaction. Cold Spring Harb Perspect Med 2018; 8.
  20. Reddington JA, Mendez GA, Ching A, et al. Imaging characteristic analysis of metastatic spine lesions from breast, prostate, lung, and renal cell carcinomas for surgical planning: Osteolytic versus osteoblastic. Surg Neurol Int 2016; 7:S361.
  21. Coleman RE, Major P, Lipton A, et al. Predictive value of bone resorption and formation markers in cancer patients with bone metastases receiving the bisphosphonate zoledronic acid. J Clin Oncol 2005; 23:4925.
  22. Sottnik JL, Keller ET. Understanding and targeting osteoclastic activity in prostate cancer bone metastases. Curr Mol Med 2013; 13:626.
  23. von Moos R, Costa L, Gonzalez-Suarez E, et al. Management of bone health in solid tumours: From bisphosphonates to a monoclonal antibody. Cancer Treat Rev 2019; 76:57.
  24. Seider MJ, Pugh SL, Langer C, et al. Randomized phase III trial to evaluate radiopharmaceuticals and zoledronic acid in the palliation of osteoblastic metastases from lung, breast, and prostate cancer: report of the NRG Oncology RTOG 0517 trial. Ann Nucl Med 2018; 32:553.
  25. Izraely S, Witz IP. Site-specific metastasis: A cooperation between cancer cells and the metastatic microenvironment. Int J Cancer 2021; 148:1308.
  26. Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev 1989; 8:98.
  27. Kang Y, Siegel PM, Shu W, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 2003; 3:537.
  28. Lu X, Wang Q, Hu G, et al. ADAMTS1 and MMP1 proteolytically engage EGF-like ligands in an osteolytic signaling cascade for bone metastasis. Genes Dev 2009; 23:1882.
  29. Yang YH, Buhamrah A, Schneider A, et al. Semaphorin 4D Promotes Skeletal Metastasis in Breast Cancer. PLoS One 2016; 11:e0150151.
  30. Ottewell PD, Wang N, Meek J, et al. Castration-induced bone loss triggers growth of disseminated prostate cancer cells in bone. Endocr Relat Cancer 2014; 21:769.
  31. Pickup M, Novitskiy S, Moses HL. The roles of TGFβ in the tumour microenvironment. Nat Rev Cancer 2013; 13:788.
  32. 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.
  33. Fournier PG, Juárez P, Jiang G, et al. The TGF-β Signaling Regulator PMEPA1 Suppresses Prostate Cancer Metastases to Bone. Cancer Cell 2015; 27:809.
  34. Zhang XH, Jin X, Malladi S, et al. Selection of bone metastasis seeds by mesenchymal signals in the primary tumor stroma. Cell 2013; 154:1060.
  35. Rieunier G, Wu X, Macaulay VM, et al. Bad to the Bone: The Role of the Insulin-Like Growth Factor Axis in Osseous Metastasis. Clin Cancer Res 2019; 25:3479.
  36. Zhang XH, Wang Q, Gerald W, et al. Latent bone metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell 2009; 16:67.
  37. Parmo-Cabañas M, Bartolomé RA, Wright N, et al. Integrin alpha4beta1 involvement in stromal cell-derived factor-1alpha-promoted myeloma cell transendothelial migration and adhesion: role of cAMP and the actin cytoskeleton in adhesion. Exp Cell Res 2004; 294:571.
  38. Cooper CR, Chay CH, Pienta KJ. The role of alpha(v)beta(3) in prostate cancer progression. Neoplasia 2002; 4:191.
  39. Michigami T, Shimizu N, Williams PJ, et al. Cell-cell contact between marrow stromal cells and myeloma cells via VCAM-1 and alpha(4)beta(1)-integrin enhances production of osteoclast-stimulating activity. Blood 2000; 96:1953.
  40. Ibrahim T, Sacanna E, Gaudio M, et al. Role of RANK, RANKL, OPG, and CXCR4 tissue markers in predicting bone metastases in breast cancer patients. Clin Breast Cancer 2011; 11:369.
  41. Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal attraction. Nat Rev Cancer 2011; 11:411.
  42. Roccaro AM, Sacco A, Maiso P, et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest 2013; 123:1542.
  43. Olkhanud PB, Damdinsuren B, Bodogai M, et al. Tumor-evoked regulatory B cells promote breast cancer metastasis by converting resting CD4⁺ T cells to T-regulatory cells. Cancer Res 2011; 71:3505.
  44. Görgün GT, Whitehill G, Anderson JL, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood 2013; 121:2975.
  45. Dai J, Escara-Wilke J, Keller JM, et al. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J Exp Med 2019; 216:2883.
  46. Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012; 18:883.
  47. Ollodart J, Contino KF, Deep G, Shiozawa Y. The impacts of exosomes on bone metastatic progression and their potential clinical utility. Bone Rep 2022; 17:101606.
  48. Batoon L, McCauley LK. Cross Talk Between Macrophages and Cancer Cells in the Bone Metastatic Environment. Front Endocrinol (Lausanne) 2021; 12:763846.
  49. Soki FN, Koh AJ, Jones JD, et al. Polarization of prostate cancer-associated macrophages is induced by milk fat globule-EGF factor 8 (MFG-E8)-mediated efferocytosis. J Biol Chem 2014; 289:24560.
  50. Soki FN, Cho SW, Kim YW, et al. Bone marrow macrophages support prostate cancer growth in bone. Oncotarget 2015; 6:35782.
  51. Zheng H, Bae Y, Kasimir-Bauer S, et al. Therapeutic Antibody Targeting Tumor- and Osteoblastic Niche-Derived Jagged1 Sensitizes Bone Metastasis to Chemotherapy. Cancer Cell 2017; 32:731.
  52. Dunn LK, Mohammad KS, Fournier PG, et al. Hypoxia and TGF-beta drive breast cancer bone metastases through parallel signaling pathways in tumor cells and the bone microenvironment. PLoS One 2009; 4:e6896.
  53. Page JM, Merkel AR, Ruppender NS, et al. Matrix rigidity regulates the transition of tumor cells to a bone-destructive phenotype through integrin β3 and TGF-β receptor type II. Biomaterials 2015; 64:33.
  54. Sottnik JL, Dai J, Zhang H, et al. Tumor-induced pressure in the bone microenvironment causes osteocytes to promote the growth of prostate cancer bone metastases. Cancer Res 2015; 75:2151.
  55. Croucher PI, McDonald MM, Martin TJ. Bone metastasis: the importance of the neighbourhood. Nat Rev Cancer 2016; 16:373.
  56. Clézardin P, Coleman R, Puppo M, et al. Bone metastasis: mechanisms, therapies, and biomarkers. Physiol Rev 2021; 101:797.
  57. Summers MA, McDonald MM, Croucher PI. Cancer Cell Dormancy in Metastasis. Cold Spring Harb Perspect Med 2020; 10.
  58. Sistigu A, Musella M, Galassi C, et al. Tuning Cancer Fate: Tumor Microenvironment's Role in Cancer Stem Cell Quiescence and Reawakening. Front Immunol 2020; 11:2166.
  59. Risson E, Nobre AR, Maguer-Satta V, Aguirre-Ghiso JA. The current paradigm and challenges ahead for the dormancy of disseminated tumor cells. Nat Cancer 2020; 1:672.
  60. Chen F, Han Y, Kang Y. Bone marrow niches in the regulation of bone metastasis. Br J Cancer 2021; 124:1912.
  61. Calvi LM, Link DC. The hematopoietic stem cell niche in homeostasis and disease. Blood 2015; 126:2443.
  62. Shiozawa Y, Pedersen EA, Havens AM, et al. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J Clin Invest 2011; 121:1298.
  63. Lawson MA, McDonald MM, Kovacic N, et al. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat Commun 2015; 6:8983.
  64. Allocca G, Hughes R, Wang N, et al. The bone metastasis niche in breast cancer-potential overlap with the haematopoietic stem cell niche in vivo. J Bone Oncol 2019; 17:100244.
  65. Capulli M, Hristova D, Valbret Z, et al. Notch2 pathway mediates breast cancer cellular dormancy and mobilisation in bone and contributes to haematopoietic stem cell mimicry. Br J Cancer 2019; 121:157.
  66. Roodman GD. Genes associate with abnormal bone cell activity in bone metastasis. Cancer Metastasis Rev 2012; 31:569.
  67. Kim JK, Jung Y, Wang J, et al. TBK1 regulates prostate cancer dormancy through mTOR inhibition. Neoplasia 2013; 15:1064.
  68. Yumoto K, Eber MR, Wang J, et al. Axl is required for TGF-β2-induced dormancy of prostate cancer cells in the bone marrow. Sci Rep 2016; 6:36520.
  69. Ghajar CM, Peinado H, Mori H, et al. The perivascular niche regulates breast tumour dormancy. Nat Cell Biol 2013; 15:807.
  70. Yu-Lee LY, Yu G, Lee YC, et al. Osteoblast-Secreted Factors Mediate Dormancy of Metastatic Prostate Cancer in the Bone via Activation of the TGFβRIII-p38MAPK-pS249/T252RB Pathway. Cancer Res 2018; 78:2911.
  71. Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med 2006; 12:657.
  72. Lu X, Mu E, Wei Y, et al. VCAM-1 promotes osteolytic expansion of indolent bone micrometastasis of breast cancer by engaging α4β1-positive osteoclast progenitors. Cancer Cell 2011; 20:701.
  73. Giancotti FG. Mechanisms governing metastatic dormancy and reactivation. Cell 2013; 155:750.
  74. Baram T, Rubinstein-Achiasaf L, Ben-Yaakov H, Ben-Baruch A. Inflammation-Driven Breast Tumor Cell Plasticity: Stemness/EMT, Therapy Resistance and Dormancy. Front Oncol 2020; 10:614468.
  75. Schneider A, Kalikin LM, Mattos AC, et al. Bone turnover mediates preferential localization of prostate cancer in the skeleton. Endocrinology 2005; 146:1727.
  76. Mundy GR, Yoneda T, Hiraga T. Preclinical studies with zoledronic acid and other bisphosphonates: impact on the bone microenvironment. Semin Oncol 2001; 28:35.
  77. Jakob T, Tesfamariam YM, Macherey S, et al. Bisphosphonates or RANK-ligand-inhibitors for men with prostate cancer and bone metastases: a network meta-analysis. Cochrane Database Syst Rev 2020; 12:CD013020.
  78. Ikeda T, Utsuyama M, Hirokawa K. Expression profiles of receptor activator of nuclear factor kappaB ligand, receptor activator of nuclear factor kappaB, and osteoprotegerin messenger RNA in aged and ovariectomized rat bones. J Bone Miner Res 2001; 16:1416.
  79. Nakashima T, Hayashi M, Fukunaga T, et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med 2011; 17:1231.
  80. Park JH, Lee NK, Lee SY. Current Understanding of RANK Signaling in Osteoclast Differentiation and Maturation. Mol Cells 2017; 40:706.
  81. Terpos E, Ntanasis-Stathopoulos I, Gavriatopoulou M, Dimopoulos MA. Pathogenesis of bone disease in multiple myeloma: from bench to bedside. Blood Cancer J 2018; 8:7.
  82. Heider U, Zavrski I, Jakob C, et al. Expression of receptor activator of NF-kappaB ligand (RANKL) mRNA in human multiple myeloma cells. J Cancer Res Clin Oncol 2004; 130:469.
  83. Schmiedel BJ, Scheible CA, Nuebling T, et al. RANKL expression, function, and therapeutic targeting in multiple myeloma and chronic lymphocytic leukemia. Cancer Res 2013; 73:683.
  84. Giuliani N, Colla S, Morandi F, et al. Lack of receptor activator of nuclear factor-kB ligand (RANKL) expression and functional production by human multiple myeloma cells. Haematologica 2005; 90:275.
  85. Pearse RN, Sordillo EM, Yaccoby S, et al. Multiple myeloma disrupts the TRANCE/ osteoprotegerin cytokine axis to trigger bone destruction and promote tumor progression. Proc Natl Acad Sci U S A 2001; 98:11581.
  86. Croucher PI, Shipman CM, Van Camp B, Vanderkerken K. Bisphosphonates and osteoprotegerin as inhibitors of myeloma bone disease. Cancer 2003; 97:818.
  87. Zhang J, Dai J, Qi Y, et al. Osteoprotegerin inhibits prostate cancer-induced osteoclastogenesis and prevents prostate tumor growth in the bone. J Clin Invest 2001; 107:1235.
  88. Zhang J, Dai J, Yao Z, et al. Soluble receptor activator of nuclear factor kappaB Fc diminishes prostate cancer progression in bone. Cancer Res 2003; 63:7883.
  89. Lynch CC, Hikosaka A, Acuff HB, et al. MMP-7 promotes prostate cancer-induced osteolysis via the solubilization of RANKL. Cancer Cell 2005; 7:485.
  90. Rabbani SA, Gladu J, Harakidas P, et al. Over-production of parathyroid hormone-related peptide results in increased osteolytic skeletal metastasis by prostate cancer cells in vivo. Int J Cancer 1999; 80:257.
  91. Bryden AA, Islam S, Freemont AJ, et al. Parathyroid hormone-related peptide: expression in prostate cancer bone metastases. Prostate Cancer Prostatic Dis 2002; 5:59.
  92. Cafforio P, Savonarola A, Stucci S, et al. PTHrP produced by myeloma plasma cells regulates their survival and pro-osteoclast activity for bone disease progression. J Bone Miner Res 2014; 29:55.
  93. Martin TJ, Johnson RW. Multiple actions of parathyroid hormone-related protein in breast cancer bone metastasis. Br J Pharmacol 2021; 178:1923.
  94. Fu J, Li S, Feng R, et al. Multiple myeloma-derived MMP-13 mediates osteoclast fusogenesis and osteolytic disease. J Clin Invest 2016; 126:1759.
  95. Delgado-Calle J, Anderson J, Cregor MD, et al. Bidirectional Notch Signaling and Osteocyte-Derived Factors in the Bone Marrow Microenvironment Promote Tumor Cell Proliferation and Bone Destruction in Multiple Myeloma. Cancer Res 2016; 76:1089.
  96. Lee C, Whang YM, Campbell P, et al. Dual targeting c-met and VEGFR2 in osteoblasts suppresses growth and osteolysis of prostate cancer bone metastasis. Cancer Lett 2018; 414:205.
  97. Xu W, Neill T, Yang Y, et al. The systemic delivery of an oncolytic adenovirus expressing decorin inhibits bone metastasis in a mouse model of human prostate cancer. Gene Ther 2015; 22:247.
  98. Koro K, Parkin S, Pohorelic B, et al. Interactions between breast cancer cells and bone marrow derived cells in vitro define a role for osteopontin in affecting breast cancer cell migration. Breast Cancer Res Treat 2011; 126:73.
  99. He F, Chiou AE, Loh HC, et al. Multiscale characterization of the mineral phase at skeletal sites of breast cancer metastasis. Proc Natl Acad Sci U S A 2017; 114:10542.
  100. Matsugaki A, Kimura Y, Watanabe R, et al. Impaired Alignment of Bone Matrix Microstructure Associated with Disorganized Osteoblast Arrangement in Malignant Melanoma Metastasis. Biomolecules 2021; 11.
  101. Clines GA, Mohammad KS, Bao Y, et al. Dickkopf homolog 1 mediates endothelin-1-stimulated new bone formation. Mol Endocrinol 2007; 21:486.
  102. Dai J, Keller J, Zhang J, et al. Bone morphogenetic protein-6 promotes osteoblastic prostate cancer bone metastases through a dual mechanism. Cancer Res 2005; 65:8274.
  103. Achbarou A, Kaiser S, Tremblay G, et al. Urokinase overproduction results in increased skeletal metastasis by prostate cancer cells in vivo. Cancer Res 1994; 54:2372.
  104. Rabbani SA, Ateeq B, Arakelian A, et al. An anti-urokinase plasminogen activator receptor antibody (ATN-658) blocks prostate cancer invasion, migration, growth, and experimental skeletal metastasis in vitro and in vivo. Neoplasia 2010; 12:778.
  105. Yu G, Corn PG, Shen P, et al. Retinoic Acid Receptor Activation Reduces Metastatic Prostate Cancer Bone Lesions by Blocking the Endothelial-to-Osteoblast Transition. Cancer Res 2022; 82:3158.
  106. David Roodman G, Silbermann R. Mechanisms of osteolytic and osteoblastic skeletal lesions. Bonekey Rep 2015; 4:753.
  107. Hall CL, Daignault SD, Shah RB, et al. Dickkopf-1 expression increases early in prostate cancer development and decreases during progression from primary tumor to metastasis. Prostate 2008; 68:1396.
  108. Bu G, Lu W, Liu CC, et al. Breast cancer-derived Dickkopf1 inhibits osteoblast differentiation and osteoprotegerin expression: implication for breast cancer osteolytic bone metastases. Int J Cancer 2008; 123:1034.
  109. Jaschke N, Hofbauer LC, Göbel A, Rachner TD. Evolving functions of Dickkopf-1 in cancer and immunity. Cancer Lett 2020; 482:1.
  110. Adamik J, Jin S, Sun Q, et al. EZH2 or HDAC1 Inhibition Reverses Multiple Myeloma-Induced Epigenetic Suppression of Osteoblast Differentiation. Mol Cancer Res 2017; 15:405.
  111. Dallas SL, Prideaux M, Bonewald LF. The osteocyte: an endocrine cell ... and more. Endocr Rev 2013; 34:658.
  112. Bonewald LF. The amazing osteocyte. J Bone Miner Res 2011; 26:229.
  113. Bonewald LF. Osteocyte messages from a bony tomb. Cell Metab 2007; 5:410.
  114. Giuliani N, Ferretti M, Bolzoni M, et al. Increased osteocyte death in multiple myeloma patients: role in myeloma-induced osteoclast formation. Leukemia 2012; 26:1391.
  115. Delgado-Calle J, Anderson J, Cregor MD, et al. Genetic deletion of Sost or pharmacological inhibition of sclerostin prevent multiple myeloma-induced bone disease without affecting tumor growth. Leukemia 2017; 31:2686.
  116. Mulcrone PL, Edwards SKE, Petrusca DN, et al. Osteocyte Vegf-a contributes to myeloma-associated angiogenesis and is regulated by Fgf23. Sci Rep 2020; 10:17319.
  117. Toscani D, Palumbo C, Dalla Palma B, et al. The Proteasome Inhibitor Bortezomib Maintains Osteocyte Viability in Multiple Myeloma Patients by Reducing Both Apoptosis and Autophagy: A New Function for Proteasome Inhibitors. J Bone Miner Res 2016; 31:815.
  118. Choi YW, Park JS, Han JH, et al. Strong immunoexpression of dickkopf-1 is associated with response to bortezomib in multiple myeloma. Leuk Lymphoma 2018; 59:2670.
  119. Heath DJ, Chantry AD, Buckle CH, et al. Inhibiting Dickkopf-1 (Dkk1) removes suppression of bone formation and prevents the development of osteolytic bone disease in multiple myeloma. J Bone Miner Res 2009; 24:425.
  120. Iyer SP, Beck JT, Stewart AK, et al. A Phase IB multicentre dose-determination study of BHQ880 in combination with anti-myeloma therapy and zoledronic acid in patients with relapsed or refractory multiple myeloma and prior skeletal-related events. Br J Haematol 2014; 167:366.
  121. Toscani D, Bolzoni M, Ferretti M, et al. Role of Osteocytes in Myeloma Bone Disease: Anti-sclerostin Antibody as New Therapeutic Strategy. Front Immunol 2018; 9:2467.
  122. Dwivedi A, Kiely PA, Hoey DA. Mechanically stimulated osteocytes promote the proliferation and migration of breast cancer cells via a potential CXCL1/2 mechanism. Biochem Biophys Res Commun 2021; 534:14.
  123. Liu S, Wu D, Sun X, et al. Overexpression of Lrp5 enhanced the anti-breast cancer effects of osteocytes in bone. Bone Res 2021; 9:32.
  124. Meunier P, Aaron J, Edouard C, Vignon G. Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies. Clin Orthop Relat Res 1971; 80:147.
  125. Lee S, Cho HY, Bui HT, Kang D. The osteogenic or adipogenic lineage commitment of human mesenchymal stem cells is determined by protein kinase C delta. BMC Cell Biol 2014; 15:42.
  126. Morris EV, Edwards CM. Bone Marrow Adipose Tissue: A New Player in Cancer Metastasis to Bone. Front Endocrinol (Lausanne) 2016; 7:90.
  127. Shin E, Koo JS. The Role of Adipokines and Bone Marrow Adipocytes in Breast Cancer Bone Metastasis. Int J Mol Sci 2020; 21.
  128. Uehara H, Kobayashi T, Matsumoto M, et al. Adipose tissue:Critical contributor to the development of prostate cancer. J Med Invest 2018; 65:9.
  129. Saxena NK, Sharma D. Metastasis suppression by adiponectin: LKB1 rises up to the challenge. Cell Adh Migr 2010; 4:358.
  130. Trotter TN, Gibson JT, Sherpa TL, et al. Adipocyte-Lineage Cells Support Growth and Dissemination of Multiple Myeloma in Bone. Am J Pathol 2016; 186:3054.
  131. Hardaway AL, Herroon MK, Rajagurubandara E, Podgorski I. Marrow adipocyte-derived CXCL1 and CXCL2 contribute to osteolysis in metastatic prostate cancer. Clin Exp Metastasis 2015; 32:353.
  132. Dirat B, Bochet L, Dabek M, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res 2011; 71:2455.
  133. Wang C, Wang J, Chen K, et al. Caprylic acid (C8:0) promotes bone metastasis of prostate cancer by dysregulated adipo-osteogenic balance in bone marrow. Cancer Sci 2020; 111:3600.
  134. Bakewell SJ, Nestor P, Prasad S, et al. Platelet and osteoclast beta3 integrins are critical for bone metastasis. Proc Natl Acad Sci U S A 2003; 100:14205.
  135. Boucharaba A, Serre CM, Grès S, et al. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J Clin Invest 2004; 114:1714.
  136. Uluçkan O, Eagleton MC, Floyd DH, et al. APT102, a novel adpase, cooperates with aspirin to disrupt bone metastasis in mice. J Cell Biochem 2008; 104:1311.
  137. Chollet ME, Brouland JP, Bal dit Sollier C, et al. Evidence of a colocalisation of osteoprotegerin (OPG) with von Willebrand factor (VWF) in platelets and megakaryocytes alpha granules. Studies from normal and grey platelets. Br J Haematol 2010; 148:805.
  138. Lemieux JM, Horowitz MC, Kacena MA. Involvement of integrins alpha(3)beta(1) and alpha(5)beta(1) and glycoprotein IIb in megakaryocyte-induced osteoblast proliferation. J Cell Biochem 2010; 109:927.
  139. Li X, Koh AJ, Wang Z, et al. Inhibitory effects of megakaryocytic cells in prostate cancer skeletal metastasis. J Bone Miner Res 2011; 26:125.
  140. Ma RY, Zhang H, Li XF, et al. Monocyte-derived macrophages promote breast cancer bone metastasis outgrowth. J Exp Med 2020; 217.
  141. Huang R, Wang S, Wang N, et al. CCL5 derived from tumor-associated macrophages promotes prostate cancer stem cells and metastasis via activating β-catenin/STAT3 signaling. Cell Death Dis 2020; 11:234.
  142. Mendoza-Reinoso V, Baek DY, Kurutz A, et al. Unique Pro-Inflammatory Response of Macrophages during Apoptotic Cancer Cell Clearance. Cells 2020; 9.
Topic 2828 Version 27.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟