Abstract
Purpose of review.
The goal of this manuscript is to review the current knowledge on the role of osteocytes in cancer in bone, discuss the potential of osteocytes as a therapeutic target, and propose future research needed to understand the crosstalk between cancer cells and osteocytes in the tumor niche.
Recent findings.
Numerous studies have established that cancer cells manipulate osteocytes to facilitate invasion and tumor progression in bone. Moreover, cancer cells dysregulate osteocyte function to disrupt physiological bone remodeling, leading to the development of bone disease. Targeting of osteocytes and their derived factors has proven to effectively interfere with the progression of cancer in bone and the associated bone disease.
Summary.
Osteocytes communicate with cancer cells and are also part of the vicious cycle of cancer in bone. Additional studies investigating the role of osteocytes on metastases to the bone and development of drug resistance are needed.
Keywords: Osteocytes, Cancer, Bone, myeloma, breast cancer
1. Introduction
Bone is a dynamic mineralized connective tissue. It provides support for body movement and protects vital internal organs. It is also considered a reservoir of calcium and phosphate, and provides an environment to the bone marrow. The bone marrow microenvironment contains mainly two different cell populations of stem cells: hematopoietic and mesenchymal. Hematopoietic stem cells differentiate into blood cells, as well as osteoclasts, the bone resorbing cells. Mesenchymal stem cells differentiate into osteoblasts, the bone forming cells, osteocytes, fat cells (adipocytes), chondrocytes, and myoblasts.
Bone is constantly being replaced by a process known as bone remodeling [1]. Bone remodeling is a complex cycle that requires the coordinated and balanced action of osteoclasts, osteoblasts, and osteocytes, which together form a temporary anatomical structure called a basic multicellular unit [2, 3, 1]. During physiological bone remodeling, old/damaged bone is replaced by the same amount of new bone. This process occurs in five phases: (1) activation, (2) bone resorption, (3) reversal, (4) formation, and (5) quiescence. Osteocytes are currently considered as key initiators and drivers of bone remodeling [4]. Apoptotic osteocytes might activate bone remodeling by sending signals to lining cells, which retract from the bone surface and to osteoclasts precursors to promote targeted recruitment. Once osteoclasts mature, they initiate the resorption of the old/damaged bone and undergo apoptosis. Osteoclasts are a known source of coupling factors that recruit osteoblast precursors to the resorption pit and enable spatial and temporal coordination of bone resorption with bone formation [5]. Osteoblasts are responsible for phase 4. They produce new bone made of collagen and other proteins, and control calcium and mineral deposition. After completion of matrix formation, osteoblasts have three possible fates: differentiate further and become osteocytes, die by programmed cell death, or transform into inactive quiescent bone lining cells covering the bone surface.
Alterations in the balance between the activity of osteoblasts or osteoclasts leads to bone mass changes, as seen in several genetic disorders favoring osteoblast formation resulting in bone gain, or in osteoporosis with a concomitant decline in osteoblasts and sustain activity of osteoclasts that results in bone loss. Different types of cancer cells grow in bone and markedly disrupt physiological bone remodeling. Primary bone tumors, such as osteosarcomas, chondrosarcomas, Ewing sarcomas, and chordomas, initiate from cells present in the bone tissue [6]. Other cancers start in bone but not from bone cells. This is the case of multiple myeloma, a hematological cancer of plasma cells that initiates in the bone marrow and causes bone lesions [7, 8]. Lastly, metastatic bone tumors develop from cancer cells that metastasize to the bone from other organs [9].
Bone is one of the most favorable sites for the growth of cancer cells [10]. Steven Paget originally proposed the seed and soil hypothesis, which stated that cancer cells metastasize to local favorable microenvironments to grow, just like seed needs a fertile soil to grow into a complete plant [11]. Bone is a highly vascularized tissue and contains multiple soluble factors. When released from the mineralized matrix, their factors promote tumor growth, initiating a vicious cycle that disrupts normal bone homeostasis and further stimulates tumor growth [12, 13]. In cancer-induced osteolytic bone disease (i.e. bone metastatic breast cancer and myeloma), tumor cells increase osteoclastogenesis and bone resorption to sustain tumor growth [12]. In osteoblastic bone lesions (i.e. osteosarcomas and prostate cancer metastasis), tumor-derived factors stimulate osteoblasts differentiation, leading to the formation of woven tissue with compromised mechanical properties [14]. In turn, osteoblasts produce growth factors that fuel tumor growth. The growth of cancer cells in bone has a strong negative impact on patients’ quality of life and represents a significant cause of morbidity and mortality.
Much is known about the contribution of communication between cancer cells and osteoblasts and osteoclasts to tumor progression in bone. However, the role of osteocytes, the most abundant cells in bone and key regulators of bone homeostasis, in cancer in bone is just starting to be revealed. In this review, we focus on the emerging data supporting that osteocytes contribute to the progression of cancer in bone.
II. Osteocytes in bone physiology
Osteocytes make up between 90–95% of the cellular component in bone and are considered permanent residents of the skeleton, where they can live for decades [15, 16]. Osteocytes belong to the mesenchymal lineage, and differentiate from osteoblasts that become entombed by the mineral they produce [17]. Mature osteocytes lived in lacunae within mineralized bone and are star-shaped looking bone cells, with lots of dendrite like structures protruding out from their cell bodies [15, 16]. Osteocytes communicate with neighboring osteocytes and other cells in the bone marrow via the lacuno-canalicular system, a large and complex network intercommunicating the lacunae and canaliculi containing both the osteocytes and their cytoplasmic processes [18]. Although initially thought to be passive cells, now we know that osteocytes play a key role in bone homeostasis. First, osteocytes are the main mechanosensing cells in bone, with a high ability to detect mechanical stimuli and send signals to other effector cells such as osteoclasts and osteoblasts [19, 20]. Second, osteocytes are endocrine cells that produce and secrete a number of factors with a strong influence on osteoblasts and osteoclasts biology. For instance, osteocytes secrete Sclerostin, a potent inhibitor of Wnt signaling that negatively regulates osteoblast differentiation and survival [21, 22]. Additionally, osteocytes are the major source of Receptor activator of nuclear factor kappa-β ligand (Rankl in adult bone, and thus regulate osteoclast differentiation and function [23, 24]. Lastly, as discussed above, osteocyte apoptosis is known to stimulate recruitment of osteoclast precursors and initiate targeted remodeling.
Accumulating evidence supports that alterations in osteocyte viability and function contributes to multiple skeletal pathologies. This is the case for osteoporosis, as well as the decline in bone mass with aging, both accompanied by increased apoptotic osteocytes [25]; hypophosphatemia, characterized by osteocyte-derived Fibroblast growth factor 23 (Fgf23) excess [26]; and several genetic high and low bone mass diseases [27, 28]. Give their importance in bone biology, osteocyte and their derived factors have become effective targets for the treatment of skeletal diseases (see section VII). As discussed below, emerging evidence shows that osteocytes also play an important role in the progression of cancer in bone, providing a microenvironment that is conducive to tumor growth and bone disease.
III. Osteocytes and hematological cancers: multiple myeloma
Multiple myeloma (MM) is a hematological cancer, caused by the uncontrolled proliferation of plasma cells in the bone marrow [7, 8]. The growth of MM cells in bone disrupts normal bone remodeling, leading to increased osteoclast formation and exacerbated bone resorption, as well prolonged suppression of osteoblast differentiation and new bone formation [29–31]. Consequently, MM patients present with osteolytic lesions that weaken the bones and favor pathological fractures. Interactions between MM cells and cells of the bone marrow microenvironment (osteoblasts, osteoclasts, stromal cells, adipocytes) lead to the formation of favorable niches that induce growth and survival of the MM cells and bone destruction [32]. The first evidence supporting the role of osteocytes in multiple myeloma was provided by Giuliani and colleagues. They showed in MM patients that osteocyte apoptosis is increased in areas of bones infiltrated with cancer cells [33]. Using a mouse model of MM-induced bone disease, our group also reported increased in osteocyte apoptosis in bones bearing MM cells [34]. Mechanistic studies demonstrated that MM cells physically interact with the osteocytes and activate Notch signaling, which together with MM-derived TNF, leads to osteocytes apoptosis [34]. Recent findings suggest that MM-induced osteocyte apoptosis could also be mediated by autophagy [35]. The increase in osteocyte apoptosis was sufficient to stimulate the recruitment of osteoclast precursors [34]. In addition, MM cells increased RANKL and IL-11 expression, and decreased OPG production in osteocytes [34, 33]. Together, these results support that both healthy and apoptotic osteocytes contribute to the exacerbated, local bone resorption seen in MM.
Accumulating evidence also supports that osteocytes contribute to osteoblasts suppression in the MM niche. Sclerostin, a potent inhibitor of bone formation [36, 37], is elevated in the circulation of MM patients [38]. Consistent with this clinical observation, Sclerostin production by osteocytes is increased in bones injected with MM cells [34]. Further, osteocytes are an important source of Dickkopf-related protein 1 (Dkk1), another Wnt signaling antagonist elevated in MM cells [39]. Genetic and pharmacologic approaches demonstrated that both Sclerostin and Dkkk-1 contribute to osteoblast suppression in MM [39–43]. More recently, research efforts from our group showed that MM cells stimulated Vascular endothelial growth factor-a (Vegf-A) production in osteocytes, and that osteocyte-derived Vegf-a contributes to the increased angiogenesis in MM [44]. Lastly, Fgf-23 in osteocytes is upregulated by MM cells, which in turn regulates the expression of osteocytic Vegf-a and heparanase, a pro-osteolytic factor, in MM cells [45].
Osteocytes are also able to communicate with MM cells and alter their function. The physical interaction between MM cells and osteocytes also results in Notch signaling activation in MM cells [34]. This activation increases cyclin D1 mRNA expression level and increases the proliferation of MM cells. Interestingly, osteocytes also altered the Notch receptor repertoire in MM cells by rapidly increases the expression of Notch receptors 3 and 4. Using genetic silencing tools, we recently showed that Notch receptor 3, but not Notch receptor 2, mediates the communication between osteocytes and MM cells via Notch [46]. Lastly, genetic ablation of osteocytes increased the homing of myeloma cells to particular areas of the bone and increased total tumor burden [47], suggesting that osteocytes may play a role in MM cell homing. Altogether, the results highlighted in this section provide compelling evidence supporting that osteocytes contribute to generate a microenvironment conducive to tumor progression, angiogenesis, and bone destruction in multiple myeloma.
IV. Osteocytes and cancer metastasis
Bone metastases are a common manifestation in patients with breast, prostate and lung cancer. Bone metastasis occurs when cancer cells migrate from a distant primary site and invade and colonize the bone [48, 49]. Metastasis to the bone is a complex process that begins with metastatic tumor cells undergoing epithelial to mesenchymal transition (EMT) and detaching from the primary tumor to enter into the circulation [50]. Once in bone, cancer cells undergo apoptosis, with only a few becoming dormant and surviving [51]. Interactions with resident bone cells such as osteoblasts, facilitate the homing and control the fate of metastatic cancer cells in bone [52, 32]. Although osteocytes have the potential to contribute to bone metastasis and homing of cancer cells to particular areas of bone, their role in these processes remains largely unknown. Below we summarize the main findings supporting a role of osteocytes in the establishment and progression of bone metastasis.
V. Osteocytes and breast cancer
Studies have suggested that the interaction between metastatic breast cancer cells and osteocytes not only contributes to bone destruction, but also to migration, invasion, EMT and its reversal, and tumor growth. For instance, treatment of breast cancer cells with conditioned medium from osteocytes induced EMT, increased migration, and stimulated proliferation of breast cancer cells [53, 54]. Mechanistically, these effects appear to be mediated by changes in Connexin 43, Snail, and Wisp signaling, as well as adenosine nucleotides released by osteocytes [55–59]. Interestingly, conditioned media from mechanically stimulated osteocytes decreased the expression of EMT-related genes and their invasion capacity [60]. These results suggest an important role of mechanical signals in the crosstalk between osteocytes and breast cancer cells. Although there is no direct evidence, osteocytes could also be involved in the early stages of breast cancer colonization of bone by releasing Cxcl12 [61], a chemokine involved in cancer cell dormancy [62, 63]. Similar to multiple myeloma cells, metastatic breast cancer cells also appear to alter osteocyte function. Conditioned media from breast cancer cells reduced osteocyte viability and increased their Rankl/Opg ratio [64]. Knowledge of the role of osteocytes on breast cancer metastasis remains limited. Thus, further studies are needed to clarify the role of osteocytes in breast cancer and the potential of mechanical signals for the treatment of breast cancer metastasis to bone (see section VII).
VI. Osteocytes and prostate cancer
In vitro experiments with conditioned media show that osteocytes also stimulate proliferation, migration, and invasion capacity of prostate cancer cells [53]. Mechanistically, Keller’s group demonstrated that osteocytes secrete Gdf15 to stimulate the growth and invasiveness of protate cancer cells [65]. Osteocyte-derived Fgf23 also enhances proliferation and invasion of prostate cancer cells, and the genetic knockdown of Fgf23 decreases tumor growth in vivo. Interestingly, FGF23 can also be produced by prostate cancer cells and could act in both paracrine and/or endocrine manner to promote prostate cancer progression. The growth of prostate cancer cells in bone also alters osteocyte biology [66]. Tumors growing in mouse tibiae increased intraosseous pressure and mechanically stimulated osteocytes, which in turn produced Ccl5 and matrix metalloproteinases to stimulate tumor proliferation and invasion. Intriguingly, the serum levels of the Wnt antagonists Sclerostin and Dkk-1 levels are elevated in patients with prostate bone metastasis [67]. This elevation might be a compensatory response to the increased osteoblasts presented at metastatic skeletal sites.
VII. Osteocytes and their derived factors as therapeutic targets
Osteocytes and their derived factors have become a valuable target for the treatment of bone disease. In this section, we will discuss the potential of targeting osteocytes for the treatment of cancer in bone and the effects of chemotherapy on osteocytes.
Sclerostin.
Sclerostin binds to Lipoprotein receptor-related proteins (Lrp) 4, 5, and 6 to downregulate Wnt/beta-catenin signaling and inhibit bone formation [68–70]. Several neutralizing antibodies against Sclerostin have been developed and have shown remarkable ability to stimulate new bone formation, and transiently decrease bone resorption, in osteoporotic patients. Thus, Sclerostin is currently recognized as a target for the treatment of osteoporotic bone loss [71]. Interestingly, Sclerostin circulating levels are elevated in patients with cancers that grow in bone [72, 38, 73, 74]. We and others demonstrated in preclinical models of MM that Sclerostin plays an important role in myeloma-induced suppression of bone formation. Inhibition of Sclerostin using genetic and pharmacologic approaches prevented the suppression of osteoblasts in bones bearing MM cells. More importantly, treatment with neutralizing antibodies stimulated osteoblast differentiation and promoted new bone formation in bones infiltrated with MM cells. Treatment with anti-Sclerostin had no effect on tumor burden across the different animal models used [42, 43, 41]. More recently, similar findings have been shown in a preclinical model of bone metastatic breast cancer [75]. Together, these results support that targeting osteocyte-derived Sclerostin is a novel therapeutic approach to combat cancer-induced bone disease. Despite the promising results obtained in preclinical models, to the best of our knowledge, there are no clinical trials studying the effects of anti-Sclerostin in cancer patients with bone involvement.
Dkk-1.
Dkk-1 is abundantly produced by osteocytes and osteoblasts, as well as by some cancer cells like MM cells, breast and prostate cancer cells. In MM, pharmacological inhibition of Dkk1 results in bone protection due to both increases in osteoblasts and decreases in osteoclasts [39, 40]. However, the in vivo effects of Dkk-1 inhibition on tumor were controversial between studies. Several clinical trials in different populations of MM patients were performed but, as of today, Dkk-1 is not an approved therapeutic target for the treatment of MM. Of note, a new bispecific antibody targeting both Sclerostin and Dkk-1 has been developed, which has more potent anabolic effects that targeting each factor alone. [76] However, the effects of this dual antibody in humans or cancer in bone remain to be determined.
Fgf-23.
Fgf-23 is an endocrine hormone secreted by the osteocytes that plays an important role in the regulation of phosphate levels [77]. Fgf-23 levels are elevated in several cancers, including prostate cancer and myeloma. [78, 45, 79, 80]. Importantly, low serum Fgf-23 levels correlate with survival in prostate cancer patients [78], suggesting that targeting it could be a therapeutic target to inhibit the progression of cancer. Earlier this year, the FDA approved the use of Burosumab, a neutralizing antibody against Fgf-23 to treat patients with treat X-linked hypophosphatemia (XLH), as well as tumor-induce osteomalacia [81]. Future research efforts are needed to define the effects of Burosumab in cancer that grows in bone.
Notch signaling.
The Notch signaling pathway mediates communication between adjacent cells and controls proliferation and programmed death programs. The dysregulation of Notch signaling leads to the progression of several cancers in bone [82, 83]. Pharmacologic inhibition of the Notch pathway with gamma secretase inhibitors (GSIs) in cancer results in multiple beneficial outcomes, including decreased tumor growth, maintenance of osteocyte viability, and inhibition of bone destruction [84, 85]. However, GSIs induce severe gut toxicity, limiting their use in the clinic [86]. Our group is developing a novel bone-targeted Notch inhibitor to bypass gut toxicity. Preliminary results showed that bone-targeted inhibition of Notch decreases tumor growth and bone destruction, without inducing gut toxicity. Although this approach is promising, identification of specific components of the Notch pathway that contribute to cancer progression in bone is paramount to avoid the off-target effects of GSIs [84]. In this line, neutralizing antibodies against Notch receptors and ligands Delta-1 and Jagged-1 are currently being studied in preclinical models [83, 87]. Future research efforts are needed to define the precise Notch receptors and ligands involved in the interactions between cancer cells and osteocytes.
Connexin 43 hemichannels.
Connexin 43 is a protein expressed in osteocytes that forms gap junctions and mediates communication among nearby osteocytes and between osteocytes other cells in the bone [88, 89]. Connexin 43 hemichannels are mechanosensitive and mediate osteocytic response to mechanical signals [88]. Osteocytic Connexin 43 hemichannels are required for the full inhibitory effects of conditioned media from mechanically stimulated osteocytes on migration, invasion, and growth of breast cancer cells [90, 59]. Further, the permeability of connexin 43 channels for small molecules makes them highly attractive for delivering drugs directly into the cytoplasm. Several small molecules targeting connexin 43 have been developed and are currently being tested for the treatment of osteosarcoma (i.e. ALMB-01683; humanized Cx43 monoclonal antibody), which could be used in the future for the treatment of other cancers that infiltrate bones.
Mechanical stimulation.
Exercise promotes skeletal preservation by regulating bone remodeling via osteocytes and is often proposed as an intervention for cancer patients [91]. For instance, tibial compression decreases osteolysis and tumor formation in a human metastatic breast cancer animal model [60, 57]. Therefore, mechanical stimulation of osteocytes might be considered as a treatment for breast cancer patients. However, it is important to note that exercise and the associated mechanical stimulation in bone might not always be safe and possible during cancer treatment. Several studies have investigated the effects of delivering low-intensity vibration as a mean to deliver mechanical signals without exercise [91]. In preclinical models, low-intensity vibration signal decreased tumor progression and bone loss in several models of cancer, including MM [92, 93]. This promising approach is currently being tested in the clinic.
Rankl.
The pro-osteoclastogenic cytokine Rankl plays a key role in cancer-induced bone loss by promoting osteoclast differentiation and bone resorption [94]. New developments demonstrate that besides bone destruction, soluble Rankl, formed after cleavage of membrane-bound Rankl, could also be involved in bone cancer metastasis [95]. Denosumab is a monoclonal antibody against Rankl with strong anti-resorptive activity [96]. Currently, this agent is only approved to prevent skeletal-related events in cancer patients with solid tumors (breast and prostate) and bone metastases. However, a recent study showed that Denosumab has non-inferiority for the prevention of skeletal-related events in multiple myeloma patients when compared with zoledronic acid [97]. It is likely that osteocytes mediate some of the protective effects of Denosumab, as they are the main source of Rankl and cancer cells increase Rankl expression in osteocytes.
Effects of current anti-cancer therapies on osteocytes:
It is clear now that anti-cancer therapies have an impact on bone and osteocytes [98]. Bisphosphonates are potent anti-resorptive agents and the gold standard approach to treat/prevent skeletal-related events induced by cancer. Besides decreasing bone loss by inhibiting osteoclast activity, bisphosphonates prevent osteocyte apoptosis, which also contributes to better bone health [99]. The synthetic glucocorticoid dexamethasone is often used in combination with other chemotherapy due to its ability to induce cancer cell apoptosis. Glucocorticoids have deleterious effects in bone partially mediated by increasing apoptosis in osteocytes and osteoblasts [100]. Bortezomib is a first-in-class proteasome inhibitor that blocks the degradation of abnormal or misfolded proteins targeted for destruction [101]. These proteins accumulate and result in MM cell apoptosis. In addition to its anti-cancer effects, proteasome inhibitors also transiently increase bone formation by acting on osteoblasts [102]. In osteocytes, Bortezomib prevents the increase in osteocyte apoptosis induced by MM cells [35]. Another anti-cancer agent with effects in osteocytes is Aplidin [103, 104]. This agent, isolated from a marine truncate Aplidium albicans, exhibits anti-MM activity. Aplidin also induces osteocyte programmed cell death [105]. Interestingly, this effect is prevented when combined with Bortezomib [105]. Lastly, aromatase inhibitors are used to treat endocrine-responsive breast cancer [106]. Aromatase inhibitors induce bone loss, a process in part mediated by increases in osteocyte-derived Sclerostin [107]. For more effects of anti-cancer therapy in osteocytes and bone, see here [108].
VIII. Conclusion
Interactions between tumor cells and bone cells contribute to tumor progression in bone. Early studies demonstrated that osteoblasts and osteoclasts are important in the vicious cycle that fuels tumor progression in the skeleton. Advances in the last decade have provided compelling evidence that osteocytes communicate with cancer cells and thus are also part of the vicious cycle of cancer in bone. Now we know that cancer cells “educate” osteocytes to promote tumor progression and bone destruction (Figure 1). Further, preclinical work in the last years have proven that targeting osteocytes and their derived factors is an attractive strategy to prevent bone destruction and slow down tumor progression.
Figure 1. Bidirectional crosstalk signaling between cancer cells and osteocytes in the bone niche.
Osteocytes and cancer cells communicate via exchange of soluble factors and physical interactions. Cancer cells induced osteocyte apoptosis and increase their production of osteoclastogenic factors to further promote osteoclast formation and bone resorption. Further, cancer cells stimulate osteocyte production of Wnt antagonists, which in turn contribute to suppress osteoblast formation and new bone formation. Lastly, cancer cells also increase Vegf-a secretion by osteocytes to enhance angiogenesis in the tumor niche. Osteocytes communication with cancer cells results in increased proliferation and tumor growth, and increased invasiveness and migration in cancer cells, ultimately favoring bone metastasis.
Yet, several questions regarding the role of osteocytes in bone metastasis and the growth of cancer in the bone remain to be determined. For instance, it is hypothesized that primary tumors modify and prime the sites of future metastasis, creating a pre-metastatic niche that favors their growth and survival in bone [109]. However, if osteocytes contribute to generate a microenvironment conducive for bone metastasis is unknown. Moreover, although pharmacological targeting of osteocytes improves cancer-induced bone disease, it is unknown whether therapies targeting osteocytes can simultaneously 1) repair the osteolytic lesions and 2) preserve the anti-cancer effects when combined with chemotherapy. Osteoblasts are key regulators of cancer cell dormancy. Yet, whether direct effects of osteocytes or indirect, through their influence on osteoblasts, contribute to the regulation of dormancy demands further investigation. Another interesting underexplored area is the role of osteocytes in the development of resistance to chemotherapy. Identification of the specific mechanisms mediating the different functions that osteocytes perform in bones colonized by cancer cells will lead to a better understanding of cancer cell dissemination, survival, and proliferation in bone and guide the development of more efficient therapeutic approaches to treat cancer patients with bone lesions.
Acknowledgments
This work was supported by the National Institutes of Health (R37-CA251763 to J.D.C., R01-CA209882 to G.D.R. and T.B., and R01CA241677 to G.D.R.), the Arkansas COBRE program (NIGMS P20GM125503) to J.D.C., a Scholar Award by the American Society of Hematology Scholar Award (to J.D.C), and a Brian D. Novis Award by the International Myeloma Foundation (to J.D.C.).
Footnotes
Compliance with Ethical Standards
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
Conflict of Interest
Manish Adhikari and Jesus Delgado-Calle declare that they have no conflict of interest.
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.
References
Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance
- 1.Allen MR, Burr DB. Bone modeling and remodeling. In: D B MA, editors. Basic and Applied Bone Biology. First ed.: Elsevier; 2014. p. 75–90. [Google Scholar]
- 2.Frost HM. Tetracycline-based histological analysis of bone remodeling. Calcif Tissue Res.1969;3(3):211–37. [DOI] [PubMed] [Google Scholar]
- 3.Andersen TL, Sondergaard TE, Skorzynska KE, gnaes-Hansen F, Plesner TL, Hauge EM et al. A physical mechanism for coupling bone resorption and formation in adult human bone. Am J Pathol. 2009;174(1):239–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bellido T. Osteocyte-Driven Bone Remodeling. Calcif Tissue Int. 2014;94(1):25–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Burr DB, Allen MR. Basic and Applied Bone Biology. 1 ed.Elsevier; 2013. [Google Scholar]
- 6.Moore DD, Luu HH. Osteosarcoma. Cancer Treat Res. 2014;162:65–92. [DOI] [PubMed] [Google Scholar]
- 7.Rajkumar SV. Myeloma today: Disease definitions and treatment advances. Am J Hematol.2016;91(1):90–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Terpos E, Berenson J, Raje N, Roodman GD. Management of bone disease in multiple myeloma. Expert Rev Hematol. 2014;7(1):113–25. [DOI] [PubMed] [Google Scholar]
- 9.Macedo F, Ladeira K, Pinho F, Saraiva N, Bonito N, Pinto L et al. Bone Metastases: An Overview. Oncol Rev. 2017;11(1):321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Coleman RE. Metastatic bone disease: clinical features, pathophysiology and treatment strategies. Cancer Treatment Reviews. 2001;27(3):165–76. [DOI] [PubMed] [Google Scholar]
- 11.Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 1989;8(2):98–101. [PubMed] [Google Scholar]
- 12.Mundy GR. Mechanisms of osteolytic bone destruction. Bone. 1991;12 Suppl 1:S1–S6. [DOI] [PubMed] [Google Scholar]
- 13.Kiriyama T, Gillespie MT, Glatz JA, Fukumoto S, Moseley JM, Martin TJ. Transforming growth factor b stimulation of parathyroid hormone-related protein (PTHrP): A paracrine regulator. Mol Cell Endocrinol. 1993;92:55–62. [DOI] [PubMed] [Google Scholar]
- 14.Guise TA, Mundy GR. Cancer and bone. Endocr Rev. 1998;19(1):18–54. [DOI] [PubMed] [Google Scholar]
- 15.Bonewald LF. The Amazing Osteocyte. J Bone Miner Res. 2011;26(2):229–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Delgado-Calle J, Bellido T. Osteocytes and Skeletal Pathophysiology. Curr Mol Biol Rep. 2015;1(4):157–67. ●This review manuscript provides a deep overview of osteocytes function under physiological and pathological conditions.
- 17.Plotkin LI, Bellido T. Osteocytic signalling pathways as therapeutic targets for bone fragility. Nat Rev Endocrinol. 2016;12(10):593–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Dallas SL, Prideaux M, Bonewald LF. The osteocyte: an endocrine cell ... and more. Endocr Rev. 2013;34(5):658–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Duncan RL, Turner CH. Mechanotransduction and the functional response of bone to mechanical strain. Calcif Tissue Int. 1995;57(5):344–58. [DOI] [PubMed] [Google Scholar]
- 20.Burger EH, Klein-Nulend J, Van Der Plas A, Nijweide PJ. Function of osteocytes in bone--their role in mechanotransduction. J Nutr. 1995;125(7 Suppl):2020S-3S. [DOI] [PubMed] [Google Scholar]
- 21.Delgado-Calle J, Sato AY, Bellido T. Role and mechanism of action of sclerostin in bone. Bone. 2017;96:29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med. 2013;19(2):179–92. [DOI] [PubMed] [Google Scholar]
- 23.Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, O’Brien CA. Matrix-embedded cells control osteoclast formation. Nat Med. 2011;17(10):1235–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nakashima T, Hayashi M, Fukunaga T, Kurata K, Oh-hora M, Feng JQ et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med. 2011;17(10):1231–4. [DOI] [PubMed] [Google Scholar]
- 25.Jilka RL, O’Brien CA. The Role of Osteocytes in Age-Related Bone Loss. Curr Osteoporos Rep. 2016;14(1):16–25. [DOI] [PubMed] [Google Scholar]
- 26.White KE, Evans WE, O’Riordan JLH, Speer MC, Econs MJ, Lorenz-Depiereux B et al. Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23. Nat Genet. 2000;26(3):345–8. [DOI] [PubMed] [Google Scholar]
- 27.Balemans W, Ebeling M, Patel N, Van Hul E, Olson P, Dioszegi M et al. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Hum Mol Genet. 2001;10(5):537–43. [DOI] [PubMed] [Google Scholar]
- 28.Balemans W, Van Den Ende J, Freire Paes-Alves A, Dikkers FG, Willems PJ, Vanhoenacker F et al. Localization of the gene for sclerosteosis to the van Buchem disease-gene region on chromosome 17q12-q21. Am J Hum Genet. 1999;64(6):1661–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Roodman GD. Pathogenesis of myeloma bone disease. Leukemia. 2009;23(3):435–41. [DOI] [PubMed] [Google Scholar]
- 30.Roodman GD. Pathogenesis of myeloma bone disease. J Cell Biochem. 2010;109(2):283–91. [DOI] [PubMed] [Google Scholar]
- 31.Roodman GD. Targeting the bone microenvironment in multiple myeloma. J Bone Miner Metab. 2010;28(3):244–50. [DOI] [PubMed] [Google Scholar]
- 32.Croucher PI, McDonald MM, Martin TJ. Bone metastasis: the importance of the neighbourhood. Nat Rev Cancer. 2016;16(6):373–86. [DOI] [PubMed] [Google Scholar]
- 33.Giuliani N, Ferretti M, Bolzoni M, Storti P, Lazzaretti M, Dalla PB et al. Increased osteocyte death in multiple myeloma patients: role in myeloma-induced osteoclast formation. Leukemia. 2012;26(6):1391–401. [DOI] [PubMed] [Google Scholar]
- 34. Delgado-Calle J, Anderson J, Cregor MD, Hiasa M, Chirgwin JM, Carlesso N 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(5):1089–100. ●●This work describes the cellular and molecular mechanisms by which osteocytes and myeloma cells communicate in the bone niche.
- 35.Toscani D, Palumbo C, Dalla PB, Ferretti M, Bolzoni M, Marchica V 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(4):815–27. [DOI] [PubMed] [Google Scholar]
- 36.Keller H, Kneissel M. SOST is a target gene for PTH in bone. Bone. 2005;37(2):148–58. [DOI] [PubMed] [Google Scholar]
- 37.Bellido T, Ali AA, Gubrij I, Plotkin LI, Fu Q, O’Brien CA et al. Chronic elevation of PTH in mice reduces expression of sclerostin by osteocytes: a novel mechanism for hormonal control of osteoblastogenesis. Endocrinology. 2005;146(11):4577–83. [DOI] [PubMed] [Google Scholar]
- 38.Terpos E, Christoulas D, Katodritou E, Bratengeier C, Gkotzamanidou M, Michalis E et al. Elevated circulating sclerostin correlates with advanced disease features and abnormal bone remodeling in symptomatic myeloma: reduction post-bortezomib monotherapy. Int J Cancer. 2012;131(6):1466–71. [DOI] [PubMed] [Google Scholar]
- 39.Yaccoby S, Ling W, Zhan F, Walker R, Barlogie B, Shaughnessy JD Jr,Antibody-based inhibition of DKK1 suppresses tumor-induced bone resorption and multiple myeloma growth in vivo. Blood. 2007;109(5):2106–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Heath DJ, Chantry AD, Buckle CH, Coulton L, Shaughnessy JD Jr., , Evans HR 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(3):425–36. [DOI] [PubMed] [Google Scholar]
- 41. Delgado-Calle J, Anderson J, Cregor MD, Condon KW, Kuhstoss SA, Plotkin LI et al. Genetic deletion of Sost or pharmacological inhibition of sclerostin prevent multiple myeloma-induced bone disease without affecting tumor growth. Leukemia. 2017;31(12):2686–94. ●● This manuscript, and the two below describe how targeting the osteocyte-derived factor sclrostin induces new bone formation and protects from myeloma-induced bnoe loss.
- 42.McDonald MM, Reagan MR, Youlten SE, Mohanty ST, Seckinger A, Terry RL et al. Inhibiting the osteocyte specific protein sclerostin increases bone mass and fracture resistance in multiple myeloma. Blood. 2017;129(26):3452–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Eda H, Santo L, Wein MN, Hu DZ, Cirstea DD, Nemani N et al. Regulation of Sclerostin Expression in Multiple Myeloma by Dkk-1; A Potential Therapeutic Strategy for Myeloma Bone Disease. J Bone Miner Res. 2016;31(6):1225–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Mulcrone PL, Edwards SKE, Petrusca DN, Haneline LS, Delgado-Calle J, Roodman GD. Osteocyte Vegf-a contributes to myeloma-associated angiogenesis and is regulated by Fgf23. Sci Rep. 2020;10(1):17319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Suvannasankha A, Tompkins DR, Edwards DF, Petyaykina KV, Crean CD, Fournier PG et al. FGF23 is elevated in multiple myeloma and increases heparanase expression by tumor cells. Oncotarget. 2015;In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sabol H, Amorim T, Halladay DL, Kurihara N, Anderson J, Cregor M et al. Autocrine and paracrine Notch receptor 3 signaling in the myeloma niche stimulates tumor growth and bone destruction. Journal of Bone and Mineral Research. 2019;32(Supl 1):https://www.asbmr.org/meetings/annualmeeting/AbstractDetail?aid=3b75df5e-8ab8-4da1-a887-05234a187e72. [Google Scholar]
- 47.Trotter TM, Fok M, Gibson JT, Peker D, Javed A, Yang Y. Osteocyte apoptosis attracts myeloma cells to bone and supports progression through regulation of the bone marrow microenvironment. Blood. 2016;128(22):484. [Google Scholar]
- 48.Roodman GD. Mechanisms of bone metastasis. N Engl J Med. 2004;350(16):1655–64. [DOI] [PubMed] [Google Scholar]
- 49.Guise TA. Molecular mechanisms of osteolytic bone metastases. Cancer. 2000;88(12 Suppl):2892–8. [DOI] [PubMed] [Google Scholar]
- 50.Jiang WG, Sanders AJ, Katoh M, Ungefroren H, Gieseler F, Prince M et al. Tissue invasion and metastasis: Molecular, biological and clinical perspectives. Semin Cancer Biol. 2015;35 Suppl:S244–S75. [DOI] [PubMed] [Google Scholar]
- 51.Kusumbe AP. Vascular niches for disseminated tumour cells in bone. J Bone Oncol.2016;5(3):112–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Tamma R, Ribatti D. Bone Niches, Hematopoietic Stem Cells, and Vessel Formation. Int J Mol Sci. 2017;18(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Cui YX, Evans BA, Jiang WG. New Roles of Osteocytes in Proliferation, Migration and Invasion of Breast and Prostate Cancer Cells. Anticancer Res. 2016;36(3):1193–201. ●●This manuscript is one of the first studies describing how osteocytes control the proloferation, migration, and invasion of bone metastatic cells.
- 54.Chen A, Wang L, Liu S, Wang Y, Liu Y, Wang M et al. Attraction and Compaction of Migratory Breast Cancer Cells by Bone Matrix Proteins through Tumor-Osteocyte Interactions. Sci Rep. 2018;8(1):5420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhou JZ, Riquelme MA, Gao X, Ellies LG, Sun LZ, Jiang JX. Differential impact of adenosine nucleotides released by osteocytes on breast cancer growth and bone metastasis. Oncogene. 2015;34(14):1831–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ma YV, Lam C, Dalmia S, Gao P, Young J, Middleton K et al. Mechanical regulation of breast cancer migration and apoptosis via direct and indirect osteocyte signaling. J Cell Biochem. 2018;119(7):5665–75. [DOI] [PubMed] [Google Scholar]
- 57.Liu S, Wu D, Sun X, Fan Y, Zha R, Jalali A et al. Mechanical stimulations can inhibit local and remote tumor progression by downregulating WISP1. Faseb j. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Liu S, Fan Y, Chen A, Jalali A, Minami K, Ogawa K et al. Osteocyte-Driven Downregulation of Snail Restrains Effects of Drd2 Inhibitors on Mammary Tumor Cells. Cancer Res. 2018;78(14):3865–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhou JZ, Riquelme MA, Gu S, Kar R, Gao X, Sun L et al. Osteocytic connexin hemichannels suppress breast cancer growth and bone metastasis. Oncogene. 2016;35(43):5597–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Fan Y, Jalali A, Chen A, Zhao X, Liu S, Teli M et al. Skeletal loading regulates breast cancer-associated osteolysis in a loading intensity-dependent fashion. Bone Res. 2020;8:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Myers TJ, Longobardi L, Willcockson H, Temple JD, Tagliafierro L, Ye P et al. BMP2 Regulation of CXCL12 Cellular, Temporal, and Spatial Expression is Essential During Fracture Repair. J Bone Miner Res. 2015;30(11):2014–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK. Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res. 2002;62(6):1832–7. [PubMed] [Google Scholar]
- 63.Wang N, Docherty FE, Brown HK, Reeves KJ, Fowles AC, Ottewell PD et al. Prostate cancer cells preferentially home to osteoblast-rich areas in the early stages of bone metastasis: evidence from in vivo models. J Bone Miner Res. 2014;29(12):2688–96. [DOI] [PubMed] [Google Scholar]
- 64.Wang W, Sarazin BA, Kornilowicz G, Lynch ME. Mechanically-Loaded Breast Cancer Cells Modify Osteocyte Mechanosensitivity by Secreting Factors That Increase Osteocyte Dendrite Formation and Downstream Resorption. Front Endocrinol (Lausanne). 2018;9:352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wang W, Yang X, Dai J, Lu Y, Zhang J, Keller ET. Prostate cancer promotes a vicious cycle of bone metastasis progression through inducing osteocytes to secrete GDF15 that stimulates prostate cancer growth and invasion. Oncogene. 2019;38(23):4540–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Sottnik JL, Dai J, Zhang H, Campbell B, Keller ET. Tumor-induced pressure in the bone microenvironment causes osteocytes to promote the growth of prostate cancer bone metastases. Cancer Res. 2015;75(11):2151–8. ●●This manuscript describes the effects of metastatic prostate cancer on osteocytes.
- 67.Aufderklamm S, Hennenlotter J, Leidenberger P, Rausch S, Hohneder A, Kühs U et al. Systemic Alterations of Wnt Inhibitors in Patients with Prostate Cancer and Bone Metastases. Dis Markers. 2018;2018:1874598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J et al. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem. 2005;280(20):19883–7. [DOI] [PubMed] [Google Scholar]
- 69.Chang MK, Kramer I, Huber T, Kinzel B, Guth-Gundel S, Leupin O et al. Disruption of Lrp4 function by genetic deletion or pharmacological blockade increases bone mass and serum sclerostin levels. Proc Natl Acad Sci U S A. 2014;111(48):E5187-E95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kim J, Han W, Park T, Kim EJ, Bang I, Lee HS et al. Sclerostin inhibits Wnt signalling through tandem interaction with two LRP6 ectodomains. Nat Commun. 2020;11(1):5357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.McClung MR, Grauer A, Boonen S, Bolognese MA, Brown JP, ez-Perez A et al. Romosozumab in Postmenopausal Women with Low Bone Mineral Density. N Engl J Med. 2014;370(5):412–20. [DOI] [PubMed] [Google Scholar]
- 72.Kim W, Chung Y, Kim SH, Park S, Bae JH, Kim G et al. Increased sclerostin levels after further ablation of remnant estrogen by aromatase inhibitors. Endocrinol Metab (Seoul ). 2015;30(1):58–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Garcia-Fontana B, Morales-Santana S, Varsavsky M, Garcia-Martin A, Garcia-Salcedo JA, Reyes-Garcia R et al. Sclerostin serum levels in prostate cancer patients and their relationship with sex steroids. Osteoporos Int. 2014;25(2):645–51. [DOI] [PubMed] [Google Scholar]
- 74.Yavropoulou MP, van Lierop AH, Hamdy NA, Rizzoli R, Papapoulos SE. Serum sclerostin levels in Paget’s disease and prostate cancer with bone metastases with a wide range of bone turnover. Bone. 2012;51(1):153–7. [DOI] [PubMed] [Google Scholar]
- 75.Hesse E, Schröder S, Brandt D, Pamperin J, Saito H, Taipaleenmäki H. Sclerostin inhibition alleviates breast cancer-induced bone metastases and muscle weakness. JCI Insight. 2019;5(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Florio M, Gunasekaran K, Stolina M, Li X, Liu L, Tipton B et al. A bispecific antibody targeting sclerostin and DKK-1 promotes bone mass accrual and fracture repair. Nat Commun. 2016;7:11505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Quarles LD. FGF23, PHEX, and MEPE regulation of phosphate homeostasis and skeletal mineralization. Am J Physiol Endocrinol Metab. 2003;285(1):E1–E9. [DOI] [PubMed] [Google Scholar]
- 78.Mansinho A, Ferreira AR, Casimiro S, Alho I, Vendrell I, Costa AL et al. Levels of Circulating Fibroblast Growth Factor 23 (FGF23) and Prognosis in Cancer Patients with Bone Metastases. Int J Mol Sci. 2019;20(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Feng S, Wang J, Zhang Y, Creighton CJ, Ittmann M. FGF23 promotes prostate cancer progression. Oncotarget. 2015;6(19):17291–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kim HJ, Kim KH, Lee J, Oh JJ, Cheong HS, Wong EL et al. Single nucleotide polymorphisms in fibroblast growth factor 23 gene, FGF23, are associated with prostate cancer risk. BJU Int. 2014;114(2):303–10. [DOI] [PubMed] [Google Scholar]
- 81.Carpenter TO, Imel EA, Ruppe MD, Weber TJ, Klausner MA, Wooddell MM et al. Randomized trial of the anti-FGF23 antibody KRN23 in X-linked hypophosphatemia. J Clin Invest. 2014;124(4):1587–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Meurette O, Mehlen P. Notch Signaling in the Tumor Microenvironment. Cancer Cell.2018;34(4):536–48. [DOI] [PubMed] [Google Scholar]
- 83.Colombo M, Galletti S, Garavelli S, Platonova N, Paoli A, Basile A et al. Notch signalling deregulation in multiple myeloma: A rational molecular target. Oncotarget. 2015;6(29):26826–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ferrari A, McAndrews K, Nelson JH, Bell J, Srinivasan V, Ebetino FH et al. Bone-Targeted Inhibition of Notch Signaling Blocks Tumor Growth and Prevents Bone Loss Without Inducing Gut Toxicity In Immunodeficient and Immunocompetent Murine Models of Established Multiple Myeloma. J Bone Miner Res. 2019;32(Suppl 1):Available at https://www.asbmr.org/ItineraryBuilder/PresentationDetail.aspx?pid=06aa6e80-775d-477b-a922-0c109ad9e7e2&ptag=WebItinerarySearch. [Google Scholar]
- 85.Schwarzer R, Nickel N, Godau J, Willie BM, Duda GN, Schwarzer R et al. Notch pathway inhibition controls myeloma bone disease in the murine MOPC315.BM model. Blood Cancer J. 2014;4:e217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Searfoss GH, Jordan WH, Calligaro DO, Galbreath EJ, Schirtzinger LM, Berridge BR et al. Adipsin, a biomarker of gastrointestinal toxicity mediated by a functional gamma-secretase inhibitor. J Biol Chem. 2003;278(46):46107–16. [DOI] [PubMed] [Google Scholar]
- 87.Colombo M, Mirandola L, Platonova N, Apicella L, Basile A, Figueroa AJ et al. Notch-directed microenvironment reprogramming in myeloma: a single path to multiple outcomes. Leukemia. 2013;27(5):1009–18. [DOI] [PubMed] [Google Scholar]
- 88.Plotkin LI, Speacht TL, Donahue HJ. Cx43 and Mechanotransduction in Bone. Curr Osteoporos Rep. 2015;13(2):67–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Xu H, Gu S, Riquelme MA, Burra S, Callaway D, Cheng H et al. Connexin 43 Channels are Essential for Normal Bone Structure and Osteocyte Viability. J Bone Miner Res. 2015;30(3):550–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Riquelme MA, Gu S, Chavez JB, An Z, Jiang JX. Abstract P5–05-15: Activation of osteocytes connexin43 hemichannels suppresses breast cancer bone metastasis, a potential therapeutic target. Cancer Research. 2020;80(4 Supplement):P5–05-15-P5–05-15. [Google Scholar]
- 91.Pagnotti GM, Styner M, Uzer G, Patel VS, Wright LE, Ness KK et al. Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity. Nat Rev Endocrinol. 2019;15(6):339–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pagnotti GM, Chan ME, Adler BJ, Shroyer KR, Rubin J, Bain SD et al. Low intensity vibration mitigates tumor progression and protects bone quantity and quality in a murine model of myeloma. Bone. 2016;90:69–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Pagnotti GM, Adler BJ, Green DE, Chan ME, Frechette DM, Shroyer KR et al. Low magnitude mechanical signals mitigate osteopenia without compromising longevity in an aged murine model of spontaneous granulosa cell ovarian cancer. Bone. 2012;51(3):570–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Tanaka S, Nakamura K, Takahasi N, Suda T. Role of RANKL in physiological and pathological bone resorption and therapeutics targeting the RANKL-RANK signaling system. Immunol Rev. 2005;208:30–49. [DOI] [PubMed] [Google Scholar]
- 95.Asano T, Okamoto K, Nakai Y, Tsutsumi M, Muro R, Suematsu A et al. Soluble RANKL is physiologically dispensable but accelerates tumour metastasis to bone. Nat Metab. 2019;1(9):868–75. [DOI] [PubMed] [Google Scholar]
- 96.McClung MR, Lewiecki EM, Cohen SB, Bolognese MA, Woodson GC, Moffett AH et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med. 2006;354(8):821–31. [DOI] [PubMed] [Google Scholar]
- 97.Raje N, Terpos E, Willenbacher W, Shimizu K, Garcia-Sanz R, Durie B et al. Denosumab versus zoledronic acid in bone disease treatment of newly diagnosed multiple myeloma: an international, double-blind, double-dummy, randomised, controlled, phase 3 study. Lancet Oncol. 2018;19(3):370–81. [DOI] [PubMed] [Google Scholar]
- 98.Silbermann R, Roodman GD. Bone effects of cancer therapies: pros and cons. Curr Opin Support Palliat Care. 2011;5(3):251–7. [DOI] [PubMed] [Google Scholar]
- 99.Bellido T, Plotkin LI. Novel actions of bisphosphonates in bone: Preservation of osteoblast and osteocyte viability. Bone. 2011;49:50–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Weinstein RS. Glucocorticoid-induced osteoporosis and osteonecrosis. Endocrinol Metab Clin North Am. 2012;41(3):595–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.LeBlanc R, Catley LP, Hideshima T, Lentzsch S, Mitsiades CS, Mitsiades N et al. Proteasome inhibitor PS-341 inhibits human myeloma cell growth in vivo and prolongs survival in a murine model. Cancer Res. 2002;62(17):4996–5000. [PubMed] [Google Scholar]
- 102.Giuliani N, Morandi F, Tagliaferri S, Lazzaretti M, Bonomini S, Crugnola M et al. The proteasome inhibitor bortezomib affects osteoblast differentiation in vitro and in vivo in multiple myeloma patients. Blood. 2007;110(1):334–8. [DOI] [PubMed] [Google Scholar]
- 103.Mitsiades CS, Ocio EM, Pandiella A, Maiso P, Gajate C, Garayoa M et al. Aplidin, a marine organism-derived compound with potent antimyeloma activity in vitro and in vivo. Cancer Res. 2008;68(13):5216–25. [DOI] [PubMed] [Google Scholar]
- 104.Caers J, Menu E, De RH, Lepage D, Van VE, Van CB et al. Antitumour and antiangiogenic effects of Aplidin in the 5TMM syngeneic models of multiple myeloma. Br J Cancer. 2008;98(12):1966–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Delgado-Calle J, Kurihara N, Atkinson EG, Nelson J, Miyagawa K, Galmarini CM et al. Aplidin (plitidepsin) is a novel anti-myeloma agent with potent anti-resorptive activity mediated by direct effects on osteoclasts. Oncotarget. 2019;10(28):2709–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Goss PE. Emerging role of aromatase inhibitors in the adjuvant setting. Am J Clin Oncol. 2003;26(4):S27–33. [DOI] [PubMed] [Google Scholar]
- 107.Gaillard S, Stearns V. Aromatase inhibitor-associated bone and musculoskeletal effects: new evidence defining etiology and strategies for management. Breast Cancer Res. 2011;13(2):205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Silbermann R, Roodman GD. Myeloma bone disease: Pathophysiology and management. J Bone Oncol. 2013;2(2):59–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Liu Y, Cao X. Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell. 2016;30(5):668–81. [DOI] [PubMed] [Google Scholar]

