<<

Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Clinical Review Research

Molecular Mechanisms of Bone Metastasis and Associated Muscle

David L. Waning and Theresa A. Guise

Abstract Bone is a preferred site for breast cancer metastasis and leads to pathologic bone loss due to increased osteoclast-induced bone resorption. The homing of tumor cells to the bone depends on the support of the bone microenvironment in which the tumor cells prime the premetastatic niche. The colonization and growth of tumor cells then depend on adaptations in the invading tumor cells to take advantage of normal physiologic responses by mimicking bone marrow cells. This concerted effort by tumor cells leads to uncoupled bone remodeling in which the balance of osteoclast-driven bone resorption and osteoblast- driven bone deposition is lost. Breast cancer bone metastases often lead to osteolytic lesions due to hyperactive bone resorption. Release of growth factors from bone matrix during resorption then feeds a "vicious cycle" of bone destruction leading to many skeletal-related events. In addition to activity in bone, some of the factors released during bone resorption are also known to be involved in regeneration and contraction. In this review, we discuss the mechanisms that lead to osteolytic breast cancer bone metastases and the potential for cancer-induced bone-muscle cross-talk leading to skeletal muscle weakness. Clin Cancer Res; 20(12); 1–7. 2014 AACR.

Introduction ing skeletal-related events but is not curative with regard to Bone metastases are common in patients with advanced tumor burden (1, 2). malignancy. Primary tumors exhibit metastatic tropism to A significant comorbidity of osteolytic bone metastases is particular organs and the skeleton is a preferred site for muscle weakness and that is often associated with breast cancer metastasis. Breast cancer that is metastatic to cancer cachexia. Cachexia is a common paraneoplastic bone causes a significant imbalance in normal bone remo- syndrome that is characterized by severe due to deling through perturbation of osteoclast-mediated bone loss of both fat and lean body mass (3, 4). Although the age resorption and osteoblast-mediated bone formation (1). and chemotherapeutic treatment regimens of patients with Bone metastases are classified on the basis of radiographic advanced and bone metastases make it difficult to appearance as either osteolytic or osteoblastic (osteosclero- assess the true incidence of malignancy-induced muscle tic). Breast cancer is typically associated with osteolytic weakness (5), a clinical perspective suggests that many lesions, but most cases involve uncoupled components of patients do experience severe muscle weakness and fatigue. both bone destruction and new bone formation. Bone Improving muscle function and mobility of patients with metastases from breast cancer affect 65% to 80% of patients cancer would have a positive impact on adherence to with advanced malignancy (2). Bone metastases cause treatment regimens and overall health (5). Therefore, a severe bone and increased risk of pathologic fracture, better understanding of the mechanism(s) of muscle weak- hypercalcemia, and nerve compression syndromes that ness associated with bone metastases and cancer cachexia significantly reduce the quality of life (1). Perhaps most will lead to targeted therapeutics. Moreover, refocusing devastating is the fact that once the primary tumor has attention to determine muscle quality in addition to spread to the bone, it is incurable. The current standard of improving muscle mass will likely provide the most ben- care for patients with bone loss due to osteolytic bone eficial treatment options for this devastating complication metastases includes antiresorptive therapy aimed at reduc- of malignancy. Molecular mechanisms of bone metastasis The initiation and progression of bone metastasis are complex multistep processes. Tumor cells must detach from Authors' Affiliation: Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis, Indiana the primary tumor and enter the systemic circulation (intra- Corresponding Author: Theresa A. Guise, 980 W. Walnut, Walther Hall R3/ vasation), evade detection by the immune system, and Room C123, Indianapolis, IN 46202. Phone: 317-278-7512; Fax: 317-278- adhere to capillaries in the bone marrow leading to extrav- 2912; E-mail: [email protected] asation into the bone marrow space (6). Tumor cells in the doi: 10.1158/1078-0432.CCR-13-1590 bone first form micrometastases that can either develop into 2014 American Association for Cancer Research. overt metastatic lesions or lay dormant for long periods

www.aacrjournals.org OF1

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Waning and Guise

before reactivating in the bone microenvironment. In either promotes adhesion of breast cancer cells in bone and is case, it is believed that the invading tumor cells prime the associated with bone metastasis (20). avb3 integrin also bone microenvironment by enriching the premetastatic cooperates with bone sialoprotein and matrix metallopro- niche (local environment) for further colonization and teinase (MMP)-2 to promote tumor cell colonization in growth of tumor cells (Fig. 1; refs. 2, 7–9). bone (21, 22). Receptor activator of NF-kB (RANK) med- The hematopoietic system plays an important role in iates osteoclast-induced bone resorption and supports development of the premetastatic niche. The bone marrow tumor cell colonization (23). The chemokine CXC ligand may serve as a protective milieu for dormant tumor cells to 12 [CXCL12; also known as stromal cell-derived factor 1 resist chemotherapeutic attack, and tumor cells may use the (SDF-1)] is a potent chemoattractant for HSCs and is highly same physiologic mechanisms as those used by hemato- expressed on osteoblasts and bone marrow stromal cells. poietic stem cells (HSC) homing to bone (10, 11). In the Expression of its receptor, CXC receptor 4 (CXCR4), on premetastatic niche, the invading tumor cells prime the cancer cells plays an important role in bringing tumor cells stroma by production of factors that elicit responses in cells to bone (24). In addition, interactions between CXCL12 of the bone microenvironment and make it conducive to and CXCR4 in the bone microenvironment lead to an tumor colonization and growth (2). In addition, bone upregulation of avb3 integrin, facilitating additional cell resorption also regulates HSC homing (12). Factors derived adhesion. CXCR4 was identified as one of a set of proteins from tumor cells include osteopontin (OPN), which pro- highly overexpressed in breast cancer cells (MDA-MB-231) motes bone marrow cell migration and tumor cell prolif- of high bone metastatic potential by serial selection in vivo eration (13, 14); heparanase (HPSE), which acts in the (10). Kang and colleagues also found that MMP-1, inter- extracellular matrix to reduce heparin sulfate chain length leukin (IL)-11, and connective tissue growth factor (CTGF) leading to increased bone resorption (15); and parathyroid were highly expressed in in vivo serially selected tumor cells hormone-related protein (PTHrP), which promotes bone that exhibited increased homing to bone compared with resorption (16) and may also enhance production of bone parental cells. IL-11 and MMP-1 stimulate bone resorption marrow chemokines such as C-C motif ligand 2 (17). by increasing osteoblast production of RANK ligand Recently, it has also been shown that the sympathetic (RANKL). Increased expression of MMP-1 and a disintegrin nervous system is also capable of stimulating stromal cells, and metalloproteinase with thrombospondin motifs thus promoting breast cancer bone metastasis (18). (ADAMTS1) suppresses osteoprotegerin (OPG) expression Tumor cells invading the bone also express factors that in osteoblasts, which leads to osteoclast differentiation facilitate further recruitment to the bone microenviron- (25). CTGF stimulates osteoblast proliferation, which leads ment, a process called osteotropism (19). avb3 integrin to further osteoclast activation and increased osteolysis (2).

Breast cancer cells

Figure 1. Premetastatic niche and αvβ3 bone homing. Modulation of the OPN RANK Tumor cell bone microenvironment by HPSE CXCR4 homing circulating breast cancer cells PTHrP MMP-1 results in priming of the bone IL-11 marrow as a premetastatic niche CTGF through tumor cell secretion of HSC homing OPN, HPSE, and PTHrP. CXCR4 Colonization of the bone and CXCL12 recruitment of HSC occur by tumor (SDF-1) cell expression of integrins (avb3), RANK, CXCR4, MMP-1, IL-11, and CTGF. CXCL12 (SDF-1) expression on osteoblasts Osteoblast facilitates homing of tumor Osteoclast Bone matrix cells to bone.

© 2014 American Association for Cancer Research CCR Reviews

OF2 Clin Cancer Res; 20(12) June 15, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Bone Metastasis and Muscle Weakness

Following osteotropism, the tumor cells adapt to the (40–43). Blocking bone resorption, especially through bone marrow space by expressing factors that allow growth modulating TGF-b signaling, offers a promising area for in their new microenvironment. Osteolytic lesions are the therapeutic intervention in bone metastasis and poten- most common type observed from breast metastatic tially its comorbidities. to bone. PTHrP secreted by breast cancer cells was the first characterized tumor-derived mediator of bone destruction Muscle dysfunction associated with breast cancer bone (16). In mice without detectable circulating PTHrP or metastasis hypercalcemia, neutralizing antibodies to PTHrP blocked Muscle and bone anabolism are tightly coupled during breast cancer bone metastases-associated bone loss and growth and development. Conversely, muscle and bone tumor growth. It was then shown that TGF-b in the bone catabolism occur during aging. Yet the cellular and molec- microenvironment induced the expression of PTHrP by ular mechanisms linking these two tissues are not well metastatic breast cancer cells that led to increased bone understood. destruction (26). Breast cancer cells in bone also express Muscle is known to secrete many factors capable of COX-2, which supports the development and progression affecting other tissues. These factors, collectively termed of bone metastases controlled by prostaglandin leading to myokines, include the bone active molecules insulin-like bone resorption (27). The osteolytic factors IL-8 and IL-11 growth factor-I (IGF-I), fibroblast growth factor (FGF)-2, are also expressed by tumor cells in the bone microenvi- (also called growth and differentiation factor; ronment and directly support osteoclast maturation (28, GDF-8), and IL-6 (44). Our current understanding of bone 29). In addition to secreted factors, tumor cells express and muscle cross-talk seems to show a predominant role of transcription factors that support growth in bone. The signaling in the direction of muscle to bone. Yet bone- transcription factors, GLI2, runt-related transcription factor derived factors are also known to modulate muscle. For 2 (RUNX2), and hypoxia-inducible growth factor-1 a (HIF- example, Indian hedgehog promotes myoblast survival and 1a) promote osteolysis. GLI2, part of the Hedgehog signal- myogenesis in both mouse and chick embryos (45) thus ing network, induces PTHrP expression leading to bone indicating bidirectional bone–muscle cross-talk. It seems destruction (30). RUNX2 regulates MMP-9 transcription likely that in cases of abnormal physiology, such as osteo- leading to increased tumor cell invasion by breaking down lytic bone metastases, the signals are co-opted and lead to a the extracellular matrix (31). HIF-1a expression inhibits shift in the homeostatic signaling balance (Fig. 3). osteoblast differentiation and promotes osteoclastogenesis, Data from our laboratory using a preclinical model of thus supporting bone resorption and tumor growth (32, breast cancer bone metastases (MDA-MB-231 cells) show 33). Tumor cells also express Jagged1 (Jag1) that activates significant reduction in forelimb grip strength and ex vivo the Notch pathway, which activates osteoclast differentia- maximum specific force generation of the extensor digi- tion (34). torum longus (EDL) muscle that cannot be explained by In the normal adult, setting bone is constantly remodeled reduction in muscle mass. Ex vivo specific force calculations to adjust for functional demands or to repair microfractures compensate for differences in size and weight of individual that occur as a part of normal activity. This process is driven muscles. Further muscle dysfunction is systemic and depen- by the coupled activity of osteoclasts that resorb mineral- dent on tumor-induced osteolytic bone resorption without ized matrix and osteoblasts that lay down new bone (35, tumor cell involvement in the muscle. Primary MDA-MB- 36). Ultimately, tumor cells in the bone microenvironment 231 tumors (mammary fat pad injection site) do not elicit disrupt this normal physiologic process and skew balance muscle dysfunction (46). Our investigation into muscle toward either bone destruction or bone formation. In most function in mice with breast cancer bone metastases was breast cancers metastatic to bone, the tumor cells produce borne out of the observation that these mice develop factors that directly or indirectly induce the formation of cachexia with advancing bone destruction. Cancer cachexia osteoclasts. In turn, bone resorption releases growth factors is one of the most common paraneoplastic conditions in from bone matrix (e.g., TGF-b) that stimulate tumor growth advanced malignancy, occurring in approximately 80% of and further osteolysis. This reciprocal interaction between patients. There is no effective treatment for cancer cachexia, breast cancer cells and the bone microenvironment results and it has been estimated to be responsible for 20% of in a "vicious cycle" that increases both bone destruction and cancer-related deaths (3, 47). However, there is a large the tumor burden (Fig. 2; ref. 2). heterogeneity in clinical presentation of cachexia that can Preclinical data suggest that reducing bone resorption vary according to tumor type, site, and individual patient prevents the development of bone metastases. Osteoclast factors. In fact, the true incidence of cancer cachexia is likely inhibitors are useful agents to slow or reverse bone loss (2), greatly underestimated (5). whereas antiresorptive therapy (bisphosphonates), OPG, Many well-established models of cancer cachexia have and other RANKL antagonists reduce growth of bone metas- used reduction of muscle size to imply muscle dysfunction. tases (37, 38). TGF-b antagonism is another mechanism for However, this does not take into account the loss of muscle reducing tumor growth in bone. TGF-b is abundant in the quality. Our laboratory has shown that mice with bone mineralized bone matrix and is released from the matrix metastases exhibited a primary defect (in addition to loss of during osteoclastic bone resorption (39). Blocking the muscle weight) that is independent of cachexia, although TGF-b pathway reduces bone metastasis and tumor burden mechanisms of cancer cachexia may also be at work (48). In

www.aacrjournals.org Clin Cancer Res; 20(12) June 15, 2014 OF3

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Waning and Guise

Breast cancer cells

GLI-2 RUNX2 HIF-1α Figure 2. Vicious cycle of osteolytic bone metastasis. Osteolytic bone PTHrP destruction due to dysregulation of COX-2 normal bone remodeling is IL-11 predominant in breast cancer metastasis. Breast cancer cells colonizing the bone secrete TGF-β osteolytic factors: PTHrP, COX-2, and IL-11. Tumor cells also express transcription factors GLI2, RUNX2, and HIF-1a that promote osteolysis. Jag1 expressed on Jag1 RANKL tumor cells activates osteoclast Notch RANK differentiation by inducing Notch signaling in preosteoclasts. Bone resorption releases TGF-b from the bone matrix, which enhances tumor cell proliferation and Osteoclast Osteoblast survival, thus feeding a vicious Bone matrix cycle leading to further bone destruction. Osteolytic metastases

© 2014 American Association for Cancer Research CCR Reviews

a mouse model of multiple myeloma that leads to osteo- The salient question therefore is what factor(s) derived lytic bone lesions but without measurable cachexia, we from bone matrix during resorption is capable of inducing observed systemic muscle dysfunction (49). In both of systemic muscle dysfunction? Bone matrix is a rich store- these mouse models of osteolytic bone loss, the severity house of growth factors that have known effects on muscle, of muscle dysfunction correlatedwithanincreaseinbone such as activin A, TGF-b, IGF-I, and bone morphogenic destruction. protein 2 (BMP-2; refs. 50, 51). It is useful to begin by

Bone Skeletal Figure 3. Bone–muscle cross-talk. muscle Bone and muscle are physically and functionally tightly coupled. IGF-I, FGF-2, myostatin, and IL-6 are factors released from muscle that have functions in muscle as Osteokines well as bone. These factors have Activin A been collectively termed myokines. b TGF-β Likewise, activin A, TGF- , IGF-I, IGF-I and BMP-2 released from bone affect bone as well as muscle, BMP-2 Myokines and thus we have called these IGF-I osteokines. During osteolytic bone FGF-2 Breast resorption due to breast cancer, Myostatin cancer bone metastasis osteokines are IL-6 cells released and may be responsible for systemic skeletal muscle weakness. In certain settings, tumor-derived factors could also lead to modulation of muscle activity (dotted line). © 2014 American Association for Cancer Research CCR Reviews

OF4 Clin Cancer Res; 20(12) June 15, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Bone Metastasis and Muscle Weakness

considering these as potentiators of muscle dysfunction due exhibited an increase in sinking episodes in a forced swim to bone destruction. test, reduced "time on" in a rotarod test (68, 69), and The high-affinity activin type 2 receptor, ActRIIB, med- reduced time before falling from a vertical screen test iates signaling of a small group of TGF-b family members (70). These results indicate an overall defect in motor (activin A, myostatin, GDF-11) and is important in regu- performance in mice lacking proper D metabolism. lating muscle mass (52). Pharmacologic blockade of In human studies, and are associated ActRIIB prevents muscle wasting, induces muscle satellite with muscle weakness. In addition to general weakness, cell mobilization and differentiation, and significantly pro- more specific muscle deficits are also commonly reported, longs survival in murine models of cachexia (53). In addi- including reduced timed up and go, 6-minute walk, stair tion, blockade of ActRIIB dramatically improves muscle climbing, and object lifting (71, 72). It should be noted that function in a Duchenne muscular dystrophy model (mdx myopathies reported with vitamin D deficiency might mice; ref. 54). However, in these studies it is not possible to also involve calcium and phosphate deficiencies, thus com- determine whether the effect is due to blocking activin A, plicating the assessment of individual factors. FGF-23 neu- myostatin, or GDF-11 signaling due to receptor usage tralizing antibody, which increases serum phosphate and overlap. Myostatin signaling antagonism has been investi- vitamin D levels, has been shown to improve murine grip gated as a way to improve muscle wasting due to cachexia strength in a model of rickets/osteomalacia (X-linked hypo- because myostatin is a potent inhibitor of skeletal muscle phosphatemic rickets/osteomalacia), suggesting that vita- differentiation and growth (55). Activin A has also been min D levels could influence muscle function (73). shown to function with myostatin to reduce muscle size microRNA (miRNA) profiling of tumors has identified (56). GDF-11 shares 90% sequence homology with myos- signatures associated with diagnosis and progression. tatin and in skeletal muscle inhibits myoblast differentia- Human miRNA Let-7 was recently shown to be elevated tion (57), suggesting that GDF-11 may act in a very similar in serum of mice harboring breast cancer bone metastases manner as myostatin. (74). miRNA Let-7 is also elevated in serum of elderly TGF-b is a potent regulator of wound healing in muscle, patients with muscle weakness and has been suggested to and persistent exposure leads to altered extracellular matrix reduce regenerative capacity in aging (75). architecture and formation of fibrotic tissue in muscle Another intriguing possibility is the role of the sympa- (58). Increased TGF-b signaling in muscle also inhibits thetic nervous system in muscle weakness due to bone satellite cell activation and impairs myocyte differentiation metastases. The sympathetic nervous system modulates (59, 60). Increased TGF-b signaling is also associated with skeletal muscle metabolism, ion transport, and contractil- skeletal muscle dysfunction in many of the muscular ity. Recent evidence has shown that the sympathetic nervous dystrophies (61, 62). In a direct assessment of the effect system is capable of promoting breast cancer bone metas- of TGF-b on muscle function, the contractile properties of tasis through stimulation of marrow stromal cells (18), yet a the EDL muscle were examined from limbs exposed to connection to muscle weakness has not been investigated. recombinant TGF-b. Muscle function from limbs receiving TGF-b treatment exhibited a significant reduction in spe- Summary b cific force (63). These experiments suggest that TGF- is a Bone and muscle functions are tightly coupled in normal capable factor in reducing muscle function independent of physiology. Recent studies have focused on muscle as an changes in muscle mass. endocrine organ with a predominant role over bone in In contrast with the negative effects possible from activin bone–muscle cross-talk. Osteolytic bone metastases from b A, myostatin, and TGF- signaling in muscle, IGF-I and breast cancer represent a severe divergence from normal BMP-2 signaling results in muscle hypertrophy (58, 64, 65). bone physiology by tipping the balance of remodeling. IGF-I is a major regulator of muscle mass due to its effect on Bone is a rich storehouse of growth factors that have activity myogenic cell proliferation and differentiation (66). Like- in bone (as a part of normal remodeling) and in other wise, BMP signaling leads to muscle hypertrophy, but, organs, including muscle. It is therefore possible that during interestingly, specific force (corrected for muscle mass) is hyperactive bone resorption, bone might have a predom- significantly lower when BMP signaling is constitutively inant role over muscle in bone–muscle cross-talk and activated (64). This result demonstrates the importance of become a source of "osteokines" that affect muscle function. interpreting muscle-specific function, not merely muscle Likewise, factors released from muscle may play an impor- mass, in murine models of skeletal muscle weakness. tant role in bone metabolism that could further exacerbate In addition to factors released from bone matrix during the role of bone in muscle dysfunction. Identification and osteoclast-driven resorption, other factors present in pat- characterization of such factors would provide new possi- ients with malignancy involving bone may play important bilities for therapeutic intervention in muscle weakness roles in muscle weakness. Serum vitamin D levels are low associated with malignancy and perhaps cancer cachexia. among patients with breast cancer with either osteoporosis or metastatic bone disease who are receiving bisphospho- nate therapy (67). Vitamin D deficiency has been studied Disclosure of Potential Conflicts of Interest T.A. Guise reports receiving commercial research grants from AstraZeneca in rodent models using vitamin D receptor knockout and Exelexis, is a consultant/advisory board member for Novartis, and has (VDRKO) mice. Functional muscle tests in VDRKO mice provided expert testimony on bisphosphonates and osteonecrosis of the jaw

www.aacrjournals.org Clin Cancer Res; 20(12) June 15, 2014 OF5

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Waning and Guise

for Novartis. No potential conflicts of interest were disclosed by the other JerryW.andPeggyS.ThrogmartinEndowment of the Indiana Univer- author. sity (IU) Simon Cancer Center, the IU Simon Cancer Center Breast Cancer Program, and a generous donation from the Withycombe family Grant Support (to T.A. Guise). This work was supported by NIH grants (U01CA143057 from the NCI Tumor Microenvironment Network; R01CA69158), the Susan G. Received November 19, 2013; revised January 30, 2014; accepted February Komen Foundation, the Indiana Economic Development Grant, the 3, 2014; published OnlineFirst March 27, 2014.

References 1. Roodman GD. Mechanisms of bone metastasis. N Engl J Med 20. Zhao Y, Bachelier R, Treilleux I, Pujuguet P, Peyruchaud O, Baron R, 2004;350:1655–64. et al. Tumor alphavbeta3 integrin is a therapeutic target for breast 2. Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal cancer bone metastases. Cancer Res 2007;67:5821–30. attraction. Nat Rev Cancer 2011;11:411–25. 21. Karadag A, Ogbureke KU, Fedarko NS, Fisher LW. Bone sialoprotein, 3. Fearon KC, Glass DJ, Guttridge DC. Cancer cachexia: media- matrix metalloproteinase 2, and alpha(v)beta3 integrin in osteotropic tors, signaling, and metabolic pathways. Cell Metab 2012;16: cancer cell invasion. J Natl Cancer Inst 2004;96:956–65. 153–66. 22. Waltregny D, Bellahcene A, de Leval X, Florkin B, Weidle U, Castronovo 4. Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev 2009;89: V. Increased expression of bone sialoprotein in bone metastases 381–410. compared with visceral metastases in human breast and prostate 5. Fox KM, Brooks JM, Gandra SR, Markus R, Chiou CF. Estimation of cancers. J Bone Miner Res 2000;15:834–43. Cachexia among Cancer Patients Based on Four Definitions. Can 23. Jones DH, Nakashima T, Sanchez OH, Kozieradzki I, Komarova SV, J Oncol 2009;2009:693458. Sarosi I, et al. Regulation of cancer cell migration and bone metastasis 6. Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and by RANKL. Nature 2006;440:692–6. evolving paradigms. Cell 2011;147:275–92. 24. Liang Z, Wu T, Lou H, Yu X, Taichman RS, Lau SK, et al. Inhibition of 7. Lu X, Mu E, Wei Y, Riethdorf S, Yang Q, Yuan M, et al. VCAM-1 breast cancer metastasis by selective synthetic polypeptide against promotes osteolytic expansion of indolent bone micrometastasis of CXCR4. Cancer Res 2004;64:4302–8. breast cancer by engaging alpha4beta1-positive osteoclast progeni- 25. Lu X, Wang Q, Hu G, Van Poznak C, Fleisher M, Reiss M, et al. ADAMTS1 tors. Cancer cell 2011;20:701–14. and MMP1 proteolytically engage EGF-like ligands in an osteolytic 8. Luzzi KJ, MacDonald IC, Schmidt EE, Kerkvliet N, Morris VL, Cham- signaling cascade for bone metastasis. Genes Dev 2009;23:1882–94. bers AF, et al. Multistep nature of metastatic inefficiency: dormancy of 26. Yin JJ, Selander K, Chirgwin JM, Dallas M, Grubbs BG, Wieser R, et al. solitary cells after successful extravasation and limited survival of early TGF-beta signaling blockade inhibits PTHrP secretion by breast can- micrometastases. Am J Pathol 1998;153:865–73. cer cells and bone metastases development. J Clin Invest 1999;103: 9. Watson MA, Ylagan LR, Trinkaus KM, Gillanders WE, Naughton MJ, 197–206. Weilbaecher KN, et al. Isolation and molecular profiling of bone 27. Singh B, Berry JA, Shoher A, Ayers GD, Wei C, Lucci A. COX-2 marrow micrometastases identifies TWIST1 as a marker of early involvement in breast cancer metastasis to bone. Oncogene 2007;26: tumor relapse in breast cancer patients. Clin Cancer Res 2007; 3789–96. 13:5001–9. 28. Zhang Y, Fujita N, Oh-hara T, Morinaga Y, Nakagawa T, Yamada M, 10. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon-Cardo et al. Production of interleukin-11 in bone-derived endothelial cells and C, et al. A multigenic program mediating breast cancer metastasis to its role in the formation of osteolytic bone metastasis. Oncogene bone. Cancer cell 2003;3:537–49. 1998;16:693–703. 11. Yoneda T. Cellular and molecular basis of preferential metastasis of 29. Bendre MS, Montague DC, Peery T, Akel NS, Gaddy D, Suva LJ. breast cancer to bone. J Orthop Sci 2000;5:75–81. Interleukin-8 stimulation of osteoclastogenesis and bone resorption is 12. Kollet O, Dar A, Shivtiel S, Kalinkovich A, Lapid K, Sztainberg Y, a mechanism for the increased osteolysis of metastatic bone disease. et al. Osteoclasts degrade endosteal components and promote Bone 2003;33:28–37. mobilization of hematopoietic progenitor cells. Nat Med 2006;12: 30. Sterling JA, Oyajobi BO, Grubbs B, Padalecki SS, Munoz SA, Gupta A, 657–64. et al. The hedgehog signaling molecule Gli2 induces parathyroid 13. Anborgh PH, Mutrie JC, Tuck AB, Chambers AF. Role of the metas- hormone-related peptide expression and osteolysis in metastatic tasis-promoting protein osteopontin in the tumour microenvironment. human breast cancer cells. Cancer Res 2006;66:7548–53. J Cell Mol Med 2010;14:2037–44. 31. Pratap J, Javed A, Languino LR, van Wijnen AJ, Stein JL, Stein GS, 14. McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince et al. The Runx2 osteogenic transcription factor regulates matrix TA, et al. Systemic endocrine instigation of indolent tumor growth metalloproteinase 9 in bone metastatic cancer cells and controls cell requires osteopontin. Cell 2008;133:994–1005. invasion. Mol Cell Biol 2005;25:8581–91. 15. Kelly T, Suva LJ, Huang Y, Macleod V, Miao HQ, Walker RC, et al. 32. Dunn LK, Mohammad KS, Fournier PG, McKenna CR, Davis HW, Expression of heparanase by primary breast tumors promotes bone Niewolna M, et al. Hypoxia and TGF-beta drive breast cancer bone resorption in the absence of detectable bone metastases. Cancer Res metastases through parallel signaling pathways in tumor cells and the 2005;65:5778–84. bone microenvironment. PloS one 2009;4:e6896. 16. Guise TA, Yin JJ, Taylor SD, Kumagai Y, Dallas M, Boyce BF, et al. 33. Hiraga T, Kizaka-Kondoh S, Hirota K, Hiraoka M, Yoneda T. Hypoxia Evidence for a causal role of parathyroid hormone-related protein in the and hypoxia-inducible factor-1 expression enhance osteolytic bone pathogenesis of human breast cancer-mediated osteolysis. J Clin metastases of breast cancer. Cancer Res 2007;67:4157–63. Invest 1996;98:1544–9. 34. Sethi N, Dai X, Winter CG, Kang Y. Tumor-derived JAGGED1 promotes 17. Li X, Loberg R, Liao J, Ying C, Snyder LA, Pienta KJ, et al. A destructive osteolytic bone metastasis of breast cancer by engaging notch sig- cascade mediated by CCL2 facilitates prostate cancer growth in bone. naling in bone cells. Cancer cell 2011;19:192–205. Cancer Res 2009;69:1685–92. 35. Raggatt LJ, Partridge NC. Cellular and molecular mechanisms of bone 18. Campbell JP, Karolak MR, Ma Y, Perrien DS, Masood-Campbell SK, remodeling. J Biol Chem 2010;285:25103–8. Penner NL, et al. Stimulation of host bone marrow stromal cells by 36. Allen MR, Burr DB. Bisphosphonate effects on bone turnover, micro- sympathetic nerves promotes breast cancer bone metastasis in mice. damage, and mechanical properties: what we think we know and what PLoS Biol 2012;10:e1001363. we know that we don't know. Bone 2011;49:56–65. 19. Clezardin P, Teti A. Bone metastasis: pathogenesis and therapeutic 37. Boissier S, Ferreras M, Peyruchaud O, Magnetto S, Ebetino FH, implications. Clin Exp Metastasis 2007;24:599–608. Colombel M, et al. Bisphosphonates inhibit breast and prostate

OF6 Clin Cancer Res; 20(12) June 15, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Bone Metastasis and Muscle Weakness

carcinoma cell invasion, an early event in the formation of bone 56. Lee SJ, Lee YS, Zimmers TA, Soleimani A, Matzuk MM, Tsuchida K, metastases. Cancer Res 2000;60:2949–54. et al. Regulation of muscle mass by follistatin and activins. Mol 38. Croucher PI, Shipman CM, Lippitt J, Perry M, Asosingh K, Hijzen A, Endocrinol 2010;24:1998–2008. et al. Osteoprotegerin inhibits the development of osteolytic bone 57. Jeanplong F, Falconer SJ, Thomas M, Matthews KG, Oldham AM, disease in multiple myeloma. Blood 2001;98:3534–40. Watson T, et al. Growth and differentiation factor-11 is developmen- 39. Korpal M, Yan J, Lu X, Xu S, Lerit DA, Kang Y. Imaging transforming tally regulated in skeletal muscle and inhibits myoblast differentiation. growth factor-beta signaling dynamics and therapeutic response in Open Int J Mol Integr Physiol 2012;2:127–38. breast cancer bone metastasis. Nat Med 2009;15:960–6. 58. Serrano AL, Munoz-Canoves P. Regulation and dysregulation of 40. Biswas S, Nyman JS, Alvarez J, Chakrabarti A, Ayres A, Sterling J, et al. fibrosis in skeletal muscle. Exp Cell Res 2010;316:3050–8. Anti-transforming growth factor ss antibody treatment rescues bone 59. Allen RE, Boxhorn LK. Inhibition of skeletal muscle satellite cell dif- loss and prevents breast cancer metastasis to bone. PloS one 2011;6: ferentiation by transforming growth factor-beta. J Cell Physiol e27090. 1987;133:567–72. 41. Buijs JT, Stayrook KR, Guise TA. The role of TGF-beta in bone 60. Allen RE, Boxhorn LK. Regulation of skeletal muscle satellite cell metastasis: novel therapeutic perspectives. BoneKEy Rep 2012; proliferation and differentiation by transforming growth factor-beta, 1:96. insulin-like growth factor I, and fibroblast growth factor. J Cell Physiol 42. Mohammad KS, Chen CG, Balooch G, Stebbins E, McKenna CR, 1989;138:311–5. Davis H, et al. Pharmacologic inhibition of the TGF-beta type I receptor 61. Chen YW, Nagaraju K, Bakay M, McIntyre O, Rawat R, Shi R, et al. Early kinase has anabolic and anti-catabolic effects on bone. PloS one onset of inflammation and later involvement of TGFbeta in Duchenne 2009;4:e5275. muscular dystrophy. Neurology 2005;65:826–34. 43. Wright LE, Frye JB, Lukefahr AL, Timmermann BN, Mohammad KS, 62. Kollias HD, McDermott JC. Transforming growth factor-beta and myos- Guise TA, et al. Curcuminoids block TGF-beta signaling in human tatin signaling in skeletal muscle. J Appl Physiol 2008;104:579–87. breast cancer cells and limit osteolysis in a murine model of breast 63. Mendias CL, Gumucio JP, Davis ME, Bromley CW, Davis CS, Brooks cancer bone metastasis. J Nat Prod 2013;76:316–21. SV. Transforming growth factor-beta induces skeletal muscle 44. DiGirolamo DJ, Kiel DP, Esser KA. Bone and skeletal muscle: neigh- and fibrosis through the induction of atrogin-1 and scleraxis. Muscle bors with close ties. J Bone Miner Res 2013;28:1509–18. Nerve 2012;45:55–9. 45. Bren-Mattison Y, Hausburg M, Olwin BB. Growth of limb muscle is 64. SartoriR,SchirwisE,BlaauwB,BortolanzaS,ZhaoJ,EnzoE,etal. dependent on skeletal-derived Indian hedgehog. Dev Biol 2011;356: BMP signaling controls muscle mass. Nat Genet 2013;45:1309–18. 486–95. 65. Schiaffino S, Mammucari C. Regulation of skeletal muscle growth by 46. WaningDL,MohammadKS,AnderssonDC,JohnSK,RiekenS,Xie the IGF1-Akt/PKB pathway: insights from genetic models. Skeletal W, et al.Role of the tumor-bone microenvironment in muscle weak- muscle 2011;1:4. ness and cachexia. Cancer-Induced Bone Disease. Miami, FL: 66. Florini JR, Ewton DZ, Coolican SA. Growth hormone and the insulin- IBMS; 2013. like growth factor system in myogenesis. Endocr Rev 1996;17:481– 47. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, 517. et al. Definition and classification of cancer cachexia: an international 67. Wang-Gillam A, Miles DA, Hutchins LF. Evaluation of vitamin D defi- consensus. lancet Oncol 2011;12:489–95. ciency in breast cancer patients on bisphosphonates. Oncologist 48. Fearon K, Arends J, Baracos V. Understanding the mechanisms and 2008;13:821–7. treatment options in cancer cachexia. Nat Rev Clin Oncol 2013;10: 68. Burne TH, Johnston AN, McGrath JJ, Mackay-Sim A. Swimming 90–9. behaviour and post-swimming activity in Vitamin D receptor knockout 49. Waning DL, Mohammad KS, Reiken S, Marks AR, Roodman GD, Guise mice. Brain Res Bull 2006;69:74–8. TA. Muscle weakness is associated with osteolysis in multiple mye- 69. Minasyan A, Keisala T, Zou J, Zhang Y, Toppila E, Syvala H, et al. loma. Cancer-Induced Bone Disease. Miami, FL: IBMS; 2013. Vestibular dysfunction in vitamin D receptor mutant mice. J Steroid 50. Sakai R, Eto Y. Involvement of activin in the regulation of bone Biochem Mol Biol 2009;114:161–6. metabolism. Mol Cell Endocrinol 2001;180:183–8. 70. Kalueff AV, Lou YR, Laaksi I, Tuohimaa P. Impaired motor performance 51. Wildemann B, Kadow-Romacker A, Haas NP, Schmidmaier G. Quan- in mice lacking neurosteroid vitamin D receptors. Brain Res Bull tification of various growth factors in different demineralized bone 2004;64:25–9. matrix preparations. J Biomed Mater Res A 2007;81:437–42 71. Russell JA. Osteomalacic myopathy. Muscle Nerve 1994;17:578–80. 52. Lee SJ, Reed LA, Davies MV, Girgenrath S, Goad ME, Tomkinson KN, 72. Schott GD, Wills MR. Muscle weakness in osteomalacia. Lancet et al. Regulation of muscle growth by multiple ligands signaling 1976;1:626–9. through activin type II receptors. Proc Natl Acad Sci U S A 2005; 73. Aono Y, Hasegawa H, Yamazaki Y, Shimada T, Fujita T, Yamashita T, 102:18117–22. et al. Anti-FGF-23 neutralizing antibodies ameliorate muscle weakness 53. Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, et al. Reversal of and decreased spontaneous movement of Hyp mice. J Bone Miner cancer cachexia and muscle wasting by ActRIIB antagonism leads to Res 2011;26:803–10. prolonged survival. Cell 2010;142:531–43. 74. Ell B, Mercatali L, Ibrahim T, Campbell N, Schwarzenbach H, Pantel K, 54. Pistilli EE, Bogdanovich S, Goncalves MD, Ahima RS, Lachey J, Seehra et al. Tumor-induced osteoclast miRNA changes as regulators and J, et al. Targeting the activin type IIB receptor to improve muscle mass biomarkers of osteolytic bone metastasis. Cancer cell 2013;24: and function in the mdx mouse model of Duchenne muscular dystro- 542–56. phy. Am J Pathol 2011;178:1287–97. 75. Drummond MJ, McCarthy JJ, Sinha M, Spratt HM, Volpi E, Esser KA, 55. Zimmers TA, Davies MV, Koniaris LG, Haynes P, Esquela AF, Tom- et al. Aging and microRNA expression in human skeletal muscle: a kinson KN, et al. Induction of cachexia in mice by systemically admin- microarray and bioinformatics analysis. Physiol Genomics 2011;43: istered myostatin. Science 2002;296:1486–8. 595–603.

www.aacrjournals.org Clin Cancer Res; 20(12) June 15, 2014 OF7

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst March 27, 2014; DOI: 10.1158/1078-0432.CCR-13-1590

Molecular Mechanisms of Bone Metastasis and Associated Muscle Weakness

David L. Waning and Theresa A. Guise

Clin Cancer Res Published OnlineFirst March 27, 2014.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-13-1590

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2014/05/22/1078-0432.CCR-13-1590. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2014 American Association for Cancer Research.