Next Article in Journal
Magnesium Is a Key Regulator of the Balance between Osteoclast and Osteoblast Differentiation in the Presence of Vitamin D3
Next Article in Special Issue
Efficient Selection of Antibodies Reactive to Homologous Epitopes on Human and Mouse Hepatocyte Growth Factors by Next-Generation Sequencing-Based Analysis of the B Cell Repertoire
Previous Article in Journal
Cytokine Modulation in Breast Cancer Patients Undergoing Radiotherapy: A Revision of the Most Recent Studies
Previous Article in Special Issue
c-Src Recruitment is Involved in c-MET-Mediated Malignant Behaviour of NT2D1 Non-Seminoma Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting the Hepatocyte Growth Factor and c-Met Signaling Axis in Bone Metastases

1
Department of Urology, Chung-Ang University College of Medicine, Seoul 06973, Korea
2
Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Anam Hospital, Seoul 02841, Korea
3
Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea
4
Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(2), 384; https://doi.org/10.3390/ijms20020384
Submission received: 11 December 2018 / Revised: 14 January 2019 / Accepted: 15 January 2019 / Published: 17 January 2019
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))

Abstract

:
Bone metastasis is the terminal stage disease of prostate, breast, renal, and lung cancers, and currently no therapeutic approach effectively cures or prevents its progression to bone metastasis. One of the hurdles to the development of new drugs for bone metastasis is the complexity and heterogeneity of the cellular components in the metastatic bone microenvironment. For example, bone cells, including osteoblasts, osteoclasts, and osteocytes, and the bone marrow cells of diverse hematopoietic lineages interact with each other via numerous cytokines and receptors. c-Met tyrosine kinase receptor and its sole ligand hepatocyte growth factor (HGF) are enriched in the bone microenvironment, and their expression correlates with the progression of bone metastasis. However, no drugs or antibodies targeting the c-Met/HGF signaling axis are currently available in bone metastatic patients. This significant discrepancy should be overcome by further investigation of the roles and regulation of c-Met and HGF in the metastatic bone microenvironment. This review paper summarizes the key findings of c-Met and HGF in the development of novel therapeutic approaches for bone metastasis.

1. Introduction

Bone metastasis is uniquely positioned in cancer research because of its diverse cellular populations comprising the metastatic microenvironment. For example, cells in the skeletal system, such as osteoblasts, osteoclasts, and osteocytes, as well as numerous subpopulations, including hematopoietic cells, in the bone marrow all play distinct roles in the progression of bone metastasis [1,2]. Bone metastasis is a major clinical problem for many types of cancers, particularly prostate, breast, lung, and kidney cancers. Bone metastasis is considered a terminal stage disease, and often results in extensive morbidities, such as severe bone pain, spinal cord compression, pathologic fracture, immobility, and ultimately death [3]. Currently, no drugs are available for curing or preventing bone metastasis. Anti-resorptives such as bisphosphonates and denosumab are used to delay or prevent the skeletal-related events (SREs), yet have not been proven to show significant overall survival benefits [4]. This conflict between important unmet clinical needs and the absence of effective therapeutic approaches is mostly due to the complexity in the signaling pathways among the diverse stromal cell types in the metastatic bone microenvironment [5]. For example, the transforming growth factor (TGF)-beta/bone morphogenetic protein (BMP) signaling pathway is the most comprehensively investigated mechanism of cancer-induced bone diseases [6,7]. In addition, the Wnt/beta-Catenin signaling pathway is frequently dysregulated in the bone lesions of multiple myeloma, prostate, and breast cancer metastasis [6,8]. Inhibitors of Wnt antagonists, such as anti-Sclerostin and anti-Dickkopf-related protein 1 (DKK1), have been investigated in the context of bone metastasis. Tyrosine kinases, such as Src family kinases, platelet-derived growth factor receptor (PDGFR), and c-Kit, have previously been highlighted as promising therapeutic targets in cancer-induced osteolysis and metastatic tumor growth in bone [9,10,11,12]. Inhibitors of chemokines and chemokine receptors, such as anti-C-C chemokine ligand 2 antibodies, have also been tested for prostate cancer bone metastasis [13,14]. However, none of these drugs or antibodies resulted in significant benefits during clinical trials in patients with bone metastasis, suggesting that the signaling pathways in the metastatic bone microenvironment are extremely heterogeneous and complex and also that no single target can effectively stop progression of the disease. Accordingly, more extensive delineation of growth factors and their related signaling pathways specific to the metastatic bone microenvironment must be clarified to develop more effective therapeutics for bone metastasis. Recent publications have highlighted hepatocyte growth factor (HGF) and its cognate receptor c-Met as promising therapeutic targets of metastatic tumor growth and osteolysis of bone metastasis. This review will summarize the new findings on the roles of hepatocyte growth factor (HGF) and c-Met receptor tyrosine kinase in the progression of bone metastasis.

2. Biology of c-Met and HGF

HGF, also known as Scatter Factor (SF), is a soluble cytokine and belongs to the plasminogen-related growth factor family. HGF/SF was identified by two groups that independently characterized a mitogenic factor for hepatocytes (named HGF) and a fibroblast-derived epithelial cell mobility factor (named SF), respectively, and later, the two factors were identified to be the same [15]. Proteolytic regulation is an important characteristic of HGF. Single chain pro-form HGF is produced and subsequently activated by proteolytic cleavage into disulfide-bonded α- and β-chain heterodimers by serine proteinases such as urokinase plasminogen activator (uPA); tissue-type plasminogen activator (tPA); coagulation factors X, XI, and XII; and a homologue of factor XII. HGF has a strong binding affinity with heparan sulfate proteoglycans abundant in the extracellular matrix, which explains the limited diffusion of HGF throughout the tissue.
c-Met is encoded by the MET gene that was cloned and identified as a proto-oncogene by George Vande Woude at the U.S. National Cancer Institute in 1984 [16]. c-Met is a receptor tyrosine kinase, and HGF is the sole ligand for c-Met. Since its discovery, the c-Met receptor has been extensively investigated for its roles in cellular functions and tumor progression, and thus ample review papers are available to read [17,18]. Therefore, only a summary on the c-Met structure and downstream signaling will be briefly covered here. c-Met is a single-pass disulfide-linked 50kDa α- and 140kDa β-subunit heterodimer. The extracellular compartment of c-Met has three domains, including semaphorin, PSI (plexins, semaphorins, and integrins), and IPT (immunoglobulin-plexin-transcription) domains. The intracellular compartment contains a kinase domain and a multifunctional docking site. c-Met activation by ligand binding leads to the phosphorylation of Y1234 and Y1235 in the kinase domain. Subsequently, Y1349 and Y1356 in the multifunctional docking site become phosphorylated, followed by the recruitment of multiple adaptor proteins, such as growth factor receptor-bound protein (Grb) 2; Grb 2-associated binding protein (Gab) 1; Src homology-2-containing (SHC); v-crk sarcoma virus CT10 oncogene homolog (CRK); and CRK like (CRKL), as well as effector molecules such as phosphatidylinositol 3-kinase (PI3K), phospholipase C (PLC) γ and Src, Src homology domain-containing 5′ inositol phosphatase (SHIP)-2, and the transcription factor signal transducer and activator of transcription (STAT)-3. In particular, Gab 1 is a multi-adaptor protein that serves binding sites for numerous downstream adaptors, further diversifying the intracellular signaling pathways. Intracellular downstream signaling pathways of c-Met include Akt/PKB (protein kinase B) regulating cell survival and growth; Src/FAK (focal adhesion kinase) regulating mobility and invasion; JNK (c-Jun N-terminal Kinase) regulating transformation; Ras/Raf regulating cell proliferation and cell cycle progression; and PIP3 (phosphatidyl inositol 3,4,5-triphosphate)/PKC (protein kinase C), which works to negatively regulate c-Met signaling, etc.

3. c-MET/HGF Signaling in Cancer Progression

c-Met was first cloned using chemically transformed human osteosarcoma cells and was identified as an oncogene as mentioned above [18]. Indeed, the activation of c-Met is frequently observed in many types of cancers. Mechanisms of c-Met activation include gain-of-function mutation, gene amplification, and constitutive over-production of HGF in the microenvironment, and therefore, c-Met can be activated in both ligand-dependent and -independent manners. c-Met can be indirectly activated by growth factors other than HGF, including insulin-like growth factor (IGF)-1, epidermal growth factor (EGF), etc. Varkaris et al. demonstrated that IGF-1 binding to its IGF receptor (IGFR) trans-activates c-Met ligand-independently via Src phosphorylation in prostate cancer tumor cells [19]. Lee et al. demonstrated that a similar mechanism (i.e., IGF-dependent trans-activation of c-Met) exists in osteoblasts [20]. Furthermore, Breindel et al. demonstrated that epidermal growth factor (EGF) trans-activates c-Met via MAPK [21].
Genetic and protein expression analyses of patient tissue samples demonstrated that c-Met activation is closely associated with tumor progression and metastasis in breast and prostate cancers, with a higher expression in the metastatic lesions compared to the primary lesions [22,23,24,25]. c-Met is predominantly expressed in epithelial cells (including carcinoma cells), hepatocytes, and endothelial cells, and HGF expression is limited by mesenchymal cells, such as fibroblasts and muscle cells. c-Met and HGF are essential to the development of the liver and the cardio-vascular system, and thus Met- or Hgf-knockout mice are both embryonic lethal [26,27,28]. The pro-tumorigenic role of c-Met can function through various downstream signaling pathways, such as Ras/Raf/MAPK, PI3K/Akt, and Wnt/β-catenin, among many others, resulting in multiple biological effects, such as cell proliferation, cell motility, invasion, metastasis, evasion of apoptosis, epithelial-to-mesenchymal transition, and angiogenesis.

4. c-MET/HGF Signaling in Bone Metastasis

One of the earliest and key findings supporting the role of c-Met and HGF in bone metastasis was reported by the Vande Woude group, who demonstrated that prostate cancer bone metastasis patient samples showed significantly increased c-Met protein expression in the metastatic lesions, and also that c-Met expression inversely correlated with androgen receptor expression [29]. The authors obtained and analyzed 90 prostatectomy surgical samples, and discovered that Met is expressed in half of the primary cancers, whereas Met expression is expressed in all metastases. In addition, Met expression inversely correlated with the expression of the androgen receptor, indicating that Met expression increases during the disease progression. Given that HGF is a stromal factor whose receptor, c-Met, is expressed in carcinoma cells, the c-Met/HGF axis was investigated in the context of tumor-stromal interactions. The study from Grano et al. was the first important report describing the expression and the roles of HGF and c-Met in osteoblasts and osteoclasts. The authors demonstrated that human osteoclasts and osteoblasts both express c-Met and that HGF stimulates both cell types. The authors also demonstrated that c-Met activation increases the intracellular calcium concentration and Src phosphorylation in osteoclasts, whereas c-Met activation in osteoblasts induced cell cycle progression, collectively suggesting that HGF is a coupling factor for osteoclasts and osteoblasts [30]. In line with this important finding, we and others have demonstrated that c-Met and HGF play important roles among three compartments in the metastatic bone microenvironments, i.e., tumor cells, osteoclasts, and osteoblasts [20,31,32]. According to the advent of novel tyrosine kinase inhibitors (TKIs), many studies regarding the role of c-Met/HGF in bone metastasis originated from the pre-clinical and clinical studies using novel c-Met TKIs.
Most recently, Smith et al. demonstrated that a novel c-Met and VEGFR dual TKI (cabozantinib) resulted in a partial or complete resolution of bone lesions measured by Tc-99m bone scans and also the improvement of cancer-induced bone pain [33,34]. Briefly, in a Phase 2 nonrandomized expansion study in castration resistance prostate cancer, the patients (n = 144) received cabozantinib 100 mg (n = 93) or 40 mg (n = 51) daily from the start until disease progression or unacceptable toxicity. Cabozantinib treatment resulted in pain relief (57% of patients) measured by a reduction or discontinuation of narcotic analgesics, as well as improvements in bone biomarkers. Both dosage group (100mg and 40mg) patients had benefits in the bone scan response in 73% and 45%, respectively, as well as reductions in measurable soft tissue disease in 80% and 79%, respectively. However, because cabozantinib is a TKI suppressing both tumor cells and bone cells, the clinical benefits observed in the phase 2 clinical trials may have been confounding effects of suppressing two compartments (i.e. tumor and stroma) at the same time. Indeed, cabozantinib reduced bone turnover blood serum markers such as alkaline phosphatase (ALP, a bone formation marker) and c-telopeptide (CTx, a bone resorption marker), within 12 weeks, indicating that cabozantinib affects the stromal compartment of the tumor microenvironment [34]. Accordingly, to dissect the net effect of c-Met suppression in the stromal compartment alone, we performed preclinical studies using cabozantinib-resistant bone metastatic prostate tumor cells, as well as in vitro studies using c-Met knockdown osteoblasts, and found that the suppression of c-Met specifically in osteoblasts suppressed osteoclastogenesis, tumor-induced osteolysis, and tumor growth in bone [20]. In parallel with our data, Tsai et al. demonstrated that HGF increased bone morphogenetic protein (BMP)-2 in human osteoblasts via c-Met, FAK, JNK, RUNX2, and p300 pathways [35], and Chen et al. demonstrated that HGF increased osteopontin in human osteoblasts via PI3K/AKT, c-Src, and AP-1 pathways [36]. These data suggest that the activation of osteoblasts in the metastatic bone microenvironment is dependent on the growth factors that can stimulate c-Met pathways. To more directly support this idea, Dai et al. showed that cabozantinib has direct anti-tumoral activity in their pre-clinical in vivo mouse models of metastatic prostate cancer, and more importantly, the data suggest that cabozantinib modulates osteoblast activity, which contributes to anti-tumoral efficacy [32,37]. Although the majority of pre-clinical and clinical results are from prostate cancer, c-Met inhibitors have been tested on other bone metastatic cancers, such as breast cancer, and showed a similar clinically efficacy [38]. Watanabe et al. used another c-Met/VEGFR2 dual kinase inhibitor (TAS-115) and showed that the novel inhibitor attenuates FMS-dependent osteoclast differentiation and prostate cancer-induced osteolysis [31,39]. Fioramonti et al. provided additional evidence that cabozantinib decreased tumor-induced osteolysis via direct effects on osteoclasts, as well as indirect effects on osteoblasts (reduction of RANKL and OPG expression), in agreement with our data [40]. Patnaik et al. demonstrated that cabozantinib induced CXCL12 and high mobility group box 1 (HMGB1) proteins, leading to increased neutrophil chemoattraction in the prostate tumor tissues of PTEN/p-53-decifient mice, suggesting that cabozantinib suppresses prostate tumors by activating anti-tumoral innate immunity [41]. Interestingly, short-term cabozantinib treatment (30 mg/kg, 5 × weekly for one or two weeks) in female Balb/c mice suppressed trabecular bone structures and reduced bone marrow cellularity, but increased the number of megakaryocytes. The effects were all transient and disappeared following cessation of the treatment [42]. Collectively, these lines of evidence indicate that HGF/c-Met signaling is important in the microenvironment of bone metastasis, and the efficacy of cabozantinib in bone metastasis is, at least in part, dependent on suppression of the stromal compartment cells, including osteoblasts and osteoclasts.

5. Limitations and Challenges Targeting c-MET in Bone Metastasis

Despite the large amount of data supporting the rationale to target c-Met/HGF in bone metastasis, clinical trial results with c-Met/HGF inhibitors, particularly TKIs, have failed to show favorable results supporting further advancement of the drugs in the clinic. For example, the COMET-1 trial, in which metastatic castration-resistant prostate cancer (CRPC) patients were treated with cabozantinib (60 mg once per day) or prednisone (5 mg twice per day), showed that the cabozantinib group had improved bone biomarkers, bone scan responses (BSR), and radiographic progression-free survival (rPFS), yet failed to satisfy the primary endpoint, i.e., improvement of the overall survival (OS) [43]. In a companion study, the COMET-2 trial, in which progressive mCRPC patients were treated with cabozantinib vs. mitoxantrone (topoisomerase inhibitor)/prednisone, showed no efficacy on the primary endpoint (more than 30% reduction in pain responses), with secondary endpoint (BSR and OS) showing trends favoring cabozantinib treatment. Based on these negative results from two clinical trials with cabozantinib, the c-MET and VEGFR2 dual TKI will not be available for mCRPC patients in the near future. Another c-Met TKI, Tivantinib (also known as ARQ 107), showed no effects in metastatic triple-negative breast cancer, but showed improved PFS in mCRPC patients in Phase 2 clinical trials [44,45]. Tivantinib alone or in combination with zoledronic acid has been shown to suppress breast cancer bone metastasis in an in vivo mouse model [46,47]. However, there are no clinical trials (registered in www.clinicaltrials.gov) ongoing for bone metastasis treated with tivantinib.
In addition to TKIs, there are several monoclonal antibodies blocking c-Met or HGF undergoing clinical development, such as onartuzumab (anti-c-Met antibody), emibetuzumab, LY3164530, JNJ-61186372, rilotuzumab (anti-HGF antibody), ficlatuzumab, etc [48]. These antibodies are currently in various phases of clinical trials or pre-clinical trials, but none of them are being tested for bone metastatic diseases. D’Amico et al. highlighted the roles of cancer stem cells expressing c-Met in renal cancer bone metastasis, and showed that the c-Met inhibitor JNJ-38877605 inhibits osteoclast activation in a preclinical NOD/SCID mouse model [49]. Recently, an interesting Phase 1 trial was registered at the clinicaltrials.gov (NCT01837602) website. The trial tests autologous c-Met redirected T cells administered intra-tumorally in metastatic breast cancer patients. c-Met-targeting chimeric antigen receptor (CAR) T cells showed promising therapeutic efficacy in a pre-clinical model [50], and thus the trial was initiated.
Even with the negative results from the multiple clinical trials, c-Met/HGF remains an important therapeutic target for metastatic cancers, including bone metastasis. Further extensive research to understand c-Met/HGF regulation in the organ-specific microenvironment is required for the clinical application of novel therapeutics. For example, roles of the extracellular matrix (ECM) and tumor-associated fibroblasts should be considered. Noriega-Guerra et al. described that the ECM is a reservoir for bioactive molecules, including HGF [51]. In addition, ECM components such as proteoglycans, matricelluar proteins, and/or proteases, can negatively regulate c-Met activation by sequestering HGF, and thus it is necessary to consider the effects of ECM components on HGF and c-Met for the development of novel therapeutic strategies. Similarly, Bendinelli et al. demonstrated that HGF and transforming growth factor (TGF)-β, both of which are enriched in the metastatic bone matrix, regulate the mesenchymal-to-epithelial transition (the opposite of the epithelial-to-mesenchymal transition) of bone metastatic breast cancer cells via the suppression of tumor suppressor Wwox (WW domain-containing oxidoreductase) [52,53]. In addition to the ECM, the hypoxic environment in bone metastasis is a key regulator in the network of the biological soluble and structural components of the matrix. In bone metastatic cells under hypoxia, similar patterns of Runx2 and SPARC are observed, with both showing downregulation. Conversely, hypoxia induces Endothelin 1, which upregulates SPARC, and these biological stimuli may be considered prognostic markers of bone metastasis in breast carcinoma patients [54]. Lastly, with the advancement of immuno-oncology, the immune system and tumor immunology warrant consideration. Papaccio et al. have extensively reviewed the relevance of c-Met/HGF in tumor immunity [55].

6. Conclusions

Since its first discovery as an oncogene, c-Met and its sole ligand HGF have been postulated to play critical roles in tumor progression, particularly tumor-stromal interactions [56,57]. Bone is a unique, complex microenvironment encompassing hard tissue, bone marrow immune cells, loose vasculature, enriched growth factors released from the matrix, and hypoxia. The c-Met and HGF axes play important functions in bone metastasis, and remain promising therapeutic targets for bone metastasis. However, as observed in the results of many clinical trials, the c-Met/HGF signaling is not simple, and the regulation and function of c-Met/HGF in the progression of bone metastasis necessitate extensive further investigation.

Author Contributions

Conceptualization: Y.M.W., I.H.C., and S.I.P.; Writing: Y.M.W. and S.I.P.; Editing: S.P.J., M.-K.K., I.H.C., and S.I.P.

Funding

This work was supported in part by the Korea University Research Grants (S.P.J. and S.I.P.), the BK21 Plus Program of the Ministry of Education of Korea (S.I.P.), the National Cancer Center of Korea (No. HA17C0040 to S.I.P.), the National Health Technology R&D Project (HI17C0710 to I.H.C.) and the National Research Foundation of Korea (Grants No. 2016R1D1A1B03933826 to Y.M.W.; 2017R1D1A1B03028103 to S.P.J.; 2018R1D1A1A02050248 to I.H.C.; and 2015R1C1A1A01051508 and 2018R1D1A1B07050329 to S.I.P.).

Acknowledgments

The authors thank Amy J. Koh for assistance with editing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Park, S.I.; Soki, F.N.; McCauley, L.K. Roles of bone marrow cells in skeletal metastases: No longer bystanders. Cancer Microenviron. 2011, 4, 237–246. [Google Scholar] [CrossRef] [PubMed]
  2. Buenrostro, D.; Park, S.I.; Sterling, J.A. Dissecting the role of bone marrow stromal cells on bone metastases. Biomed. Res. Int. 2014, 2014, 875305. [Google Scholar] [CrossRef] [PubMed]
  3. Croucher, P.I.; McDonald, M.M.; Martin, T.J. Bone metastasis: The importance of the neighbourhood. Nat. Rev. Cancer 2016, 16, 373. [Google Scholar] [CrossRef] [PubMed]
  4. Sousa, S.; Clézardin, P. Bone-Targeted Therapies in Cancer-Induced Bone Disease. Calcif. Tissue Int. 2018, 102, 227–250. [Google Scholar] [CrossRef] [PubMed]
  5. Park, S.H.; Keller, E.T.; Shiozawa, Y. Bone Marrow Microenvironment as a Regulator and Therapeutic Target for Prostate Cancer Bone Metastasis. Calcif. Tissue Int. 2018, 102, 152–162. [Google Scholar] [CrossRef] [PubMed]
  6. Sottnik, J.L.; Hall, C.L.; Zhang, J.; Keller, E.T. Wnt and Wnt inhibitors in bone metastasis. BoneKEy Rep. 2012, 1, 101. [Google Scholar] [CrossRef] [PubMed]
  7. Xu, J.; Acharya, S.; Sahin, O.; Zhang, Q.; Saito, Y.; Yao, J.; Wang, H.; Li, P.; Zhang, L.; Lowery, F.J.; et al. 14-3-3ζ turns TGF-β’s function from tumor suppressor to metastasis promoter in breast cancer by contextual changes of Smad partners from p53 to Gli2. Cancer Cell 2015, 27, 177–192. [Google Scholar] [CrossRef] [PubMed]
  8. Zhuang, X.; Zhang, H.; Li, X.; Li, X.; Cong, M.; Peng, F.; Yu, J.; Zhang, X.; Yang, Q.; Hu, G. Differential effects on lung and bone metastasis of breast cancer by Wnt signalling inhibitor DKK1. Nat. Cell. Biol. 2017, 19, 1274–1285. [Google Scholar] [CrossRef] [PubMed]
  9. Park, S.I.; Zhang, J.; Phillips, K.A.; Araujo, J.C.; Najjar, A.M.; Volgin, A.Y.; Gelovani, J.G.; Kim, S.J.; Wang, Z.; Gallick, G.E. Targeting SRC family kinases inhibits growth and lymph node metastases of prostate cancer in an orthotopic nude mouse model. Cancer Res. 2008, 68, 3323–3333. [Google Scholar] [CrossRef]
  10. Summy, J.M.; Gallick, G.E. Treatment for advanced tumors: SRC reclaims center stage. Clin. Cancer Res. 2006, 12, 1398–1401. [Google Scholar] [CrossRef] [PubMed]
  11. Kim, S.J.; Uehara, H.; Yazici, S.; Langley, R.R.; He, J.; Tsan, R.; Fan, D.; Killion, J.J.; Fidler, I.J. Simultaneous blockade of platelet-derived growth factor-receptor and epidermal growth factor-receptor signaling and systemic administration of paclitaxel as therapy for human prostate cancer metastasis in bone of nude mice. Cancer Res. 2004, 64, 4201–4208. [Google Scholar] [CrossRef] [PubMed]
  12. Mathew, P.; Thall, P.F.; Bucana, C.D.; Oh, W.K.; Morris, M.J.; Jones, D.M.; Johnson, M.M.; Wen, S.; Pagliaro, L.C.; Tannir, N.M.; et al. Platelet-derived growth factor receptor inhibition and chemotherapy for castration-resistant prostate cancer with bone metastases. Clin. Cancer Res. 2007, 13, 5816–5824. [Google Scholar] [CrossRef] [PubMed]
  13. Mizutani, K.; Sud, S.; McGregor, N.A.; Martinovski, G.; Rice, B.T.; Craig, M.J.; Varsos, Z.S.; Roca, H.; Pienta, K.J. The chemokine CCL2 increases prostate tumor growth and bone metastasis through macrophage and osteoclast recruitment. Neoplasia 2009, 11, 1235–1242. [Google Scholar] [CrossRef] [PubMed]
  14. Bonapace, L.; Coissieux, M.-M.; Wyckoff, J.; Mertz, K.D.; Varga, Z.; Junt, T.; Bentires-Alj, M. Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature 2014, 515, 130–133. [Google Scholar] [CrossRef] [PubMed]
  15. Stoker, M.; Gherardi, E.; Perryman, M.; Gray, J. Scatter factor is a fibroblast-derived modulator of epithelial cell mobility. Nature 1987, 327, 239–242. [Google Scholar] [CrossRef] [PubMed]
  16. Cooper, C.S.; Park, M.; Blair, D.G.; Tainsky, M.A.; Huebner, K.; Croce, C.M.; Vande Woude, G.F. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature 1984, 311, 29–33. [Google Scholar] [CrossRef] [PubMed]
  17. Organ, S.L.; Tsao, M.-S. An overview of the c-MET signaling pathway. Ther. Adv. Med. Oncol. 2011, 3, S7–S19. [Google Scholar] [CrossRef]
  18. Birchmeier, C.; Birchmeier, W.; Gherardi, E.; Vande Woude, G.F. Met, metastasis, motility and more. Nat. Rev. Mol. Cell Biol. 2003, 4, 915–925. [Google Scholar] [CrossRef]
  19. Varkaris, A.; Gaur, S.; Parikh, N.U.; Song, J.H.; Dayyani, F.; Jin, J.-K.; Logothetis, C.J.; Gallick, G.E. Ligand-independent activation of MET through IGF-1/IGF-1R signaling. Int. J. Cancer 2013, 133, 1536–1546. [Google Scholar] [CrossRef] [Green Version]
  20. Lee, C.; Whang, Y.M.; Campbell, P.; Mulcrone, P.L.; Elefteriou, F.; Cho, S.W.; Park, S.I. Dual targeting c-met and VEGFR2 in osteoblasts suppresses growth and osteolysis of prostate cancer bone metastasis. Cancer Lett. 2018, 414, 205–213. [Google Scholar] [CrossRef]
  21. Breindel, J.L.; Haskins, J.W.; Cowell, E.P.; Zhao, M.; Nguyen, D.X.; Stern, D.F. EGF receptor activates MET through MAPK to enhance non-small cell lung carcinoma invasion and brain metastasis. Cancer Res. 2013, 73, 5053–5065. [Google Scholar] [CrossRef] [PubMed]
  22. Jardim, D.L.F.; de Melo Gagliato, D.; Falchook, G.; Zinner, R.; Wheler, J.J.; Janku, F.; Subbiah, V.; Piha-Paul, S.A.; Fu, S.; Tannir, N.; et al. MET abnormalities in patients with genitourinary malignancies and outcomes with c-MET inhibitors. Clin. Genitourin. Cancer 2015, 13, e19–e26. [Google Scholar] [CrossRef] [PubMed]
  23. Verhoef, E.I.; Kolijn, K.; De Herdt, M.J.; van der Steen, B.; Hoogland, A.M.; Sleddens, H.F.B.M.; Looijenga, L.H.J.; van Leenders, G.J.L.H. MET expression during prostate cancer progression. Oncotarget 2016, 7, 31029–31036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. De Melo Gagliato, D.; Jardim, D.L.F.; Falchook, G.; Tang, C.; Zinner, R.; Wheler, J.J.; Janku, F.; Subbiah, V.; Piha-Paul, S.A.; Fu, S.; et al. Analysis of MET genetic aberrations in patients with breast cancer at MD Anderson Phase I unit. Clin. Breast Cancer 2014, 14, 468–474. [Google Scholar] [CrossRef] [PubMed]
  25. Colombel, M.; Eaton, C.L.; Hamdy, F.; Ricci, E.; van der Pluijm, G.; Cecchini, M.; Mege-Lechevallier, F.; Clézardin, P.; Thalmann, G. Increased expression of putative cancer stem cell markers in primary prostate cancer is associated with progression of bone metastases. Prostate 2012, 72, 713–720. [Google Scholar] [CrossRef] [PubMed]
  26. Bladt, F.; Riethmacher, D.; Isenmann, S.; Aguzzi, A.; Birchmeier, C. Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud. Nature 1995, 376, 768–771. [Google Scholar] [CrossRef]
  27. Schmidt, C.; Bladt, F.; Goedecke, S.; Brinkmann, V.; Zschiesche, W.; Sharpe, M.; Gherardi, E.; Birchmeier, C. Scatter factor/hepatocyte growth factor is essential for liver development. Nature 1995, 373, 699–702. [Google Scholar] [CrossRef]
  28. Uehara, Y.; Minowa, O.; Mori, C.; Shiota, K.; Kuno, J.; Noda, T.; Kitamura, N. Placental defect and embryonic lethality in mice lacking hepatocyte growth factor/scatter factor. Nature 1995, 373, 702–705. [Google Scholar] [CrossRef]
  29. Knudsen, B.S.; Gmyrek, G.A.; Inra, J.; Scherr, D.S.; Vaughan, E.D.; Nanus, D.M.; Kattan, M.W.; Gerald, W.L.; Vande Woude, G.F. High expression of the Met receptor in prostate cancer metastasis to bone. Urology 2002, 60, 1113–1117. [Google Scholar] [CrossRef]
  30. Grano, M.; Galimi, F.; Zambonin, G.; Colucci, S.; Cottone, E.; Zallone, A.Z.; Comoglio, P.M. Hepatocyte growth factor is a coupling factor for osteoclasts and osteoblasts in vitro. Proc. Natl. Acad. Sci. USA 1996, 93, 7644–7648. [Google Scholar] [CrossRef]
  31. Watanabe, K.; Hirata, M.; Tominari, T.; Matsumoto, C.; Fujita, H.; Yonekura, K.; Murphy, G.; Nagase, H.; Miyaura, C.; Inada, M. The MET/Vascular Endothelial Growth Factor Receptor (VEGFR)-targeted Tyrosine Kinase Inhibitor Also Attenuates FMS-dependent Osteoclast Differentiation and Bone Destruction Induced by Prostate Cancer. J. Biol. Chem. 2016, 291, 20891–20899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Dai, J.; Zhang, H.; Karatsinides, A.; Keller, J.M.; Kozloff, K.M.; Aftab, D.T.; Schimmoller, F.; Keller, E.T. Cabozantinib inhibits prostate cancer growth and prevents tumor-induced bone lesions. Clin. Cancer Res. 2014, 20, 617–630. [Google Scholar] [CrossRef] [PubMed]
  33. Smith, D.C.; Smith, M.R.; Sweeney, C.; Elfiky, A.A.; Logothetis, C.; Corn, P.G.; Vogelzang, N.J.; Small, E.J.; Harzstark, A.L.; Gordon, M.S.; et al. Cabozantinib in patients with advanced prostate cancer: Results of a phase II randomized discontinuation trial. J. Clin. Oncol. 2013, 31, 412–419. [Google Scholar] [CrossRef] [PubMed]
  34. Basch, E.; Autio, K.A.; Smith, M.R.; Bennett, A.V.; Weitzman, A.L.; Scheffold, C.; Sweeney, C.; Rathkopf, D.E.; Smith, D.C.; George, D.J.; et al. Effects of cabozantinib on pain and narcotic use in patients with castration-resistant prostate cancer: Results from a phase 2 nonrandomized expansion cohort. Eur. Urol. 2015, 67, 310–318. [Google Scholar] [CrossRef] [PubMed]
  35. Tsai, S.-Y.; Huang, Y.-L.; Yang, W.-H.; Tang, C.-H. Hepatocyte growth factor-induced BMP-2 expression is mediated by c-Met receptor, FAK, JNK, Runx2, and p300 pathways in human osteoblasts. Int. Immunopharmacol. 2012, 13, 156–162. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, H.-T.; Tsou, H.-K.; Chang, C.-H.; Tang, C.-H. Hepatocyte Growth Factor Increases Osteopontin Expression in Human Osteoblasts through PI3K, Akt, c-Src, and AP-1 Signaling Pathway. PLoS ONE 2012, 7, e38378. [Google Scholar] [CrossRef] [PubMed]
  37. Lee, R.J.; Smith, M.R. Cabozantinib and prostate cancer: Inhibiting seed and disrupting soil? Clin. Cancer Res. 2014, 20, 525–527. [Google Scholar] [CrossRef]
  38. Tolaney, S.M.; Nechushtan, H.; Ron, I.-G.; Schöffski, P.; Awada, A.; Yasenchak, C.A.; Laird, A.D.; O’Keeffe, B.; Shapiro, G.I.; Winer, E.P. Cabozantinib for metastatic breast carcinoma: Results of a phase II placebo-controlled randomized discontinuation study. Breast Cancer Res. Treat. 2016, 160, 305–312. [Google Scholar] [CrossRef]
  39. Fujita, H.; Miyadera, K.; Kato, M.; Fujioka, Y.; Ochiiwa, H.; Huang, J.; Ito, K.; Aoyagi, Y.; Takenaka, T.; Suzuki, T.; et al. The novel VEGF receptor/MET-targeted kinase inhibitor TAS-115 has marked in vivo antitumor properties and a favorable tolerability profile. Mol. Cancer Ther. 2013, 12, 2685–2696. [Google Scholar] [CrossRef]
  40. Fioramonti, M.; Santini, D.; Iuliani, M.; Ribelli, G.; Manca, P.; Papapietro, N.; Spiezia, F.; Vincenzi, B.; Denaro, V.; Russo, A.; et al. Cabozantinib targets bone microenvironment modulating human osteoclast and osteoblast functions. Oncotarget 2017, 8, 20113–20121. [Google Scholar] [CrossRef] [Green Version]
  41. Patnaik, A.; Swanson, K.D.; Csizmadia, E.; Solanki, A.; Landon-Brace, N.; Gehring, M.P.; Helenius, K.; Olson, B.M.; Pyzer, A.R.; Wang, L.C.; et al. Cabozantinib Eradicates Advanced Murine Prostate Cancer by Activating Antitumor Innate Immunity. Cancer Discov. 2017, 7, 750–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Haider, M.-T.; Hunter, K.D.; Robinson, S.P.; Graham, T.J.; Corey, E.; Dear, T.N.; Hughes, R.; Brown, N.J.; Holen, I. Rapid modification of the bone microenvironment following short-term treatment with Cabozantinib in vivo. Bone 2015, 81, 581–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Smith, M.; de Bono, J.; Sternberg, C.; Le Moulec, S.; Oudard, S.; De Giorgi, U.; Krainer, M.; Bergman, A.; Hoelzer, W.; De Wit, R.; et al. Phase III Study of Cabozantinib in Previously Treated Metastatic Castration-Resistant Prostate Cancer: COMET-1. J. Clin. Oncol. 2016, 34, 3005–3013. [Google Scholar] [CrossRef] [PubMed]
  44. Tolaney, S.M.; Tan, S.; Guo, H.; Barry, W.; Van Allen, E.; Wagle, N.; Brock, J.; Larrabee, K.; Paweletz, C.; Ivanova, E.; et al. Phase II study of tivantinib (ARQ 197) in patients with metastatic triple-negative breast cancer. Invest. New Drugs 2015, 33, 1108–1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Monk, P.; Liu, G.; Stadler, W.M.; Geyer, S.; Huang, Y.; Wright, J.; Villalona-Calero, M.; Wade, J.; Szmulewitz, R.; Gupta, S.; et al. Phase II randomized, double-blind, placebo-controlled study of tivantinib in men with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). Invest. New Drugs 2018, 36, 919–926. [Google Scholar] [CrossRef]
  46. Previdi, S.; Abbadessa, G.; Dalò, F.; France, D.S.; Broggini, M. Breast cancer-derived bone metastasis can be effectively reduced through specific c-MET inhibitor tivantinib (ARQ 197) and shRNA c-MET knockdown. Mol. Cancer Ther. 2012, 11, 214–223. [Google Scholar] [CrossRef]
  47. Previdi, S.; Scolari, F.; Chilà, R.; Ricci, F.; Abbadessa, G.; Broggini, M. Combination of the c-Met inhibitor tivantinib and zoledronic acid prevents tumor bone engraftment and inhibits progression of established bone metastases in a breast xenograft model. PLoS ONE 2013, 8, e79101. [Google Scholar] [CrossRef]
  48. Kim, K.-H.; Kim, H. Progress of antibody-based inhibitors of the HGF–cMET axis in cancer therapy. Exp. Mol. Med. 2017, 49, e307. [Google Scholar] [CrossRef]
  49. D’Amico, L.; Belisario, D.; Migliardi, G.; Grange, C.; Bussolati, B.; D’Amelio, P.; Perera, T.; Dalmasso, E.; Dalle Carbonare, L.; Godio, L.; et al. C-met inhibition blocks bone metastasis development induced by renal cancer stem cells. Oncotarget 2016, 7, 45525–45537. [Google Scholar] [CrossRef] [Green Version]
  50. Tchou, J.; Zhao, Y.; Levine, B.L.; Zhang, P.J.; Davis, M.M.; Melenhorst, J.J.; Kulikovskaya, I.; Brennan, A.L.; Liu, X.; Lacey, S.F.; et al. Safety and Efficacy of Intratumoral Injections of Chimeric Antigen Receptor (CAR) T Cells in Metastatic Breast Cancer. Cancer Immunol. Res. 2017, 5, 1152–1161. [Google Scholar] [CrossRef] [Green Version]
  51. Noriega-Guerra, H.; Freitas, V. Extracellular Matrix Influencing HGF/c-MET Signaling Pathway: Impact on Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3300. [Google Scholar] [CrossRef] [PubMed]
  52. Maroni, P.; Matteucci, E.; Bendinelli, P.; Desiderio, M.A. Functions and Epigenetic Regulation of Wwox in Bone Metastasis from Breast Carcinoma: Comparison with Primary Tumors. Int. J. Mol. Sci. 2017, 18, 75. [Google Scholar] [CrossRef] [PubMed]
  53. Bendinelli, P.; Maroni, P.; Matteucci, E.; Desiderio, M.A. HGF and TGFβ1 differently influenced Wwox regulatory function on Twist program for mesenchymal-epithelial transition in bone metastatic versus parental breast carcinoma cells. Mol. Cancer 2015, 14, 112. [Google Scholar] [CrossRef] [PubMed]
  54. Bendinelli, P.; Maroni, P.; Matteucci, E.; Desiderio, M.A. Cell and Signal Components of the Microenvironment of Bone Metastasis Are Affected by Hypoxia. Int. J. Mol. Sci. 2016, 17, 706. [Google Scholar] [CrossRef] [PubMed]
  55. Papaccio, F.; Della Corte, C.M.; Viscardi, G.; Di Liello, R.; Esposito, G.; Sparano, F.; Ciardiello, F.; Morgillo, F. HGF/MET and the Immune System: Relevance for Cancer Immunotherapy. Int. J. Mol. Sci. 2018, 19, 3595. [Google Scholar] [CrossRef]
  56. Matsumoto, K.; Nakamura, T. Hepatocyte growth factor and the Met system as a mediator of tumor-stromal interactions. Int. J. Cancer 2006, 119, 477–483. [Google Scholar] [CrossRef] [PubMed]
  57. Masuya, D.; Huang, C.; Liu, D.; Nakashima, T.; Kameyama, K.; Haba, R.; Ueno, M.; Yokomise, H. The tumour-stromal interaction between intratumoral c-Met and stromal hepatocyte growth factor associated with tumour growth and prognosis in non-small-cell lung cancer patients. Br. J. Cancer 2004, 90, 1555–1562. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Whang, Y.M.; Jung, S.P.; Kim, M.-K.; Chang, I.H.; Park, S.I. Targeting the Hepatocyte Growth Factor and c-Met Signaling Axis in Bone Metastases. Int. J. Mol. Sci. 2019, 20, 384. https://doi.org/10.3390/ijms20020384

AMA Style

Whang YM, Jung SP, Kim M-K, Chang IH, Park SI. Targeting the Hepatocyte Growth Factor and c-Met Signaling Axis in Bone Metastases. International Journal of Molecular Sciences. 2019; 20(2):384. https://doi.org/10.3390/ijms20020384

Chicago/Turabian Style

Whang, Young Mi, Seung Pil Jung, Meyoung-Kon Kim, In Ho Chang, and Serk In Park. 2019. "Targeting the Hepatocyte Growth Factor and c-Met Signaling Axis in Bone Metastases" International Journal of Molecular Sciences 20, no. 2: 384. https://doi.org/10.3390/ijms20020384

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop