GET THE APP

The MET Receptor Tyrosine Kinase as a Therapeutic Target in Multi
Journal of Leukemia

Journal of Leukemia
Open Access

ISSN: 2329-6917

+44 1300 500008

Editorial - (2014) Volume 2, Issue 2

The MET Receptor Tyrosine Kinase as a Therapeutic Target in Multiple Myeloma

Christine M Stellrecht*
Department of Experimental Therapeutics, UT MD Anderson Cancer Center, Houston, Texas 77030, USA
*Corresponding Author: Christine M Stellrecht, Department of Experimental Therapeutics, UT MD Anderson Cancer Center, Houston, Texas 77030, USA, Tel: 281-648-8112 Email:

Abstract

Multiple Myeloma (MM) is a progressive and debilitating B-cell disorder typified by the accumulation and dissemination of malignant plasma cells in the bone marrow which subsequently induce osteolytic lesions.Despite recent advancement of new therapies for MM, patients ultimately develop drug resistanceand succumb to the disease. This highlights the need to increase our understanding of the disease and to identify new myeloma targets for drug development.

Multiple Myeloma (MM) is a progressive and debilitating B-cell disorder typified by the accumulation and dissemination of malignant plasma cells in the bone marrow which subsequently induce osteolytic lesions [1]. Despite recent advancement of new therapies for MM, patients ultimately develop drug resistance and succumb to the disease [2]. This highlights the need to increase our understanding of the disease and to identify new myeloma targets for drug development.

The growth and progression of myeloma is multifaceted and signaling through several cytokine/receptor pathways have been shown to be vital for the pathobiology of this disease [3-6]. Evidence for the MET receptor signaling pathway being an important contributor to the pathogenesis of this disease has been mounting. Both MET and its ligand, hepatocyte growth factor (HGF), are expressed in an autocrine loop in most myeloma cell lines and primary patient samples, as discerned at both the mRNA and protein level [7-10]. This is compounded by the high expression of the proteoglycan, syndecan 1 (CD138), on plasma cells as it binds HGF and boosts HGF-mediated signaling [11,12]. Additionally, malignant plasma cells convert HGF into its active form by secreting HGF-activator (HGFA), a serine protease specific for HGF activation [13].

The effects of HGF/MET signaling on myeloma cells are diverse. It has been shown to promote myeloma-cell growth [3,12,14], and survival [3,15]. Moreover, HGF/MET uniquely signals a morphogenic invasivegrowth program [16-18] that is associated with the dissemination of myeloma cells in the bone marrow, leading to disease progression [14,19]. HGF/MET signaling boosts myeloma cell adherence to the bone marrow matrix through enhanced binding to fibronectin and other components of the microenvironment [20]. Additionally, it is involved in the up-regulation of matrix metalloproteinases and urokinase plasminogen activator [21], further enhancing migration and invasion [22]. Characteristically, MET signaling is able to induce angiogenesis by stimulating the production of VEGF and inhibiting thrombospondin 1 expression [23], broadening its support of tumor survival.

In addition to promoting growth and progression of myeloma, HGF/MET signaling may play a role in the focalization of myelomarelated bone disease. HGF levels are increased in patients with extensive bone lesions, and correlates with increases in cytokines, interleukin-6 (IL-6) and tumor necrosis factor-alpha, involved in osteoclast stimulation [24]. HGF stimulation induces IL-11 secretion from osteoblasts [25], and HGF inhibits bone morphogenetic proteininduced osteoblastogenesis [26].

Clinically, several studies have correlated HGF/MET pathway with myeloma disease parameters. HGF is elevated at diagnosis in the serum and marrows of patients with multiple myeloma [11,27-29] and activated HGFA has been found to be present at high levels in the serum and bone marrow of myeloma patients [30]. Moreover, serum HGF levels tend to be further elevated in patients with advanced stage or aggressive disease [24,28,31-34] and have been associated with a poorer prognosis [27,29,31,35,36]. Similarly, a higher level of MET mRNA in MM patients’ plasma cells is also associated with a poorer prognosis [37]. Conversely, patients with low HGF levels have been shown to be more likely to achieve high-quality responses [38,39], and the serum levels of a soluble, inhibiting form of MET negatively correlates with disease stage [40], and declining HGF levels are seen in patients who are responding to anti-myeloma therapies [39,41,42].

Targeting the HGF/MET signaling pathway is a rational approach to multiple myeloma therapy. Genetically or pharmacologically depleting MET is fatal to myeloma cells [19,43,44], further underscoring the role of MET/HGF axis in myeloma and potential as a therapeutic target. Moreover, a number of translational studies using varying strategies to suppress the HGF/MET axis further bolster the concept of MET being a therapeutic target for MM. PHA-665752, a MET inhibitor, blocked HGF-mediated signaling and inhibited MM cell proliferation, adhesion, and migration [14]. Similarly, an anti- MET Nanobody was shown to not only inhibit MET signaling, growth, adhesion, and migration, but also showed evidence of abolished inhibitory effect of HGF on bone morphogenetic protein-2-induced osteoblastogenesis [45]. NK4, an internal fragment of HGF that acts as a competitive inhibitor, not only inhibited MET signaling and growth in MM cells in culture, but also significantly inhibited the in vivo growth of myelomatous tumors in a murine model [46] as does the MET inhibitor, SU11274 [47]. The MET inhibitor amuvatinib was shown to be tumoricidal to primary myeloma cells and cell lines Additionally, these studies suggest that myeloma survival may be dependent on HGF levels [9].

Preclinical studies of the MET inhibitor, tivantinib was also performed in myeloma cell lines and patient samples. Tivantinib is unique in that it is not an ATP-mimetic, but instead is an inhibitor which binds to a part of the MET receptor that is close to the ATPbinding site [48]. This atypical interaction is believed to provide this small-molecule inhibitor with a very unique and high degree of kinase selectivity. In addition to being a kinase inhibitor, recent evidence indicates tivantinib also has activity independent of MET as well [49,50]. The preclinical analysis showed that at clinically achievable concentrations, tivantinib induced apoptosis by >50% in 12 myeloma cell lines [51]. The cytotoxicity in these cells was associated with a loss of MET activity and an inhibition of downstream signaling through the MAPK and PI3K pathways. Tivantinib was equally effective in inducing apoptosis in myeloma cell lines resistant to standard chemotherapy. Furthermore, tivantinib also induced apoptosis in primary myeloma plasma cells and demonstrated efficacy in a myeloma mouse xenograft model. Based on the preclinical studies, a small Phase II clinical trial of tivantinib (NCT01447914), was performed in unselected relapsed/ refractory myeloma patients [52,53]. In the clinical trial, 16 patients were enrolled and 11 were evaluated for response. Bone disease was not evaluated in this study. In this study, tivantinib was well tolerated. Even though none of these patients had high HGF levels (data not shown), the agent was able to achieve stable disease in 4 of 11 (36%) patients with previously progressing myeloma [53]. Overall, these data demonstrate the promise of targeting HGF/MET signaling axis for the treatment of multiple myeloma and highlights increased need for biomarker testing for selection of candidates for future trials.

Recently, two clinical phase I/II trials (NCT0186629 and NCT01582295) to assess the safety and efficacy of Cabozantinib (XL184) in relapsed/refractory myeloma patients have opened. Cabozantinib is a pan-kinase inhibitor which shows potent activity against MET, vascular endothelial growth factor receptor 2 (VEGFR2/KDR), VEGFR3/FLT4 VEGFR1/FLT1, and RET with IC50 values of 1.8, 0.035, 12.2, 6.0 and 9.8 nM, respectively [54]. Additional targets of XL184 include FLT3, TIE2, AXL, TRKB, and KIT with IC50 values of 14.4, 14.3, 7.7, and 4.6 nM, respectively. Thus cabozantinib is able to target both VEGF signaling as well as MET, both important for myeloma progression. The primary assessment in both trials will be to determine safety in a dose escalation study. Patients in NCT01582295 will also be selected for bone disease and an assessment of various markers of bone turnover will also be included in the study, including serum bone specific alkaline phosphatase, osteocalcin, sclerostin, procollagen type 1N propeptide, and urine N-telopeptide. Additionally, changes on whole body FDGPET/ CT scans as well as pain will be monitored.

In conclusion, preclinical studies show strong indication for targeting MET in myeloma for both reducing tumor burden in METdependent tumors and for alleviating bone disease. The clinical trial with tivantinib shows promise of MET being a viable therapeutic target. Future clinical trials need to be planned where myeloma patient populations are selected which have high serum HGF levels or possibly high plasma-cell MET levels for targeting this pathway. Additionally, monitoring of bone disease may also prove advantageous for evaluating the treatment benefit. The use of these criteria will allow for a more applicable means to effectively assess MET inhibitors as a therapeutic strategy in myeloma.

References

  1. Hallek M, Bergsagel PL, Anderson KC (1998) Multiple myeloma: increasing evidence for a multistep transformation process. Blood 91: 3-21.
  2. Ocio EM, Mitsiades CS, Orlowski RZ, Anderson KC (2014) Future agents and treatment directions in multiple myeloma. Expert Rev Hematol 7: 127-141.
  3. Derksen PW, de Gorter DJ, Meijer HP, Bende RJ, van Dijk M, et al. (2003) The hepatocyte growth factor/Met pathway controls proliferation and apoptosis in multiple myeloma. Leukemia 17: 764-774.
  4. Yasui H, Hideshima T, Richardson PG, Anderson KC (2006) Novel therapeutic strategies targeting growth factor signalling cascades in multiple myeloma. Br J Haematol 132: 385-397.
  5. Hideshima T, Podar K, Chauhan D, Anderson KC (2005) Cytokines and signal transduction. Best Pract Res ClinHaematol 18: 509-524.
  6. Song L, Li Y, Sun YX, Yu M, Shen BF (2002) IL-6 inhibits apoptosis of human myeloma cell line XG-7 through activation of JAK/STAT pathway and up-regulation of Mcl-1. Ai Zheng 21: 113-116.
  7. Börset M, Hjorth-Hansen H, Seidel C, Sundan A, Waage A (1996) Hepatocyte growth factor and its receptor c-met in multiple myeloma. Blood 88: 3998-4004.
  8. Borset M, Lien E, Espevik T, Helseth E, Waage A, et al. (1996) Concomitant expression of hepatocyte growth factor/scatter factor and the receptor c-MET in human myeloma cell lines. J BiolChem 271: 24655-24661.
  9. Phillip CJ, Zaman S, Shentu S, Balakrishnan K, Zhang J, et al. (2013) Targeting MET kinase with the small-molecule inhibitor amuvatinib induces cytotoxicity in primary myeloma cells and cell lines. Journal of hematology& oncology 6: 93.
  10. Seidel C, Borset M, Hjorth-Hansen H, Sundan A, Waage A (1998) Role of hepatocyte growth factor and its receptor c-met in multiple myeloma. Med Oncol 15: 145-53.
  11. Seidel C, Børset M, Hjertner O, Cao D, Abildgaard N, et al. (2000) High levels of soluble syndecan-1 in myeloma-derived bone marrow: modulation of hepatocyte growth factor activity. Blood 96: 3139-3146.
  12. Derksen PW, Keehnen RM, Evers LM, van Oers MH, Spaargaren M, et al. (2002) Cell surface proteoglycan syndecan-1 mediates hepatocyte growth factor binding and promotes Metsignaling in multiple myeloma. Blood 99: 1405-1410.
  13. Tjin EP, Derksen PW, Kataoka H, Spaargaren M, Pals ST (2004) Multiple myeloma cells catalyze hepatocyte growth factor (HGF) activation by secreting the serine protease HGF-activator. Blood 104: 2172-2175.
  14. Hov H, Holt RU, Rø TB, Fagerli UM, Hjorth-Hansen H, et al. (2004) A selective c-met inhibitor blocks an autocrine hepatocyte growth factor growth loop in ANBL-6 cells and prevents migration and adhesion of myeloma cells. Clin Cancer Res 10: 6686-6694.
  15. Derenne S, Monia B, Dean NM, Taylor JK, Rapp MJ, et al. (2002) Antisense strategy shows that Mcl-1 rather than Bcl-2 or Bcl-x(L) is an essential survival protein of human myeloma cells. Blood 100: 194-199.
  16. Birchmeier C, Birchmeier W, Gherardi E, VandeWoude GF (2003) Met, metastasis, motility and more. Nat Rev Mol Cell Biol 4: 915-925.
  17. Comoglio PM, Boccaccio C (2001) Scatter factors and invasive growth. Semin Cancer Biol 11: 153-165.
  18. Stellrecht CM, Gandhi V (2009) MET receptor tyrosine kinase as a therapeutic anticancer target. Cancer Lett 280: 1-14.
  19. Stellrecht CM, Phillip CJ, Cervantes-Gomez F, Gandhi V (2007) Multiple myeloma cell killing by depletion of the MET receptor tyrosine kinase. Cancer Res 67: 9913-9920.
  20. Holt RU, Baykov V, Rø TB, Brabrand S, Waage A, et al. (2005) Human myeloma cells adhere to fibronectin in response to hepatocyte growth factor. Haematologica 90: 479-488.
  21. Hecht M, Heider U, Kaiser M, von Metzler I, Sterz J, et al. (2007) Osteoblasts promote migration and invasion of myeloma cells through upregulation of matrix metalloproteinases, urokinase plasminogen activator, hepatocyte growth factor and activation of p38 MAPK. Br J Haematol 138: 446-458.
  22. Holt RU, Fagerli UM, Baykov V, Rø TB, Hov H, et al. (2008) Hepatocyte growth factor promotes migration of human myeloma cells. Haematologica 93: 619-622.
  23. Zhang YW, Su Y, Volpert OV, VandeWoude GF (2003) Hepatocyte growth factor/scatter factor mediates angiogenesis through positive VEGF and negative thrombospondin 1 regulation. ProcNatlAcadSci U S A 100: 12718-12723.
  24. Alexandrakis MG, Passam FH, Sfiridaki A, Kandidaki E, Roussou P, et al. (2003) Elevated serum concentration of hepatocyte growth factor in patients with multiple myeloma: correlation with markers of disease activity. Am J Hematol 72: 229-233.
  25. Hjertner O, Torgersen ML, Seidel C, Hjorth-Hansen H, Waage A, et al. (1999) Hepatocyte growth factor (HGF) induces interleukin-11 secretion from osteoblasts: a possible role for HGF in myeloma-associated osteolytic bone disease. Blood 94: 3883-3888.
  26. Standal T, Abildgaard N, Fagerli UM, Stordal B, Hjertner O, et al. (2007) HGF inhibits BMP-induced osteoblastogenesis: possible implications for the bone disease of multiple myeloma. Blood 109: 3024-3030.
  27. Seidel C, Børset M, Turesson I, Abildgaard N, Sundan A, et al. (1998) Elevated serum concentrations of hepatocyte growth factor in patients with multiple myeloma. The Nordic Myeloma Study Group. Blood 91: 806-812.
  28. Iwasaki T, Hamano T, Ogata A, Hashimoto N, Kitano M, et al. (2002) Clinical significance of vascular endothelial growth factor and hepatocyte growth factor in multiple myeloma. Br J Haematol 116: 796-802.
  29. Seidel C, Lenhoff S, Brabrand S, Anderson G, Standal T, et al. (2002) Hepatocyte growth factor in myeloma patients treated with high-dose chemotherapy. Br J Haematol 119: 672-676.
  30. Wader KF, Fagerli UM, Holt RU, Stordal B, Børset M, et al. (2008) Elevated serum concentrations of activated hepatocyte growth factor activator in patients with multiple myeloma. Eur J Haematol 81: 380-383.
  31. Kara IO, Sahin B, Gunesacar R, Unsal C (2006) Clinical significance of hepatocyte growth factor, platelet-derived growth factor-AB, and transforming growth factor-alpha in bone marrow and peripheral blood of patients with multiple myeloma. Advances in therapy 23: 635-45.
  32. Alexandrakis MG, Passam FJ, Ganotakis E, Dafnis E, Dambaki C, et al. (2004) Bone marrow microvascular density and angiogenic growth factors in multiple myeloma. ClinChem Lab Med 42: 1122-1126.
  33. Urba ska-Rys H, Wierzbowska A, Robak T (2003) Circulating angiogenic cytokines in multiple myeloma and related disorders. Eur Cytokine Netw 14: 40-51.
  34. Di Raimondo F, Azzaro MP, Palumbo G, Bagnato S, Giustolisi G, et al. (2000) Angiogenic factors in multiple myeloma: higher levels in bone marrow than in peripheral blood. Haematologica 85: 800-805.
  35. Turesson I, Abildgaard N, Ahlgren T, Dahl I, Holmberg E, et al. (1999) Prognostic evaluation in multiple myeloma: an analysis of the impact of new prognostic factors. Br J Haematol 106: 1005-1012.
  36. Iwasaki T, Sano H (2003) Predicting treatment responses and disease progression in myeloma using serum vascular endothelial growth factor and hepatocyte growth factor levels. Leuk Lymphoma 44: 1275-1279.
  37. Rocci A, Gambella M, Aschero S, Baldi I, Trusolino L, et al. (2014) MET dysregulation is a hallmark of aggressive disease in multiple myeloma patients. Br J Haematol 164: 841-850.
  38. Pour L, Svachova H, Adam Z, Mikulkova Z, Buresova L, et al. (2010) Pretreatment hepatocyte growth factor and thrombospondin-1 levels predict response to high-dose chemotherapy for multiple myeloma. Neoplasma 57: 29-34.
  39. Ludek P, Hana S, Zdenek A, Martina A, Dana K, et al. (2010) Treatment response to bortezomib in multiple myeloma correlates with plasma hepatocyte growth factor concentration and bone marrow thrombospondin concentration. Eur J Haematol 84: 332-336.
  40. Wader KF, Fagerli UM, Holt RU, Børset M, Sundan A, et al. (2011) Soluble c-Met in serum of patients with multiple myeloma: correlation with clinical parameters. Eur J Haematol 87: 394-399.
  41. Sezer O, Jakob C, Eucker J, Niemoller K, Gatz F, et al. (2001) Serum levels of the angiogenic cytokines basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) in multiple myeloma. Eur J Haematol 66: 83-88.
  42. Pour L, Svachova H, Adam Z, Almasi M, Buresova L, et al. (2010) Levels of angiogenic factors in patients with multiple myeloma correlate with treatment response. Ann Hematol 89: 385-389.
  43. Chen LS, Stellrecht CM, Gandhi V (2008) RNA-directed agent, cordycepin, induces cell death in multiple myeloma cells. Br J Haematol 140: 682-391.
  44. Phillip CJ, Stellrecht CM, Nimmanapalli R, Gandhi V (2009) Targeting MET transcription as a therapeutic strategy in multiple myeloma. Cancer ChemotherPharmacol 63: 587-597.
  45. Schmidt Slørdahl T, Denayer T, Helen Moen S, Standal T, Børset M, et al. (2013) Anti-c-MET Nanobody - a new potential drug in multiple myeloma treatment. Eur J Haematol 91: 399-410.
  46. Du W1, Hattori Y, Yamada T, Matsumoto K, Nakamura T, et al. (2007) NK4, an antagonist of hepatocyte growth factor (HGF), inhibits growth of multiple myeloma cells: molecular targeting of angiogenic growth factor. Blood 109: 3042-3049.
  47. Moschetta M, Basile A, Ferrucci A, Frassanito MA, Rao L, et al. (2013) Novel targeting of phospho-cMET overcomes drug resistance and induces antitumor activity in multiple myeloma. Clin Cancer Res 19: 4371-4382.
  48. Munshi N1, Jeay S, Li Y, Chen CR, France DS, et al. (2010) ARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activity. Mol Cancer Ther 9: 1544-1553.
  49. Basilico C, Pennacchietti S, Vigna E, Chiriaco C, Arena S, et al. (2013) Tivantinib (ARQ197) displays cytotoxic activity that is independent of its ability to bind MET. Clin Cancer Res 19: 2381-2392.
  50. Katayama R, Aoyama A, Yamori T, Qi J, Oh-hara T, et al. (2013) Cytotoxic activity of tivantinib (ARQ 197) is not due solely to c-MET inhibition. Cancer Res 73: 3087-3096.
  51. Zaman S, Stellrecht CM, Shentu S, Orlowski R, Gandhi V (2012) Targeting the pro-survival protein c-MET with ARQ 197 inhibits growth of multiple myeloma cells. Proc Am Assoc Cancer Res 53: 844.
  52. Dorkhom SJ, Zaman S, Thomas SK, Alexanian R, Shah JJ, et al. (2012) Phase II study of the c-Met inhibitor ARQ 197 (tivantinib) in patients with relapsed multiple myeloma. Blood 120: 2976.
  53. Orlowski RZ, Zaman S, Thomas SK, Alexanian R, Shah JJ, et al. (2013) Phase II Study Of The c-MET Inhibitor ARQ 197 (Tivantinib) In Patients With Relapsed Or Relapsed/Refractory Multiple Myeloma (RRMM). Blood 122: 1953.
  54. Pharmacology/toxicology NDA review and evaluation; Cabozantinib (S)-malate; Application number: 203,756. Department of Health And Human Services Public Health Service Food and Drug Administration Center for Drug Evaluation and Research.
Citation: Stellrecht CM (2014) The MET Receptor Tyrosine Kinase in Multiple Myeloma. J Leuk (Los Angel) 2:e107.

Copyright: © 2014 Stellrecht CM. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author andsource are credited.
Top