ISSN 1866-8836
Клеточная терапия и трансплантация

Hepatocyte growth factor (HGF) in the pathogenesis of multiple myeloma

Magne Børset1,2, Therese Standal1, Anders Waage1,3 and Anders Sundan1

1Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; 2Department of Immunology and Transfusion medicine, St. Olavs University Hospital, Trondheim, Norway; 3 Department of Hematology, St. Olavs University Hospital, Trondheim, Norway.

Corresponding author:
Magne Børset, Norwegian University of Science and Technology, Faculty of Medicine, Department of Cancer Research and Molecular Medicine, Medical Technical Research Center, N-7489 Trondheim, Norway

Telephone: + 47 72573038,
Fax: + 47 73598801,
E-mail: magne.borset@ntnu.no

doi 10.3205/ctt2008-05-23-001-en
Submitted 04 February 2008
Accepted 20 March 2008
Published 23 May 2008

Summary

HGF is emerging as a cytokine with an important role in the pathophysiology of multiple myeloma. Originally identified and described as a growth factor for hepatocytes, HGF was later found to have mitogenic, motogenic, or morphogenic effects on several cell types through its interaction with the tyrosine kinase receptor c-Met. This cytokine–receptor pair is implicated in the development and promotion of several types of cancer. The expression of both HGF and c-Met by myeloma cells is one of the traits distinguishing these cells from healthy plasma cells, and seems to be an early step in tumor development. HGF and c-Met have an effect on proliferation, migration, and adhesion of myeloma cells; and research suggests that myeloma cell-produced HGF is an important factor in angiogenesis and bone destruction seen in the majority of patients with multiple myeloma.

Keywords

Hepatocyte growth factor, rank, rankl, opg, carcinogenesis, bone resorption, bone formation, overview, multiple myeloma, blood malignancies


Multiple myeloma (MM)

MM is malignant growth of plasma cells, which are terminally differentiated B lymphocytes 17. The disease is characterized by production of monoclonal immunoglobulin, by anemia, and by destruction of bone. The malignant myeloma cells are usually located in the bone marrow (BM). 

Despite some advances in treatment in recent years, MM still is a persistently fatal disease with a median patient survival time of three to four years from the time of diagnosis. In addition to the dismal prognosis, patients also experience substantial morbidity during the course of the disease. MM therefore continues to be a serious health problem. Standard front-line therapy for patients with MM includes the chemotherapeutic agents melphalan and prednisone, drugs that have been used for this purpose for more than 40 years. Recently, immunomodulatory drugs with putative effect against formation of new blood vessels (e.g., thalidomide), as well as botezomib, a member of an entirely new class of drugs: so-called protesome inhibitors, have shown effects in subsets of patients with MM18. However, there is an urgent need for better therapy that is targeted at the weak points of MM.

Our understanding of the molecular etiology of MM has increased enormously over the past ten years5;19. Approximately 50% of patients have translocations affecting the immunoglobulin heavy chain locus on chromosome 14. The translocation partner varies, but close to the breakpoint in the partner chromosome, there is a (putative) oncogene that is placed under transcriptional control of an enhancer normally controlling the immunoglobuline heavy chain gene. The ch14 translocations are believed to be early events in the development of MM, and are also present with roughly the same frequency in the premalignant condition “monoclonal gammopathy of undetermined significance”(MGUS) as in overt MM. (There is only a one percent chance per year of progressing from MGUS to MM, so the majority of MGUS cases never develop into MM.) The other 50% of MM cases do not have ch14 translocations, and the early and crucial genetic aberrations in this group of patients are still unknown, but overall, these cases tend to have hyperdiploidy with trisomy of a several recurrent chromosomes. A common theme in all patients with MM is the expression of various isoforms of the cell cycle regulatory protein cyclin-D5.

Myeloma cells usually do not grow for longer periods in vitro, despite addition of rich growth medium and growth factors. It is generally believed that the cells are dependent on currently unknown factors in the microenvironment of the bone marrow for growth and for protection against apoptosis. At the same time the malignant cells exert a profound influence on the same microenvironment. In overt myeloma there is increased bone marrow angiogenesis40 and – in most cases – perturbed bone homeostasis2;3. It has been known for more than three decades that myeloma cells stimulate osteoclasts, the bone-resorbing cells28. A simultaneous reduction in bone formation, leading to an unbalanced bone metabolism with ensuing erosion of bone substance4 was also observed many years ago. Factors produced by myeloma cells and either secreted or presented on the cell surface, are believed to be responsible for the perturbed microenvironment, and many candidate factors have been proposed as being responsible for increased bone resorption, with variable experimental documentation.

Role of HGF in the pathogenesis of MM: a mediator of autocrine loops

In 1996 we showed for the first time that myeloma cells express the receptor for HGF, c-Met, and at the same time often produce the ligand HGF7. This simultaneous expression of a cytokine and its receptor in the same cell was suggestive of an autocrine stimulatory loop, and we were able to demonstrate that c-Met was indeed activated in the myeloma cell line JJN-3 in an autocrine fashion8. Autocrine HGF-driven growth loops have also been demonstrated in other MM cell lines25. Later we showed that high levels of HGF in the serum of a patient with MM at the time of diagnosis was an adverse prognostic sign, a finding that has been confirmed by others26;32. Recently, we and others have demonstrated that HGF stimulates growth and survival of myeloma cells, and that HGF uses the myeloma marker protein syndecan-1 (CD138) as a co-receptor13;31. It has also been shown that myeloma cells express HGF activator, an enzyme converting pre-HGF into the active form of the growth factor39. Pre-HGF can also be converted to HGF by urokinase plasminogen activator (uPA)29. This enzyme is also produced by MM cells20. HGF could also be involved in the migration of myeloma cells to the bone marrow41. HGF is important in promoting adherence of MM cells to fibronectin, a matrix protein in the bone marrow environment23. Such adhesion is beneficial to the MM cells in the sense that it increases cell proliferation. HGF is also a potent angiogenic factor, and there is a positive correlation between HGF levels in serum and bone marrow angiogenesis in patients with MM, suggesting HGF's role in the excessive angiogenesis seen in these patients1. In an abstract presented to the 2006 ASH meeting, D. Hose and colleagues presented data showing that among 89 proangiogic genes only HGF was significantly overexpressed in MM cells compared to normal bone marrow plasma cells24.

HGF expression is characteristic of malignant plasma cells and distinguishes MM from other closely related diseases

The first comprehensive gene array study of MM by Zhan et al., comparing gene expression in MM cells with that in normal plasma cells, showed that HGF was the only secreted growth factor on the list of the 70 genes that were the most up-regulated of more than 5000 examined genes44. Interestingly, HGF was not only expressed in overt MM, but also in BM plasma cells from a majority of patients with MGUS, indicating that initiation of HGF expression is an early event in the transformation of healthy cells into malignant MM cells (Erming Tian and John D. Shaughnessy Jr., personal communication). A recently published gene array study by Chng et al. showed that expression of HGF together with IL-6, a potent growth factor for MM cells, was characteristic for a subgroup of patients with hyperdiploid MM10. In another study, using comparative genomic hybridization (aCGH) analysis, recurrent gene copy number alterations were identified, and 47 areas of recurrent gene amplifications were found9. HGF was located within a small recurrent amplification that included a total of four genes, and HGF was found to be the only one of those genes with an oncogene-like expression pattern. This amplification was present in more than 40% of patients, and the finding indicates that gene aberrations leading to HGF expression are part of the oncogenic development leading to MM.

In order to identify the gene expression that is important for the specific clinical manifestations of MM, a logical approach would be to compare the gene expression profile of MM cells with that of cells from closely related diseases. This was done with gene array analysis of purified malignant cells from patients with chronic lymphocytic leukemia, Waldenstom macroglobulinemia and MM.11 Again, HGF stood out as one of the genes that characterized MM as opposed to the two other diseases. Interestingly, the HGF receptor c-Met was also on this list of MM-related genes.

Disturbance of key regulators of bone homeostasis in patients with MM

Skeletal tissue in healthy people is constantly undergoing a balanced remodeling process, where osteoclasts resorb bone and are followed by osteoblasts forming bone. Central to this regulation are specific factors that act directly on osteoclasts and are downstream mediators for many of the systemic bone-active factors. It has become clear that osteoblasts play a crucial role in the direct regulation of osteoclast activity. Osteoblasts express the cell surface protein RANKL, which is necessary for osteoclast differentiation43. Furthermore, the osteoblasts express a soluble decoy receptor for RANKL, osteoprotegerin (OPG)35. The balance between the two osteoblast products, RANKL and OPG, seems to be critical for the regulation of bone homeostasis.

We have found that multiple myeloma patients have reduced levels of soluble OPG in bone marrow plasma compared to healthy controls33 and others have found that there is also an increased expression of RANKL in the MM bone marrow30. OPG contains a heparin binding site and may bind to heparan sulfates on cells in the bone marrow. We have shown that MM cells bind OPG, presumably via the heparan sulfate-containing protein Syndecan-16. Moreover, we found that this binding led to internalization and degradation of OPG by the myeloma cells37, thereby providing one possible explanation for the reduced OPG levels in the bone marrow of multiple myeloma patients.

Inhibition of bone formation is important for skeletal destruction in patients with MM

In patients with MM, the balanced process of bone remodeling is upset, leading to degradation of bone and to skeletal morbidity. Intensive research has been conducted to try to unravel the mechanism causing this bone disease. For several decades, the research focus was on factors leading to untimely activation of osteoclasts, although it had long beenrealized that perturbationin osteoblast function might be equally important4. In a mouse model of MM with severe bone disease, osteoblasts were virtually non-existent22. Lately, the focus has moved from osteoclasts to osteoblasts, and several papers have contributed to our understanding of osteoblast inhibition34;38. Searching for a correlation between gene expression in purified primary MM cells and level of bone disease in the patients, Tian and colleagues identified DKK1, an inhibitor of Wnt signaling, as a prime suspect for the destruction of bone in MM patients38. They found that DKK1 inhibited the differentiation of osteoblast precursors into mature bone-forming osteoblasts. Later studies seem to confirm that DKK1 is linked to excessive bone disease and that it works by inhibiting the formation of osteoblasts16;42. Interestingly, genes that encode known osteoclast-regulating factors, such as RANKL, RANK, OPG, MIP1, PTHrP, and IL-1, did not show a significant relationto the presence of bone disease27. This is not a proof against these factors as important for bone destruction in MM, but argues against MM cells as the source of them. Similarly, osteoclast-activating factors were conspicuously absent from the list of gene expression that was characteristic for MM cells; as opposed to cells from chronic lymphocytic leukemia and Waldenstom macroglobulinemia11. Bone disease is not a common clinical trait of the latter two diseases, and one would expect the genes that are responsible for this hallmark of MM to be present on the list of genes that define the specific cancer phenotype of MM. Like HGF and c-Met, DKK1 was high up on this list, further supporting the role of DKK1 in promoting the bone disease that is linked to MM.

HGF inhibits bone morphogenetic protein-induced differentiation of mesenchymal stem cells into bone-forming osteoblasts

Since HGF is one of the genes distinguishing malignant plasma cells from healthy plasma cells, and also defines malignant plasma cells as opposed to other closely related malignant cells, it was logical to see whether HGF played a role in bone homeostasis. It had been previously published that HGF induces bone resorption by osteoclasts, but only in the presence of osteoblasts15. This indirect effect on osteoclasts could be partly through HGF-induced production of IL-11, an osteoclast-stimulating cytokine21. Bone morphogenetic proteins (BMP) promote differentiation of osteoblast precursors from mesenchymal stem cells (MSCs) and further maturation into bone forming osteoblasts. Experiments by our group showed that HGF inhibited BMP-induced expression of alkaline phosphatase in human MSCs and in the murine myoid cell line C2C1236. HGF also prevented BMP-induced mineralization by human MSCs. Furthermore, the expression of the osteoblast-specific transcription factors Runx2 and Osterix was reduced by HGF treatment. Interestingly, HGF promoted proliferation of human MSCs, whereas BMP halted the proliferation. Again, HGF was a key regulator, keeping the cells in a proliferative, undifferentiating state despite the presence of BMP. BMP-induced nuclear translocation of receptor-activated Smads was inhibited by HGF, providing a possible explanation as to how HGF inhibits BMP signaling. These findings support a role of HGF similar to that of DKK1. By preventing MSCs from becoming mature osteoblasts, the osteoblast precursors are arrested in an intermediate stage of differentiation, where they express RANKL, an osteoclast-stimulating protein. Therefore, instead of contributing to bone repair, these cells promote the bone-destruction process: bone homeostasis is no longer balanced. Was there any clinical evidence that HGF really played this role as an osteoblast inhibitor in patients? Yes, the in vitro data were supported by the observation of a significant negative correlation between HGF and a marker of osteoblast activity, bone-specific alkaline phosphatase, in sera from 34 patients with myeloma36.

Targeting HGF hepatocyte growth factor and its receptor c-Met in multiple myeloma

Since expression of HGF seems to be an early oncogenic event in the development of MM, and due to HGF’s many effects on disease manifestations, it might be an attractive target in treatment of MM. A host of new pharmacological inhibitors of c-Met are in the pipelines of the pharmaceutical industry. Possible HGF inhibitors include small molecular drugs and antibodies, as well as naturally occurring splice variants of HGF with antagonistic or partially antagonistic effects on c-Met. NK4 belongs to the latter group: a variant of HGF that lacks part of the full molecule12. This molecule was shown to block growth of MM cell line cells in a mouse model, an effect that was believed to be a combination of direct anti-proliferative effect of the drug on MM cells, as well as an indirect, anti-angiogenic effect on formation of new blood vessels14. PHA-665752, a novel pharmacological inhibitor of c-Met from Pfizer belongs to the group of small molecular inhibitors. This molecule prevented HGF-driven autocrine loops in an MM cell line, as well as in freshly isolated MM cells from myeloma patients25. No in vivo data on effects on MM cells of small-molecule inhibitors of c-Met have been published yet, and no clinical trial with c-Met inhibitors has been started so far. However, the data from such studies are sure to be met with great anticipation.

References

1.   Andersen NF, Standal T, Nielsen JL et al. Syndecan-1 and angiogenic cytokines in multiple myeloma: correlation with bone marrow angiogenesis and survival. Br.J.Haematol. 2005;128:210-217.

2.   Bataille R, Chappard D, Basle M. Excessive bone resorption in human plasmacytomas: direct induction by tumour cells in vivo. Br.J.Haematol. 1995;90:721-724.

3.   Bataille R, Chappard D, Klein B. Mechanisms of bone lesions in multiple myeloma. Hematol.Oncol.Clin.North Am. 1992;6:285-295.

4.   Bataille R, Chappard D, Marcelli C et al. Mechanisms of bone destruction in multiple myeloma: the importance of an unbalanced process in determining the severity of lytic bone disease. J.Clin.Oncol. 1989;7:1909-1914.

5.   Bergsagel PL, Kuehl WM. Molecular pathogenesis and a consequent classification of multiple myeloma. J.Clin.Oncol. 2005;23:6333-6338.

6.   Borset M, Hjertner O, Yaccoby S, Epstein J, Sanderson RD. Syndecan-1 is targeted to the uropods of polarized myeloma cells where it promotes adhesion and sequesters heparin-binding proteins. Blood 2000;96:2528-2536.

7.   Borset M, Hjorth-Hansen H, Seidel C, Sundan A, Waage A. Hepatocyte growth factor and its receptor c-met in multiple myeloma. Blood 1996;88:3998-4004.

8.   Borset M, Lien E, Espevik T et al. Concomitant expression of hepatocyte growth factor/scatter factor and the receptor c-MET in human myeloma cell lines. J.Biol.Chem. 1996;271:24655-24661.

9.   Carrasco DR, Tonon G, Huang Y et al. High-resolution genomic profiles define distinct clinico-pathogenetic subgroups of multiple myeloma patients. Cancer Cell 2006;9:313-325.

10.   Chng WJ, Kumar S, Vanwier S et al. Molecular dissection of hyperdiploid multiple myeloma by gene expression profiling. Cancer Res. 2007;67:2982-2989.

11.   Chng WJ, Schop RF, Price-Troska T et al. Gene-expression profiling of Waldenstrom macroglobulinemia reveals a phenotype more similar to chronic lymphocytic leukemia than multiple myeloma. Blood 2006;108:2755-2763.

12.   Date K, Matsumoto K, Shimura H, Tanaka M, Nakamura T. HGF/NK4 is a specific antagonist for pleiotrophic actions of hepatocyte growth factor. FEBS Lett. 1997;420:1-6.

13.   Derksen PW, Keehnen RM, Evers LM et al. Cell surface proteoglycan syndecan-1 mediates hepatocyte growth factor binding and promotes Met signaling in multiple myeloma. Blood 2002;99:1405-1410.

14.   Du W, Hattori Y, Yamada T et al. NK4, an antagonist of hepatocyte growth factor (HGF), inhibits growth of multiple myeloma cells in vivo; molecular targeting of angiogenic growth factor. Blood 2006

15.   Grano M, Galimi F, Zambonin G et al. Hepatocyte growth factor is a coupling factor for osteoclasts and osteoblasts in vitro. Proc Natl Acad Sci USA 1996;93:7644-7648.

16.   Haaber J, Abildgaard N, Knudsen LM et al. Myeloma cell expression of 10 candidate genes for osteolytic bone disease. Only overexpression of DKK1 correlates with clinical bone involvement at diagnosis. Br.J.Haematol. 2007

17.   Hallek, M., Bergsagel, P. L., and Anderson, K. C. Multiple myeloma: Increasing evidence for a multistep transformation process. Blood 91, 3-21. 1998.

18.   Hayden PJ, Mitsiades CS, Anderson KC, Richardson PG. Novel therapies in myeloma. Curr.Opin.Hematol. 2007;14:609-615.

19.   Hideshima T, Bergsagel PL, Kuehl WM, Anderson KC. Advances in biology of multiple myeloma: clinical applications. Blood 2004;104:607-618.

20.   Hjertner O, Qvigstad G, Hjorth-Hansen H et al. Expression of urokinase plasminogen activator and the urokinase plasminogen activator receptor in myeloma cells. Br.J.Haematol. 2000;109:815-822.

21.   Hjertner O, Torgersen ML, Seidel C et al. Hepatocyte growth factor (HGF) induces interleukin-11 secretion from osteoblasts: a possible role for HGF in myeloma-associated osteolytic bone disease. Blood 1999;94:3883-3888.

22.   Hjorth-Hansen H, Seifert MF, Borset M et al. Marked osteoblastopenia and reduced bone formation in a model of multiple myeloma bone disease in severe combined immunodeficiency mice. J.Bone Miner.Res. 1999;14:256-263.

23.   Holt RU, Baykov V, Ro TB et al. Human myeloma cells adhere to fibronectin in response to hepatocyte growth factor. Haematologica 2005;90:479-488.

24.   Hose D, Devos J, Heib C et al. Angiogenesis in multiple myeloma (MM): Angiogenic switch or reflexion of plasma cell number? A gene expression based survey in primary myeloma cells and the bone marrow microenvironment. Blood 2006;108:972A-973A.

25.   Hov H, Holt RU, Ro TB et al. 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. 2004;10:6686-6694.

26.   Iwasaki T, Hamano T, Ogata A et al. Clinical significance of vascular endothelial growth factor and hepatocyte growth factor in multiple myeloma. Br.J.Haematol. 2002;116:796-802.

27.   Lu CM. DKK1 in multiple myeloma. N.Engl.J.Med. 2004;350:1464-1466.

28.   Mundy GR, Raisz LG, Cooper RA, Schechter GP, Salmon SE. Evidence for the Secretion of an Osteoclast Stimulating Factor in Myeloma. N.Engl.J.Med. 1974;291:1041-1046.

29.   Naldini L, Vigna E, Bardelli A et al. Biological activation of pro-HGF (hepatocyte growth factor) by urokinase is controlled by a stoichiometric reaction. J.Biol.Chem. 1995;270:603-611.

30.   Pearse RN, Sordillo EM, Yaccoby S et al. Multiple myeloma disrupts the TRANCE/ osteoprotegerin cytokine axis to trigger bone destruction and promote tumor progression. Proc.Natl.Acad.Sci.U.S.A 2001;98:11581-11586.

31.   Seidel C, Borset M, Hjertner O et al. High levels of soluble syndecan-1 in myeloma-derived bone marrow: modulation of hepatocyte growth factor activity. Blood 2000;96:3139-3146.

32.   Seidel C, Borset M, Turesson I et al. Elevated serum concentrations of hepatocyte growth factor in patients with multiple myeloma. The Nordic Myeloma Study Group. Blood 1998;91:806-812.

33.   Seidel C, Hjertner O, Abildgaard N et al. Serum osteoprotegerin levels are reduced in patients with multiple myeloma with lytic bone disease. Blood 2001;98:2269-2271.

34.   Silvestris F, Cafforio P, Tucci M, Grinello D, Dammacco F. Upregulation of osteoblast apoptosis by malignant plasma cells: a role in myeloma bone disease. Br.J.Haematol. 2003;122:39-52.

35.   Simonet WS, Lacey DL, Dunstan CR et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997;89:309-319.

36.   Standal T, Abildgaard N, Fagerli UM et al. HGF inhibits BMP-induced osteoblastogenesis: possible implications for the bone disease of multiple myeloma. Blood 2006

37.   Standal T, Seidel C, Hjertner O et al. Osteoprotegerin is bound, internalized, and degraded by multiple myeloma cells. Blood 2002;100:3002-3007.

38.   Tian E, Zhan F, Walker R et al. The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N.Engl.J.Med. 2003;349:2483-2494.

39.   Tjin EP, Derksen PW, Kataoka H, Spaargaren M, Pals ST. Multiple myeloma cells catalyze hepatocyte growth factor (HGF) activation by secreting the serine protease HGF-activator. Blood 2004;104:2172-2175.

40.   Vacca A, Ribatti D, Roncali L et al. Bone-Marrow Angiogenesis and Progression in Multiple Myeloma. Br.J.Haematol. 1994;87:503-508.

41.   Vande B, I, Asosingh K, Allegaert V et al. Bone marrow endothelial cells increase the invasiveness of human multiple myeloma cells through upregulation of MMP-9: evidence for a role of hepatocyte growth factor. Leukemia 2004;18:976-982.

42.   Yaccoby S, Ling W, Zhan F et al. Antibody-based inhibition of DKK1 suppresses tumor-induced bone resorption and multiple myeloma growth in vivo. Blood 2007;109:2106-2111.

43.   Yasuda H, Shima N, Nakagawa N et al. Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc.Natl.Acad.Sci.U.S.A 1998;95:3597-3602.

44.   Zhan F, Hardin J, Kordsmeier B et al. Global gene expression profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and normal bone marrow plasma cells. Blood 2002;99:1745-1757.

Volume 1, Number 1
06/18/2008

Download PDF version

doi 10.3205/ctt2008-05-23-001-en
Submitted 04 February 2008
Accepted 20 March 2008
Published 23 May 2008

Back to the list