Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2013

Open Access 01.12.2013 | Research

Targeting MET kinase with the small-molecule inhibitor amuvatinib induces cytotoxicity in primary myeloma cells and cell lines

verfasst von: Cornel Joseph Phillip, Shadia Zaman, Shujun Shentu, Kumudha Balakrishnan, Jiexin Zhang, Veera Baladandayuthapani, Pietro Taverna, Sanjeev Redkar, Michael Wang, Christine Marie Stellrecht, Varsha Gandhi

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2013

Abstract

Background

MET is a receptor tyrosine kinase that is activated by the ligand HGF and this pathway promotes cell survival, migration, and motility. In accordance with its oncogenic role, MET is constitutively active, mutated, or over-expressed in many cancers. Corollary to its impact, inhibition of MET kinase activity causes reduction of the downstream signaling and demise of cells. In myeloma, a B-cell plasma malignancy, MET is neither mutated nor over-expressed, however, HGF is increased in plasma or serum obtained from myeloma patients and this was associated with poor prognosis. The small-molecule, amuvatinib, inhibits MET receptor tyrosine kinase. Based on this background, we hypothesized that targeting the HGF/MET signaling pathway is a rational approach to myeloma therapy and that myeloma cells would be sensitive to amuvatinib.

Methods

Expression of MET and HGF mRNAs in normal versus malignant plasma cells was compared during disease progression. Cell death and growth as well as MET signaling pathway were assessed in amuvatinib treated primary myeloma cells and cell lines.

Results

There was a progressive increase in the transcript levels of HGF (but not MET) from normal plasma cells to refractory malignant plasma cells. Amuvatinib readily inhibited MET phosphorylation in primary CD138+ cells from myeloma patients and in concordance, increased cell death. A 48-hr amuvatinib treatment in high HGF-expressing myeloma cell line, U266, resulted in growth inhibition. Levels of cytotoxicity were time-dependent; at 24, 48, and 72 h, amuvatinib (25 μM) resulted in 28%, 40%, and 55% cell death. Consistent with these data, there was an amuvatinib-mediated decrease in MET phosphorylation in the cell line. Amuvatinib at concentrations of 5, 10, or 25 μM readily inhibited HGF-dependent MET, AKT, ERK and GSK-3-beta phosphorylation. MET-mediated effects were not observed in myeloma cell line that has low MET and/or HGF expression.

Conclusions

These data suggest that at the cellular level MET/HGF pathway inclines with myeloma disease progression. Amuvatinib, a small molecule MET kinase inhibitor, is effective in inducing growth inhibition and cell death in myeloma cell lines as well as primary malignant plasma cells. These cytostatic and cytotoxic effects were associated with an impact on MET/HGF pathway.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1756-8722-6-92) contains supplementary material, which is available to authorized users.
Cornel Joseph Phillip, Shadia Zaman, Christine Marie Stellrecht and Varsha Gandhi contributed equally to this work.

Competing interest

SR and PT are employed by Astex Pharmaceuticals, Inc., Dublin, CA. For the remaining authors, none was declared.

Authors’ contributions

CJP, SZ, KB, and CMS designed and performed experiments; CJP, SZ, JZ, VB, and CMS analyzed data; SS performed experiments; PT and SR provided amuvatinib; MW provide sorted bone marrow samples; CJP and CMS wrote the manuscript; CMS and VG directed study; VG provided laboratory resources; All authors critically read and approved manuscript.
Abkürzungen
DMSO
Dimethyl sulfoxide
ERK
Extracellular signal-regulated kinases
FBS
Fetal bovine serum
GSK3β
Glycogen synthase kinase 3 β
HGF
Hepatocyte growth factor
IL-6
Interleukin-6
MAPK
Mitogen-activated protein kinase
MGUS
Monoclonal gammopathy of undetermined significance
MM
Multiple myeloma
MM-N
Newly-diagnosed multiple myeloma
MM-R
Relapsed/refractory disease
PARP
Poly ADP ribose polymerase
PDGFR
Platelet derived growth factor receptor
PI
Propidium iodide
SMM
Smoldering multiple myeloma.

Introduction

Multiple myeloma (MM) is an indolent B-cell disease that develops in the bone marrow and is associated with osteolytic lesions in the advanced stages[1]. Despite progress in prolonging myeloma patient survival, current therapies are not curative; thus, it is imperative that new treatments be developed for this debilitating disease[2, 3].
Survival and proliferation of myeloma cells are dependent on the presence of a permissive microenvironment, which includes bone marrow stroma and soluble cytokines[49] such as IL-6 and HGF[8, 10]. HGF is the ligand for MET receptor tyrosine kinase. When HGF binds to and activates MET, MET is autophosphorylated on Tyr1230, Tyr1234 and Tyr1235 located in the activation loop[1114]. In addition, MET has a multisubstrate docking site that is activated at Tyr1349 and Tyr1356. The phosphorylation of this region results in the induction of MET signaling through the activation of several downstream target pathways, including the mitogen-activated protein kinase (MAPK) and AKT signaling pathways[11]. HGF/MET-induced MAPK signaling has been shown to be essential for proliferation, migration and invasion[7, 11, 15, 16] while the induction of AKT signaling promotes tumor cell survival[17].
HGF/MET signaling is increasingly recognized as an important contributor to the pathogenesis of myeloma. Expression of both HGF and MET has been demonstrated in most myeloma cell lines and primary patient samples[18, 19]. Studies correlating HGF levels with MM clinical parameters such as diagnosis[2023] disease stage, aggressiveness[22, 24, 25], prognosis[22, 23, 26], and response[2629]. Besides its effects on the malignant myeloma cells, HGF is involved in the pathogenesis of myeloma-related bone disease. HGF levels are increased in patients with extensive bone lesions, and correlates with expression of osteoclast stimulating cytokines[24]. IL-11 secretion from osteoblasts is induced by HGF[30], and HGF inhibits bone morphogenetic protein-induced osteoblastogenesis[31].
Taken together, these clinical findings strongly support our hypothesis that targeting the HGF/MET signaling pathway is a rational approach to myeloma therapy. In line with this postulate, our laboratory studies demonstrated that genetically knocking down MET in myeloma cell lines using short hairpin RNA and ribozyme approaches resulted in growth inhibition and demise of the myeloma cells[32, 33]. Consistent with these observations, a decline in MET transcript and protein levels induced by treatment with any of the transcription inhibitors flavopiridol, cordycepin, or 8-chloro-adenosine, promoted myeloma cell death[3234]. Collectively, these data demonstrate MET’s pivotal role in myeloma cell biology and underscore the importance of MET targeting as a therapeutic strategy in MM[35].
While these genetic and pharmacologic strategies suggest utility of MET/HGF inhibition as therapeutic targets, these interventions are not pragmatic for clinical use. Amuvatinib (previously known as MP470, Astex Pharmaceuticals, Inc.) is a synthetic carbothioamide that inhibits MET, cKIT and platelet derived growth factor receptor (PDGFR). This small-molecule inhibitor competes with ATP for binding at the catalytic site. In solid cancers, amuvatinib has been shown to be effective in inhibiting MET at low micromolar concentrations (IC50 ~5 μM)[36]. Amuvatinib is a well-tolerated, orally bioavailable drug currently in phase II clinical trials[37, 38]. Availability of a clinical candidate, its inhibitory potential for MET kinase, and the role of MET in myeloma cell survival provided compelling rationales for testing the effects of amuvatinib on myeloma cells.
In the present study, we compared mRNA levels of MET and HGF in normal and primary myeloma plasma cells. We investigated amuvatinib’s actions and cytotoxic effects in primary plasma cells obtained from patients with myeloma. To elucidate in more detail the mechanism of action of amuvatinib in myeloma cells, we evaluated its effect on MET activity and downstream signaling in the myeloma cell line U266, which over-expresses HGF. Our data demonstrate that MET receptor tyrosine kinase may be targeted in myeloma and support the investigation of small-molecule inhibitors such as amuvatinib as possible therapeutic agents against this disease.

Results

Expression levels of MET and HGF mRNA in bone marrow plasma cells of healthy donors and patients

Previously studies have correlated plasma HGF levels with MM clinical parameters such as diagnosis[2023] disease stage, aggressiveness[22, 24, 25], prognosis[22, 23, 26], and response[2629]. While expression of both HGF and MET transcripts has been shown to be present in myeloma cells[18, 19] and HGF mRNA has also been demonstrated to be expressed in bone marrow stromal cells[39] the levels of HGF and MET mRNA in patient plasma cells have not been well evaluated nor correlated with disease status.
To determine the levels of MET and HGF gene expression in malignant and normal plasma cells, we analyzed data from the Mayo Clinic Patient Dataset available in the public domain[40, 41]. The 162 samples evaluated represented 15 healthy individuals (normal), 22 patients with monoclonal gammopathy of undetermined significance (MGUS), 24 with smoldering MM (SMM), 74 with newly diagnosed MM (MM-N), and 27 with relapsed/refractory MM (MM-R). Among these five groups, there was no significant difference (P = 0.708) in the expression of MET in the CD138+ cells (Figure 1A). In contrast, there was a significant trend (P = 2.5 × 10-06) for increases in HGF mRNA levels in CD138+ plasma cells, with progressive severity of disease from healthy donors to patients with relapsed or refractory MM (Figure 1B). Within each group, there was heterogeneity in HGF expression as evinced by the 75th percentile mark. It is interesting to note that even samples with lower HGF mRNA levels in the plasma cells, typically had higher levels than the samples from healthy individuals; the 25th percentile for the myeloma patient HGF levels was > the 75th percentile for healthy individuals.

Induction of apoptosis by amuvatinib in primary CD138+ and CD138– Cells

Gene array analysis along with numerous other studies by us[32, 33] and others[22, 23, 26], identified the HGF/MET axis as a therapeutic target in myeloma. To test this, we assessed the sensitivity of primary myeloma cells to the MET-kinase inhibitor, amuvatinib. CD138+ (myeloma plasma cells) and CD138– (non-malignant cells) cells were isolated from bone marrow samples obtained from eight myeloma patients (Table 1) and treated with 25 μM amuvatinib for 24 h. This dose was chosen based on findings that >95% of the compound is bound and sequestered by serum proteins (unpublished data) as well as a small preliminary screen in myeloma cell lines (data not shown). CD138+ cells from six out of eight patient samples showed a cell death induction in the amuvatinib treated cells compared to time-matched dimethyl sulfoxide (DMSO)-treated control cells as measured by annexin V/propidium iodide (PI) staining (Figure 2A). Cell death in these six samples increased by 20% to 67% compared to time-matched controls; patients 3 and 5 showed <10% increase in cell death.
Table 1
Myeloma patient characteristics
Pt #
Age (yrs)
Sex
Ethnicity
WBC a (103/μl)
Plasma cells (%)
BM b aspirate done
Outcome of treatment
1
62
Male
White
3.6
7
At diagnosis
NA c
2
53
Female
Black
2.0
74
Post bortezomib + doxorubicin + lenalidomide + dexamethasone
Progressive
3
53
Female
White
6.3
4
Post 3 cycles bortezomib + dexamethasone
Near complete remission
4
76
Female
White
4.4
2
Post cyclophosphamide + lenalidomide + dexamethasone
Progressive
5
58
Female
Hispanic
9.1
8
At diagnosis
NA
6
64
Female
White
12.5
49
At diagnosis
NA
7
71
Male
White
3.9
32
NA
NA
8
76
Male
White
4.9
36
post 1 yr auto stem cell transplant
Progressive
a white blood cells; b bone marrow; c not applicable.
An assessment of clinical characteristics of the myeloma patients did not reveal any correlations with amuvatinib-induced cell death, including prior treatment, patient age, or cellularity of the bone marrow (Table 1). We were able to measure the HGF levels in plasma samples from patients 2, 4, and 5 which were 11.9, 1.7, and 1.4 ng/mL, respectively. Although the number of total samples is small, there seems to be a relationship between levels of HGF and amuvatinib-induced apoptosis. Additional studies are needed to determine if there is any correlation between HGF level and sensitivity of CD138+ cells to MET inhibition.
In contrast to the sensitivity of CD138+ cells to amuvatinib, CD138– cells did not show any sizeable inductions (all, <10%) of death compared to time-matched controls (Figure 2B), suggesting that non-malignant bone marrow cells are not affected by amuvatinib. Overall, these results indicate that treatment of CD138+ MM cells with a MET inhibitor is detrimental to their survival.

Effect of Amuvatinib on MET signaling in CD138+ and CD138- cells

To determine whether death of CD138+ cells was associated with an effect on the target, total and phosphorylated-MET (p-MET) expression levels were determined by flow cytometry. There were sufficient numbers of cells from patients 6 and 8 to perform this assessment. In both samples, MET phosphorylation was reduced on Tyr 1234/1235 in the CD138+ cells by 40% and 50%, respectively, as compared to the time-matched controls (Figure 3A and B). In contrast, there was no detectable level of p-MET in CD138– cells from patient 8 as compared to isotype control (Figure 3C). The lack of p-MET in CD138– cells likely explains why this population of cells was not affected by amuvatinib treatment in any of the eight patient samples.

Effect of Amuvatinib on Growth Inhibition

Since the number of the primary CD138+ cells in eight patient samples were insufficient for performing a detailed investigation of HGF/MET signaling, we decided to further investigate the effects of amuvatinib in a myeloma cell line. Because our results in the patient samples suggested that higher levels of HGF may be associated with an increased sensitivity to MET inhibition, we used the U266 cell line which expresses high levels of HGF[19]. Compared to DMSO-treated control cells, amuvatinib-treated U266 cells showed a dose- and time-dependent decrease in growth (Figure 4A). The growth inhibition was ~40% at a dose of 5 μM after 48 and 72 h of incubation and 50% at a dose of ~7 μM at 72 h.

Effect of Amuvatinib on cell cycle arrest and DNA synthesis

We also tested whether the observed amuvatinib-induced growth inhibition was associated with an alteration in the cell cycle. At low micromolar doses (3 and 5 μM), U266 cells were arrested at G1 after 48 and 72 h (Figure 4B and C). In the DMSO-treated controls, approximately 65% of cells were in G1 phase at 72 h, while cells treated 3 μM amuvatinib significantly increased to 75% in G1 phase at the same time point (p < 0.05). Incubation with a higher level of amuvatinib (25 μM) resulted in a lower percentage of cells in G1 phase (Figure 4C), with a concomitant increase in the subG1 fraction (data not shown).
To determine whether the observed cell cycle changes were associated with an effect on DNA synthesis, we measured incorporation of thymidine in total DNA. Compared to DMSO-treated (control cells), amuvatinib-treated cells had decreased thymidine incorporation at doses of both 5 and 25 μM (Figure 4D), which was significantly higher for cells treated with 5 μM amuvatinib for 24 and 72 h, (P = 0.008 and P = 0.048, respectively). At 24 h, the inhibition of thymidine incorporation was greater than 50% with 5 μM amuvatinib. Additionally, as expected, the decrease in the cells’ S-phase DNA replicative capacity was discernible 24 h before there was a measurable change in the cell cycle profile as the doubling time for U266 cells is ~36 hours.

Induction of Apoptosis by Amuvatinib

Similar to what was seen in the primary CD138+ cells, U266 cells treated with 25 μM amuvatinib exhibited significantly greater cell death (28%, 40%, and 54%) than DMSO-treated controls (6%, 7%, and 7%) for 24, 48, and 72 h, respectively (Figure 5A and B) (P = 0.045, 0.015, and 0.018 respectively). This apoptotic induction was blocked by a pan-caspase inhibitor, ZVAD, suggesting a role for caspases in amuvatinib-mediated cell death (data not shown). Consistent with the annexin V/PI staining results was our finding that amuvatinib induced poly ADP ribose polymerase (PARP) cleavage in these cells in a dose-dependent manner. Under full serum conditions (10% fetal bovine serum (FBS)), an induction of PARP cleavage was seen after 24 h with doses as low as 3 μM amuvatinib with the highest level of cleaved PARP product was seen at 25 μM (Figure 5C).
Because 95% of amuvatinib binds serum proteins and is unavailable to the cells, we also examined the efficacy of amuvatinib under low serum (0.1% FBS) conditions. When cells were treated with 5 μM amuvatinib for 16 hours in the presence of low serum, but in the presence of endogenous autocrine HGF, we found maximum induction of PARP cleavage (Figure 5D). To further confirm the cytotoxic effects of amuvatinib in myeloma cells is associated with inhibiting the HGF/MET signaling axis, we compared the efficacy of amuvatinib to induce apoptosis in U266 cells versus RPMI-8226/S, a myeloma cell line which express 75% lower levels of HGF and 95% lower levels of MET (Additional file1: Figure S1). As expected, there was a significant amuvatinib dose-dependent apoptosis-induction in the U266 cells after 48 h treatment (Figure 5E) (25 μM versus 0, 2.5, 5, and 10; P = 0.022, 0.018, 0.013, and 0.029, respectively). In contrast there was only a minor apoptosis induction in the RPMI-8226/S cells which was not statistically significant (P = 0.053, 0.300, 0.427, and 0.503, respectively). These results suggest that the apoptosis induction is due to targeting MET kinase which U266 cells are addicted to while RPMI-8226/S cells are not.

Tumoricidal Effects of Amuvatinib in Myeloma Cells Grown in a Protective Stromal Environment

Bone marrow stroma provides a protective environment for MM cells[42]; thus, it is important to assess the efficacy of therapeutic agents in the context of a stromal environment. To assess this, we treated U266 cells co-cultured with and without stromal cells with amuvatinib for 48 h and measured viability by using flow cytometry analysis of annexin V/PI staining. Under these conditions, the U266 do not attach to the stromal cells, but are protected by them through both cell to cell contact and by various soluble factors produced by the stromal cells[43]. Amuvatinib induced ~50% cell killing during this time period and co-culture with the stromal cells provided no protection from this effect (Figure 5F). In contrast, these stromal cells were able to protect U266 cells from bortezomib treatment as they reduced the amount of bortezomib-induced apoptosis from ~75% to ~40% (Additional file1: Figure S2A). To determine whether amuvatinib had an effect on the survival of stromal cells, stromal cells cultured alone were treated with amuvatinib, harvested by trypsinization, and similarly assessed for viability. Interestingly, amuvatinib had a very minimal effect on the survival of this population of cells (Figure 5F), though they express MET (Additional file1: Figure S2B). These results indicate that the tumoricidal action of amuvatinib was largely restricted to the U266 myeloma cells, whereas the stromal cells, which are not addicted to MET, are not affected by this inhibitor. Furthermore, the stromal cells were not able to protect the U266 cells from amuvatinib’s tumoricidal activity.
Since amuvatinib also inhibits PDGFR and KIT, we validated MET kinase inhibition as the primary cause of cell death by using imatinib as a negative control. In addition to ABL, imatinib is known to also inhibit PDGFR and KIT but not MET[44]. In contrast to amuvatinib, 25 μM imatinib did not induce significant cell death (Figure 5G; P = 0.07) indicating that amuvatinib-mediated cell death is not due to its effects on PDGFR and KIT.

Effect of Amuvatinib on MET Protein

To further investigate the effect of amuvatinib on MET signaling, we first measured MET receptor tyrosine kinase activity in U266 cells by flow cytometry. Similar to the results seen with the CD138+ cells from the patient samples, a reduction in MET phosphorylation was detected in cells treated for 24 hr with 25 μM amuvatinib when cells are grown in normal growth conditions (10% FBS) (Figure 6A and B). This decrease of p-MET was associated with cell death as cell death was induced with 25 μM amuvatinib when cells are grown in normal growth conditions (10% FBS) (Figure 5B).
To assess the effects of amuvatinib on HGF-specific signaling, protein lysates from U266 cells serum starved in 0.1% FBS for 16 h with and without various concentrations of amuvatinib followed with 15 min HGF stimulation were examined by immunoblot analysis. The results showed that under serum starved conditions, treatment with 5 μM amuvatinib, decreased phosphorylation of the processed ~140 kDa MET β-chain at Tyr1349 by ~60% (Figure 6C and D). Because of the autocrine stimulation of MET by the endogenous HGF produced in these cells, MET was phosphorylated under serum-starved conditions even without the addition of exogenous HGF. Furthermore, an amuvatinib-dependent decrease of total MET levels of ~30% was also observed. A ~170 kDa phosphorylated MET band was detected at ~2 fold higher levels than the 140 kDa band in untreated U266 cells. A comparison with total MET shows both bands were present but the levels of the total 140 kDa band was ~4 times greater than the levels of the 170 kDa band. Although unprocessed pro-MET, containing both the α and β subunits, has been detected by SDS PAGE as a 170 kDa band, it has not been associated with kinase activity. Conversely, a splice variant of MET containing an additional 54 nt of exon 10 has been reported to be expressed at low levels[45]. This splice form produces a MET isoform that has kinase activity, though it cannot be processed into α and β subunits. In U266 cells, amuvatinib inhibited phosphorylation of a 170 kDa MET by ~70% (Figure 6C and D, B). Again, the decrease of HGF-specific phosphorylation of both isoforms of MET under low serum conditions is associated with cell death under low serum conditions (Figure 5D). The lower concentration of amuvatinib needed to decrease MET phosphorylation under 0.1% serum versus 10% serum conditions is in agreement with binding of the drug by serum proteins. Additionally, the concentration of amuvatinib required to decrease MET phosphorylation correlates with the concentration required to induce cell killing under either growth conditions. These results suggest the amuvatinib-induced cell death was associated with reduced MET activity.

Effect of Amuvatinib on downstream targets of MET

Previous studies have shown that inhibition of MET causes a reduction in the phosphorylation of both AKT and extracellular signal-regulated kinases (ERK)1/2 in the MAPK signaling pathway[11]. The regulation of AKT activity by MET plays a prominent role in promoting cell survival. Moreover, MET regulation of the ERK pathway is important for proliferation and both the ERK1/2 and AKT pathways are involved in MET-induced cell spreading and motility. To examine AKT activity, p-AKT (S473) levels were measured in U266 cells by flow cytometry. Similar to the results seen with p-MET, a reduction in AKT phosphorylation was detected in cells treated for 24 hr with 25 μM amuvatinib when cells were grown in normal growth conditions (10% FBS) (Figure 7A and B). An assessment of amuvatinib’s effects on HGF-specific signaling was also performed in the U266 cells cultured in 0.1% FBS for 16 h with and without various concentrations of amuvatinib followed with 15 min HGF. Immunoblot analysis again showed that lower concentrations amuvatinib is needed to decreased AKT phosphorylation at Ser473 (Figure 7C and D), even though in these cells the levels were low and difficult to detect. Interestingly, total AKT decreased by 60% with amuvatinib treatment. To better assess the effect of amuvatinib on the AKT pathway, we examined the phosphorylation of an AKT target, glycogen synthase kinase 3 β (GSK3β) on Ser9. Amuvatinib-treated cells showed, in addition to reduction of AKT, a 65% decrease in phosphorylation of GSK3β, with a 24% decrease in total GSK3β (Figure 7C and E).
A similar assessment of phospho-ERK1/2 levels under HGF specific signaling demonstrated that amuvatinib inhibited phosphorylation of both the 44-kDa and the 42-kDa ERK isoforms by 55% and 50%, respectively, while total ERK1/2 levels did not significantly change (Figure 7C and F). These results demonstrate that amuvatinib treatment inhibits both ERK1/2 and AKT signaling through the MET pathway.

Discussion

MET is a receptor tyrosine kinase that is activated by the ligand HGF and has been shown to be constitutively expressed, mutated, or over-expressed in many different cancer cell types. It serves as an important factor for cell survival, migration, and motility[7, 11, 15]. Corollary to that, inhibition of MET kinase activity causes reduction of the downstream signaling that is necessary for these cells to maintain their oncogenic properties[46]. Previous studies in our laboratory showed that while MET receptor tyrosine kinase acts as a survival factor for myeloma cells[32, 33], it is neither mutated nor, for the most part, over-expressed in MM. However, its ligand HGF is increased in plasma or serum obtained from myeloma patients and higher HGF level has been associated with poor prognosis[18, 20, 22, 26]. Furthermore, HGF not only promotes growth, migration, and survival of myeloma cells, it also potentiates IL-6 effects[46].
While levels of plasma HGF have been associated with myeloma, levels of HGF and MET mRNA in patient plasma cells have not been well evaluated nor correlated with disease status. Our analyses of mRNA array data[40, 41] demonstrated autocrine expression of HGF in CD138+ plasma cells from MM patients. This was consistent with previous report in 7 myeloma patient samples[18]. Our results further elucidated that the level of the HGF expression was directly associated with disease progression.
Together, these findings provide a rationale for targeting the HGF/MET signaling axis in myeloma. Targeting HGF directly may prove difficult, since therapeutic targeting of HGF would need to be effective at elevated levels to successfully compete and inhibit the high serum HGF concentrations in myeloma patients. Therefore, using a small-molecule MET suppressor such as amuvatinib may be a viable option to target the HGF/MET pathway. Additionally, several MET inhibitors are available for clinical testing[11].
Amuvatinib is an orally available drug that is currently in clinical trials for the treatment of solid tumors[37, 38, 47]. This compound was designed, developed, and selected via a computation-driven in silico process whereby drug scaffolds were screened, docked, and fitted against a homologous model of KIT. After additional screening in biochemical and cell-based assays, amuvatinib was selected as a tyrosine kinase inhibitor with activity against wild-type and mutant KIT, MET, RET, FLT3 and PDGFRα[48, 49]. Later, amuvatinib inhibition of MET activity was found to lead to reduction of RAD51 expression and to radiosensitization of tumor cells[50].
Since amuvatinib is a small-molecule inhibitor that suppresses MET activity, we tested this agent as a proof-of-concept to therapeutically target MET in myeloma. Our study demonstrated that amuvatinib was effective in inhibiting growth and DNA synthesis at low micromolar concentrations in cell lines grown under normal conditions (10% FBS). Moreover, amuvatinib treatment resulted in cell death in U266 myeloma cell line dependent on MET/HGF signaling, as measured by annexin V/PI staining and PARP cleavage. This cytotoxic effect remained even when these MET-addicted cells were grown on bone marrow stromal cells. In contrast, the drug did not induce apoptosis in another myeloma cell line (RPMI-8226/S) that is not dependent on the MET/HGF signaling axis due to lower levels of HGF (75% less) and MET (95% less).
Because amuvatinib also impairs KIT and PDGFR signaling, we tested impact of imatinib (an established KIT and PDGFR inhibitor) in myeloma cells. Imatinib induced no significant amount of cell death in U266 cells demonstrating that amuvatinib’s effect was due to MET inhibition. This statement was in line with the data regarding decreased phosphorylation of MET after amuvatinib treatment. Because >95% of the compound is bound and sequestered by serum proteins (Unpublished data), the dose required to achieve maximum inhibition of MET phosphorylation in serum starved conditions was lower than the dose to induce apoptosis in full serum conditions. Likewise, under serum starved conditions, the maximum induction of apoptosis was seen at the same dose which achieved maximum inhibition of MET phosphorylation. As expected, in imatinib treated cells, there was no reduction of p-MET (data not shown) as well as no significant reduction in survival. These correlation data suggest that amuvatinib mediated growth inhibition and cell death is due to its action on MET and not its action on KIT or PDGFR.
In conjunction with a decrease in MET phosphorylation, there was a decline in HGF-dependent ERK1/2 and AKT phosphorylation as well as the phosphorylation of the AKT targets GSK3β and caspase-9 (data not shown). Diminution of phosphorylated MET and associated decreases in ERK1/2 and AKT phosphorylation has been shown to be important in growth, migration and cell survival pathways for other cancer cell types[11].
Amuvatinib proved to be effective in inducing cell death not only in a MET dependent myeloma cell line but also in primary CD138+ malignant plasma cells obtained from patients with myeloma. In contrast, amuvatinib did not cause cell death in normal CD138– cells obtained from the same individuals (Figure 2). These data provide evidence of the selectivity of amuvatinib, suggesting that it may be used specifically for myeloma treatment without impairing other normal hematological cells in the bone marrow. In line with this selective cytotoxic effect on CD138 plasma cells, MET phosphorylation was reduced by amuvatinib treatment in primary plasma cells but not CD138– cells.
The effects of amuvatinib described here provide proof-of-concept that MET is important for the survival of myeloma cells and that reduction of its kinase activity may prove to be an effective targeted therapy. The 25 μM dose of amuvatinib needed to robustly induce apoptosis in cell lines and plasma cells under full serum conditions may not be achievable in vivo. Pharmacokinetic studies of amuvatinib during a phase I trial indicated that plasma levels reached between 1 and 2 μM[51]. Hence, newer generation and more potent MET tyrosine kinase inhibitors are needed[11]. ARQ 197 (tivantinib) is a small-molecule, non-ATP-competitive inhibitor which is highly specific for MET[52, 53]. This drug is well tolerated in clinical trials and has shown efficacy in solid tumors[5458]. Pharmacodynamic studies from a phase I trial indicated that at an oral dosing of 360 mg, twice daily, ARQ 197 reached steady-state plasma concentrations of 6–7 μM[55]. This correlated with decreases in total MET and phospho-FAK (Tyr861) and increases in TUNEL-positive cells in patients’ tumors.
Our results with amuvatinib provided the impetus to pursue testing of ARQ 197 in myeloma cells. Our preclinical studies indicated that treatment with ARQ 197 for 48 hours was cytotoxic to myeloma cell lines (≥ 60% increase in annexin V/PI-positive cells) at clinically achievable doses[59]. Moreover, these studies provided the foundation for a Cancer Therapy Evaluation Program, National Cancer Institute sponsored phase 2 clinical trial of ARQ 197 in myeloma patients, which is currently underway at MD Anderson Cancer Center[60].

Conclusions

Our finding provides proof-of-principle that MET is important for the survival of myeloma cells and using a MET inhibitor such as amuvatinib may prove to be an effective strategy for treatment of MM. Amuvatinib exhibited tumoricidal activity in myeloma cells which was associated with inhibition of MET signaling. Amuvatinib’s lack of effect on CD138– cells from the same patients further establishes the selectivity of this agent. The clinical success of other targeted therapeutics for cytoplasmic and receptor tyrosine kinases, further underscores a need for testing a small-molecule inhibitor that targets MET kinase activity for patients with myeloma.

Methods

Materials

Amuvatinib (MP470) was obtained from Astex Pharmaceuticals, Inc. (Dublin, CA) and was dissolved in DMSO (Sigma Aldrich, St. Louis, MO). Because of stability constraints, amuvatinib solution was prepared fresh for each experiment. Imatinib was purchased from Novartis (St. Louis, MO) and was dissolved in DMSO and stored in aliquots at -20°C. [3H] thymidine (60 Ci/mmol) was obtained from Moravek Biochemical Inc. (Brea, CA).

Cell culture and growth analysis

The myeloma cell lines U266[61] and RPMI-8226/S[62] were obtained from Dr. William Dalton at H. Lee Moffitt Cancer Center (Tampa, FL). NK-tert human bone marrow stromal cells were obtained from Dr. Jan Burger at UT MD Anderson Cancer Center[63]. The cell lines were maintained as described[32, 63] and routinely tested for Mycoplasma infection and authenticated by short tandem repeat analysis by UT MD Anderson Cancer Center’s Characterized Cell Line Core facility.
Myeloma cell-stromal co-cultures were performed using U266 cells and NK-tert cells at a ratio of 20 to 1. Stromal cells were plated at a concentration of ~2 × 102 cells/mm2 surface area 5 hours before adding U266 cells at a 20 fold higher concentration. The cells were co-cultured for 2 h prior to treatment with or without amuvatinib or bortezomib for 48 h. At the end of incubation, the U266 cells, which are free floating in these cultures, were carefully removed for analysis, leaving the adherent stromal layer undisturbed. Additionally, the stromal cells were also harvested by trypsinization and similarly assessed.
The effect of amuvatinib treatment on cell growth inhibition was measured in exponentially growing U266 cells. Cells were counted using a Coulter counter (Beckman Coulter, Fullerton, CA). DNA synthesis was measured using [3H]thymidine incorporation as described[64].

Gene expression array analyses

Expression data from 162 CD138+ bone marrow plasma cell samples from healthy individuals as well as patients with MGUS, SMM, MM-N, and MM-R, which were measured by using Affymetrix U133A microarrays, were downloaded from GEO (GSE6477)[40, 41]. Robust Multichip Average (RMA) algorithm was used for normalization/quantification of the data. The maximal values for the respective probe-sets of MET and HGF were used for gene expression profiling. The Kruskal-Wallis test was applied to assess whether expression of MET and HGF was associated with defined clinical groups, and results are presented as box-plots.

Isolation of CD138+ AND CD138– cells from primary bone marrow aspirates from MM patients

Primary samples were obtained from both male and female myeloma patients being treated at MD Anderson Cancer Center (Table 1). Patient samples were obtained using an MD Anderson Cancer Center Institutional Review Board approved protocol. All patients signed an informed consent form to provide peripheral blood and bone marrow samples. After collection of bone marrow samples, CD138+ cells were isolated as described[65], suspended in RPMI 1640 with 10% human AB serum (Cambrex Biosciences, East Rutherford, NJ) and used immediately for experiments. Peripheral blood samples were collected from patients 2, 4, and 5 for assessment of plasma HGF levels.

Immunoblot analysis

The effects of amuvatinib on HGF dependent signaling were assessed in U266 cells that had been serum starved for 24 h in RPMI 1640 containing 0.1% FBS; for the last 16 h of starvation the cells were treated with various concentrations of amuvatinib or DMSO. They were then treated with 50 ng/ml HGF for 15 min to stimulate MET. Amuvatinib-mediated induction of PARP cleavage was performed on U266 cells cultured in full serum (10% FBS) as well as under low serum conditions (0.1% FBS). Protein lysates and immunoblots were prepared as previously described[66]. Experiments were performed in triplicates, and bands were quantified by using an Odyssey Infrared Imaging System (LI-COR Biosciences, Lincoln, NE). Primary antibodies were: mouse monoclonal antibodies to MET clone 3D4 (Invitrogen, Carlsbad, CA); GSK-3β clone 7/GSK-3b, PARP clone C2-10, cleaved PARP Asp 214 clone F21-852, AKT clone 9Q7 (BD Biosciences Pharmingen, San Diego, CA); GAPDH clone 6C6 (Abcam, Inc, Cambridge, MA); phospho-ERK1/2 (Thr202/Tyr204) clone E10 (Cell Signaling Technology, Danvers, MA); β-actin clone AC-15 (Sigma Aldrich); rabbit monoclonal antibodies to phospho-GSK-3β (Ser9) clone 5B3 (Cell Signaling Technology); rabbit polyclonal antibodies to phospho-MET (Tyr1349) (Invitrogen); ERK1/2, and phospho-AKT (Ser473) (Cell Signaling Technology).

Flow cytometry

Intracellular protein expression in U266 cells was measured using BD Cytofix/Cytoperm Fixation/Permeabilization Kit (BD Biosciences). Primary antibodies used were anti-phospho-HGF R/c-MET (Tyr1234/1235) (R&D Systems, Minneapolis, MN), MET (C-12): sc-10 (Santa Cruz Biotechnology, Santa Cruz, CA), phospho-AKT (Ser473), AKT antibody (Cell Signaling Technology); and caspase-9 (Ser196) (Santa Cruz Biotechnology). Secondary antibody was a fluorescein isothiocyanate–conjugated (FITC) Affinipure goat anti-rabbit (Jackson ImmunoResearch, West Grove, PA). Cell cycle analysis and annexin V/propidium iodide (PI) staining were performed, respectively, as described[32, 33]. All flow cytometry analysis was performed using a Becton Dickinson FACSCalibur flow cytometer (San Jose, CA, USA). Statistical significance of changes was assessed by paired t-test analysis using Prism software (Graphpad, San Diego, CA).

Enzyme linked-immuno-sorbent assay for HGF levels

HGF levels in primary patient plasma were determined using the Human HGF Immunoassay Kit as per the manufacturer’s protocol (Invitrogen). The absorbance of this horseradish-peroxidase based assay was measured at 450 nm. Each sample was assayed in triplicate.

Acknowledgements

This work was supported in part by grants from the National Institutes of Health [Minority Graduate Student Supplement NIH RO1-CA85915 to CJP], the National Cancer Institute [Cancer Center Support Grant NIH P50 CA16672; and MD Anderson Cancer Center SPORE in Multiple Myeloma Research NIH P50 CA142509 to VB and MW], the Center for Targeted Therapy to CMS and VG, and Multiple Myeloma Research Foundation Senior Research Award to CMS.
The authors are grateful to David Graber for processing the bone marrow samples and separating the CD138+ cells and to Jairo A Matthews for coordinating patient sample collection distributing these samples to our laboratory.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​ ) applies to the data made available in this article, unless otherwise stated.

Competing interest

SR and PT are employed by Astex Pharmaceuticals, Inc., Dublin, CA. For the remaining authors, none was declared.

Authors’ contributions

CJP, SZ, KB, and CMS designed and performed experiments; CJP, SZ, JZ, VB, and CMS analyzed data; SS performed experiments; PT and SR provided amuvatinib; MW provide sorted bone marrow samples; CJP and CMS wrote the manuscript; CMS and VG directed study; VG provided laboratory resources; All authors critically read and approved manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Hallek M, Bergsagel PL, Anderson KC: Multiple myeloma: increasing evidence for a multistep transformation process. Blood. 1998, 91: 3-21.PubMedCentralPubMed Hallek M, Bergsagel PL, Anderson KC: Multiple myeloma: increasing evidence for a multistep transformation process. Blood. 1998, 91: 3-21.PubMedCentralPubMed
2.
Zurück zum Zitat Piazza FA, Gurrieri C, Trentin L, Semenzato G: Towards a new age in the treatment of multiple myeloma. Ann Hematol. 2007, 86: 159-172. 10.1007/s00277-006-0239-5.CrossRefPubMed Piazza FA, Gurrieri C, Trentin L, Semenzato G: Towards a new age in the treatment of multiple myeloma. Ann Hematol. 2007, 86: 159-172. 10.1007/s00277-006-0239-5.CrossRefPubMed
3.
Zurück zum Zitat Mahindra A, Laubach J, Raje N, Munshi N, Richardson PG, Anderson K: Latest advances and current challenges in the treatment of multiple myeloma. Nat Rev Clin Oncol. 2012, 9: 135-143. 10.1038/nrclinonc.2012.15.CrossRefPubMed Mahindra A, Laubach J, Raje N, Munshi N, Richardson PG, Anderson K: Latest advances and current challenges in the treatment of multiple myeloma. Nat Rev Clin Oncol. 2012, 9: 135-143. 10.1038/nrclinonc.2012.15.CrossRefPubMed
4.
Zurück zum Zitat Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC: Multiple myeloma. Lancet. 2009, 374: 324-339. 10.1016/S0140-6736(09)60221-X.CrossRefPubMed Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC: Multiple myeloma. Lancet. 2009, 374: 324-339. 10.1016/S0140-6736(09)60221-X.CrossRefPubMed
5.
Zurück zum Zitat Roodman GD: Targeting the bone microenvironment in multiple myeloma. J Bone Miner Metab. 2010, 28: 244-250. 10.1007/s00774-009-0154-7.CrossRefPubMed Roodman GD: Targeting the bone microenvironment in multiple myeloma. J Bone Miner Metab. 2010, 28: 244-250. 10.1007/s00774-009-0154-7.CrossRefPubMed
6.
Zurück zum Zitat Podar K, Anderson KC: The pathophysiologic role of VEGF in hematologic malignancies: therapeutic implications. Blood. 2005, 105: 1383-1395. 10.1182/blood-2004-07-2909.CrossRefPubMed Podar K, Anderson KC: The pathophysiologic role of VEGF in hematologic malignancies: therapeutic implications. Blood. 2005, 105: 1383-1395. 10.1182/blood-2004-07-2909.CrossRefPubMed
7.
Zurück zum Zitat Derksen PW, De Gorter DJ, Meijer HP, Bende RJ, Van Dijk M, Lokhorst HM, Bloem AC, Spaargaren M, Pals ST: The hepatocyte growth factor/Met pathway controls proliferation and apoptosis in multiple myeloma. Leukemia. 2003, 17: 764-774. 10.1038/sj.leu.2402875.CrossRefPubMed Derksen PW, De Gorter DJ, Meijer HP, Bende RJ, Van Dijk M, Lokhorst HM, Bloem AC, Spaargaren M, Pals ST: The hepatocyte growth factor/Met pathway controls proliferation and apoptosis in multiple myeloma. Leukemia. 2003, 17: 764-774. 10.1038/sj.leu.2402875.CrossRefPubMed
8.
Zurück zum Zitat Yasui H, Hideshima T, Richardson PG, Anderson KC: Novel therapeutic strategies targeting growth factor signalling cascades in multiple myeloma. Br J Haematol. 2006, 132: 385-397.PubMed Yasui H, Hideshima T, Richardson PG, Anderson KC: Novel therapeutic strategies targeting growth factor signalling cascades in multiple myeloma. Br J Haematol. 2006, 132: 385-397.PubMed
9.
Zurück zum Zitat Hideshima T, Podar K, Chauhan D, Anderson KC: Cytokines and signal transduction. Best Pract Res Clin Haematol. 2005, 18: 509-524. 10.1016/j.beha.2005.01.003.CrossRefPubMed Hideshima T, Podar K, Chauhan D, Anderson KC: Cytokines and signal transduction. Best Pract Res Clin Haematol. 2005, 18: 509-524. 10.1016/j.beha.2005.01.003.CrossRefPubMed
10.
Zurück zum Zitat Song L, Li Y, Sun YX, Yu M, Shen BF: 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. 2002, 21: 113-116.PubMed Song L, Li Y, Sun YX, Yu M, Shen BF: 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. 2002, 21: 113-116.PubMed
11.
Zurück zum Zitat Stellrecht CM, Gandhi V: MET receptor tyrosine kinase as a therapeutic anticancer target. Cancer Lett. 2009, 280: 1-14. 10.1016/j.canlet.2008.10.045.CrossRefPubMed Stellrecht CM, Gandhi V: MET receptor tyrosine kinase as a therapeutic anticancer target. Cancer Lett. 2009, 280: 1-14. 10.1016/j.canlet.2008.10.045.CrossRefPubMed
12.
Zurück zum Zitat Naldini L, Vigna E, Ferracini R, Longati P, Gandino L, Prat M, Comoglio PM: The tyrosine kinase encoded by the MET proto-oncogene is activated by autophosphorylation. Mol Cell Biol. 1991, 11: 1793-1803.PubMedCentralCrossRefPubMed Naldini L, Vigna E, Ferracini R, Longati P, Gandino L, Prat M, Comoglio PM: The tyrosine kinase encoded by the MET proto-oncogene is activated by autophosphorylation. Mol Cell Biol. 1991, 11: 1793-1803.PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Naldini L, Vigna E, Narsimhan RP, Gaudino G, Zarnegar R, Michalopoulos GK, Comoglio PM: Hepatocyte growth factor (HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-MET. Oncogene. 1991, 6: 501-504.PubMed Naldini L, Vigna E, Narsimhan RP, Gaudino G, Zarnegar R, Michalopoulos GK, Comoglio PM: Hepatocyte growth factor (HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-MET. Oncogene. 1991, 6: 501-504.PubMed
14.
Zurück zum Zitat Rodrigues GA, Park M: Autophosphorylation modulates the kinase activity and oncogenic potential of the Met receptor tyrosine kinase. Oncogene. 1994, 9: 2019-2027.PubMed Rodrigues GA, Park M: Autophosphorylation modulates the kinase activity and oncogenic potential of the Met receptor tyrosine kinase. Oncogene. 1994, 9: 2019-2027.PubMed
15.
Zurück zum Zitat Derksen PW, Keehnen RM, Evers LM, van Oers MH, Spaargaren M, Pals ST: Cell surface proteoglycan syndecan-1 mediates hepatocyte growth factor binding and promotes Met signaling in multiple myeloma. Blood. 2002, 99: 1405-1410. 10.1182/blood.V99.4.1405.CrossRefPubMed Derksen PW, Keehnen RM, Evers LM, van Oers MH, Spaargaren M, Pals ST: Cell surface proteoglycan syndecan-1 mediates hepatocyte growth factor binding and promotes Met signaling in multiple myeloma. Blood. 2002, 99: 1405-1410. 10.1182/blood.V99.4.1405.CrossRefPubMed
16.
Zurück zum Zitat Tjin EPM, Groen RWJ, Vogelzang I, Derksen PWB, Klok MD, Meijer HP, van Eeden S, Pals ST, Spaargaren M: Functional analysis of HGF/MET signaling and aberrant HGF-activator expression in diffuse large B-cell lymphoma. Blood. 2006, 107: 760-768. 10.1182/blood-2005-05-1929.CrossRefPubMed Tjin EPM, Groen RWJ, Vogelzang I, Derksen PWB, Klok MD, Meijer HP, van Eeden S, Pals ST, Spaargaren M: Functional analysis of HGF/MET signaling and aberrant HGF-activator expression in diffuse large B-cell lymphoma. Blood. 2006, 107: 760-768. 10.1182/blood-2005-05-1929.CrossRefPubMed
17.
Zurück zum Zitat Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB: Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005, 4: 988-1004. 10.1038/nrd1902.CrossRefPubMed Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB: Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005, 4: 988-1004. 10.1038/nrd1902.CrossRefPubMed
18.
Zurück zum Zitat 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.PubMed 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.PubMed
19.
Zurück zum Zitat Borset M, Lien E, Espevik T, Helseth E, Waage A, Sundan A: 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. 10.1074/jbc.271.40.24655.CrossRefPubMed Borset M, Lien E, Espevik T, Helseth E, Waage A, Sundan A: 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. 10.1074/jbc.271.40.24655.CrossRefPubMed
20.
Zurück zum Zitat Seidel C, Borset M, Turesson I, Abildgaard N, Sundan A, Waage A: Elevated serum concentrations of hepatocyte growth factor in patients with multiple myeloma. The nordic myeloma study group. Blood. 1998, 91: 806-812.PubMed Seidel C, Borset M, Turesson I, Abildgaard N, Sundan A, Waage A: Elevated serum concentrations of hepatocyte growth factor in patients with multiple myeloma. The nordic myeloma study group. Blood. 1998, 91: 806-812.PubMed
21.
Zurück zum Zitat Seidel C, Borset M, Hjertner O, Cao D, Abildgaard N, Hjorth-Hansen H, Sanderson RD, Waage A, Sundan A: High levels of soluble syndecan-1 in myeloma-derived bone marrow: modulation of hepatocyte growth factor activity. Blood. 2000, 96: 3139-3146.PubMed Seidel C, Borset M, Hjertner O, Cao D, Abildgaard N, Hjorth-Hansen H, Sanderson RD, Waage A, Sundan A: High levels of soluble syndecan-1 in myeloma-derived bone marrow: modulation of hepatocyte growth factor activity. Blood. 2000, 96: 3139-3146.PubMed
22.
Zurück zum Zitat Iwasaki T, Hamano T, Ogata A, Hashimoto N, Kitano M, Kakishita E: Clinical significance of vascular endothelial growth factor and hepatocyte growth factor in multiple myeloma. Br J Haematol. 2002, 116: 796-802. 10.1046/j.0007-1048.2002.03364.x.CrossRefPubMed Iwasaki T, Hamano T, Ogata A, Hashimoto N, Kitano M, Kakishita E: Clinical significance of vascular endothelial growth factor and hepatocyte growth factor in multiple myeloma. Br J Haematol. 2002, 116: 796-802. 10.1046/j.0007-1048.2002.03364.x.CrossRefPubMed
23.
Zurück zum Zitat Seidel C, Lenhoff S, Brabrand S, Anderson G, Standal T, Lanng-Nielsen J, Turesson I, Borset M, Waage A: Hepatocyte growth factor in myeloma patients treated with high-dose chemotherapy. Br J Haematol. 2002, 119: 672-676. 10.1046/j.1365-2141.2002.03898.x.CrossRefPubMed Seidel C, Lenhoff S, Brabrand S, Anderson G, Standal T, Lanng-Nielsen J, Turesson I, Borset M, Waage A: Hepatocyte growth factor in myeloma patients treated with high-dose chemotherapy. Br J Haematol. 2002, 119: 672-676. 10.1046/j.1365-2141.2002.03898.x.CrossRefPubMed
24.
Zurück zum Zitat Alexandrakis MG, Passam FH, Sfiridaki A, Kandidaki E, Roussou P, Kyriakou DS: Elevated serum concentration of hepatocyte growth factor in patients with multiple myeloma: correlation with markers of disease activity. Am J Hematol. 2003, 72: 229-233. 10.1002/ajh.10304.CrossRefPubMed Alexandrakis MG, Passam FH, Sfiridaki A, Kandidaki E, Roussou P, Kyriakou DS: Elevated serum concentration of hepatocyte growth factor in patients with multiple myeloma: correlation with markers of disease activity. Am J Hematol. 2003, 72: 229-233. 10.1002/ajh.10304.CrossRefPubMed
25.
Zurück zum Zitat Kara IO, Sahin B, Gunesacar R, Unsal C: 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. Adv Ther. 2006, 23: 635-645. 10.1007/BF02850052.CrossRefPubMed Kara IO, Sahin B, Gunesacar R, Unsal C: 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. Adv Ther. 2006, 23: 635-645. 10.1007/BF02850052.CrossRefPubMed
26.
Zurück zum Zitat Sezer O, Jakob C, Eucker J, Niemoller K, Gatz F, Wernecke K, Possinger K: 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. 2001, 66: 83-88. 10.1034/j.1600-0609.2001.00348.x.CrossRefPubMed Sezer O, Jakob C, Eucker J, Niemoller K, Gatz F, Wernecke K, Possinger K: 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. 2001, 66: 83-88. 10.1034/j.1600-0609.2001.00348.x.CrossRefPubMed
27.
Zurück zum Zitat Pour L, Svachova H, Adam Z, Mikulkova Z, Buresova L, Kovarova L, Buchler T, Penka M, Vorlicek J, Hajek R: Pretreatment hepatocyte growth factor and thrombospondin-1 levels predict response to high-dose chemotherapy for multiple myeloma. Neoplasma. 2010, 57: 29-34. 10.4149/neo_2010_01_029.CrossRefPubMed Pour L, Svachova H, Adam Z, Mikulkova Z, Buresova L, Kovarova L, Buchler T, Penka M, Vorlicek J, Hajek R: Pretreatment hepatocyte growth factor and thrombospondin-1 levels predict response to high-dose chemotherapy for multiple myeloma. Neoplasma. 2010, 57: 29-34. 10.4149/neo_2010_01_029.CrossRefPubMed
28.
Zurück zum Zitat Ludek P, Hana S, Zdenek A, Martina A, Dana K, Tomas B, Lucie K, Marta K, Jaroslav M, Miroslav P, et al: Treatment response to bortezomib in multiple myeloma correlates with plasma hepatocyte growth factor concentration and bone marrow thrombospondin concentration. Eur J Haematol. 2010, 84: 332-336. 10.1111/j.1600-0609.2009.01396.x.CrossRefPubMed Ludek P, Hana S, Zdenek A, Martina A, Dana K, Tomas B, Lucie K, Marta K, Jaroslav M, Miroslav P, et al: Treatment response to bortezomib in multiple myeloma correlates with plasma hepatocyte growth factor concentration and bone marrow thrombospondin concentration. Eur J Haematol. 2010, 84: 332-336. 10.1111/j.1600-0609.2009.01396.x.CrossRefPubMed
29.
Zurück zum Zitat Pour L, Svachova H, Adam Z, Almasi M, Buresova L, Buchler T, Kovarova L, Nemec P, Penka M, Vorlicek J, Hajek R: Levels of angiogenic factors in patients with multiple myeloma correlate with treatment response. Ann Hematol. 2010, 89: 385-389. 10.1007/s00277-009-0834-3.CrossRefPubMed Pour L, Svachova H, Adam Z, Almasi M, Buresova L, Buchler T, Kovarova L, Nemec P, Penka M, Vorlicek J, Hajek R: Levels of angiogenic factors in patients with multiple myeloma correlate with treatment response. Ann Hematol. 2010, 89: 385-389. 10.1007/s00277-009-0834-3.CrossRefPubMed
30.
Zurück zum Zitat Hjertner O, Torgersen ML, Seidel C, Hjorth-Hansen H, Waage A, Borset M, Sundan A: 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.PubMed Hjertner O, Torgersen ML, Seidel C, Hjorth-Hansen H, Waage A, Borset M, Sundan A: 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.PubMed
31.
Zurück zum Zitat Standal T, Abildgaard N, Fagerli UM, Stordal B, Hjertner O, Borset M, Sundan A: HGF inhibits BMP-induced osteoblastogenesis: possible implications for the bone disease of multiple myeloma. Blood. 2007, 109: 3024-3030.PubMed Standal T, Abildgaard N, Fagerli UM, Stordal B, Hjertner O, Borset M, Sundan A: HGF inhibits BMP-induced osteoblastogenesis: possible implications for the bone disease of multiple myeloma. Blood. 2007, 109: 3024-3030.PubMed
32.
Zurück zum Zitat Stellrecht CM, Phillip CJ, Cervantes-Gomez F, Gandhi V: Multiple myeloma cell killing by depletion of the MET receptor tyrosine kinase. Cancer Res. 2007, 67: 9913-9920. 10.1158/0008-5472.CAN-07-0770.CrossRefPubMed Stellrecht CM, Phillip CJ, Cervantes-Gomez F, Gandhi V: Multiple myeloma cell killing by depletion of the MET receptor tyrosine kinase. Cancer Res. 2007, 67: 9913-9920. 10.1158/0008-5472.CAN-07-0770.CrossRefPubMed
33.
Zurück zum Zitat Phillip CJ, Stellrecht CM, Nimmanapalli R, Gandhi V: Targeting MET transcription as a therapeutic strategy in multiple myeloma. Cancer Chemother Pharmacol. 2009, 63: 587-597. 10.1007/s00280-008-0770-2.PubMedCentralCrossRefPubMed Phillip CJ, Stellrecht CM, Nimmanapalli R, Gandhi V: Targeting MET transcription as a therapeutic strategy in multiple myeloma. Cancer Chemother Pharmacol. 2009, 63: 587-597. 10.1007/s00280-008-0770-2.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Chen LS, Stellrecht CM, Gandhi V: RNA-directed agent, cordycepin, induces cell death in multiple myeloma cells. Br J Haematol. 2008, 140: 682-691. 10.1111/j.1365-2141.2007.06955.x.CrossRefPubMed Chen LS, Stellrecht CM, Gandhi V: RNA-directed agent, cordycepin, induces cell death in multiple myeloma cells. Br J Haematol. 2008, 140: 682-691. 10.1111/j.1365-2141.2007.06955.x.CrossRefPubMed
35.
Zurück zum Zitat Mahtouk K, Tjin EP, Spaargaren M, Pals ST: The HGF/MET pathway as target for the treatment of multiple myeloma and B-cell lymphomas. Biochim Biophys Acta. 1806, 2010: 208-219. Mahtouk K, Tjin EP, Spaargaren M, Pals ST: The HGF/MET pathway as target for the treatment of multiple myeloma and B-cell lymphomas. Biochim Biophys Acta. 1806, 2010: 208-219.
36.
Zurück zum Zitat Qi W, Cooke L, Calaluce R, Bearss D, Mahadevan D: Dual inhibition of receptor tyrosine kinases of PDGFR and EGFR abolishes prostate cancer cell growth in a mouse xenograft model by complete dephosphorylation of PKB/Akt. Proc Am Assoc Cancer Res. 2007, 48: 5421. Qi W, Cooke L, Calaluce R, Bearss D, Mahadevan D: Dual inhibition of receptor tyrosine kinases of PDGFR and EGFR abolishes prostate cancer cell growth in a mouse xenograft model by complete dephosphorylation of PKB/Akt. Proc Am Assoc Cancer Res. 2007, 48: 5421.
37.
Zurück zum Zitat Tibes R, Berk GI, Fine GD, Choy GS, Tolcher AW: A phase-1 study of MP-470, a novel orally bioavailable small molecule with Rad51 suppression activity. Proc Am Assoc Cancer Res. 2008, 49: 4083. Tibes R, Berk GI, Fine GD, Choy GS, Tolcher AW: A phase-1 study of MP-470, a novel orally bioavailable small molecule with Rad51 suppression activity. Proc Am Assoc Cancer Res. 2008, 49: 4083.
38.
Zurück zum Zitat Tolcher AW, Berk GI, Fine GD, Choy GD, Bearss DJ, Redkar S, Tibes R: MP470, a potent oral Rad51 suppressor is safe and tolerable in first-in-human study. Proc Am Assoc Cancer Res. 2008, 49: 4083. Tolcher AW, Berk GI, Fine GD, Choy GD, Bearss DJ, Redkar S, Tibes R: MP470, a potent oral Rad51 suppressor is safe and tolerable in first-in-human study. Proc Am Assoc Cancer Res. 2008, 49: 4083.
39.
Zurück zum Zitat Mahtouk K, Moreaux J, Hose D, Reme T, Meissner T, Jourdan M, Rossi JF, Pals ST, Goldschmidt H, Klein B: Growth factors in multiple myeloma: a comprehensive analysis of their expression in tumor cells and bone marrow environment using Affymetrix microarrays. BMC Cancer. 2010, 10: 198-10.1186/1471-2407-10-198.PubMedCentralCrossRefPubMed Mahtouk K, Moreaux J, Hose D, Reme T, Meissner T, Jourdan M, Rossi JF, Pals ST, Goldschmidt H, Klein B: Growth factors in multiple myeloma: a comprehensive analysis of their expression in tumor cells and bone marrow environment using Affymetrix microarrays. BMC Cancer. 2010, 10: 198-10.1186/1471-2407-10-198.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Chng WJ, Kumar S, Vanwier S, Ahmann G, Price-Troska T, Henderson K, Chung TH, Kim S, Mulligan G, Bryant B, et al: Molecular dissection of hyperdiploid multiple myeloma by gene expression profiling. Cancer Res. 2007, 67: 2982-2989. 10.1158/0008-5472.CAN-06-4046.CrossRefPubMed Chng WJ, Kumar S, Vanwier S, Ahmann G, Price-Troska T, Henderson K, Chung TH, Kim S, Mulligan G, Bryant B, et al: Molecular dissection of hyperdiploid multiple myeloma by gene expression profiling. Cancer Res. 2007, 67: 2982-2989. 10.1158/0008-5472.CAN-06-4046.CrossRefPubMed
41.
Zurück zum Zitat Keats JJ, Fonseca R, Chesi M, Schop R, Baker A, Chng WJ, Van Wier S, Tiedemann R, Shi CX, Sebag M, et al: Promiscuous mutations activate the noncanonical NF-kappaB pathway in multiple myeloma. Cancer Cell. 2007, 12: 131-144. 10.1016/j.ccr.2007.07.003.PubMedCentralCrossRefPubMed Keats JJ, Fonseca R, Chesi M, Schop R, Baker A, Chng WJ, Van Wier S, Tiedemann R, Shi CX, Sebag M, et al: Promiscuous mutations activate the noncanonical NF-kappaB pathway in multiple myeloma. Cancer Cell. 2007, 12: 131-144. 10.1016/j.ccr.2007.07.003.PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Meads MB, Hazlehurst LA, Dalton WS: The bone marrow microenvironment as a tumor sanctuary and contributor to drug resistance. Clin Cancer Res. 2008, 14: 2519-2526. 10.1158/1078-0432.CCR-07-2223.CrossRefPubMed Meads MB, Hazlehurst LA, Dalton WS: The bone marrow microenvironment as a tumor sanctuary and contributor to drug resistance. Clin Cancer Res. 2008, 14: 2519-2526. 10.1158/1078-0432.CCR-07-2223.CrossRefPubMed
43.
Zurück zum Zitat Zhang W, Huang P: Cancer-stromal interactions: role in cell survival, metabolism and drug sensitivity. Cancer Biol Ther. 2011, 11: 150-156. 10.4161/cbt.11.2.14623.PubMedCentralCrossRefPubMed Zhang W, Huang P: Cancer-stromal interactions: role in cell survival, metabolism and drug sensitivity. Cancer Biol Ther. 2011, 11: 150-156. 10.4161/cbt.11.2.14623.PubMedCentralCrossRefPubMed
44.
45.
Zurück zum Zitat Rodrigues GA, Naujokas MA, Park M: Alternative splicing generates isoforms of the met receptor tyrosine kinase which undergo differential processing. Mol Cell Biol. 1991, 11: 2962-2970.PubMedCentralCrossRefPubMed Rodrigues GA, Naujokas MA, Park M: Alternative splicing generates isoforms of the met receptor tyrosine kinase which undergo differential processing. Mol Cell Biol. 1991, 11: 2962-2970.PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Hov H, Holt RU, Ro TB, Fagerli UM, Hjorth-Hansen H, Baykov V, Christensen JG, Waage A, Sundan A, Borset M: 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. 10.1158/1078-0432.CCR-04-0874.CrossRefPubMed Hov H, Holt RU, Ro TB, Fagerli UM, Hjorth-Hansen H, Baykov V, Christensen JG, Waage A, Sundan A, Borset M: 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. 10.1158/1078-0432.CCR-04-0874.CrossRefPubMed
47.
Zurück zum Zitat Tolcher AW, Mita M, Gordon M, Rosen L, Patnaik A, Mita A, Fine GD, Choy GS, Berk GI: Clinical responses in highly refractory solid tumor patients with oral MP-470, a multi-targeted tyrosine kinase inhibitor, in combination with standard of care chemotherapy regimens: preliminary report from a multi-institutional phase-Ib clinical trial. Proc Am Assoc Cancer Res. 2008, 49: 403. Tolcher AW, Mita M, Gordon M, Rosen L, Patnaik A, Mita A, Fine GD, Choy GS, Berk GI: Clinical responses in highly refractory solid tumor patients with oral MP-470, a multi-targeted tyrosine kinase inhibitor, in combination with standard of care chemotherapy regimens: preliminary report from a multi-institutional phase-Ib clinical trial. Proc Am Assoc Cancer Res. 2008, 49: 403.
48.
Zurück zum Zitat Mahadevan D, Cooke L, Riley C, Swart R, Simons B, Della Croce K, Wisner L, Iorio M, Shakalya K, Garewal H, et al: A novel tyrosine kinase switch is a mechanism of imatinib resistance in gastrointestinal stromal tumors. Oncogene. 2007, 26: 3909-3919. 10.1038/sj.onc.1210173.CrossRefPubMed Mahadevan D, Cooke L, Riley C, Swart R, Simons B, Della Croce K, Wisner L, Iorio M, Shakalya K, Garewal H, et al: A novel tyrosine kinase switch is a mechanism of imatinib resistance in gastrointestinal stromal tumors. Oncogene. 2007, 26: 3909-3919. 10.1038/sj.onc.1210173.CrossRefPubMed
49.
Zurück zum Zitat Trevor K, Combs D, Mahadevan D, Bearss D, Cranmer L: Activity of the multi-targeted, receptor tyrosine kinase inhibitor MP470 against synovial sarcoma cells. Proc Am Assoc Cancer Res. 2008, 49: 4891. Trevor K, Combs D, Mahadevan D, Bearss D, Cranmer L: Activity of the multi-targeted, receptor tyrosine kinase inhibitor MP470 against synovial sarcoma cells. Proc Am Assoc Cancer Res. 2008, 49: 4891.
50.
Zurück zum Zitat Welsh JW, Mahadevan D, Ellsworth R, Cooke L, Bearss D, Stea B: The c-Met receptor tyrosine kinase inhibitor MP470 radiosensitizes glioblastoma cells. Radiat Oncol. 2009, 4: 69-10.1186/1748-717X-4-69.PubMedCentralCrossRefPubMed Welsh JW, Mahadevan D, Ellsworth R, Cooke L, Bearss D, Stea B: The c-Met receptor tyrosine kinase inhibitor MP470 radiosensitizes glioblastoma cells. Radiat Oncol. 2009, 4: 69-10.1186/1748-717X-4-69.PubMedCentralCrossRefPubMed
51.
Zurück zum Zitat Tibes R, Fine G, Choy G, Redkar S, Taverna P, Oganesian A, Sahai A, Azab M, Tolcher AW: A phase I, first-in-human dose-escalation study of amuvatinib, a multi-targeted tyrosine kinase inhibitor, in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2013, 71: 463-471. 10.1007/s00280-012-2019-3.CrossRefPubMed Tibes R, Fine G, Choy G, Redkar S, Taverna P, Oganesian A, Sahai A, Azab M, Tolcher AW: A phase I, first-in-human dose-escalation study of amuvatinib, a multi-targeted tyrosine kinase inhibitor, in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2013, 71: 463-471. 10.1007/s00280-012-2019-3.CrossRefPubMed
52.
Zurück zum Zitat Munshi N, Jeay S, Li Y, Chen CR, France DS, Ashwell MA, Hill J, Moussa MM, Leggett DS, Li CJ: ARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activity. Mol Cancer Ther. 2010, 9: 1544-1553. 10.1158/1535-7163.MCT-09-1173.CrossRefPubMed Munshi N, Jeay S, Li Y, Chen CR, France DS, Ashwell MA, Hill J, Moussa MM, Leggett DS, Li CJ: ARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activity. Mol Cancer Ther. 2010, 9: 1544-1553. 10.1158/1535-7163.MCT-09-1173.CrossRefPubMed
53.
Zurück zum Zitat Eathiraj S, Palma R, Volckova E, Hirschi M, France DS, Ashwell MA, Chan TC: Discovery of a novel mode of protein kinase inhibition characterized by the mechanism of inhibition of human mesenchymal-epithelial transition factor (c-Met) protein autophosphorylation by ARQ 197. J Biol Chem. 2011, 286: 20666-20676. 10.1074/jbc.M110.213801.PubMedCentralCrossRefPubMed Eathiraj S, Palma R, Volckova E, Hirschi M, France DS, Ashwell MA, Chan TC: Discovery of a novel mode of protein kinase inhibition characterized by the mechanism of inhibition of human mesenchymal-epithelial transition factor (c-Met) protein autophosphorylation by ARQ 197. J Biol Chem. 2011, 286: 20666-20676. 10.1074/jbc.M110.213801.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W, Ferrari D, Chen Y, Costa DB, Gerber DE, Orlov S, et al: Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011, 29: 3307-3315. 10.1200/JCO.2010.34.0570.CrossRefPubMed Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W, Ferrari D, Chen Y, Costa DB, Gerber DE, Orlov S, et al: Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011, 29: 3307-3315. 10.1200/JCO.2010.34.0570.CrossRefPubMed
55.
Zurück zum Zitat Yap TA, Olmos D, Brunetto AT, Tunariu N, Barriuso J, Riisnaes R, Pope L, Clark J, Futreal A, Germuska M, et al: Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. J Clin Oncol. 2011, 29: 1271-1279. 10.1200/JCO.2010.31.0367.CrossRefPubMed Yap TA, Olmos D, Brunetto AT, Tunariu N, Barriuso J, Riisnaes R, Pope L, Clark J, Futreal A, Germuska M, et al: Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. J Clin Oncol. 2011, 29: 1271-1279. 10.1200/JCO.2010.31.0367.CrossRefPubMed
56.
Zurück zum Zitat Goldman JW, Laux I, Chai F, Savage RE, Ferrari D, Garmey EG, Just RG, Rosen LS: Phase 1 dose-escalation trial evaluating the combination of the selective MET (mesenchymal-epithelial transition factor) inhibitor tivantinib (ARQ 197) plus erlotinib. Cancer. 2012, 118: 5903-5911. 10.1002/cncr.27575.CrossRefPubMed Goldman JW, Laux I, Chai F, Savage RE, Ferrari D, Garmey EG, Just RG, Rosen LS: Phase 1 dose-escalation trial evaluating the combination of the selective MET (mesenchymal-epithelial transition factor) inhibitor tivantinib (ARQ 197) plus erlotinib. Cancer. 2012, 118: 5903-5911. 10.1002/cncr.27575.CrossRefPubMed
57.
Zurück zum Zitat Previdi S, Abbadessa G, Dalo F, France DS, 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. 10.1158/1535-7163.MCT-11-0277.CrossRefPubMed Previdi S, Abbadessa G, Dalo F, France DS, 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. 10.1158/1535-7163.MCT-11-0277.CrossRefPubMed
58.
Zurück zum Zitat Wagner AJ, Goldberg JM, Dubois SG, Choy E, Rosen L, Pappo A, Geller J, Judson I, Hogg D, Senzer N, et al: Tivantinib (ARQ 197), a selective inhibitor of MET, in patients with microphthalmia transcription factor-associated tumors: results of a multicenter phase 2 trial. Cancer. 2012, 118: 5894-5902. 10.1002/cncr.27582.CrossRefPubMed Wagner AJ, Goldberg JM, Dubois SG, Choy E, Rosen L, Pappo A, Geller J, Judson I, Hogg D, Senzer N, et al: Tivantinib (ARQ 197), a selective inhibitor of MET, in patients with microphthalmia transcription factor-associated tumors: results of a multicenter phase 2 trial. Cancer. 2012, 118: 5894-5902. 10.1002/cncr.27582.CrossRefPubMed
59.
Zurück zum Zitat Zaman S, Stellrecht CM, Shentu S, Orlowski R, Gandhi V: Targeting the pro-survival protein c-MET with ARQ 197 inhibits growth of multiple myeloma cells. Proc Am Assoc Cancer Res. 2012, 53: 844. Zaman S, Stellrecht CM, Shentu S, Orlowski R, Gandhi V: Targeting the pro-survival protein c-MET with ARQ 197 inhibits growth of multiple myeloma cells. Proc Am Assoc Cancer Res. 2012, 53: 844.
60.
Zurück zum Zitat Dorkhom SJ, Zaman S, Thomas SK, Alexanian R, Shah JJ, Weber DM, Wang M, Anderson ML, Baladandayuthapani V, Lin YH, et al: Phase II study of the c-Met inhibitor ARQ 197 (tivantinib) in patients with relapsed multiple myeloma. Blood (ASH Annual Meeting Abstracts). 2012, 120: 2976. Dorkhom SJ, Zaman S, Thomas SK, Alexanian R, Shah JJ, Weber DM, Wang M, Anderson ML, Baladandayuthapani V, Lin YH, et al: Phase II study of the c-Met inhibitor ARQ 197 (tivantinib) in patients with relapsed multiple myeloma. Blood (ASH Annual Meeting Abstracts). 2012, 120: 2976.
61.
Zurück zum Zitat Catlett-Falcone R, Landowski TH, Oshiro MM, Turkson J, Levitzki A, Savino R, Ciliberto G, Moscinski L, Fernandez-Luna JL, Nunez G, et al: Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 1999, 10: 105-115. 10.1016/S1074-7613(00)80011-4.CrossRefPubMed Catlett-Falcone R, Landowski TH, Oshiro MM, Turkson J, Levitzki A, Savino R, Ciliberto G, Moscinski L, Fernandez-Luna JL, Nunez G, et al: Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 1999, 10: 105-115. 10.1016/S1074-7613(00)80011-4.CrossRefPubMed
62.
Zurück zum Zitat Bellamy WT, Dalton WS, Gleason MC, Grogan TM, Trent JM: Development and characterization of a Melphalan-resistant human multiple myeloma cell line. Cancer Res. 1991, 51: 995-1002.PubMed Bellamy WT, Dalton WS, Gleason MC, Grogan TM, Trent JM: Development and characterization of a Melphalan-resistant human multiple myeloma cell line. Cancer Res. 1991, 51: 995-1002.PubMed
63.
Zurück zum Zitat Manshouri T, Estrov Z, Quintas-Cardama A, Burger J, Zhang Y, Livun A, Knez L, Harris D, Creighton CJ, Kantarjian HM, Verstovsek S: Bone marrow stroma-secreted cytokines protect JAK2(V617F)-mutated cells from the effects of a JAK2 inhibitor. Cancer Res. 2011, 71: 3831-3840. 10.1158/0008-5472.CAN-10-4002.PubMedCentralCrossRefPubMed Manshouri T, Estrov Z, Quintas-Cardama A, Burger J, Zhang Y, Livun A, Knez L, Harris D, Creighton CJ, Kantarjian HM, Verstovsek S: Bone marrow stroma-secreted cytokines protect JAK2(V617F)-mutated cells from the effects of a JAK2 inhibitor. Cancer Res. 2011, 71: 3831-3840. 10.1158/0008-5472.CAN-10-4002.PubMedCentralCrossRefPubMed
64.
Zurück zum Zitat Gandhi V, Ayres M, Halgren RG, Krett NL, Newman RA, Rosen ST: 8-chloro-cAMP and 8-chloro-adenosine act by the same mechanism in multiple myeloma cells. Cancer Res. 2001, 61: 5474-5479.PubMed Gandhi V, Ayres M, Halgren RG, Krett NL, Newman RA, Rosen ST: 8-chloro-cAMP and 8-chloro-adenosine act by the same mechanism in multiple myeloma cells. Cancer Res. 2001, 61: 5474-5479.PubMed
65.
Zurück zum Zitat Bjorklund CC, Ma W, Wang ZQ, Davis RE, Kuhn DJ, Kornblau SM, Wang ML, Shah JJ, Orlowski RZ: Evidence of a role for activation of Wnt/b-catenin signaling in the resistance of plasma cells to lenalidomide. J Biol Chem. 2011, 286: 11009-11020. 10.1074/jbc.M110.180208.PubMedCentralCrossRefPubMed Bjorklund CC, Ma W, Wang ZQ, Davis RE, Kuhn DJ, Kornblau SM, Wang ML, Shah JJ, Orlowski RZ: Evidence of a role for activation of Wnt/b-catenin signaling in the resistance of plasma cells to lenalidomide. J Biol Chem. 2011, 286: 11009-11020. 10.1074/jbc.M110.180208.PubMedCentralCrossRefPubMed
66.
Zurück zum Zitat Stellrecht CM, Ayres M, Arya R, Gandhi V: A unique RNA-directed nucleoside analog is cytotoxic to breast cancer cells and depletes cyclin E levels. Breast Cancer Res Treat. 2010, 121: 355-364. 10.1007/s10549-009-0481-3.CrossRefPubMed Stellrecht CM, Ayres M, Arya R, Gandhi V: A unique RNA-directed nucleoside analog is cytotoxic to breast cancer cells and depletes cyclin E levels. Breast Cancer Res Treat. 2010, 121: 355-364. 10.1007/s10549-009-0481-3.CrossRefPubMed
Metadaten
Titel
Targeting MET kinase with the small-molecule inhibitor amuvatinib induces cytotoxicity in primary myeloma cells and cell lines
verfasst von
Cornel Joseph Phillip
Shadia Zaman
Shujun Shentu
Kumudha Balakrishnan
Jiexin Zhang
Veera Baladandayuthapani
Pietro Taverna
Sanjeev Redkar
Michael Wang
Christine Marie Stellrecht
Varsha Gandhi
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2013
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/1756-8722-6-92

Weitere Artikel der Ausgabe 1/2013

Journal of Hematology & Oncology 1/2013 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.