p38 mitogen-activated protein kinase (MAPK) is a member of the MAPK family which is activated by cytokines and growth factors, but its role in pathogenesis of multiple myeloma (MM) is unknown. In this study, we demonstrate that the specific p38 MAPK inhibitor VX-745 inhibits interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF) secretion in bone marrow stromal cells (BMSCs), without affecting their viability. Tumor necrosis factor alpha (TNF-α)–induced IL-6 secretion in BMSCs is also inhibited by VX-745. Importantly, VX-745 inhibits both MM cell proliferation and IL-6 secretion in BMSCs triggered by adherence of MM cells to BMSCs, suggesting that it can inhibit paracrine MM cell growth in the BM milieu and overcome cell adhesion–related drug resistance. These studies therefore identify p38 MAPK as a novel therapeutic target to overcome drug resistance and improve patient outcome in MM.

Multiple myeloma (MM) cell growth, survival, drug resistance against conventional chemotherapies, and migration are mediated via cytokines in the bone marrow (BM) milieu. Specifically, interleukin 6 (IL-6)1-4 promotes multiple myeloma (MM) cell growth, survival, and drug resistance; whereas vascular endothelial growth factor (VEGF)5 induces MM cell migration. Importantly, adherence of MM cells to bone marrow stromal cells (BMSCs) up-regulates cytokine (IL-6, VEGF) secretion from both MM and BMSCs.6-8 Most excitingly, novel agents including proteasome inhibitor PS-341 and the immunodulatory derivatives of thalidomide (IMiDs) can overcome drug resistance in vitro9and in animal models10 by targeting not only tumor cells, but also host tumor cell interactions and cytokines in the BM milieu. Already these agents are achieving responses, even complete responses, in the setting of relapse and refractory MM.11,12 

Recent studies have demonstrated that activation of p38 MAPK is associated with IL-6 gene expression and/or protein secretion in Sertoli cells,13 myocardial cells,14 and osteoblasts15; however, the role of p38 MAPK activation in modulating IL-6 secretion in BMSCs is undefined. In the present study, we demonstrate that the p38 MAPK inhibitor VX-745 decreases IL-6 secretion in the BMSCs of patients with MM, proliferation of MM cells adherent to BMSCs, and IL-6 secretion in BMSCs triggered by adherence of MM cells to BMSCs. These studies therefore identify p38 MAPK as a novel therapeutic target in MM, and provide the framework for clinical trials of VX-745 to improve patient outcome.

MM-derived cell lines and patient MM cells

The MM.1S human MM cell line was kindly provided by Dr Steven Rosen (Northwestern University, Chicago, IL). RPMI8226 and U266 human MM cells were obtained from American Type Culture Collection (Rockville, MD). All MM cell lines were maintained as described.16,17 

BMSC cultures

BM specimens were obtained from patients with MM. Mononuclear cells (MNCs) separated by Ficoll-Hipaque density sedimentation were used to establish long-term BMSC cultures, as described.16,18 

p38 MAPK inhibitor and cytokines

A potent p38 MAPK–specific inhibitor, VX-745 (Vertex Pharmaceuticals, Cambridge, MA), was dissolved in dimethyl sulfoxide (DMSO) and kept at −20°C until use. Tumor necrosis factor alpha (TNF-α) was purchased from R&D Systems (Minneapolis, MN). SB203580 (Calbiochem, San Diego, CA) was used as a positive control for p38 MAPK inhibition.

DNA synthesis and growth inhibition assay

Proliferation was measured as previously described.16 MM cells (3 × 104cells/well) or BMSCs (5 × 104cells/well) were incubated in 96-well culture plates (Costar, Cambridge, MA) in the presence or absence of VX-745 for 48 hours at 37°C. DNA synthesis was measured by [3H]-thymidine ([3H]TdR; NEN Perkin Elmer Life Sciences, Boston, MA) uptake. Cells were pulsed with [3H]TdR (0.5 μCi/well [.0185 MBq]) during the last 8 hours of 48-hour cultures. Growth inhibition of both MM cells and BMSCs by VX-745 was also assessed by measuring 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) dye absorbance, as previously described.16 All experiments were performed in quadruplicate.

Effect of VX-745 on paracrine MM cell growth in the BM

To evaluate growth stimulation and signaling in MM cells adherent to BMSCs, 3 × 104 MM.1S cells were cultured in BMSC-coated 96-well plates for 48 hours, in the presence or absence of VX-745. DNA synthesis was measured as described above. The Duoset enzyme-linked immunosorbent assay (ELISA) (R&D Systems) was used to measure IL-6 and VEGF in supernatants of 48-hour cultures of BMSCs with or without MM.1S cells, and in the presence or absence of VX-745, as previously described.19 

Statistical analysis

Statistical significance of differences observed in drug-treated versus control cultures was determined using the Wilcoxon signed-rank test. The minimal level of significance was P < .05.

VX-745 inhibits IL-6 and VEGF secretion in BMSCs

We first examined the direct effect of VX-745 on the growth of BMSCs in patients with MM, assessed by MTT assay. The growth of BMSCs in 6 patients with MM was not inhibited by VX-745 (0.05 μM-20 μM; Figure 1A). We have shown that BMSCs secrete IL-6 and VEGF,7,8,19which mediated MM cell growth, survival, drug resistance, and migration, and we therefore next examined whether VX-745 inhibits IL-6 and/or VEGF secretion from BMSCs. VX-745 significantly (P < .01) decreases IL-6 secretion in BMSCs in a dose-dependent fashion (Figure 1B), with peak inhibitory effect at 2 μM VX-745. VX-745 similarly inhibits VEGF secretion from BMSCs (Figure 1B) and MM cell lines (data not shown). To confirm that this inhibitory effect was mediated via inhibition of p38 MAPK, we used SB203580. As seen in Figure 1C, SB203580 also inhibits secretion of both IL-6 and VEGF in a dose-dependent fashion from BMSCs, suggesting that p38 MAPK regulates secretion of these cytokines from BMSCs.

Fig. 1.

Effect of VX-745 on IL-6 and VEGF secretion in BMSCs.

(A) BMSCs from patients with MM were cultured in the presence (0.06 μM-20 μM) or absence of VX-745 for 48 hours, and cell viability was assessed by MTT assay. The figure represents means (± SDs) from BMSCs of 6 patients with MM. (B) BMSCs were cultured in the presence (0.06 μM-20 μM) or absence of VX-745 for 48 hours. IL-6 (▪) and VEGF (■) in culture supernatants was measured by ELISA. (C) BMSCs were cultured in the presence or absence of SB203580 (2.5 μM-20 μM) for 48 hours. IL-6 (▪) and VEGF (■) in culture supernatants was measured by ELISA. (D) BMSCs were cultured in the presence (0.06 μM-20 μM) or absence of VX-745 for 48 hours. VEGF in culture supernatants was measured by ELISA. (D) BMSCs were cultured with TNF-α (5 ng/mL) in the absence (■) or presence of 0.05 μM (▧), 0.5 μM (▤), and 5 μM (▪) VX-745 for 48 hours. IL-6 in culture supernatants was measured by ELISA.

Fig. 1.

Effect of VX-745 on IL-6 and VEGF secretion in BMSCs.

(A) BMSCs from patients with MM were cultured in the presence (0.06 μM-20 μM) or absence of VX-745 for 48 hours, and cell viability was assessed by MTT assay. The figure represents means (± SDs) from BMSCs of 6 patients with MM. (B) BMSCs were cultured in the presence (0.06 μM-20 μM) or absence of VX-745 for 48 hours. IL-6 (▪) and VEGF (■) in culture supernatants was measured by ELISA. (C) BMSCs were cultured in the presence or absence of SB203580 (2.5 μM-20 μM) for 48 hours. IL-6 (▪) and VEGF (■) in culture supernatants was measured by ELISA. (D) BMSCs were cultured in the presence (0.06 μM-20 μM) or absence of VX-745 for 48 hours. VEGF in culture supernatants was measured by ELISA. (D) BMSCs were cultured with TNF-α (5 ng/mL) in the absence (■) or presence of 0.05 μM (▧), 0.5 μM (▤), and 5 μM (▪) VX-745 for 48 hours. IL-6 in culture supernatants was measured by ELISA.

Close modal

We next examined whether VX-745 inhibits IL-6 secretion in BMSCs in the presence of TNF-α, since TNF-α is secreted by MM cells,19 activates p38 MAPK,20,21 and induces IL-6 secretion in BMSCs.19 Importantly, VX-745 inhibits TNF-α–induced up-regulation of IL-6 in BMSCs (Figure 1D). Similar results are observed in the presence of VEGF (data not shown), which is secreted by MM cells and BMSCs8 and in turn induces further IL-6 transcription22 and secretion from BMSCs.8 Since transcription and protein expression of IL-6 is regulated by NF-κB activation,6,16 these results suggest that p38 MAPK may mediate TNF-α–induced nuclear factor (NF)–κB activation and related induction of IL-6 in BMSCs, consistent with previous reports in myocytes14 and osteoblasts.15 Importantly, since both TNF-α and VEGF are secreted by MM cells,8,19 these results suggest that VX-745 can inhibit cytokine (TNF-α, IL-6, VEGF)–induced paracrine MM cell growth, survival, and drug resistance in the BM microenvironment. Ongoing studies are delineating whether VX-745 inhibits constitutive and/or TNF-α–stimulated NF-κB activation in BMSCs.

VX-745 inhibits paracrine MM cell growth in the BM

We next examined whether VX-745 inhibits MM cell growth in the BMSC microenvironment. As seen in Figure2A, VX-745 induces modest growth inhibition of MM.1S, RPMI8226, and U266 cell lines in a dose-dependent fashion, with inhibitory concentration of 50% (IC50) of 10 μM as assessed by MTT assay. This result suggests that p38 MAPK only partially mediates MM cell proliferation. Since adherence of MM cells to BMSCs promotes tumor cell growth,7,8,16 we next examined whether VX-745 overcomes growth of MM cells adherent to BMSCs. As expected, VX-745 in a dose-dependent fashion significantly (P < .01) inhibited proliferation of MM.1S cells induced by adherence to BMSCs (Figure 2B). Since binding of MM cells to BMSCs also triggers IL-6 transcription and secretion in the BM milieu, we next determined whether VX-745 similarly inhibited IL-6 secretion in MM.1S/BMSCs cultures. As seen in Figure 2C, VX-745 completely (P < .01) abrogates IL-6 secretion triggered by adherence of MM cells to BMSCs. These results demonstrate that VX-745 inhibits MM cell growth, at least in part, due to inhibition of IL-6 secretion in BMSCs. Importantly, these studies suggest that VX-745 may overcome cell adhesion–related drug resistance. They provide the framework for clinical evaluation of this agent, alone or combined with conventional or other novel therapies, to improve patient outcome in MM.

Fig. 2.

Effect of VX-745 on paracrine MM cell growth in the BM.

(A) MM.1S (●), RPMI8226 (▪), and U266 (▴) (3 × 104) cells were cultured in the presence of VX-745 (0.06 μM-20 μM) for 48 hours, and cell viability was assessed by MTT assay. (B) MM.1S cells were cultured in BMSC-coated 96-well plates for 48 hours, in the absence (■) or presence of 0.2 μM (▤), 1 μM (▧), and 5 μM (▪) VX-745. DNA synthesis was assessed by [3H]TdR uptake. (C) BMSCs were cultured with 3 × 104 MM.1S cells in the absence (■) or presence of 0.2 μM (▤), 1 μM (▧), and 5 μM (▪) VX-745 for 24 hours. IL-6 in culture supernatants was measured by ELISA.

Fig. 2.

Effect of VX-745 on paracrine MM cell growth in the BM.

(A) MM.1S (●), RPMI8226 (▪), and U266 (▴) (3 × 104) cells were cultured in the presence of VX-745 (0.06 μM-20 μM) for 48 hours, and cell viability was assessed by MTT assay. (B) MM.1S cells were cultured in BMSC-coated 96-well plates for 48 hours, in the absence (■) or presence of 0.2 μM (▤), 1 μM (▧), and 5 μM (▪) VX-745. DNA synthesis was assessed by [3H]TdR uptake. (C) BMSCs were cultured with 3 × 104 MM.1S cells in the absence (■) or presence of 0.2 μM (▤), 1 μM (▧), and 5 μM (▪) VX-745 for 24 hours. IL-6 in culture supernatants was measured by ELISA.

Close modal

Prepublished online as Blood First Edition Paper, September 5, 2002; DOI 10.1182/blood-2002-06-1874.

Supported by National Institutes of Health grants PO-1 78378 and RO-1 50947 and the Doris Duke Distinguished Clinical Research Scientist Award (K.C.A.), and the Multiple Myeloma Research Foundation Senior Award (T.H.).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Lichtenstein
A
Tu
Y
Fady
C
Vescio
R
Berenson
J
Interleukin-6 inhibits apoptosis of malignant plasma cells.
Cell Immunol.
162
1995
248
255
2
Ogata
A
Chauhan
D
Teoh
G
et al
Interleukin-6 triggers cell growth via the ras-dependent mitogen-activated protein kinase cascade.
J Immunol.
159
1997
2212
2221
3
Chauhan
D
Kharbanda
S
Ogata
A
et al
Interleukin-6 inhibits Fas-induced apoptosis and stress-activated protein kinase activation in multiple myeloma cells.
Blood.
89
1997
227
234
4
Hideshima
T
Nakamura
N
Chauhan
D
Anderson
KC
Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling in multiple myeloma.
Oncogene.
20
2001
5991
6000
5
Podar
K
Tai
YT
Davies
FE
et al
Vascular endothelial growth factor triggers signaling cascades mediating multiple myeloma cell growth and migration.
Blood.
98
2001
428
435
6
Chauhan
D
Uchiyama
H
Akbarali
Y
et al
Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-κB.
Blood.
87
1996
1104
1112
7
Hideshima
T
Richardson
P
Chauhan
D
et al
The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma.
Cancer Res.
61
2001
3071
3076
8
Gupta
D
Treon
SP
Shima
Y
et al
Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: therapeutic applications.
Leukemia.
15
2001
1950
1961
9
Hideshima
T
Chauhan
D
Shima
Y
et al
Thalidomide and its analogues overcome drug resistance of human multiple myeloma cells to conventional therapy.
Blood.
96
2000
2943
2950
10
Lentzsch
S
Rogers
MS
LeBlanc
R
et al
S-3-amino-phthalimido-glutarimide inhibits angiogenesis and growth of B-cell neoplasias in mice.
Cancer Res.
62
2002
2300
2305
11
Richardson
PG
Berenson
J
Irwin
D
et al
Phase II trial of PS-341, a novel proteasome inhibitor, alone or in combination with dexamethasone, in patients with multiple myeloma who have relapsed following front-line therapy and are refractory to their most recent therapy [abstract].
Blood.
98
2001
774a
12
Richardson
P
Schlosman
R
Weller
E
et al
Immunomodulatory derivative of thalidomide CC-5013 overcomes drug resistance and is well tolerated in patients with relapsed multiple myeloma.
Blood.
100
2002
3063
3067
13
De Cesaris
P
Starace
D
Riccioli
A
Padula
F
Filippin
A
Tumor necrosis factor-α induces interleukin-6 production and integrin ligand expression by distinct transcription pathways.
J Biol Chem.
273
1998
7566
7571
14
Craig
R
Larkin
A
Mingo
AM
et al
p38 MAPK and NF-κB collaborate to induce interleukin-6 gene expression and release.
J Biol Chem.
275
2000
23814
23824
15
Chae
HJ
Chae
SW
Chin
HY
et al
The p38 mitogen-activated protein kinase pathway regulates interleukin-6 synthesis in response to tumor necrosis factor in osteoblasts.
Bone.
28
2001
45
53
16
Hideshima
T
Chauhan
D
Richardson
P
et al
NF-κB as a therapeutic target in multipe myeloma.
J Biol Chem.
277
2002
6639
6647
17
Hideshima
T
Chauhan
D
Hayashi
T
et al
The biological sequelae of stromal cell-derived factor-1α in multiple myeloma.
Mol Cancer Ther.
1
2002
539
544
18
Uchiyama
H
Barut
BA
Mohrbacher
AF
Chauhan
D
Anderson
KC
Adhesion of human myeloma-derived cell lines to bone marrow stromal cells stimulates IL-6 secretion.
Blood.
82
1993
3712
3720
19
Hideshima
T
Chauhan
D
Schlossman
R
Richardson
P
Anderson
KC
The role of tumor necrosis factor α in the pathogenesis of human multiple myeloma: therapeutic applications.
Oncogene.
20
2001
4519
4527
20
Read
MA
Whitley
MZ
Gupta
S
et al
Tumor necrosis factor α-induced E-selectin expression is activated by the nuclear factor-kappaB and c-JUN N-terminal kinase/p38 mitogen-activated protein kinase pathways.
J Biol Chem.
272
1997
2753
2761
21
Barbin
G
Roisin
MP
Zalc
B
Tumor necrosis factor alpha activates the phosphorylation of ERK, SAPK/JNK, and P38 kinase in primary cultures of neurons.
Neurochem Res.
26
2001
107
112
22
Dankbar
B
Padro
T
Leo
R
et al
Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma.
Blood.
95
2000
2630
2636

Author notes

Kenneth C. Anderson, Dana-Farber Cancer Institute, M557, 44 Binney St, Boston, MA 02115; e-mail:kenneth_anderson@dfci.harvard.edu.

Sign in via your Institution