Multiple myeloma (MM) is a highly molecularly heterogeneous hematologic malignancy and remains mostly incurable despite recent improvement of treatment outcome by novel agents. Therefore, the identification of novel and universal targetable therapeutic molecules is a core component for the therapeutic development. Herein, we identified that 3-phosphoinositide-dependent protein kinase 1 (PDPK1), a member of serine threonine kinase, is a rationalistic candidate of a novel therapeutic target against MM. PDPK1 is universally phosphorylated in all eleven MM-derived cell lines examined regardless of type of cytogenetic/genetic abnormalities or the activation/mutation state of FGFR3, RAS, ERK and AKT. PDPK1 promotes the cell proliferation of myeloma cells by activating RSK2Ser227 at N-terminal kinase domain which is a pivotal regulator of molecules that are essential for myelomagenesis, such as c-MYC, IRF4, or Cyclin Ds (Shimura Y, Mol Cancer Ther 2011), and AKTThr308. In contrast, PDPK1 inhibition by a selective inhibitor, BX-912, caused G2/M arrest, which was accompanied by the inactivation of PLK1, and resulted in cell death via induction of apoptosis which was accompanied by the activation of pro-apoptotic BH3-only proteins, BIM and BAD, in myeloma-derived cell lines. These molecular and cytological effects of PDPK1 inhibition in myeloma cells were also validated by gene knockdown by means of RNA interference using two different siRNAs specific for PDPK1. In addition, the cytotoxic effect of BX-912 was not hampered in two cell lines acquiring resistance to bortezomib (BTZ) (Ri M, Leukemia 2010), and PDPK1 inhibition by BX-912 showed additive to synergistic in vitro cytotoxic effects on myeloma cells with melphalan, etoposide, bortezomib or BAY11-7085, an inhibitor for NF-κB. BX-912 also induced cell death in eleven patient-derived primary myeloma cells those were positively isolated by CD138-labelling from bone marrow aspirates, while normal peripheral lymphocytes were significantly less sensitive to PDPK1 inhibitor compared with MM cells. In the clinical setting, PDPK1 was active in myeloma cells of 57 of 65 (87.7%) symptomatic MM patients at diagnosis. While patients backgrounds, such as age, gender, and the type of M-protein, were not significantly different between PDPK1-negative (PDPK1(-)) and PDPK1-positive (PDPK1(+)) patients, patients with the disease stage III according to International Staging System were significantly more frequent in the PDPK1(+) cohort compared with PDPK1(-) cohort. The PDPK1(-) patients tended to exhibit longer overall survival (OS) than the patients with PDPK1(+) (median OS: 2925 days vs. 2155 days, p=0.069) with a median follow-up period of 1310 days (range: 228–3317 days). Of particular, the 8-year OS of PDPK1(-) patients was statistically significantly more favorable than those of PDPK1(+) patients (83% vs. 17%, p=0.041). In addition, when we analyzed the impact of PDPK1 activity on the treatment outcome of patients treated by BTZ and dexamethasone therapy (BD), our results revealed that progression free survival (PFS) of patients with PDPK1(-) was a significantly longer than those of patients with PDPK1(+) (median PFS: PDPK1(-) vs. PDPK1(+), not reached vs. 167 days, p=0.049). The PFS of PDPK1(-) patients treated by high-dose therapy/autologous stem cell transplantation (HDT/ASCT) tended to be longer than those of PDPK1(+) (median PFS: PDPK1(-) vs. PDPK1(+), 972 days vs. 567 days, p=0.125). In conclusion, our study provides the rationale for PDPK1 as a possible universal molecular target for MM with various molecular/cytogenetic features. Especially, PDPK1 is a potential therapeutic target for not only newly diagnosed patients but also patients who are resistant or refractory to currently available anti-myeloma therapies. This study was conducted in accordance with the Declaration of Helsinki and with the approval of the Institutional Review Board. Patient-derived samples were obtained with informed consent.

Disclosures

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution