scholarly journals 133P The mTOR kinase inhibitor AZD-2014 enhances the antitumor effects of XPO1 antagonist KPT-185 in mantle cell lymphoma

2015 ◽  
Vol 26 ◽  
pp. ix37
Author(s):  
K. Sekihara ◽  
S. Yamamoto ◽  
M. Kikkawa ◽  
H. Taka ◽  
N. Kaga ◽  
...  
Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 925-925 ◽  
Author(s):  
Yoko Tabe ◽  
Masako Harada ◽  
Yuka Miyamae ◽  
Hiromichi Matsushita ◽  
Kensuke Kojima ◽  
...  

Abstract Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma that frequently demonstrates chemoresistance. Since a number of signaling pathways are dysregulated in MCL, novel strategies for restoring multiple tumor suppressors and pathways are of considerable interest. Exportin 1 (XPO1/CRM1) mediates nuclear export of numerous molecules, including oncogenic transcription factors, ribosomal subunits, and RNAs, and is critical for cancer survival and proliferation. We previously reported that single-agent XPO1 antagonist KPT-185 exhibited antiproliferative and proapoptotic activities against MCL cells via inhibiting synthesis of proteins, such as chaperone proteins (HSP70), through ribosomal biogenesis and via nuclear export of transcription factors and oncogenic mRNAs, including cyclin D1, c-Myc, and PIM1 (Tabe et al. ASH 2013). Intriguingly, proteomic analysis detected significant upregulation of glycolysis and gluconeogenesis pathways in KPT-185–treated MCL cells. Aerobic glycolysis plays an important role in sustaining tumor metabolism and may negatively affect the antitumor activity of KPT-185. We therefore assessed the efficacy of combining this regulator of nucleocytoplasmic shuttling with an inhibitor of mTOR signaling, which is a central regulator of cell metabolism integrating nutrients, with KPT-185 targeting the altered metabolism. We first investigated the antitumor effects and molecular mechanisms of simultaneous treatment with KPT-185 and the ATP-competitive second-generation mTOR kinase inhibitor AZD-2014 in three MCL cell lines: JVM2, Jeko-1, and MINO (KPT-185 IC50 values: 92, 103 and 96 nM, respectively, at 48 h by MTT). AZD-2014 treatment resulted in downregulation of p-S6K and c-Myc and upregulation of p27KIP and cleaved caspase-9, which translated into concentration-dependent reduction of cell proliferation (IC50: JVM2, 247 nM; Jeko-1, 86 nM; MINO, 370 nM, at 48 h by MTT). The KPT-185/AZD-2014 combination inhibited cell growth (% of control absorbance; values given are for KPT-185 [100nM], AZD-2014 [100nM for JVM2 and MINO, 50nM for Jeko-1], and KPT-185/AZD-2014: JVM2 49.4±2.0, 61.6±3.7, 25.2±0.2; Jeko-1 46.7±4.8, 66.9±3.3, 28.6±3.4; MINO 55.0±5.6, 79.6±0.6, 21.6±2.5, at 48 h by MTT). We next investigated changes of protein expression and signaling pathways induced by AZD-2014 or the KPT-185/AZD-2014 combination (24 h) in Jeko-1 cells (KPT-100nM, AZD-2014 200nM). The proteomic technology of isobaric tags for relative and absolute quantitation (iTRAQ) demonstrated that AZD-2014 affected expression of 68 proteins (42 upregulated / 26 downregulated) and caused downregulation of fatty acid synthase expression (P<0.001). We also observed repression of importin-9, a transporter from cytoplasm to nucleus, by AZD-2014 (P<0.05) and of exportin-1, a transporter from nucleus to cytoplasm by KPT-185/AZD-2014 (P<0.001), indicating that the combination of KPT-185/AZD-2014 may disrupt bidirectional nucleocytoplasmic shuttling in MCL cells. We then performed comprehensive and quantitative analysis of charged metabolites by capillary electrophoresis mass spectrometry in Jeko-1 cells after treatment with KPT-185 or KPT-185/AZD-2014 (24 h) to detect differences in 52 polar metabolites (P<0.05). We observed that KPT-185–stimulated glutamate metabolism was effectively reversed by AZD-2014 (fold change of L-glutamic acid compared to control: KPT-185, 1.3; AZD-2014, 0.5; KPT185/AZD-2014, 0.8; P<0.001, contol vs KPT185/AZD-2014). AZD-2014 enhanced the repression of fatty acid synthesis by KPT-185 (0.3 fold) with significant downregulation of citric acid (0.3 fold, P<0.001). KPT-185/AZD-2014 combination further decreased succinic acid (0.04 fold, P<0.001, compared to control) and malic acid (0.1 fold, P<0.001), and both of these effects were associated with gluconeogenesis downregulation (Figure 1). Taken together, our findings suggest that inhibition of mTOR kinase enhances the antitumor effects of the XPO1 antagonist KPT-185 with effective repression of XPO1 blockage–induced glycolysis/gluconeogenesis upregulation and of fatty acid synthesis and with possible disruption of bidirectional nucleocytoplasmic shuttling in MCL cells. These findings suggest a novel, rationally designed combinatorial strategy targeting pro-survival metabolism in MCL. Figure 1 Figure 1. Disclosures Andreeff: Karyopharm: Research Funding.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1579-1579
Author(s):  
Weiming Xu ◽  
Christine Kang ◽  
Mercedes Delgado ◽  
Sophie M Perrin-Ninkovic ◽  
Patrick W Papa ◽  
...  

Abstract Mantle cell lymphoma (MCL) is a distinct sub-type non-Hodgkin lymphoma characterized by overexpression of cyclin D1 (CCND1) in 95% of patients due to the cytogenetic change of chromosome translocation t(11;14) (q13;q32). It remains one of the most challenging lymphomas associated with shorter response duration to conventional chemotherapy as well as continuous relapses and refractory to current drugs. However, dysregulation of cyclin D1 biology alone is insufficient to develop MCL. The emerging data suggest that the mammalian target of rapamycin (mTOR) plays a crucial role in the proper transmission of proliferative and anti-apoptotic signals through the PI3K/AKT pathway that makes it an attractive therapeutic target for hematological malignances including mantle cell lymphoma. As a single agent, rapamycin analogs such as temsirolimus (CCI-779) achieved 38% overall response rate in heavily pretreated MCL and prolonged progression free survival (PFS) in relapsed and refractory mantle cell lymphoma (4.8 months in temsirolimus vs. 1.9 months in investigator’s choice, ASCO 2008). mTOR regulates two distinct complexes TORC1 and TORC2. TORC1 complex is involved in cell cycle regulation by phosphorylating p70S6K and 4E-BP1, two molecules that are important for translational control of cyclin D1 and c-myc as well as ribosomal biogenesis whereas TORC2 complex mainly regulates phospho- AKT serine 473 leading to cell survival and proliferation. mTOR kinase also negatively regulates autophagy, a process of cellular bulk protein degradation by fusion to lysosomes upon the nutrient deprivation. We have developed mTOR kinase selective inhibitors which exhibit distinct biological profile from rapamycin in many cancer cell lines. Here we demonstrate that a selective mTOR kinase inhibitor displays potent anti-proliferative activity in JeKo-1 and Mino cells associated with decreased phosphorylation of S6, p70S6K, AKT S473, 4E-BP1 as well as decreased cyclin D1 levels leading to G1 arrest. The inhibitor also promotes autophagic cell death at 72h and 96h post-treatment. Furthermore a selective mTOR kinase inhibitor but not rapamycin induces a significant apoptosis in JeKo-1 and Mino cells. The observed apoptosis is correlated with caspases mediated PARP cleavage as well as inhibition of anti-apoptotic protein Mcl-1, suggesting TORC2/AKT S473 complex may provide survival signaling for mantle cell lymphoma. A timecourse study demonstrated that JeKo-1 and Mino cells undergo apoptosis at 24h and 48h followed by significant autophagic cell death at 72h and 96h in a dose dependent manner when exposed to our mTOR kinase inhibitor. In conclusion, mTOR kinase inhibitors are able to induce G1 cell cycle arrest, caspase-dependent apoptosis and autophagic cell death that contribute to the anti-tumor activity. Therefore it may provide a powerful alternative targeted therapy for mantle cell lymphoma.


Author(s):  
Nuchjira Takheaw ◽  
Gunya Sittithumcharee ◽  
Ryusho Kariya ◽  
Watchara Kasinrerk ◽  
Seiji Okada

Blood ◽  
2000 ◽  
Vol 95 (2) ◽  
pp. 619-626 ◽  
Author(s):  
Roberto Chiarle ◽  
Leo M. Budel ◽  
Jeffrey Skolnik ◽  
Glauco Frizzera ◽  
Marco Chilosi ◽  
...  

Mantle cell lymphoma (MCL) is an aggressive neoplasm characterized by the deregulated expression of cyclin D1 by t(11;14). The molecular mechanisms responsible for MCL's clinical behavior remain unclear. The authors have investigated the expression of p53, E2F-1, and the CDK inhibitors p27 and p21 in 110 MCLs, relating their expression to proliferative activity (Ki-67). For comparison, they have similarly analyzed low-grade (12 MALT, 16 CLL/SLL) and high-grade (19 DLCL) lymphomas. p53 was detected more frequently in large-cell MCL (l-MCL; 5 of 7) than in classical MCL (s-MCL; 13 of 103) and DLCL (8 of 19). In MCL and DLCL, the percentage of E2F-1+ nuclei was high, correlating with high Ki-67 expression. Most MCLs (91 of 112) and DLCLs (12 of 19) showed a loss of p27; MALT and CLL/SLL, however, were p27 positive. Reverse transcription–polymerase chain reaction and in vitro protein degradation assays demonstrated that MCLs have normal p27 mRNA expression but increased p27 protein degradation activity via the proteasome pathway. Correlation of MCL p53 and p27 expression with clinical data showed an association between reduced overall survival rates and the overexpression of p53 (P = .001), the loss of p27 (P = .002), or both. Loss of p27 identified patients with a worse clinical outcome among p53 negative cases (P = .002). These findings demonstrated that MCL has a distinct cell cycle protein expression similar to that of high-grade lymphoma. The loss of p27 and the overexpression of p53 in MCL are prognostic markers that identify patients at high risk. The demonstration that low levels of p27 in MCL result from enhanced proteasome-mediated degradation should encourage additional clinical trials. (Blood. 2000;95:619-626)


Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2120 ◽  
Author(s):  
Diana Malarikova ◽  
Adela Berkova ◽  
Ales Obr ◽  
Petra Blahovcova ◽  
Michael Svaton ◽  
...  

Mantle cell lymphoma (MCL) is a subtype of B-cell lymphoma with a large number of recurrent cytogenetic/molecular aberrations. Approximately 5–10% of patients do not respond to frontline immunochemotherapy. Despite many useful prognostic indexes, a reliable marker of chemoresistance is not available. We evaluated the prognostic impact of seven recurrent gene aberrations including tumor suppressor protein P53 (TP53) and cyclin dependent kinase inhibitor 2A (CDKN2A) in the cohort of 126 newly diagnosed consecutive MCL patients with bone marrow involvement ≥5% using fluorescent in-situ hybridization (FISH) and next-generation sequencing (NGS). In contrast to TP53, no pathologic mutations of CDKN2A were detected by NGS. CDKN2A deletions were found exclusively in the context of other gene aberrations suggesting it represents a later event (after translocation t(11;14) and aberrations of TP53, or ataxia telangiectasia mutated (ATM)). Concurrent deletion of CDKN2A and aberration of TP53 (deletion and/or mutation) represented the most significant predictor of short EFS (median 3 months) and OS (median 10 months). Concurrent aberration of TP53 and CDKN2A is a new, simple, and relevant index of chemoresistance in MCL. Patients with concurrent aberration of TP53 and CDKN2A should be offered innovative anti-lymphoma therapy and upfront consolidation with allogeneic stem cell transplantation.


Sign in / Sign up

Export Citation Format

Share Document