MTOR Inhibitor MLN0128 has Antitumor Efficacy in Cell Lines With Intrinsic and Acquired Rapamycin-Resistance.

2013 ◽  
Vol 179 (2) ◽  
pp. 313
Author(s):  
B. Hassan ◽  
A. Akcakanat ◽  
S. Takafumi ◽  
K. Evans ◽  
F. Adkins ◽  
...  
2013 ◽  
Author(s):  
Burhan Hassan ◽  
Argun Akcakanat ◽  
Takafumi Sangai ◽  
Farrell Adkins ◽  
Kurt Evans ◽  
...  

Digestion ◽  
2006 ◽  
Vol 74 (1) ◽  
pp. 19-27 ◽  
Author(s):  
Jinyu Gu ◽  
Hirofumi Yamamoto ◽  
Xueying Lu ◽  
Chew Yee Ngan ◽  
Tadashi Tsujino ◽  
...  

2018 ◽  
Vol 115 (16) ◽  
pp. 4206-4211 ◽  
Author(s):  
Amanda J. DuBose ◽  
Stephen T. Lichtenstein ◽  
Noreen M. Petrash ◽  
Michael R. Erdos ◽  
Leslie B. Gordon ◽  
...  

LMNA encodes the A-type lamins that are part of the nuclear scaffold. Mutations in LMNA can cause a variety of disorders called laminopathies, including Hutchinson-Gilford progeria syndrome (HGPS), atypical Werner syndrome, and Emery-Dreifuss muscular dystrophy. Previous work has shown that treatment of HGPS cells with the mTOR inhibitor rapamycin or with the rapamycin analog everolimus corrects several of the phenotypes seen at the cellular level—at least in part by increasing autophagy and reducing the amount of progerin, the toxic form of lamin A that is overproduced in HGPS patients. Since other laminopathies also result in production of abnormal and potentially toxic lamin proteins, we hypothesized that everolimus would also be beneficial in those disorders. To test this, we applied everolimus to fibroblast cell lines from six laminopathy patients, each with a different mutation in LMNA. Everolimus treatment increased proliferative ability and delayed senescence in all cell lines. In several cell lines, we observed that with treatment, there is a significant improvement in nuclear blebbing, which is a cellular hallmark of HGPS and other lamin disorders. These preclinical results suggest that everolimus might have clinical benefit for multiple laminopathy syndromes.


2015 ◽  
Author(s):  
Susan Heavey ◽  
Paul Dowling ◽  
Sinéad Toomey ◽  
Aoife Carr ◽  
Bryan Hennessy ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2025-2025
Author(s):  
Francesca Chiarini ◽  
Cecilia Grimaldi ◽  
Francesca Ricci ◽  
Pierluigi Tazzari ◽  
Camilla Evangelisti ◽  
...  

Abstract Abstract 2025 Poster Board II-2 Introduction: Recent findings have highlighted that constitutively active phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian Target of Rapamycin (mTOR) signaling is a common feature of T-cell acute lymphoblastic leukemia (T-ALL) where it strongly influences cell proliferation and survival. Pathway activation could be due to several reasons which include Notch1 activation leading to HES1-mediated transcriptional suppression of PTEN gene, PTEN phosphorylation or oxidation, and inactivation of SHIP1 phosphatase. These findings lend compelling weight for the application of PI3K/Akt/mTOR inhibitors in T-ALL. Rapamycin and its analogues have shown some promising effects in pre-clinical models of T-ALL. However, mTOR inhibitors are mainly cytostatic and could hyperactivate Akt due to the existence of feedback loops between mTOR, p70 S6 kinase, PI3K, and Akt. Recently, dual PI3K/mTOR inhibitors have been synthesized. Here, we have analyzed the therapeutic potential of the novel, dual PI3K/mTOR inhibitor, NVP-BEZ235, an orally bioavailable imidazoquinoline derivative, which has entered clinical trials for solid tumors, on both T-ALL cell lines and patient samples. Methods and Patients: We employed a panel of cell lines with up-regulated PI3K/Akt/mTOR signaling, including CEM-R cells [which overexpress high levels of the membrane transporter, 170-kDa P-glycoprotein (P-gp)], MOLT-4 and CEM-S cells (which lack PTEN expression), Jurkat cells (which do not express both PTEN and SHIP1), and RPMI-8402 and BE-13 cells. MOLT-4, CEM, and Jurkat cells have a non-functional p53 pathway. Moreover, both Jurkat and MOLT-4 cells have aberrant Notch1 signaling. Patients samples displayed pathway activation as documented by increased levels of p-Akt, p-4E-BP1, and p-S6 ribosomal protein, as well as low/absent PTEN expression. Results: NVP-BEZ235 was cytotoxic to the panel of cell lines as documented by MTT assays. NVP-BEZ235 IC50 ranged from 80 to 280 nM at 24 h. A comparison between NVP-BEZ235 and the dual PI3K/mTOR inhibitor PI-103, a small synthetic molecule of the pyridofuropyrimidine class with the same targets, demonstrated that NVP-BEZ235 was more effective than PI-103 when employed at equimolar concentrations. NVP-BEZ235 did not significantly affect the proliferation of peripheral blood T-lymphocytes from healthy donors stimulated with phytohemagglutinin and interleukin-2, whereas it blocked leukemic cells in the G1 phase of the cell cycle, and this was accompanied by decreased levels of phosphorylated Retinoblastoma protein. NVP-BEZ235 treatment also resulted in apoptotic cell death (about 20-30% at 6 h of exposure, when employed at 200 nM), as documented by Annexin V/propidium iodide staining and cytofluorimetric analysis. Moreover, NVP-BEZ235 activated caspase-8 and caspase-3, as demonstrated by western blot. Western blot documented a dose- and time-dependent dephosphorylation of Akt and its downstream target, GSK-3β, in response to NVP-BEZ235. mTOR downstream targets were also efficiently dephosphorylated, including p70S6 kinase, S6 ribosomal protein, and 4E-BP1. Remarkably, NVP-BEZ235 targeted the side population (SP, identified by Hoechst 33342 staining and ABCG2 expression) of T-ALL cell lines, which might correspond to leukemia initiating cells, and synergized with several chemotherapeutic agents (dexamethasone, vincristine, cyclophosphamide, Ara-C) currently employed for treating T-ALL patients. NVP-BEZ235 reduced chemoresistance to vincristine induced in Jurkat cells by co-culturing with MS-5 stromal cells which mimic the bone marrow microenvironment. NVP-BEZ235 was cytotoxic (IC50: 10-15 nM at 96 h) to primary lymphoblasts from patients with T-ALL, where the drug dephosphorylated 4E-BP1, at variance with rapamycin. Of note, NVP-BEZ235 targeted the SP also in T-ALL patient samples. Conclusions: NVP-BEZ235 was cytotoxic to T-ALL cell lines and patient lymphoblasts (including SP cells) at concentrations that have been previously reported to be achievable in vivo. Taken together, our findings indicate that longitudinal inhibition at two nodes of the PI3K/Akt/mTOR network with NVP-BEZ235, either alone or in combination with other drugs, may serve as an efficient treatment towards T-ALL cells (including those overexpressing P-gp and independently from p53 status) which require upregulation of this signaling pathway for their survival and growth. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 413-413
Author(s):  
Alexander E Perl ◽  
Kasner Margaret ◽  
Shank Doris ◽  
Selina Luger ◽  
Martin Carroll

Abstract Abstract 413 The mammalian target of rapamycin (mTOR) is an emerging molecular target in cancer therapy, however the relationship between target activation in individual tumors as well as inhibition of target and clinical response is poorly defined. This is in large part due to the difficulty of real-time monitoring of mTOR activation in patient samples. mTOR is activated in acute myelogneous leukemia (AML) cells and the mTOR inhibitor rapamycin enhances cytotoxic effects of chemotherapy in vitro and in vivo. We therefore developed a new application of a recently-developed, whole blood fixation/permeabilization technique for intracellular flow cytometry (Chow, et al. Cytometry A 2005). Using this approach, we sought to serially monitor S6 ribosomal protein (S6) phosphorylation in peripheral blood leukemic blasts during clinical trials of mTOR inhibitors. S6 is a known target of mTOR and its phosphorylation is a surrogate marker for mTOR kinase activation. We applied this methodology during a recent pilot trial in which an oral mTOR inhibitor, sirolimus (rapamycin), was administered in sequence with intensive combination chemotherapy (mitoxantrone, etoposide, and cytarabine, or MEC) in patients with relapsed, refractory, or secondary AML. The whole blood fixation process sufficiently preserved surface epitopes and light scatter properties for immunophenotyping, allowing specific signaling analysis of leukemic blasts as well as non-malignant cell populations. Importantly, even leukopenic trial samples containing as few as 20% blasts provided robust signaling data in malignant cells. S6 phosphorylation was readily apparent in leukemic blasts prior to therapy and, consistent with prior reports, occurred only in a subset of blasts. Exposing aliquots of pre-treatment whole blood samples to increasing concentrations of rapamycin ex vivo determined that leukemic blasts from most samples showed inhibition of S6 phosphorylation at clinically achievable concentrations (between 10-20 nM). Notably, some subjects' leukemic blasts showed no inhibition to >50 nM rapamycin, which far exceeded trough concentrations measured on our studies. To examine rapamycin's in vivo biochemical effects, we performed a paired analysis of clinical samples drawn at study entry and after 72 hours of oral sirolimus. 10 subjects provided paired samples, of which 2 did not show baseline S6 phosphorylation, 6 showed baseline S6 phosphorylation that inhibited during therapy, and 2 showed baseline S6 phosphorylation but no inhibition. Trough rapamycin levels were similar among rapamycin responsive and resistant subjects. Considering the 6 subjects with in vivo mTOR inhibition, 3 subjects achieved complete or partial remissions from the regimen. Neither subject with in vivo rapamycin resistance had a clinical response. Overall, we conclude that effective inhibition of mTOR signaling in AML blasts occurs in the majority of subjects during sirolimus treatment at the dose studied. However, cell-intrinsic rapamycin resistance occurs in a minority of patients and requires further study to clarify its mechanism and effects upon concurrent chemotherapy response. These data demonstrate the feasibility of real-time, intra-tumoral pharmacodynamic monitoring of S6 phosphorylation by flow cytometry during clinical trials combining intensive chemotherapy and signal transduction inhibitors for leukemia. Our approach greatly clarifies pharmacokinetic/pharmacodynamic relationships and has broad application to pre-clinical and clinical testing of drugs whose direct or downstream effects disrupt PI3K/AKT/mTOR signaling. Such compounds include inhibitors of FLT3, c-KIT, BCR-ABL, JAK2, and ras/raf/MAPK. Multicenter/cooperative group phase II testing of sirolimus plus MEC in AML has been initiated to establish the regimen's response rate and test the extent to which our pharmacodynamic studies predict clinical response. Disclosures: Off Label Use: The use of sirolimus in the therapy of AML is investigational and off-label. Carroll:Cephalon consultancy: Consultancy; Sanofi Aventis Corporation: Research Funding; Kyowa Hakko Kirin Pharmaceutical: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 436-436 ◽  
Author(s):  
Adam M. Petrich ◽  
Violetta V. Leshchenko ◽  
Pei-Yu Kuo ◽  
B. Hilda Ye ◽  
Joseph A Sparano ◽  
...  

Abstract Abstract 436 mTOR inhibitors have been used with clinical success in solid tumors and non-Hodgkin lymphoma (NHL), and are attractive therapeutic options for DLBCL (diffuse large B-cell lymphoma, which has been shown to have constitutively active mTOR signaling). However, resistance to this class of agents remains problematic, and mechanisms of resistance are poorly understood. We performed candidate drug discovery using connectivity mapping and global gene expression profiling (GEP) to understand the pathways and genes responsible for resistance to the mTOR inhibitor Rapamycin (Sirolimus), which is the active metabolite of several clinically available mTOR inhibitors (eg, Temsirolimus, Everolimus). Treatment of DLBCL cell lines by Rapamycin at varying doses permitted stratification of cell lines into 2 groups of 3 cell lines each: sensitive (SU-DHL6, WSU-NHL, and Karpas-422) and resistant (SU-DHL4, OCI-Ly19, and Farage). Using the Affymetrix Human Gene 1.0 ST Array, we generated a profile of 1164 differentially-expressed genes (P<0.01) in the resistant cell lines. Pathway analysis of this particular gene expression signature enriched most strongly for the networks “EIF2 signaling” and “Regulation of eIF4 and p70S6K,” both of which are known to be involved in the PI3K and mTOR/AKT pathway. The genes thus identified present novel opportunities to understand and overcome resistance to mTOR inhibitors in DLBCL and other cancers. The Connectivity Map (CMAP) database contains a reference collection of more than 7,000 expression profiles from cultured human cells treated with bioactive small molecules, together with pattern-matching software to mine these data. We next analyzed the differentially-expressed genes associated with mTOR inhibitor resistance with the CMAP database in order to identify compounds likely to reverse the profile associated with resistance. From over 6,000 agents, the top 2% (by connectivity score) contained two PI3K inhibitors (Wortmannin and LY-294002), the protease inhibitor Saquinavir, and multiple HDAC inhibitors (including both Vorinostat and Trichostatin-A in the top 40 drugs). Among protease inhibitors, Nelfinavir (and to a lesser extent Saquinavir), has been shown to have potent cytotoxicity in a variety of solid tumors, by inhibition of the AKT signaling pathway. To validate the hypothesis that modulation of AKT might help overcome mTOR inhibitor resistance, we targeted AKT with two agents: Nelfinavir and MK-2206. We found that Nelfinavir demonstrated significant cytotoxicity at clinically achievable levels in all DLBCL cell lines tested (including those resistant to Rapamycin), and inhibited phosphorylation of AKT and downstream proteins (including p70S6 kinase; S6 ribosomal protein; 4-EBP-1) in a dose-dependent fashion. Baseline total AKT and phosphorylated AKT levels correlated with degree of sensitivity to Nelfinavir. Inhibition of downstream mTOR signaling by Rapamycin synergized with Nelfinavir in cell kill and inhibition of cell cycle progression. MK-2206, an AKT inhibitor which has shown success in early-phase clinical trials, was evaluated in the same panel of cell lines and likewise demonstrated synergism with Rapamycin in cytotoxicity and cell cycle inhibition. The degree of synergism between Rapamycin and either Nelfinavir and MK-2206, as calculated using the Chou-Talalay equation, was comparable. We have also demonstrated synergy between Nelfinavir and doxorubicin, a key component in commonly utilized regimens for AIDS lymphoma patients such as CHOP (Cytoxan, Adriamycin, Oncovin and Prednisone). We are now validating these in vitro results in a mouse xenograft model of DLBCL. In conclusion, our study demonstrates that AKT inhibition by Nelfinavir results in potent cytotoxicity in DLBCL cell lines at clinically relevant doses. Our results may have implications for combination therapy beyond NHL in non-hematologic malignancies where mTOR inhibitors and MK-2206 are being used independently with clinical success. Furthermore, the synergistic combination of either Nelfinavir or MK-2206, along with Rapamycin, may permit use of lower doses of each drug to therapeutically inhibit mTOR/AKT signaling while potentially reducing toxicity from off-target effects from the individual drugs. Finally, the use of Nelfinavir has particular relevance in AIDS patients with DLBCL, where the drug has both anti-viral and anti-lymphoma potential. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3017-3017
Author(s):  
Chiara Tarantelli ◽  
Eugenio Gaudio ◽  
Petra Hillmann ◽  
Filippo Spriano ◽  
Ivo Kwee ◽  
...  

Abstract Background. The PI3K/AKT/mTOR pathway is an important therapeutic target in lymphomas. PQR309 is a dual PI3K/mTOR inhibitor that has shown in vitroanti-lymphoma activity (Tarantelli et al, ASH2015) and is in phase 2 trial (NCT02249429, , NCT02723877, NCT02669511). PQR620 is a novel mTORC1/2 inhibitor that has shown preclinical activity in solid tumor models (Beaufils et al, AACR 2016). Here, we present the in vitro and in vivo anti-lymphoma activity of PQR620 as single agent and also the in vivo results of PQR620 or PQR309 containing combinations with the BCL2 inhibitor venetoclax. Materials and Methods. The drug concentration causing 50% inhibition of cell proliferation (IC50) was obtained in lymphoma cell lines [diffuse large B cell lymphoma (DLBCL), no.=26; mantle cell lymphoma (MCL), no.=8; anaplastic large T-cell lymphoma, no.=5; others, no=5] exposed to increasing doses of PQR620 for 72h using a Tecan D300e Digital Dispenser on 384well plates. For in vivo experiments, NOD-Scid (NOD.CB17-Prkdcscid/J) mice were subcutaneously inoculated with 10 x106 (RIVA) or with 5 x106(SU-DHL-6) cells. Results. PQR620 had a median IC50 of 250 nM (95%CI, 200-269 nM) when tested on 44 lymphoma cell lines. Activity was higher in B cell (no.=36) than in T cell tumors (no.=8) (median IC50s: 250 nM vs 450 nM; P=0.002). At 72h, anti-tumor activityof PQR620 was mostly cytostatic and apoptosis induction was seen only in 6/44 cell lines (13%), Sensitivity to PQR620 or apoptosis induction did not differ between DLBCL and MCL, and they were not affected by the DLBCL cell of origin, by TP53 status or by the presence of MYC or BCL2 translocations. The activity of PQR620 as single agent underwent in vivo evaluation in two DLBCL models, the germinal center B cell type DLBCL (GCB-DLBCL) SU-DHL-6 and the acivated B cell-like DLBCL (ABC-DLBCL) RIVA. Treatments with PQR620 (100mg/kg dose per day, Qdx7/w) started with 100-150 mm3 tumors and were carried for 14 (SU-DHL-6) or 21 days (RIVA). In both models, PQR620 determined a 2-fold decrease of the tumor volumes in comparison with control, with significant differences in both SU-DHL-6 (D7, D9, D11, D14; P < 0.005) and RIVA (D14, D16, D19, D21; P < 0.005). Based on the previously reported synergy between the dual PI3K/mTOR inhibitor PQR309 and venetoclax (Tarantelli et al, ASH 2015), we evaluated the combination of the PQR620 or PQR309 with the BCL2 inhibitor venetoclax (100 mg/kg, Qdx7/w) in the SU-DHL-6 model. Both the venetoclax combination with the dual PI3K/mTOR inhibitor and the venetoclax combination with mTORC1/2 inhibitor were superior to the compounds given as single agents, leading to the eradication of the xenografts. The combination of PQR620 with venetoclax showed highly significant differences either versus control or single agents during all days of the experiment (D4, D7, D9, D11, D14; P < 0.001). Similarly, the combination of PQR309 with venetoclax showed highly significant differences versus venetoclax (D7, D9, D11, D14; P < 0.001) and PQR309 (D7, D9, D11; P < 0.005) alone. Conclusions. The novel mTORC1/2 inhibitor PQR620 had in vitro and in vivo anti-lymphoma activity as single agent. In vivo experiments showed that both PQR620 and the dual PI3K/mTOR inhibitor PQR309 can strongly benefit from the combination with the BCL2 inhibitor venetoclax. Disclosures Hillmann: PIQUR Therapeutics AG: Employment. Fabbro:PIQUR Therapeutics AG: Employment. Cmiljanovic:PIQUR Therapeutics AG: Employment, Membership on an entity's Board of Directors or advisory committees.


Sign in / Sign up

Export Citation Format

Share Document