scholarly journals Targeted inhibition of mTORC1 and mTORC2 by active-site mTOR inhibitors has cytotoxic effects in T-cell acute lymphoblastic leukemia

Leukemia ◽  
2011 ◽  
Vol 25 (5) ◽  
pp. 781-791 ◽  
Author(s):  
C Evangelisti ◽  
F Ricci ◽  
P Tazzari ◽  
G Tabellini ◽  
M Battistelli ◽  
...  
Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3242-3242
Author(s):  
Camilla Evangelisti ◽  
Francesca Chiarini ◽  
Francesca Ricci ◽  
Pierluigi Tazzari ◽  
Andrea Pession ◽  
...  

Abstract Abstract 3242 T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive form of leukemia characterized by uncontrolled proliferation of lymphoblasts committed to the T-cell lineage. Despite improvements in therapy, 25% of children and 50%–70% of adults with T-ALL relapse and develop a polychemotherapy-resistant disorder, which carries a poor prognosis. Thus, major efforts are being made to develop targeted molecules against deregulated signaling pathways that sustain T-ALL cell growth and survival. The mTOR serine/threonine kinase belongs to two separate multi-protein complexes, referred to as mTORC1 and mTORC2. The mTORC1 complex, which is inhibited by rapamycin/rapalogs, controls mRNA translation through the phosphorylation of the translation repressor 4E-BP1, while mTORC2 could be involved in regulating proliferation and survival through phosphorylation of Akt on Ser 473. mTORC2 is usually considered to be rapamycin-resistant. Both mTORC1 and mTORC2 are activated in most of T-ALL patients underscoring these complexes as major targets for T-ALL treatment. However, the allosteric inhibition of mTORC1 by rapamycin/rapalogs has only modest effects in T-ALL cells. This could be due, among other things, to the fact that rapamycin/rapalogs are mainly cytostatic and do not dephosphorylate 4E-BP1 in preclinical models of T-ALL. Recently, however, ATP-competitive inhibitors specific for the mTOR kinase active site have been synthesized. These compounds target both mTORC1 and mTORC2 and are more active than rapamycin/rapalogs against p-4E-BP1. Here, we have explored the therapeutic potential of two active-site mTOR inhibitors, PP-242 and OSI-027, against both T-ALL cell lines and primary samples from T-ALL patients displaying activation of mTORC1 and mTORC2. OSI-027 is being now tested in clinical trials in patients with solid malignancies. PP-242 and OSI-027 affected T-ALL cell line viability as documented by MTT assays. IC50 for PP-242 ranged from 0.15 to 0.50 μM at 24 hours, whereas OSI-027 was less potent (IC50 ranging from 0.6 to 1.3 μM at 48 h). Of note, both the drugs were effective against CEM T-ALL cells overexpressing 170-kDa P-glycoprotein, one of the major determinants of drug-resistance. Overall, PP-242 was slightly more powerful than the dual PI3K/mTOR inhibitor, PI-103, whose IC50 ranged from 0.2 to 1.0 μM. When T-ALL patient samples were examined, PP-242 displayed an IC50 of 0.5–1.0 μM, while the IC50 for OSI-027 ranged between 2.3 and 4.8 μM. Both the inhibitors affected to a much lower extent the proliferation of peripheral blood T-lymphocytes from healthy donors stimulated with phytohemagglutinin and interleukin-2. Remarkably, PP-242 targeted the side population of T-ALL cell lines, which might correspond to leukemic stem cells. The drugs induced both cell cycle arrest in G0/G1 phase and apoptosis, as documented by flow cytometric analysis, western blot analysis for cleaved caspase-3, and transmission electron microscopy. Western blot analysis demonstrated that the inhibitors induced a dose- and time-dependent dephosphorylation of Akt on Ser 473 (indicative of mTORC2 inhibition) as well as a dephosphorylation of mTORC1 downstream targets including Thr 389 p70S6 kinase, Ser 235/236 S6 ribosomal protein, Thr 37/46 4E-BP1, and Ser 209 eIF4E. Also a dephosphorylation of the Akt downstream target GSK3β on Ser 21/9 was observed. In contrast, rapamycin failed to dephosphorylate Thr 37/46 4E-BP1 and Ser 209 eIF4E. At variance with rapamycin, we also found a marked inhibition of mRNA translation in T-ALL cell lines treated with active-site mTOR inhibitors, as attested by a reduction in the incorporation of 3H-leucine and a shift from large to small polysomes.In MOLT-4 and Jurkat cells, the inhibitors, when used at subcytotoxic concentrations, strongly synergized with vincristine (combination index = 0.2–0.6), a traditional chemotherapeutic drug currently used for treating T-ALL patients, and with the Bcl-2 inhibitor, ABT-263 (combination index =0.15-0.19). In conclusion, both the inhibitors displayed a remarkable anti-leukemic activity through inhibition of mTORC1 and mTORC2 activity. The lower cytotoxicity against healthy T-lymphocytes ex-vivo suggests a favourable therapeutic index, which emphasizes the development of active-site mTOR inhibitors as clinical candidates for therapy in T-ALL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 445-445
Author(s):  
Kirsten Canté-Barrett ◽  
Jill AP Spijkers-Hagelstein ◽  
Jessica GCAM Buijs-Gladdines ◽  
Wilco K Smits ◽  
Rogier C Buijsman ◽  
...  

Abstract Background: Pediatric T-cell acute lymphoblastic leukemia patients frequently harbor mutations in IL7Ra or downstream molecules encoded by JAK1, JAK3, N-RAS, K-RAS, NF1, AKT, and PTEN. These mutated signaling molecules can contribute to leukemia by disturbing a multitude of cellular processes such as the cell cycle, epigenetics, apoptosis, or affecting other important signal transduction pathways. Aims: We aimed to determine the overall incidence of mutations in IL7Ra and downstream signaling components in a large cohort of pediatric T-ALL patients. In order to find better treatment options for patients with these mutations, we analyzed the effect of selected IL7Ra-pathway inhibitors-individually and in combinations-on downstream signaling and cytotoxicity in Ba/F3 cells expressing each of the mutations. Methods: We sequenced 146 pediatric T-ALL patient samples for mutations in the FERM, pseudokinase and kinase domains of the Janus kinase gene family (JAK1, JAK2, JAK3, TYK2) and hotspot regions of N-RAS and K-RAS. We adapted the IL3-dependent Ba/F3 cell line to express mutant or wild type genes upon induction by doxycycline and assessed cell viability and signaling in the absence of IL3. Various IL7Ra-pathway inhibitors were tested using this system, and the synergy of combined inhibitors was determined by comparing the dose-response curve of different ratios of IC50-based inhibitor concentrations to the curves for each of the single inhibitors. The Combination Index was calculated using Calcusyn™ software. Results: IL7Ra, JAK, RAS, AKT and PTEN mutations are present in approximately 45% of patients and occur in a predominantly mutually exclusive fashion, suggesting they share aberrant activation of similar downstream targets. We found JAK1, JAK3 and RAS mutations as previously reported, but also identified new JAK1 mutations including V427M, L624YPILKV, E668Q, P815S, and T901G. A novel three-dimensional model of JAK1 reveals that mutations in JAK molecules affect important amino acids that are involved in the interaction between the pseudokinase and kinase domains, facilitating constitutive kinase activity. In our doxycycline-inducible IL3-dependent Ba/F3 system, expression of mutant genes-in contrast to the wild type genes-transforms Ba/F3 cells by supporting IL3-independent growth through activation of the RAS-MEK-ERK and PI3K-AKT pathways. We used this system to test the sensitivity to pharmacological inhibitors; IL7Ra and JAK mutant Ba/F3 cells are sensitive to JAK inhibition, so JAK inhibitors such as ruxolitinib may offer therapeutic potential for IL7Ra, JAK1 or most JAK3 mutated T-ALL patients. The RAS and AKT mutants respond to RAS-MEK and PI3K-AKT-mTOR inhibition, respectively, but are-as expected-insensitive to JAK inhibition. Remarkably, IL7Ra and JAK mutants are relatively resistant to downstream RAS-MEK-ERK or PI3K-AKT-mTOR inhibition, indicating that inhibiting just one of these downstream pathways is insufficient. We provide evidence of (cross-)activation of the alternate pathway when one of these pathways is inhibited. Combined inhibition of MEK and PI3K/AKT synergistically prevents proliferation of the IL7Ra- and JAK-mutants by efficiently blocking both downstream signaling pathways. Furthermore, this combined inhibition is cytotoxic in two out of five tested primary T-ALL specimens. Summary/Conclusion: We show that the combined inhibition of MEK and PI3K/AKT leads to strong and synergistic cytotoxic effects in the IL7Ra and JAK mutants and efficiently blocks signaling downstream of both pathways. This inhibitor combination is effective in two out of five primary T-ALL samples. Therefore, the cytotoxic effects of synergistic MEK and PI3K/AKT inhibition should be further explored as a therapeutic option for (relapsed) ALL patients. Disclosures Buijsman: Netherlands Translational Research Center B.V.: Equity Ownership, Other: founder and shareholder. Zaman:Netherlands Translational Research Center B.V.: Equity Ownership, Other: founder and shareholder.


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Kehan Li ◽  
Cunte Chen ◽  
Rili Gao ◽  
Xibao Yu ◽  
Youxue Huang ◽  
...  

AbstractT-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of leukemia with poor prognosis, and biomarkers and novel therapeutic targets are urgently needed for this disease. Our previous studies have found that inhibition of the B-cell leukemia/lymphoma 11B (BCL11B) gene could significantly promote the apoptosis and growth retardation of T-ALL cells, but the molecular mechanism underlying this effect remains unclear. This study intends to investigate genes downstream of BCL11B and further explore its function in T-ALL cells. We found that PTK7 was a potential downstream target of BCL11B in T-ALL. Compared with the healthy individuals (HIs), PTK7 was overexpressed in T-ALL cells, and BCL11B expression was positively correlated with PTK7 expression. Importantly, BCL11B knockdown reduced PTK7 expression in T-ALL cells. Similar to the effects of BCL11B silencing, downregulation of PTK7 inhibited cell proliferation and induced apoptosis in Molt-4 cells via up-regulating the expression of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and p27. Altogether, our studies suggest that PTK7 is a potential downstream target of BCL11B, and downregulation of PTK7 expression via inhibition of the BCL11B pathway induces growth retardation and apoptosis in T-ALL cells.


2015 ◽  
Vol 208 (1-2) ◽  
pp. 52-53 ◽  
Author(s):  
Xiaolin Ma ◽  
Lijun Wen ◽  
Lili Wu ◽  
Qingrong Wang ◽  
Hong Yao ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document