Targeted disruption of signaling pathways essential for tumor cell survival and proliferation offer great promise for the design of more efficient and specific therapies. mTOR plays a central role in sensing mitogenic, nutrient and energy signals, and in the integration of different signaling pathways. Moreover, increasing evidence implicates mTOR in tumorigenesis and in the maintenance of transformed phenotypes. Therefore, we hypothesized that the mTOR pathway may play a significant role in T-cell acute lymphoblastic leukemia (ALL). BM and peripheral blood specimens from 15 pediatric patients were analyzed for the phosphorylation status of two downstream substrates of mTOR, S6 Ribosomal Protein and eIF4G. In all cases, primary leukemia T cells showed marked phosphorylation of S6 Ribosomal Protein (Ser235/236) and phosphorylation of the eukaryotic initiation factor eIF4G (Ser1108), showing that the mTOR pathway is constitutively active in T-ALL cells. Since leukemia T cells did not show significant basal phosphorylation of Akt/PKB, this observation suggests that the mTOR activation observed in T-ALL is Akt-independent. Specific blockade of mTOR by Rapamycin (100nM) reduced or abrogated phosphorylation of S6 ribosomal protein. Moreover, the mTOR blockers Rapamycin (10–100nM) and CCI-779 (10–100nM) significantly inhibited cell survival, and IL-7-promoted proliferation (40% to 85% inhibition) and cell cycle progression of T-ALL cells. Since we have shown previously that PI3K/Akt is critically involved in T-ALL responses to exogenous stimuli, we next evaluated whether inhibition of PI3K/Akt signals would synergize or potentiate the effects of mTOR blockade. We observed that the PI3K-inhibitor LY294002 (10μM) synergized with Rapamycin to abrogate IL-7-promoted leukemia T-cell proliferation. Finally, we explored the possibility that mTOR blockade can enhance the cytotoxic effects on T-ALL of the conventional drugs Dexamethasone (100nM) and Doxorubicin (100nM). Rapamycin potentiated the cytotoxic effects of both Dexamethasone (1.3 to 10 fold) and Doxorubicin (1.7 to 7.3 fold), suggesting that the combination of mTOR blockade with chemotherapy can improve the anti-leukemia efficacy of conventional regimens. In conclusion, we observed that the mTOR pathway is constitutively activated in primary leukemia T cells, and that the mTOR blockers Rapamycin and CCI-779 significantly inhibit T-ALL survival and proliferation. In addition, Rapamycin potentiates the inhibitory activity of other signaling inhibitors and the cytotoxic effects of the conventional drugs Dexamethasone and Doxorubicin. This study suggests that the mTOR pathway is a valid target for the treatment of T-ALL and lay the groundwork for the inclusion of mTOR blockade in the current therapeutic regimens.

Author notes

Corresponding author

Sign in via your Institution