IL-7R is essential for leukemia-initiating cell activity of T-cell acute lymphoblastic leukemia

S González-García, M Mosquera… - Blood, The Journal …, 2019 - ashpublications.org
S González-García, M Mosquera, P Fuentes, T Palumbo, A Escudero, A Pérez-Martínez
Blood, The Journal of the American Society of Hematology, 2019ashpublications.org
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy
resulting from the dysregulation of signaling pathways that control intrathymic T-cell
development. Relapse rates are still significant, and prognosis is particularly bleak for
relapsed patients. Therefore, development of novel therapies specifically targeting pathways
controlling leukemia-initiating cell (LIC) activity is mandatory for fighting refractory T-ALL.
The interleukin-7 receptor (IL-7R) is a crucial T-cell developmental pathway that is …
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy resulting from the dysregulation of signaling pathways that control intrathymic T-cell development. Relapse rates are still significant, and prognosis is particularly bleak for relapsed patients. Therefore, development of novel therapies specifically targeting pathways controlling leukemia-initiating cell (LIC) activity is mandatory for fighting refractory T-ALL. The interleukin-7 receptor (IL-7R) is a crucial T-cell developmental pathway that is commonly expressed in T-ALL and has been implicated in leukemia progression; however, the significance of IL-7R/IL-7 signaling in T-ALL pathogenesis and its contribution to disease relapse remain unknown. To directly explore whether IL-7R targeting may be therapeutically efficient against T-ALL relapse, we focused on a known Notch1-induced T-ALL model, because a majority of T-ALL patients harbor activating mutations in NOTCH1, which is a transcriptional regulator of IL-7R expression. Using loss-of-function approaches, we show that Il7r-deficient, but not wild-type, mouse hematopoietic progenitors transduced with constitutively active Notch1 failed to generate leukemia upon transplantation into immunodeficient mice, thus providing formal evidence that IL-7R function is essential for Notch1-induced T-cell leukemogenesis. Moreover, we demonstrate that IL-7R expression is an early functional biomarker of T-ALL cells with LIC potential and report that impaired IL-7R signaling hampers engraftment and progression of patient-derived T-ALL xenografts. Notably, we show that IL-7R–dependent LIC activity and leukemia progression can be extended to human B-cell acute lymphoblastic leukemia (B-ALL). These results have important therapeutic implications, highlighting the relevance that targeting normal IL-7R signaling may have in future therapeutic interventions, particularly for preventing T-ALL (and B-ALL) relapse.
ashpublications.org