EHA Library - The official digital education library of European Hematology Association (EHA)

MEK INHIBITION IS A PROMISING THERAPEUTIC STRATEGY FOR MLL-REARRANGED INFANT ALL PATIENTS CARRYING RAS MUTATIONS
Author(s): ,
Mark JB Kerstjens
Affiliations:
Pediatric Oncology/Hematology,Erasmus MC - Sophia Children's Hospital,Rotterdam,Netherlands
,
Emma MC Driessen
Affiliations:
Pediatric Oncology/Hematology,Erasmus MC - Sophia Children's Hospital,Rotterdam,Netherlands
,
Merel Willekes
Affiliations:
Pediatric Oncology/Hematology,Erasmus MC - Sophia Children's Hospital,Rotterdam,Netherlands
,
Sandra Mimoso Pinhancos
Affiliations:
Pediatric Oncology/Hematology,Erasmus MC - Sophia Children's Hospital,Rotterdam,Netherlands
,
Rob Pieters
Affiliations:
Princess Maxima Center for Pediatric Oncology,Utrecht,Netherlands
Ronald W Stam
Affiliations:
Pediatric Oncology/Hematology,Erasmus MC - Sophia Children's Hospital,Rotterdam,Netherlands
(Abstract release date: 05/21/15) EHA Library. Kerstjens M. 06/14/15; 103095; S823 Disclosure(s): Erasmus MC - Sophia Children's Hospital
Pediatric Oncology/Hematology
Mark Kerstjens
Mark Kerstjens
Contributions
Abstract
Abstract: S823

Type: Oral Presentation + travel grant

Presentation during EHA20: From 14.06.2015 08:45 to 14.06.2015 09:00

Location: Room Stolz 1

Background

Acute Lymphoblastic Leukemia (ALL) in infants is characterized by a high incidence (~80%) of chromosomal rearrangements of the Mixed Lineage Leukemia (MLL) gene. MLL-rearranged (MLL-r) infant ALL patients are challenged by a very poor prognosis (i.e. 30-40% 5-year EFS), hence the need for better risk stratification and improved therapeutic solutions is evident. We recently screened a relatively large cohort (n=109) infant ALL patients (all enrolled in INTERFANT treatment protocols) for the presence of KRAS and NRAS mutations and found that the incidence of such mutations ranges between 14-24%, depending on the type of MLL translocation. Moreover, these mutations were found to represent independent predictors of exceedingly poor prognosis; all RAS mutation-positive MLL-r infant ALL patients deceased within 3 years from diagnosis.



Aims

We aimed to identify a therapeutic strategy to improve the prognosis of MLL-r infant ALL patients carrying RAS mutations.



Methods
8 small molecule inhibitors against different RAS-pathway components were selected and tested for anti-leukemic activity against the MLL-r ALL cell lines SEM and RS4;11 (RASwt) and KOPN8 (RASmut) using MTS cell viability assays. Next, primary infant ALL samples (n=20) all carrying MLL translocation t(4;11) (giving rise to the MLL-AF4 fusion protein) either with (n=6) or without (n=14) RAS mutations were exposed to these inhibitors in MTT cytotoxicity assays. In addition, we assessed the RAS activity in RAS mutated and wild-type MLL-r infant ALL cells, and analysed downstream MEK and ERK activation both in the absence and presence of the MEK inhibitors. Furthermore, combination treatment of MEK inhibitors and prednisolone, the spearhead drug for ALL treatment regimes, was investigated in SEM and KOPN8 using the MTS assay.

Results
We found that the MEK inhibitors MEK162, Selumetinib, and Trametinib effectively reduced the viability of KOPN8 cells (RASmut), whereas SEM and RS4;11 cells (RASwt) largely remained unaffected. In line with this, MLL-AF4+ infant ALL patient samples carrying RAS mutations  were significantly more sensitive to these MEK inhibitors when compared with patients carrying wild-type RAS genes: LC50 values for MEK162 were 0.04 vs. 26.9 µM (p<0.01), for Selumetinib 0.04 and 23.7 µM (p<0.01), and for Trametinib 0.01 vs. 26.5 µM (p<0.01), respectively. Furthermore, the presence of RAS mutations in primary MLL-r infant ALL samples was associated with significantly increased RAS activity, as determined by immunoprecipitation of GTP-bound RAS. Remarkably, however, enhanced RAS activation was not demonstrated by increased downstream ERK phosphorylation, while a slight increase of MEK phosphorylation was observed. Yet, MEK inhibitor exposure in both KOPN8 and SEM cells resulted in nearly complete abrogation of ERK phosphorylation, without affecting total ERK protein levels, suggesting that the loss of ERK activation plays an important role in the observed anti-leukemic effects. Furthermore, MEK162 and Selumetinib, seemed to induce accumulation of phosphorylated MEK. Additionally, MEK inhibitor treatment in both SEM and KOPN8 synergistically enhanced sensitivity towards prednisolone. Interestingly, a subgroup of patient samples (n=5) with wild-type RAS also showed sensitivity towards MEK inhibition, similar to the primary cells with RAS mutations. However, this observation could not be explained by increased RAS activation, nor by the phosphorylation levels of either MEK or ERK.

Summary
Our data show that MEK inhibition represents a promising therapeutic approach for MLL-r infant ALL patients carrying additional RAS mutations. Furthermore, the mechanism of action provoked by these MEK inhibitors seems to involve abrogation of ERK phosphorylation, but the initial levels of ERK phosphorylation did not correlate with MEK inhibitor sensitivity, and has no predictive value. Moreover, the prednisolone sensitization induced by MEK inhibitor exposure, further illustrates the potential value of MEK inhibition for the treatment of MLL-r infant ALL. Currently we are in the process of testing the efficacy of the above mentioned MEK inhibitors in vivo using a xenograft mouse model, while further elucidation of the molecular mechanisms underlying the anti-leukemic effects of these inhibitors in MLL-r ALL cells are in progress.

Keyword(s): Acute lymphoblastic leukemia, Drug sensitivity, MLL, Ras

Session topic: Towards targeted therapy in ALL
Abstract: S823

Type: Oral Presentation + travel grant

Presentation during EHA20: From 14.06.2015 08:45 to 14.06.2015 09:00

Location: Room Stolz 1

Background

Acute Lymphoblastic Leukemia (ALL) in infants is characterized by a high incidence (~80%) of chromosomal rearrangements of the Mixed Lineage Leukemia (MLL) gene. MLL-rearranged (MLL-r) infant ALL patients are challenged by a very poor prognosis (i.e. 30-40% 5-year EFS), hence the need for better risk stratification and improved therapeutic solutions is evident. We recently screened a relatively large cohort (n=109) infant ALL patients (all enrolled in INTERFANT treatment protocols) for the presence of KRAS and NRAS mutations and found that the incidence of such mutations ranges between 14-24%, depending on the type of MLL translocation. Moreover, these mutations were found to represent independent predictors of exceedingly poor prognosis; all RAS mutation-positive MLL-r infant ALL patients deceased within 3 years from diagnosis.



Aims

We aimed to identify a therapeutic strategy to improve the prognosis of MLL-r infant ALL patients carrying RAS mutations.



Methods
8 small molecule inhibitors against different RAS-pathway components were selected and tested for anti-leukemic activity against the MLL-r ALL cell lines SEM and RS4;11 (RASwt) and KOPN8 (RASmut) using MTS cell viability assays. Next, primary infant ALL samples (n=20) all carrying MLL translocation t(4;11) (giving rise to the MLL-AF4 fusion protein) either with (n=6) or without (n=14) RAS mutations were exposed to these inhibitors in MTT cytotoxicity assays. In addition, we assessed the RAS activity in RAS mutated and wild-type MLL-r infant ALL cells, and analysed downstream MEK and ERK activation both in the absence and presence of the MEK inhibitors. Furthermore, combination treatment of MEK inhibitors and prednisolone, the spearhead drug for ALL treatment regimes, was investigated in SEM and KOPN8 using the MTS assay.

Results
We found that the MEK inhibitors MEK162, Selumetinib, and Trametinib effectively reduced the viability of KOPN8 cells (RASmut), whereas SEM and RS4;11 cells (RASwt) largely remained unaffected. In line with this, MLL-AF4+ infant ALL patient samples carrying RAS mutations  were significantly more sensitive to these MEK inhibitors when compared with patients carrying wild-type RAS genes: LC50 values for MEK162 were 0.04 vs. 26.9 µM (p<0.01), for Selumetinib 0.04 and 23.7 µM (p<0.01), and for Trametinib 0.01 vs. 26.5 µM (p<0.01), respectively. Furthermore, the presence of RAS mutations in primary MLL-r infant ALL samples was associated with significantly increased RAS activity, as determined by immunoprecipitation of GTP-bound RAS. Remarkably, however, enhanced RAS activation was not demonstrated by increased downstream ERK phosphorylation, while a slight increase of MEK phosphorylation was observed. Yet, MEK inhibitor exposure in both KOPN8 and SEM cells resulted in nearly complete abrogation of ERK phosphorylation, without affecting total ERK protein levels, suggesting that the loss of ERK activation plays an important role in the observed anti-leukemic effects. Furthermore, MEK162 and Selumetinib, seemed to induce accumulation of phosphorylated MEK. Additionally, MEK inhibitor treatment in both SEM and KOPN8 synergistically enhanced sensitivity towards prednisolone. Interestingly, a subgroup of patient samples (n=5) with wild-type RAS also showed sensitivity towards MEK inhibition, similar to the primary cells with RAS mutations. However, this observation could not be explained by increased RAS activation, nor by the phosphorylation levels of either MEK or ERK.

Summary
Our data show that MEK inhibition represents a promising therapeutic approach for MLL-r infant ALL patients carrying additional RAS mutations. Furthermore, the mechanism of action provoked by these MEK inhibitors seems to involve abrogation of ERK phosphorylation, but the initial levels of ERK phosphorylation did not correlate with MEK inhibitor sensitivity, and has no predictive value. Moreover, the prednisolone sensitization induced by MEK inhibitor exposure, further illustrates the potential value of MEK inhibition for the treatment of MLL-r infant ALL. Currently we are in the process of testing the efficacy of the above mentioned MEK inhibitors in vivo using a xenograft mouse model, while further elucidation of the molecular mechanisms underlying the anti-leukemic effects of these inhibitors in MLL-r ALL cells are in progress.

Keyword(s): Acute lymphoblastic leukemia, Drug sensitivity, MLL, Ras

Session topic: Towards targeted therapy in ALL

By clicking “Accept Terms & all Cookies” or by continuing to browse, you agree to the storing of third-party cookies on your device to enhance your user experience and agree to the user terms and conditions of this learning management system (LMS).

Cookie Settings
Accept Terms & all Cookies