scholarly journals Fusion of EML1 to ABL1 in T-cell acute lymphoblastic leukemia with cryptic t(9;14)(q34;q32)

Blood ◽  
2005 ◽  
Vol 105 (12) ◽  
pp. 4849-4852 ◽  
Author(s):  
Kim De Keersmaecker ◽  
Carlos Graux ◽  
Maria D. Odero ◽  
Nicole Mentens ◽  
Riet Somers ◽  
...  

Abstract The BCR-ABL1 fusion kinase is frequently associated with chronic myeloid leukemia and B-cell acute lymphoblastic leukemia but is rare in T-cell acute lymphoblastic leukemia (T-ALL). We recently identified NUP214-ABL1 as a variant ABL1 fusion gene in 6% of T-ALL patients. Here we describe the identification of another ABL1 fusion, EML1-ABL1, in a T-ALL patient with a cryptic t(9;14)(q34;q32) associated with deletion of CDKN2A (p16) and expression of TLX1 (HOX11). Echinoderm microtubule-associated protein-like 1-Abelson 1 (EML1-ABL1) is a constitutively phosphorylated tyrosine kinase that transforms Ba/F3 cells to growth factor-independent growth through activation of survival and proliferation pathways, including extracellular signal-related kinase 1/2 (Erk1/2), signal transducers and activators of transcription 5 (Stat5), and Lyn kinase. Deletion of the coiled-coil domain of EML1 abrogated the transforming properties of the fusion kinase. EML1-ABL1 and breakpoint cluster region (BCR)-ABL1 were equally sensitive to the tyrosine kinase inhibitor imatinib. These data further demonstrate the involvement of ABL1 fusions in the pathogenesis of T-ALL and identify EML1-ABL1 as a novel therapeutic target of imatinib. (Blood. 2005;105:4849-4852)

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 141-141
Author(s):  
Jan Cools ◽  
Carlos Graux ◽  
Cindy Melotte ◽  
Hilmar Quentmeier ◽  
Adolfo A. Ferrando ◽  
...  

Abstract The Philadelphia translocation, encoding the BCR-ABL1 (BCR-ABL) fusion gene, is typically found in chronic myeloid leukemia (CML) and precursor B-cell acute lymphoblastic leukemia (B-ALL), but is exceptionally rare in T-cell acute lymphoblastic leukemia (T-ALL). To study the potential involvement of ABL1 gene rearrangements in T-cell malignancies, we screened 90 T-ALL cases by fluorescence in situ hybridization (FISH), using BCR and ABL1 probes. No BCR-ABL1 fusion signals were observed, confirming the low frequency of this rearrangement in T-ALL, but we did observe marked amplification (> 10 signals per nucleus) ABL1 of in 5 of 90 (5.5 %) T-ALL patients. Amplification of ABL1 occurred on small extrachromosomal elements that were not detectable by conventional cytogenetics. and hence are referred to as episomes. FISH, and array-CGH analyses delineated the amplicon as a 500 kb region from chromosome band 9q34, containing the oncogenes ABL1 and NUP214 (CAN). Molecular analysis led to the identification of a NUP214-ABL1 fusion gene, which is generated as result of the circularization of the genomic region between ABL1 and NUP214 to form the episomes. This is the first example of an oncogenic fusion gene generated by extrachromosomal amplification. The NUP214-ABL1 transcript was detected in 5 patients with ABL1 amplification, in 5 of 85 (5.8 %) additional T-ALL patients, and in 3 of 22 T-ALL cell lines. The constitutively phosphorylated tyrosine kinase NUP214-ABL1 is sensitive to the tyrosine kinase inhibitor imatinib mesylate (STI-571). The recurrent cryptic NUP214-ABL1 rearrangement is associated with increased expression TLX1 of (HOX11) or TLX3 (HOX11L2), and with deletion of CDKN2A (p16), consistent with a multi-step pathogenesis of T-ALL. Our results identify a novel mechanism for the generation of a fusion gene on extrachromosomal elements, and indicate the importance of activated tyrosine kinase signaling in the pathogenesis of T-ALL. NUP214-ABL1 expression defines a new subgroup of T-ALL patients that could benefit from imatinib treatment.


Leukemia ◽  
2008 ◽  
Vol 23 (1) ◽  
pp. 125-133 ◽  
Author(s):  
C Graux ◽  
◽  
M Stevens-Kroef ◽  
M Lafage ◽  
N Dastugue ◽  
...  

2011 ◽  
Author(s):  
Takaomi Sanda ◽  
Jeffrey W. Tyner ◽  
Alejandro Gutierrez ◽  
Vu N. Ngo ◽  
Richard Moriggl ◽  
...  

Leukemia ◽  
2013 ◽  
Vol 28 (6) ◽  
pp. 1196-1206 ◽  
Author(s):  
A Lonetti ◽  
I L Antunes ◽  
F Chiarini ◽  
E Orsini ◽  
F Buontempo ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3155-3155
Author(s):  
Takaomi Sanda ◽  
Jeffrey W Tyner ◽  
Alejandro Gutierrez ◽  
Vu N Ngo ◽  
Richard Moriggl ◽  
...  

Abstract Abstract 3155 The often aggressive and unpredictable behavior of T-cell lymphoblastic malignancies continues to pose both major clinical challenges in children and adults. To discover oncogenic pathways downstream of critical genetic abnormalities that are characteristically deregulated in T-cell acute lymphoblastic leukemia (T-ALL), and to identify novel molecular targets for anti-leukemic agents with T-cell specificity, we carried out a genome-wide functional screen in T-ALL cell lines using a retroviral library of inducible short-hairpin RNAs (shRNAs). Among the genes that are required for the growth of T-ALL cells, we found that loss of TYK2, a JAK family tyrosine kinase, was specifically lethal in each of three T-ALL cell lines that we tested in this screen. By contrast, TYK2 knock-down did not affect the growth of cell lines from diffuse large B-cell lymphoma or multiple myeloma, indicating that TYK2 is specifically required for the growth of T-ALL cells. We confirmed by knock-down with multiple independent shRNAs that the loss of TYK2 induces apoptosis in T-ALL, whereas knock-down of other JAK proteins (JAK1, JAK2 or JAK3) had no effect. We found that the TYK2 protein is constitutively phosphorylated in many T-ALL cell lines, and that these cells are sensitive to small molecule JAK/TYK2 inhibitors, including JAK inhibitor-I, AG-490 and CEP-701. To identify upstream receptors involved in TYK2 activation, we knocked down IFNAR, IFNGR, IL10R and IL12RB, and found that loss of IFNAR1 and IFNAR2 specifically inhibit the growth of T-ALL cells, as strongly as TYK2 knock-down. In addition, targeted knock-down analysis of downstream STAT proteins revealed that loss of STAT1 also inhibits the growth of T-ALL cells, indicating that this transcription factor is involved in the TYK2 pathway and required for cell survival. In fact, STAT1 protein was constitutively phosphorylated in many T-ALL cell lines and this phosphorylation was inhibited by both TYK2 knock-down and treatment with JAK/TYK2 inhibitors. Although interferon-mediated pathways are cytostatic in many cell types, our results indicate a requirement for the IFNAR-TYK2-STAT1 pathway in promoting the growth and survival of T-ALL cells. Dependence on this pathway confers unique sensitivity of T-ALL cells to TYK2 inhibition by small molecule inhibitors, thus providing a novel therapeutic target for clinical testing in patients with this disease. Disclosures: Druker: Molecular MD: Equity Ownership.


Leukemia ◽  
2019 ◽  
Vol 34 (5) ◽  
pp. 1241-1252 ◽  
Author(s):  
Sonia Rodriguez ◽  
Christina Abundis ◽  
Francesco Boccalatte ◽  
Purvi Mehrotra ◽  
Mark Y. Chiang ◽  
...  

AbstractTimed degradation of the cyclin-dependent kinase inhibitor p27Kip1 by the E3 ubiquitin ligase F-box protein SKP2 is critical for T-cell progression into cell cycle, coordinating proliferation and differentiation processes. SKP2 expression is regulated by mitogenic stimuli and by Notch signaling, a key pathway in T-cell development and in T-cell acute lymphoblastic leukemia (T-ALL); however, it is not known whether SKP2 plays a role in the development of T-ALL. Here, we determined that SKP2 function is relevant for T-ALL leukemogenesis, whereas is dispensable for T-cell development. Targeted inhibition of SKP2 by genetic deletion or pharmacological blockade markedly inhibited proliferation of human T-ALL cells in vitro and antagonized disease in vivo in murine and xenograft leukemia models, with little effect on normal tissues. We also demonstrate a novel feed forward feedback loop by which Notch and IL-7 signaling cooperatively converge on SKP2 induction and cell cycle activation. These studies show that the Notch/SKP2/p27Kip1 pathway plays a unique role in T-ALL development and provide a proof-of-concept for the use of SKP2 as a new therapeutic target in T-cell acute lymphoblastic leukemia (T-ALL).


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1596-1596
Author(s):  
Koshi Akahane ◽  
Zhaodong Li ◽  
Julia Etchin ◽  
Alla Berezovskaya ◽  
Evisa Gjini ◽  
...  

Abstract T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy resulting from the transformation of T-cell progenitors. Although the prognosis of this disease has substantially improved due to the introduction of intensified chemotherapy, the clinical outcome of T-ALL patients with primary resistant or relapsed disease remains poor, indicating that further therapeutic improvement is urgently needed. We have previously demonstrated that activation of tyrosine kinase 2 (TYK2) contributes to aberrant survival of human T-ALL cells. TYK2 is a member of the Janus-activated kinase (JAK) tyrosine kinase family and our report was the first to implicate TYK2 in T-ALL pathogenesis. Indeed, our gene knockdown experiments showed TYK2 dependency in 14 of 16 (88%) T-ALL cell lines and 5 of 8 (63%) patient-derived T-ALL cells tested, suggesting that inhibition of TYK2 would be beneficial in most patients with T-ALL. Based on these findings, we investigated the therapeutic potential of a novel small-molecule TYK2 kinase inhibitor NDI-031301 in T-ALL. We found that NDI-031301 shows potent and selective inhibitory activity against TYK2 in a cellular context, because this compound strongly inhibited the growth of TYK2-transfomed Ba/F3 cells when compared to the JAK inhibitors tofacitinib and baricitinib, whereas Ba/F3 cells transformed by other tyrosine kinases showed decreased sensitivity to NDI-031301. NDI-031301 induced robust growth inhibition in each of 4 human T-ALL cell lines representing different molecular subtypes of the disease (DU.528, KOPT-K1, HPB-ALL and SKW-3), with IC50 values of 0.8186 - 2.380 μM after 72 hours of exposure. NDI-031301 treatment of human T-ALL cell lines resulted in induction of apoptosis that was not observed with tofacitinib and baricitinib. To elucidate the mechanism of apoptosis induced by NDI-031301 in T-ALL cells, we next investigated cellular signaling pathways that are associated with cell survival and specifically affected by TYK2 inhibition with NDI-031301. Western blotting analysis demonstrated that treatment with 3 μM of NDI-031301 resulted in reduction of STAT1 Tyr-701 phosphorylation and BCL2 levels in KOPT-K1 cells, consistent with our previous finding that TYK2 phosphorylates STAT1 and upregulates BCL2 expression in most T-ALL cells. Surprisingly, the treatment also uniquely led to activation of three mitogen-activated protein kinases (MAPKs), resulting in phosphorylation of ERK, SAPK/JNK and p38 MAPK coincident with PARP cleavage, which was not observed with tofacitinib and baricitinib. NDI-031301-mediated activation of SAPK/JNK and p38 MAPK pathways are likely mediated through inhibition of TYK2, because increased phosphorylation levels of SAPK/JNK and p38 MAPK were observed in the cells transfected with TYK2-targeting shRNAs, while the levels of ERK1/2 phosphorylation were not upregulated. Further investigation revealed that activation of p38 MAPK occurred within 1 hour of NDI-031301 treatment and was responsible for NDI-031301-induced T-ALL cell death, as pharmacologic inhibition of p38 MAPK by SB203580 partially rescued apoptosis induced by TYK2 inhibitor, while inhibition of ERK or SAPK/JNK showed no rescue effects. Finally, we found that daily oral administration of NDI-031301 at 100mg/kg BID to immunodeficient mice engrafted with KOPT-K1 T-ALL cells was well tolerated, and led to decreased tumor burden and a significant survival benefit. After 29 days of treatment, the mice receiving NDI-031301 had marked reductions in infiltration of leukemia cells into spleen and bone marrow by comparison with controls. Thus, our findings clearly support TYK2 inhibition with NDI-031301 or a related compound as a potential therapeutic strategy for patients with T-ALL, and also raise the possibility that enhancing p38 MAPK activation in T-ALL cells may be an approach to accentuate its anti-leukemic activity. Disclosures Masse: Nimbus Therapeutics: Employment. Miao:Nimbus Therapeutics: Employment. Rocnik:Nimbus Therapeutics: Employment. Kapeller:Nimbus Therapeutics: Employment.


Sign in / Sign up

Export Citation Format

Share Document