Increased understanding of pediatric acute lymphoblastic leukemia (ALL) pathobiology has led to dramatic improvements in patient survival. However, there is still a need to develop targeted therapies to enable reduced chemotherapy intensity and to treat relapsed patients. The interleukin-7 receptor α (IL-7Rα) signaling pathways are prime therapeutic targets because these pathways harbor genetic aberrations in both T-cell ALL and B-cell precursor ALL. Therapeutic targeting of the IL-7Rα signaling pathways may lead to improved outcomes in a subset of patients.

Treatment of pediatric acute lymphoblastic leukemia (ALL) is a major success of modern medicine. Fifty years ago, children diagnosed with ALL survived for a median of 2 months.1  Today, the 5-year overall survival rate is >90% in some series.2  However, survival rates are much lower in children who experience relapse, varying between 21% and 39%.3,4  In addition, although this overall improvement in outcome is staggering, it comes at a cost. To achieve remission, patients typically undergo 2 to 3 years of chemotherapy.1  Acute side effects include infections, allergic reactions, thrombosis, pancreatitis, and neurologic impairment. Chronic side effects include osteonecrosis, obesity, secondary malignancies, and neurocognitive deficits.1 

As we characterize the pathophysiology of ALL, we must use our improved understanding to develop targeted therapies.5  Targeting molecular lesions in specific patients may enable reduced-intensity chemotherapy and may help to rescue children whom current protocols fail to cure. Although there are many potential targets for therapy, this spotlight will focus on genetic aberrations involving interleukin-7 receptor α (IL-7Rα) signaling pathways.

In health, the IL-7Rα chain forms 2 heterodimeric cytokine receptors (reviewed by Tal et al6 ). The receptor for IL-7 is formed by IL-7Rα pairing with γc, and the receptor for thymic stromal lymphopoietin (TSLP) is formed by IL-7Rα pairing with TSLP receptor (TSLPR) (encoded by the CRLF2 gene).6  IL-7 signaling is vital for normal T-cell development and survival of most mature T cells (reviewed by Mazzuchelli and Durum7 ). Conversely, TSLP signaling is involved in stimulation of growth and differentiation of B1 B-cell progenitors.8  In addition, TSLP is involved with CD4+ T-cell homeostasis, regulatory T-cell development, and dendritic cell activation (reviewed by Liu et al9 ). Finally, TSLP plays a prominent role in the pathogenesis of allergy and asthma (reviewed by Ziegler and Artis10 ). Binding of IL-7 or TSLP to their respective receptors causes activation of the JAK/STAT and PI3K/AKT/mTOR intracellular signaling cascades.6  The MAPK cascade could also be activated with normal TSLP signaling and with IL-7 signaling in T-cell ALL (T-ALL) and immature B and T cells.11-13  However, MAPK signaling is not required for the prosurvival and proliferation effects of IL-7 in T-ALL cells, and it does not seem to play a role in IL-7 signaling in normal T cells.12,14  Because the JAK/STAT and PI3K/AKT/mTOR intracellular signaling cascades seem to be the major players in IL-7Rα signaling, we will focus on them for the remainder of the review.

As might be expected for a receptor that is vital to T-cell development, mutations in IL-7Rα have recently been identified in a subset of pediatric T-ALL cases.15-17  These mutations generally occur in exon 6 of the gene and lead to insertion of multiple amino acids, usually including cysteine. The insertions induce IL-7Rα homodimerization and constitutively activate JAK1 in the absence of IL-7, γc, or JAK3.16 IL-7Rα mutations seem to be more prevalent in HOXA, TLX3, and early T-cell precursor (ETP-ALL) patient subgroups.16,17  The mutation does not seem to have prognostic implications.16 

Although mutations in IL-7Rα occur in ∼10% of pediatric patients, mutations in an upstream regulator of IL-7Rα, NOTCH1, occur in >50% of T-ALL cases.18 NOTCH1 is the most commonly mutated gene in T-ALL and is known to regulate IL-7Rα transcription and expression. Increases in IL-7R signaling promoted by NOTCH1 mutations may play a role in NOTCH1 oncogenicity.19 

Mutations downstream of IL-7R also occur in T-ALL. The JAK/STAT pathway can be activated by several genetic aberrations. Activating mutations in JAK1 and JAK3 have been reported, and a TEL-JAK2 fusion has also been described.17,20-26  Notably, JAK2 mutations generally seem to be more associated with B-cell precursor ALL (BCP-ALL) than T-ALL. Further downstream, T-ALL patients have also had mutations in STAT5B.27,28  In addition, a negative regulator of JAK-STAT signaling, PTPN2, is deleted in some cases of T-ALL.29 

Activation of the PI3K/AKT/mTOR pathway also occurs in T-ALL secondary to genetic aberrations, and an estimated 47.7% of pediatric cases have deletion or mutation of PTEN, PI3K, or AKT.30,31  The PI3K/AKT/mTOR-negative regulator PTEN is mutated in an estimated 8.7% to 22% of T-ALL cases or deleted in 27.3%.30,31  PTEN activity can also be decreased by posttranslational effects such as casein kinase 2 (CK2) overexpression, high levels of reactive oxygen species (ROS), or miRNAs.32,33 

Furthermore, T-ALL cells from patients proliferate in response to IL-7 signaling, suggesting that targeting this pathway may be useful for many T-ALL patients, not only those with IL-7Rα genetic aberrations.34 

In BCP-ALL patients, IL-7Rα pathway perturbations are most commonly caused by genetic aberrations affecting the CRLF2 gene, though mutations in IL-7Rα also occur. In most cases, chromosomal translocations, rearrangements, or gene duplications caused overexpression of CRLF2. Translocation leads to IGH-CRLF2 fusion, whereas interstitial deletion causes P2RY8-CRLF2 fusion.35-37 P2RY8-CRLF2 fusions have been found at a much higher rate (53%) in patients with Down syndrome–associated ALL (DS-ALL).35  Other chromosomal abnormalities can also lead to CRLF2 deregulation.36,37  It appears that rearranged TSLPRs signal conventionally through JAK/STAT and PI3K/AKT/mTOR pathways.38  Less commonly, the CRLF2 gene can have a F232C mutation that enables constitutive activation of TSLPR in the absence of IL-7Rα.39,40  Mutant TSLPR may not signal through the same kinases as wild-type TSLPR, particularly with regards to MAPK, which is downregulated in response to mutant TSLPR/mutant JAK2 signaling.11 

Notably, CRLF2 overexpression is not always attributable to structural rearrangement involving the CRLF2 gene.41,42  In all, ∼15% of patients with BCP-ALL have CRLF2 overexpression (excluding those with MLL, TCF3, TEL, and BCR/ABL rearrangements).37,40,43  Genetic aberrations of CRLF2 seem to confer a poorer prognosis in most studies.40,41,44,45  Fusion type may be associated with prognosis.45  However, not all studies support a poorer prognosis associated with CRLF2 aberration.37,46 

Patients with CRLF2 aberrations often have additional genetic abnormalities including deletion/mutation of IKZF1 (encoding IKAROS) and mutation or translocation of JAK2. Of note, CRLF2 aberrations can also be paired with concurrent mutations in IL-7Rα as well as JAK1, JAK3, or SH2B3 (which encodes the JAK2-negative regulator LNK).40,41,43  Patients with combined CRLF2 and IKZF1 aberrations often have gene expression profiles similar to patients with the BCR-ABL fusion protein, leading them to be considered “Philadelphia chromosome–like” (along with other genetic lesions inducing similar gene expression profiles). Patients with a Ph-like gene expression profile have a poor prognosis.40,47  Mutation or translocation of JAK2 also occurs independently of CRLF2 mutations in BCP-ALL.47-49 

Receptor targeting

The mutant IL-7Rα homodimers found in T-ALL depend on formation of disulfide bonds for stability. These bonds could potentially be targeted therapeutically using the reducing agent N-acetylcysteine (NAC). In T-ALL cell lines, NAC disrupts homodimerization, and it slows progression of disease in a xenograft model. Although NAC is inexpensive, it requires frequent dosing.50  In addition to using NAC, development of antibodies or T cells engineered to express chimeric antigen receptors (CAR-T-cells) against IL-7Rα and/ or against mutant homodimers would be potentially therapeutic.6 

Although antibodies and CAR-T-cells against normal IL-7Rα might be expected to target normal T cells as well as leukemic cells, promising results have been shown with anti-CD19 CAR-T-cells in the treatment of B-ALL. An anti–IL-7Rα CAR-T-cell may have similar effects. Anti-CD19 CAR-T-cell therapy induces severe, temporary B-cell lymphopenia, but B-cell populations rebound after therapy. Similar rebound of T-cell populations might be expected after anti–IL-7Rα CAR-T-cell therapy, given the active thymic function in these patients. In anti-CD19 CAR-T-cell therapy, treatment induces minimal residual disease-negative complete response in 60% of relapsed and refractory B-ALL patients. Therapy serves as an effective bridge to hematopoietic stem cell transplantation, resulting in disease-free survival.51  Perhaps anti–IL-7Rα CAR-T-cells or antibodies could also be used as a “bridge to transplant.” In addition, development of a mutant-specific antibody or CAR-T-cell may reduce potential off-target effects on normal T cells.

For treating CRLF2 genetic aberrations in BCP-ALL, CAR-T-cells against TSLPR have been shown to be effective in treating multiple xenograft models of BCP-ALL.52  In addition, antibodies against TSLPR have been developed, though these have mostly focused on applications in allergy/asthma to date.53 

NOTCH1 targeting

In T-ALL, mutated NOTCH1 could potentially be targeted using γ-secretase inhibitors, but these have shown significant side effects in a T-ALL clinical trial (reviewed by Tosello and Ferrando54 ). Alternative approaches to targeting NOTCH1 include using antibodies and proteasome inhibitors.54-56 

JAK/STAT targeting

Efforts investigating JAK inhibitors to treat neoplasia have focused mainly on the effects of blocking JAK2 in myeloproliferative neoplasms (MPN), polycythemia vera, and myelofibrosis (reviewed by Santos and Verstovsek57 ). In ALL, most studies of JAK inhibition have explored its effects on BCP-ALL. These studies have used primary patient samples or cell lines, including CRLF2-overexpressing cells and Ph-like cells with IL7Rα-activating mutation and SH2B3 mutation. Results from these in vitro and in vivo experiments suggest JAK inhibition may have some efficacy against BCP-ALL.38,40,43,48,58,59  A recent phase 1 clinical trial of the JAK1/2 inhibitor ruxolitinib suggested that the drug is well tolerated in children.60  Fewer experimental studies have focused on JAK inhibition in experimental models of T-ALL and ETP-ALL, though results from these studies are promising.61 

Experience with MPN suggests that resistance to JAK inhibition may develop, fueled by additional mutations in JAK proteins, increased JAK2 expression, or shifting transphosphorylation partners; resistance has been identified in both MPN and T-ALL.62,63  There are several potential approaches to overcoming resistance to JAK inhibition. New generations of JAK inhibitors are being developed to bind inactive forms of JAK to overcome resistance.64  Alternatively, JAK inhibitors may be combined with histone deacetylase (HDAC) inhibitors or heat shock protein 90 (HSP90) inhibitors.65,66  HDAC inhibitors induce hyperacetylation of HSP90, block its chaperone function, and promote JAK2 degradation.66  Inhibition of HSP90 increases degradation of both wild-type and mutant JAK2 and shows some efficacy against xenograft models of CRLF2-overexpressing BCP-ALL.65  However, both HSP90 inhibitors and HDAC inhibitors have multiple side effects (reviewed by Hong et al67 ). Phase 1 clinical trials of the HDAC inhibitor panobinostat included patients with myeloid and lymphoid malignancies, though none with ALL.68 

STAT5B targeting is less advanced than JAK targeting (reviewed by Dorritie et al69 ). The drug pimozide targets STAT5B and induces apoptosis in cultures of chronic myelogenous leukemia cells. Alternative efforts to target STAT5B include the use of decoy oligonucleotides in chronic myeloid leukemia and potentially the use of siRNA.70,71  In an experimental model of BCP-ALL, STAT5B gene expression was targeted by epigenetics-based therapy using BET bromodomain inhibitors. The study indicated that bromodomain inhibition induces apoptosis in vitro, improves survival in a xenograft model, and promotes downregulation of IL-7Rα.72  In addition, use of a pan–BCL-2 inhibitor, navitoclax, has demonstrated efficacy against STAT5B-mutated cells in vitro.27 

PI3K/AKT/mTOR targeting

The PI3K/AKT/mTOR pathway can be targeted by inhibitors of PI3K, AKT, and mTOR, dual inhibitors targeting both PI3K and mTOR, or inhibition of other components of the pathway including eukaryotic translation initiation factor 4E and phosphoinositide-dependent protein kinase I (reviewed by Rodon et al73 ).

Studies of PI3K and AKT inhibition have focused on effects in experimental models of T-ALL. PI3K inhibitor NVP-BKM120 induces apoptosis in cell lines and primary T-ALL cells and synergizes with vincristine and doxorubicin in vitro and in vivo.74  Conversely, although the PI3K inhibitor AS605240 synergizes with glucocorticoids, it antagonizes methotrexate and daunorubicin activity in cell lines, primary T-ALL cells, and murine xenograft models.75  Dual PI3K and mTOR inhibitors are also effective against T-ALL cell lines and patient samples in vitro and are more effective than individual inhibition of PI3K, AKT, or mTOR.76  Dual inhibitor BEZ235 synergizes with dexamethasone in vitro and in vivo, suggesting that targeting the PI3K/AKT/mTOR pathway could modulate glucocorticoid resistance in T-ALL.77  However, dual inhibitor PI-103 has been shown to cause upregulation of NOTCH1 and c-myc; combining PI-103 with c-myc or short-term NOTCH1 inhibition is synergistic and leads to increased death of T-ALL cell lines.78  In addition, inhibition of the PI3K pathway can lead to activation of the FAK/NF-κB/Bcl-2 pathway, suggesting that inhibition of PI3K may be paired with FAK inhibition for greater efficacy.79 

AKT inhibition by GSK690693 is effective against both T-ALL and BCP-ALL cell lines in vitro.80  AKT inhibitor MK-2206 decreases T-ALL cell viability and induces apoptosis in leukemia-initiating cell populations.81  Combining 3 AKT inhibitors with different mechanisms of action leads to synergistic effects against T-ALL cell lines.82  AKT inhibition can also lead to reversal of T-ALL glucocorticoid resistance, as demonstrated in cell lines and primografts in vitro as well as in several mouse models.83 

Inhibition of mTOR has included studies in both T-ALL and BCP-ALL. Rapamycin, the first mTOR inhibitor, is an allosteric inhibitor that does not fully inhibit the mTORC1 and mTORC2 enzymes.84  Rapamycin shows efficacy against xenografted BCP-ALL with mutated IL-7Rα and altered CRLF2 expression, as well as Ph-like BCP-ALL and a transgenic model of BCP-ALL.58,85  However, treatment with rapamycin alone is not considered highly effective against T-ALL.76 

To improve on the efficacy of rapamycin, several other drug types have been developed to target mTOR. Drugs targeting the active site of mTOR are effective against both T-ALL and BCP-ALL cell lines.86,87  More specific targeting of mTORC1 also induces apoptosis and autophagy in BCP-ALL cell lines and synergizes with AKT inhibitor MK-2206.87,88  Alternatively, mTORC1 activity can be targeted with metformin, an activator of LKB1/AMPK, which down-modulates mTORC1 activity. Metformin is effective in vitro against T-ALL cell lines, primary cells, and leukemia-initiating cells. There is currently a clinical trial recruiting pediatric patients with relapsed ALL for treatment with a combination of metformin and chemotherapy (NCT01324180).89  In both T-ALL and BCP-ALL cell lines, inhibition of mTOR synergizes with methotrexate and vincristine, potentially targeting leukemia-initiating cells.86,90  In addition, inhibition of mTOR may synergize with NOTCH1 or Bcl-2 inhibition, as demonstrated in murine xenografts and T-ALL cell lines and primary samples, respectively.91,92  Recently, inhibition of the PI3K/AKT/mTOR pathway has been shown to synergize with MEK inhibition in human T-ALL primary cells and BaF3 cells transduced with mutated IL-7Rα signaling pathway members.93 

In those T-ALL cases where PTEN inhibition by CK2 or ROS leads to activation of the PI3K/AKT/mTOR pathway, antagonists of CK2 or ROS scavengers may help to increase PTEN activity, as suggested by results in T-ALL cell lines and primary cells.32,94 

In the search for targeted ALL therapies, genetic aberrations in the IL-7Rα signaling pathways offer many potential opportunities for therapeutic intervention (Figure 1). Because drugs targeting IL-7Rα signaling pathways have the potential to reduce cellular proliferation and survival, we believe these drugs could currently be used as adjunctive therapies for many leukemia patients, even those without genetic aberrations involving the IL-7Rα signaling pathways. However, the real promise of these therapies lies in the (it is hoped) near future, when each new case of pediatric leukemia will be sequenced. This will enable clinicians to target an individual leukemia’s specific genetic lesions with specific IL-7Rα pathway–directed therapies. The appropriate therapy or therapies will vary depending on the location of the genetic lesion within the signaling pathway. For example, a BCP-ALL patient with overexpressed CRLF2 might be treated by targeting multiple levels of the signaling pathway, including use of anti-TSLPR CAR-T-cells, monoclonal antibodies, JAK inhibitors, and/or dual PI3K/mTOR inhibitors. A patient with AKT mutation might benefit from AKT or mTOR inhibition. As we move forward, we will need to determine the best use of these therapies to minimize side effects and maximize patient benefit. Perhaps someday, targeted therapy will enable patients with IL-7Rα signaling pathway mutations to be treated with less intensive chemotherapy, and relapsed patients or those patients whom current therapies fail will be successfully treated.

Figure 1

IL-7Rα pathway mutations provide potential targets for acute lymphoblastic leukemia therapy. In both T-ALL and BCP-ALL, mutations can occur at many points within the IL-7Rα signaling pathways (A). Aberrant signaling through these pathways offers multiple potential therapeutic targets (B).

Figure 1

IL-7Rα pathway mutations provide potential targets for acute lymphoblastic leukemia therapy. In both T-ALL and BCP-ALL, mutations can occur at many points within the IL-7Rα signaling pathways (A). Aberrant signaling through these pathways offers multiple potential therapeutic targets (B).

Close modal

The authors are grateful to Wenqing Li, Julie Hixon, and Emilee Senkevitch for their extensive review of the manuscript.

This work was supported by grants from the Children’s Cancer Foundation, Inc.; the National Multiple Sclerosis Society (PP1882) (S.K.D.), the Comparative Biomedical Scientist Training Program (Z01 BC 010931), National Institutes of Health (S.D.C.); and the Intramural Research Program of the National Cancer Institute, National Institutes of Health (ZIA SC 010378 [P.D.A.] and ZIA BC 009287 [S.K.D.]).

Contribution: S.D.C. researched and wrote the initial manuscript; and P.D.A. and S.K.D. contributed expertise and guidance and edited the manuscript.

Conflict-of-interest disclosure: S.K.D. has applied for a patent titled “IL-7Rα specific antibodies for treating acute lymphoblastic leukemia” (US patent application #62/238 612). The remaining authors declare no competing financial interests.

Correspondence: Scott K. Durum, Center for Cancer Research, National Cancer Institute, Building 560, Room 31-71, Frederick, MD 21702-1201; e-mail: durums@mail.nih.gov.

1
Gutierrez
 
A
Silverman
 
L
Orkin
 
S
Fisher
 
D
Ginsburg
 
D
Look
 
A
Lux
 
S
Nathan
 
D
Acute lymphoblastic leukemia.
Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. Vol. 2
2015
Philadelphia
Elsevier Saunders
(pg. 
1527
-
1554
)
2
Pui
 
CH
Campana
 
D
Pei
 
D
, et al. 
Treating childhood acute lymphoblastic leukemia without cranial irradiation.
N Engl J Med
2009
, vol. 
360
 
26
(pg. 
2730
-
2741
)
3
Nguyen
 
K
Devidas
 
M
Cheng
 
SC
, et al. 
Children’s Oncology Group
Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children’s Oncology Group study.
Leukemia
2008
, vol. 
22
 
12
(pg. 
2142
-
2150
)
4
Freyer
 
DR
Devidas
 
M
La
 
M
, et al. 
Postrelapse survival in childhood acute lymphoblastic leukemia is independent of initial treatment intensity: a report from the Children’s Oncology Group.
Blood
2011
, vol. 
117
 
11
(pg. 
3010
-
3015
)
5
Mullighan
 
CG
Hunger
 
SP
Therapy of pediatric ALL: from Bowie to Obama.
Blood
2013
, vol. 
122
 
15
(pg. 
2531
-
2532
)
6
Tal
 
N
Shochat
 
C
Geron
 
I
Bercovich
 
D
Izraeli
 
S
Interleukin 7 and thymic stromal lymphopoietin: from immunity to leukemia.
Cell Mol Life Sci
2014
, vol. 
71
 
3
(pg. 
365
-
378
)
7
Mazzucchelli
 
RI
Riva
 
A
Durum
 
SK
The human IL-7 receptor gene: deletions, polymorphisms and mutations.
Semin Immunol
2012
, vol. 
24
 
3
(pg. 
225
-
230
)
8
Montecino-Rodriguez
 
E
Leathers
 
H
Dorshkind
 
K
Identification of a B-1 B cell-specified progenitor.
Nat Immunol
2006
, vol. 
7
 
3
(pg. 
293
-
301
)
9
Liu
 
YJ
Soumelis
 
V
Watanabe
 
N
, et al. 
TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation.
Annu Rev Immunol
2007
, vol. 
25
 (pg. 
193
-
219
)
10
Ziegler
 
SF
Artis
 
D
Sensing the outside world: TSLP regulates barrier immunity.
Nat Immunol
2010
, vol. 
11
 
4
(pg. 
289
-
293
)
11
van Bodegom
 
D
Zhong
 
J
Kopp
 
N
, et al. 
Differences in signaling through the B-cell leukemia oncoprotein CRLF2 in response to TSLP and through mutant JAK2.
Blood
2012
, vol. 
120
 
14
(pg. 
2853
-
2863
)
12
Barata
 
JT
Silva
 
A
Brandao
 
JG
Nadler
 
LM
Cardoso
 
AA
Boussiotis
 
VA
Activation of PI3K is indispensable for interleukin 7-mediated viability, proliferation, glucose use, and growth of T cell acute lymphoblastic leukemia cells.
J Exp Med
2004
, vol. 
200
 
5
(pg. 
659
-
669
)
13
Johnson
 
SE
Shah
 
N
Bajer
 
AA
LeBien
 
TW
IL-7 activates the phosphatidylinositol 3-kinase/AKT pathway in normal human thymocytes but not normal human B cell precursors.
J Immunol
2008
, vol. 
180
 
12
(pg. 
8109
-
8117
)
14
Crawley
 
JB
Willcocks
 
J
Foxwell
 
BMJ
Interleukin-7 induces T cell proliferation in the absence of Erk/MAP kinase activity.
Eur J Immunol
1996
, vol. 
26
 
11
(pg. 
2717
-
2723
)
15
Shochat
 
C
Tal
 
N
Bandapalli
 
OR
, et al. 
Gain-of-function mutations in interleukin-7 receptor-α (IL7R) in childhood acute lymphoblastic leukemias.
J Exp Med
2011
, vol. 
208
 
5
(pg. 
901
-
908
)
16
Zenatti
 
PP
Ribeiro
 
D
Li
 
W
, et al. 
Oncogenic IL7R gain-of-function mutations in childhood T-cell acute lymphoblastic leukemia.
Nat Genet
2011
, vol. 
43
 
10
(pg. 
932
-
939
)
17
Zhang
 
J
Ding
 
L
Holmfeldt
 
L
, et al. 
The genetic basis of early T-cell precursor acute lymphoblastic leukaemia.
Nature
2012
, vol. 
481
 
7380
(pg. 
157
-
163
)
18
Weng
 
AP
Ferrando
 
AA
Lee
 
W
, et al. 
Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia.
Science
2004
, vol. 
306
 
5694
(pg. 
269
-
271
)
19
González-García
 
S
García-Peydró
 
M
Martín-Gayo
 
E
, et al. 
CSL-MAML-dependent Notch1 signaling controls T lineage-specific IL-7Ralpha gene expression in early human thymopoiesis and leukemia.
J Exp Med
2009
, vol. 
206
 
4
(pg. 
779
-
791
)
20
Bains
 
T
Heinrich
 
MC
Loriaux
 
MM
, et al. 
Newly described activating JAK3 mutations in T-cell acute lymphoblastic leukemia.
Leukemia
2012
, vol. 
26
 
9
(pg. 
2144
-
2146
)
21
Lacronique
 
V
Boureux
 
A
Valle
 
VD
, et al. 
A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia.
Science
1997
, vol. 
278
 
5341
(pg. 
1309
-
1312
)
22
Flex
 
E
Petrangeli
 
V
Stella
 
L
, et al. 
Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia.
J Exp Med
2008
, vol. 
205
 
4
(pg. 
751
-
758
)
23
Kalender Atak
 
Z
De Keersmaecker
 
K
Gianfelici
 
V
, et al. 
High accuracy mutation detection in leukemia on a selected panel of cancer genes.
PLoS One
2012
, vol. 
7
 
6
pg. 
e38463
 
24
Jeong
 
EG
Kim
 
MS
Nam
 
HK
, et al. 
Somatic mutations of JAK1 and JAK3 in acute leukemias and solid cancers.
Clin Cancer Res
2008
, vol. 
14
 
12
(pg. 
3716
-
3721
)
25
Asnafi
 
V
Le Noir
 
S
Lhermitte
 
L
, et al. 
JAK1 mutations are not frequent events in adult T-ALL: a GRAALL study.
Br J Haematol
2010
, vol. 
148
 
1
(pg. 
178
-
179
)
26
Canté-Barrett
 
K
Uitdehaag
 
JC
Meijerink
 
JP
Structural modeling of JAK1 mutations in T-cell acute lymphoblastic leukemia reveals a second contact site between pseudokinase and kinase domains.
Haematologica
2016
, vol. 
101
 
5
(pg. 
e189
-
e191
)
27
Kontro
 
M
Kuusanmäki
 
H
Eldfors
 
S
, et al. 
Novel activating STAT5B mutations as putative drivers of T-cell acute lymphoblastic leukemia.
Leukemia
2014
, vol. 
28
 
8
(pg. 
1738
-
1742
)
28
Bandapalli
 
OR
Schuessele
 
S
Kunz
 
JB
, et al. 
The activating STAT5B N642H mutation is a common abnormality in pediatric T-cell acute lymphoblastic leukemia and confers a higher risk of relapse.
Haematologica
2014
, vol. 
99
 
10
(pg. 
e188
-
e192
)
29
Kleppe
 
M
Lahortiga
 
I
El Chaar
 
T
, et al. 
Deletion of the protein tyrosine phosphatase gene PTPN2 in T-cell acute lymphoblastic leukemia.
Nat Genet
2010
, vol. 
42
 
6
(pg. 
530
-
535
)
30
Gutierrez
 
A
Sanda
 
T
Grebliunaite
 
R
, et al. 
High frequency of PTEN, PI3K, and AKT abnormalities in T-cell acute lymphoblastic leukemia.
Blood
2009
, vol. 
114
 
3
(pg. 
647
-
650
)
31
Remke
 
M
Pfister
 
S
Kox
 
C
, et al. 
High-resolution genomic profiling of childhood T-ALL reveals frequent copy-number alterations affecting the TGF-beta and PI3K-AKT pathways and deletions at 6q15-16.1 as a genomic marker for unfavorable early treatment response.
Blood
2009
, vol. 
114
 
5
(pg. 
1053
-
1062
)
32
Silva
 
A
Yunes
 
JA
Cardoso
 
BA
, et al. 
PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability.
J Clin Invest
2008
, vol. 
118
 
11
(pg. 
3762
-
3774
)
33
Mavrakis
 
KJ
Van Der Meulen
 
J
Wolfe
 
AL
, et al. 
A cooperative microRNA-tumor suppressor gene network in acute T-cell lymphoblastic leukemia (T-ALL).
Nat Genet
2011
, vol. 
43
 
7
(pg. 
673
-
678
)
34
Barata
 
JT
Cardoso
 
AA
Nadler
 
LM
Boussiotis
 
VA
Interleukin-7 promotes survival and cell cycle progression of T-cell acute lymphoblastic leukemia cells by down-regulating the cyclin-dependent kinase inhibitor p27(kip1).
Blood
2001
, vol. 
98
 
5
(pg. 
1524
-
1531
)
35
Mullighan
 
CG
Collins-Underwood
 
JR
Phillips
 
LA
, et al. 
Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia.
Nat Genet
2009
, vol. 
41
 
11
(pg. 
1243
-
1246
)
36
Russell
 
LJ
Capasso
 
M
Vater
 
I
, et al. 
Deregulated expression of cytokine receptor gene, CRLF2, is involved in lymphoid transformation in B-cell precursor acute lymphoblastic leukemia.
Blood
2009
, vol. 
114
 
13
(pg. 
2688
-
2698
)
37
Yano
 
M
Imamura
 
T
Asai
 
D
, et al. 
Japan Association of Childhood Leukemia Study
An overall characterization of pediatric acute lymphoblastic leukemia with CRLF2 overexpression.
Genes Chromosomes Cancer
2014
, vol. 
53
 
10
(pg. 
815
-
823
)
38
Tasian
 
SK
Doral
 
MY
Borowitz
 
MJ
, et al. 
Aberrant STAT5 and PI3K/mTOR pathway signaling occurs in human CRLF2-rearranged B-precursor acute lymphoblastic leukemia.
Blood
2012
, vol. 
120
 
4
(pg. 
833
-
842
)
39
Chapiro
 
E
Russell
 
L
Lainey
 
E
, et al. 
Activating mutation in the TSLPR gene in B-cell precursor lymphoblastic leukemia.
Leukemia
2010
, vol. 
24
 
3
(pg. 
642
-
645
)
40
Yoda
 
A
Yoda
 
Y
Chiaretti
 
S
, et al. 
Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
1
(pg. 
252
-
257
)
41
Chen
 
IM
Harvey
 
RC
Mullighan
 
CG
, et al. 
Outcome modeling with CRLF2, IKZF1, JAK, and minimal residual disease in pediatric acute lymphoblastic leukemia: a Children’s Oncology Group study.
Blood
2012
, vol. 
119
 
15
(pg. 
3512
-
3522
)
42
Bugarin
 
C
Sarno
 
J
Palmi
 
C
, et al. 
I-BFM study group
Fine tuning of surface CRLF2 expression and its associated signaling profile in childhood B-cell precursor acute lymphoblastic leukemia.
Haematologica
2015
, vol. 
100
 
6
(pg. 
e229
-
e232
)
43
Roberts
 
KG
Morin
 
RD
Zhang
 
J
, et al. 
Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia.
Cancer Cell
2012
, vol. 
22
 
2
(pg. 
153
-
166
)
44
Harvey
 
RC
Mullighan
 
CG
Chen
 
IM
, et al. 
Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia.
Blood
2010
, vol. 
115
 
26
(pg. 
5312
-
5321
)
45
Palmi
 
C
Vendramini
 
E
Silvestri
 
D
, et al. 
Poor prognosis for P2RY8-CRLF2 fusion but not for CRLF2 over-expression in children with intermediate risk B-cell precursor acute lymphoblastic leukemia.
Leukemia
2012
, vol. 
26
 
10
(pg. 
2245
-
2253
)
46
van der Veer
 
A
Waanders
 
E
Pieters
 
R
, et al. 
Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in children with B-cell precursor ALL.
Blood
2013
, vol. 
122
 
15
(pg. 
2622
-
2629
)
47
Mullighan
 
CG
Zhang
 
J
Harvey
 
RC
, et al. 
JAK mutations in high-risk childhood acute lymphoblastic leukemia.
Proc Natl Acad Sci USA
2009
, vol. 
106
 
23
(pg. 
9414
-
9418
)
48
Bercovich
 
D
Ganmore
 
I
Scott
 
LM
, et al. 
Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down’s syndrome.
Lancet
2008
, vol. 
372
 
9648
(pg. 
1484
-
1492
)
49
Yano
 
M
Imamura
 
T
Asai
 
D
, et al. 
Identification of novel kinase fusion transcripts in paediatric B cell precursor acute lymphoblastic leukaemia with IKZF1 deletion.
Br J Haematol
2015
, vol. 
171
 
5
(pg. 
813
-
817
)
50
Mansour
 
MR
Reed
 
C
Eisenberg
 
AR
, et al. 
Targeting oncogenic interleukin-7 receptor signalling with N-acetylcysteine in T cell acute lymphoblastic leukaemia.
Br J Haematol
2015
, vol. 
168
 
2
(pg. 
230
-
238
)
51
Lee
 
DW
Kochenderfer
 
JN
Stetler-Stevenson
 
M
, et al. 
T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial.
Lancet
2015
, vol. 
385
 
9967
(pg. 
517
-
528
)
52
Qin
 
H
Cho
 
M
Haso
 
W
, et al. 
Eradication of B-ALL using chimeric antigen receptor-expressing T cells targeting the TSLPR oncoprotein.
Blood
2015
, vol. 
126
 
5
(pg. 
629
-
639
)
53
Borowski
 
A
Vetter
 
T
Kuepper
 
M
, et al. 
Expression analysis and specific blockade of the receptor for human thymic stromal lymphopoietin (TSLP) by novel antibodies to the human TSLPRα receptor chain.
Cytokine
2013
, vol. 
61
 
2
(pg. 
546
-
555
)
54
Tosello
 
V
Ferrando
 
AA
The NOTCH signaling pathway: role in the pathogenesis of T-cell acute lymphoblastic leukemia and implication for therapy.
Ther Adv Hematol
2013
, vol. 
4
 
3
(pg. 
199
-
210
)
55
Agnusdei
 
V
Minuzzo
 
S
Frasson
 
C
, et al. 
Therapeutic antibody targeting of Notch1 in T-acute lymphoblastic leukemia xenografts.
Leukemia
2014
, vol. 
28
 
2
(pg. 
278
-
288
)
56
Koyama
 
D
Kikuchi
 
J
Hiraoka
 
N
, et al. 
Proteasome inhibitors exert cytotoxicity and increase chemosensitivity via transcriptional repression of Notch1 in T-cell acute lymphoblastic leukemia.
Leukemia
2014
, vol. 
28
 
6
(pg. 
1216
-
1226
)
57
Santos
 
FP
Verstovsek
 
S
JAK2 inhibitors: what’s the true therapeutic potential?
Blood Rev
2011
, vol. 
25
 
2
(pg. 
53
-
63
)
58
Maude
 
SL
Tasian
 
SK
Vincent
 
T
, et al. 
Targeting JAK1/2 and mTOR in murine xenograft models of Ph-like acute lymphoblastic leukemia.
Blood
2012
, vol. 
120
 
17
(pg. 
3510
-
3518
)
59
Treanor
 
LM
Zhou
 
S
Janke
 
L
, et al. 
Interleukin-7 receptor mutants initiate early T cell precursor leukemia in murine thymocyte progenitors with multipotent potential.
J Exp Med
2014
, vol. 
211
 
4
(pg. 
701
-
713
)
60
Loh
 
ML
Tasian
 
SK
Rabin
 
KR
, et al. 
A phase 1 dosing study of ruxolitinib in children with relapsed or refractory solid tumors, leukemias, or myeloproliferative neoplasms: A Children’s Oncology Group phase 1 consortium study (ADVL1011).
Pediatr Blood Cancer
2015
, vol. 
62
 
10
(pg. 
1717
-
1724
)
61
Maude
 
SL
Dolai
 
S
Delgado-Martin
 
C
, et al. 
Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia.
Blood
2015
, vol. 
125
 
11
(pg. 
1759
-
1767
)
62
Koppikar
 
P
Bhagwat
 
N
Kilpivaara
 
O
, et al. 
Heterodimeric JAK-STAT activation as a mechanism of persistence to JAK2 inhibitor therapy.
Nature
2012
, vol. 
489
 
7414
(pg. 
155
-
159
)
63
Springuel
 
L
Hornakova
 
T
Losdyck
 
E
, et al. 
Cooperating JAK1 and JAK3 mutants increase resistance to JAK inhibitors.
Blood
2014
, vol. 
124
 
26
(pg. 
3924
-
3931
)
64
Meyer
 
SC
Levine
 
RL
Molecular pathways: molecular basis for sensitivity and resistance to JAK kinase inhibitors.
Clin Cancer Res
2014
, vol. 
20
 
8
(pg. 
2051
-
2059
)
65
Weigert
 
O
Lane
 
AA
Bird
 
L
, et al. 
Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition.
J Exp Med
2012
, vol. 
209
 
2
(pg. 
259
-
273
)
66
Wang
 
Y
Fiskus
 
W
Chong
 
DG
, et al. 
Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells.
Blood
2009
, vol. 
114
 
24
(pg. 
5024
-
5033
)
67
Hong
 
DS
Banerji
 
U
Tavana
 
B
George
 
GC
Aaron
 
J
Kurzrock
 
R
Targeting the molecular chaperone heat shock protein 90 (HSP90): lessons learned and future directions.
Cancer Treat Rev
2013
, vol. 
39
 
4
(pg. 
375
-
387
)
68
DeAngelo
 
DJ
Spencer
 
A
Bhalla
 
KN
, et al. 
Phase Ia/II, two-arm, open-label, dose-escalation study of oral panobinostat administered via two dosing schedules in patients with advanced hematologic malignancies.
Leukemia
2013
, vol. 
27
 
8
(pg. 
1628
-
1636
)
69
Dorritie
 
KA
McCubrey
 
JA
Johnson
 
DE
STAT transcription factors in hematopoiesis and leukemogenesis: opportunities for therapeutic intervention.
Leukemia
2014
, vol. 
28
 
2
(pg. 
248
-
257
)
70
Wang
 
X
Zeng
 
J
Shi
 
M
, et al. 
Targeted blockage of signal transducer and activator of transcription 5 signaling pathway with decoy oligodeoxynucleotides suppresses leukemic K562 cell growth.
DNA Cell Biol
2011
, vol. 
30
 
2
(pg. 
71
-
78
)
71
Zhang
 
Q
Hossain
 
DM
Nechaev
 
S
, et al. 
TLR9-mediated siRNA delivery for targeting of normal and malignant human hematopoietic cells in vivo.
Blood
2013
, vol. 
121
 
8
(pg. 
1304
-
1315
)
72
Ott
 
CJ
Kopp
 
N
Bird
 
L
, et al. 
BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia.
Blood
2012
, vol. 
120
 
14
(pg. 
2843
-
2852
)
73
Rodon
 
J
Dienstmann
 
R
Serra
 
V
Tabernero
 
J
Development of PI3K inhibitors: lessons learned from early clinical trials.
Nat Rev Clin Oncol
2013
, vol. 
10
 
3
(pg. 
143
-
153
)
74
Lonetti
 
A
Antunes
 
IL
Chiarini
 
F
, et al. 
Activity of the pan-class I phosphoinositide 3-kinase inhibitor NVP-BKM120 in T-cell acute lymphoblastic leukemia.
Leukemia
2014
, vol. 
28
 
6
(pg. 
1196
-
1206
)
75
Silveira
 
AB
Laranjeira
 
AB
Rodrigues
 
GO
, et al. 
PI3K inhibition synergizes with glucocorticoids but antagonizes with methotrexate in T-cell acute lymphoblastic leukemia.
Oncotarget
2015
, vol. 
6
 
15
(pg. 
13105
-
13118
)
76
Chiarini
 
F
Grimaldi
 
C
Ricci
 
F
, et al. 
Activity of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 against T-cell acute lymphoblastic leukemia.
Cancer Res
2010
, vol. 
70
 
20
(pg. 
8097
-
8107
)
77
Hall
 
CP
Reynolds
 
CP
Kang
 
MH
Modulation of glucocorticoid resistance in pediatric T-cell acute lymphoblastic leukemia by increasing BIM expression with the PI3K/mTOR inhibitor BEZ235.
Clin Cancer Res
2016
, vol. 
22
 
3
(pg. 
621
-
632
)
78
Shepherd
 
C
Banerjee
 
L
Cheung
 
CW
, et al. 
PI3K/mTOR inhibition upregulates NOTCH-MYC signalling leading to an impaired cytotoxic response.
Leukemia
2013
, vol. 
27
 
3
(pg. 
650
-
660
)
79
You
 
D
Xin
 
J
Volk
 
A
, et al. 
FAK mediates a compensatory survival signal parallel to PI3K-AKT in PTEN-null T-ALL cells.
Cell Reports
2015
, vol. 
10
 
12
(pg. 
2055
-
2068
)
80
Levy
 
DS
Kahana
 
JA
Kumar
 
R
AKT inhibitor, GSK690693, induces growth inhibition and apoptosis in acute lymphoblastic leukemia cell lines.
Blood
2009
, vol. 
113
 
8
(pg. 
1723
-
1729
)
81
Simioni
 
C
Neri
 
LM
Tabellini
 
G
, et al. 
Cytotoxic activity of the novel Akt inhibitor, MK-2206, in T-cell acute lymphoblastic leukemia.
Leukemia
2012
, vol. 
26
 
11
(pg. 
2336
-
2342
)
82
Cani
 
A
Simioni
 
C
Martelli
 
AM
, et al. 
Triple Akt inhibition as a new therapeutic strategy in T-cell acute lymphoblastic leukemia.
Oncotarget
2015
, vol. 
6
 
9
(pg. 
6597
-
6610
)
83
Piovan
 
E
Yu
 
J
Tosello
 
V
, et al. 
Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia.
Cancer Cell
2013
, vol. 
24
 
6
(pg. 
766
-
776
)
84
Janes
 
MR
Vu
 
C
Mallya
 
S
, et al. 
Efficacy of the investigational mTOR kinase inhibitor MLN0128/INK128 in models of B-cell acute lymphoblastic leukemia.
Leukemia
2013
, vol. 
27
 
3
(pg. 
586
-
594
)
85
Brown
 
VI
Fang
 
J
Alcorn
 
K
, et al. 
Rapamycin is active against B-precursor leukemia in vitro and in vivo, an effect that is modulated by IL-7-mediated signaling.
Proc Natl Acad Sci USA
2003
, vol. 
100
 
25
(pg. 
15113
-
15118
)
86
Evangelisti
 
C
Ricci
 
F
Tazzari
 
P
, et al. 
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
(pg. 
781
-
791
)
87
Simioni
 
C
Cani
 
A
Martelli
 
AM
, et al. 
Activity of the novel mTOR inhibitor Torin-2 in B-precursor acute lymphoblastic leukemia and its therapeutic potential to prevent Akt reactivation.
Oncotarget
2014
, vol. 
5
 
20
(pg. 
10034
-
10047
)
88
Neri
 
LM
Cani
 
A
Martelli
 
AM
, et al. 
Targeting the PI3K/Akt/mTOR signaling pathway in B-precursor acute lymphoblastic leukemia and its therapeutic potential.
Leukemia
2014
, vol. 
28
 
4
(pg. 
739
-
748
)
89
Grimaldi
 
C
Chiarini
 
F
Tabellini
 
G
, et al. 
AMP-dependent kinase/mammalian target of rapamycin complex 1 signaling in T-cell acute lymphoblastic leukemia: therapeutic implications.
Leukemia
2012
, vol. 
26
 
1
(pg. 
91
-
100
)
90
Teachey
 
DT
Sheen
 
C
Hall
 
J
, et al. 
mTOR inhibitors are synergistic with methotrexate: an effective combination to treat acute lymphoblastic leukemia.
Blood
2008
, vol. 
112
 
5
(pg. 
2020
-
2023
)
91
Cullion
 
K
Draheim
 
KM
Hermance
 
N
, et al. 
Targeting the Notch1 and mTOR pathways in a mouse T-ALL model.
Blood
2009
, vol. 
113
 
24
(pg. 
6172
-
6181
)
92
Iacovelli
 
S
Ricciardi
 
MR
Allegretti
 
M
, et al. 
Co-targeting of Bcl-2 and mTOR pathway triggers synergistic apoptosis in BH3 mimetics resistant acute lymphoblastic leukemia.
Oncotarget
2015
, vol. 
6
 
31
(pg. 
32089
-
32103
)
93
Canté-Barrett
 
K
Spijkers-Hagelstein
 
JA
Buijs-Gladdines
 
JG
, et al. 
MEK and PI3K-AKT inhibitors synergistically block activated IL7 receptor signaling in T-cell acute lymphoblastic leukemia.
Leukemia
2016
 
May 13
94
Buontempo
 
F
Orsini
 
E
Martins
 
LR
, et al. 
Cytotoxic activity of the casein kinase 2 inhibitor CX-4945 against T-cell acute lymphoblastic leukemia: targeting the unfolded protein response signaling.
Leukemia
2014
, vol. 
28
 
3
(pg. 
543
-
553
)
Sign in via your Institution