The transcription factor signal transducers and activators of transcription 5 (STAT5) has an important and unique role in Breakpoint Cluster Region - Abelson 1 (BCR-ABL1)–driven neoplasias. STAT5 is an essential component in the signaling network that maintains the survival and growth of chronic myeloid leukemia (CML) cells. In contrast, the function of the prototypical upstream kinase of STAT5, the Janus kinase JAK2, in CML is still under debate. Although there is widespread agreement that JAK2 is part of the signaling network downstream of BCR-ABL1, it is unclear whether and under what circumstances JAK2 inhibitors may be beneficial for CML patients. Recent studies in murine models have cast doubt on the importance of JAK2 in CML maintenance. Nevertheless, JAK2 has been proposed to have a central role in the cytokine signaling machinery that allows the survival of CML stem cells in the presence of BCR-ABL1 tyrosine kinase inhibitors. In this review, we summarize the current debate and provide an overview of the arguments on both sides of the fence. We present recent evidence showing that CML stem cells do not depend on BCR-ABL1 kinase activity but require the continuous support of the hematopoietic niche and its distinct cytokine environment and suggest that it has the potential to resolve the dispute.

The Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway represents one of the best-characterized signaling pathways in cell biology. JAK-STAT signaling was only discovered ∼20 years ago, and subsequent study has provided many valuable insights into the process by which extracellular information is transmitted through the cell membrane to the nucleus.1  We now know that the JAK-STAT pathway is involved in signaling downstream of >50 growth factors and cytokines, thereby participating in vital cellular functions such as proliferation, differentiation, apoptosis, survival, and migration.2,3  The mammalian family of Janus kinases is composed of 4 members, JAK1, JAK2, JAK3, and the tyrosine kinase 2 (Tyk2), all of which share a structure characterized by 7 JAK homology domains.4  JAK2 was initially shown to play a crucial part in immune cell development and hematopoiesis.5  Shortly afterward, it was found to be activated in the initiation and maintenance of cancer, but the exact mechanisms by which it contributes to pathogenesis remain obscure.6  The discovery that a single point mutation within the nonreceptor tyrosine kinase JAK2, leading to the substitution of a valine residue by phenylalanine at amino acid 617 (JAK2V617F), is responsible for driving a subset of myeloproliferative neoplasia (MPN) dramatically increased interest in JAK2.7-10  Point mutations and insertions/deletions within exon 12 of JAK2 have subsequently been identified in nearly all patients with JAK2V617F-negative polycythemia vera, as well as in some cases of acute myeloid leukemia, systemic mastocytosis, chronic myelomonocytic leukemia, and myelodysplastic syndrome.11  JAK2 has also been implicated in the formation of tyrosine kinase fusion genes in a variety of hematologic malignancies, mainly acute leukemias.12  The fusion proteins show a common mechanism of constitutive activation, in which JAK2’s 3′ kinase domain is translocated to a partner gene that confers oligomerization properties, namely BCR, PCM1, ETV6, PAX5, RPN1, or SSBP2.13  Increasing evidence of the involvement of JAK2 in various forms of leukemia has suggested that JAK2 might be an essential component of Breakpoint Cluster Region - Abelson 1 (BCR-ABL1)–driven leukemogenesis.14 

The BCR-ABL1 oncogene results from the t(9;22)(q34;q11) reciprocal translocation generating the Philadelphia chromosome.15,16  BCR-ABL1+ chronic myeloid leukemia (CML) is a stem cell–derived disease that progresses in 3 distinct phases: chronic phase (CP), which may last for several years; accelerated phase (AP); and finally blast crisis (BC), which is refractory to therapy.17  CML patients have an excellent treatment option based on the small molecule inhibitor imatinib mesylate and related substances. However, these substances largely prevent expression of symptoms rather than addressing the cause of the disease. Curing CML would require the eradication of the cancer stem cell expressing BCR-ABL1. Remarkably, 6 patients initially treated with interferon (IFN)-α but subsequently switched to imatinib mesylate showed a surprisingly high rate of complete long-term remission,18  suggesting that there are unknown mechanisms of disease eradication. Support for this idea stems from studies that described patients who remained free of symptoms on tyrosine kinase inhibitor (TKI) discontinuation.19  These patients may be cured in the sense of eradication of all CML cells, but this is impossible to prove. Although eradication of all CML cells remains the ideal, operational cure could be achieved even with detectable residual disease, if the relapse risk is close to zero. Despite this glimmer of hope, a considerable number of patients are resistant to the inhibitors imatinib, nilotinib, and dasatinib, which have been approved for first-line therapy of BCR-ABL1. The appearance of the T315I gatekeeper mutation puts an end to these treatment options because it is resistant to all first- and second-generation TKIs. The recent approval of the third-generation inhibitor ponatinib by the US Food and Drug Administration offers the possibility to treat such cases, although ponatinib is also likely to suffer from limitations: its effectiveness may be constrained by the development of multi-TKI resistance or BCR-ABL1–independent resistance.20  The search for additional therapeutic targets will thus remain an important task.

The recent introduction of JAK2-specific inhibitors has coincided with the appearance of a number of excellent reviews summarizing the state-of-the-art knowledge of how JAK2V617F-mutated (and nonmutated) patients may benefit.21-24  The reports have raised the issue of whether and how the new substances may influence BCR-ABL1–targeted therapy in CML. The precise role of the JAK2-STAT5 proteins in the pathogenesis, maintenance, and progression of CML has been a matter of debate for more than a decade. In this review, we summarize the current understanding, focusing on the role of JAK2 as this protein is the object of intense discussions as a possible therapeutic option for CML patients.

The longstanding lack of appropriate transgenic Stat5 knockout mouse models has limited experimental efforts because deletion of the Stat5 gene is associated with high perinatal lethality from anemia and lung abnormalities.25  The few surviving Stat5-deficient mice show normal levels of hematopoietic stem cells (HSCs) but exhibit severe lymphoid and moderate myeloid repopulation defects.26  Overexpression of a constitutively active STAT5 protein in total bone marrow and long-term HSCs suffices to induce CML that closely resembles a BCR-ABL1–induced disease.27,28  The expression of dominant negative STAT529-32  and RNA interference–mediated knockdown of STAT533-35  in cell lines and primary patient samples strengthened the evidence that STAT5 has an essential role in CML.

Only with the introduction of more advanced molecular methods did it become possible to recombine the Stat5 locus in adult mice using a unique conditional-null allele.25  Deletion of Stat5 is well tolerated in adult mice and has almost no effect on hematopoiesis. The availability of inducible Stat5-deleted mice enabled us to investigate the role of STAT5 in BCR-ABL1–mediated CML leukemogenesis.

A study conducted in the Van Etten laboratory describes the transplantation of bone marrow transduced with a retrovirus encoding for BCR-ABL1 into lethally irradiated recipient mice.36  This well-characterized procedure induces a CML-like leukemia that originates from stem/progenitor cells with multilineage repopulating activity and can progress to BC. Complete deletion of the Stat5 gene locus using the conditional-null allele prevented the development of myeloid or lymphoid leukemia in primary recipients,37  despite the persistence of BCR-ABL1–expressing HSCs. The self-renewal capacity of the BCR-ABL1–expressing Stat5-deficient HSCs was tested by serial transplantation. The BCR-ABL1+Stat5-null bone marrow conferred radioprotection and allowed myeloid engraftment, although all secondary recipients succumbed to fatal acute lymphoblastic leukemia. This indicated that BCR-ABL1–expressing Stat5-deficient HSCs possess the ability to self-renew and that loss of Stat5 does not prevent the outgrowth of transformed lymphoid cells.37 

An independent study by Hoelbl et al38  used a slightly different technical approach. The disease was established via BCR-ABL1 transduction/transplantation before inducing deletion of Stat5 and led to a massive reduction of BCR-ABL1–expressing cells, to the point where neoplastic cells could no longer be detected and signs of disease vanished. The disease eventually reappeared, caused by the outgrowth of STAT5-expressing “escaper” clones. Secondary recipients only engrafted with Stat5 wild-type cells and failed to engraft with the Stat5-deleted population. The result may be interpreted in 2 ways. Either STAT5 is required for the engraftment and repopulation of BCR-ABL1+ leukemia in secondary recipients, in contrast to previous results, or Stat5 wild-type leukemic stem cells harbor a survival advantage and rapidly outcompete Stat5-null leukemic stem cells. Hoelbl et al38  also found no outgrowth of Stat5-deficient BCR-ABL1+ lymphoid cells: deletion of Stat5 in lymphoid BCR-ABL1+ cells was incompatible with cell viability.

There is little doubt of the requirement for STAT5 in the establishment of a CML-like leukemia. However, it is not clear whether deletion of Stat5 leads to eradication of the BCR-ABL1+ HSCs or whether the stem cells persist and allow progression to lymphoid BC. The discrepancy in the results from the 2 groups may stem from differing experimental setups (Table 1) such as susceptibilities to leukemogenesis in the mouse strains used: whereas the Sexl laboratory worked with C57BL/6J mice, the Van Etten laboratory used Balb/c animals, which are more prone to lymphoid malignancies. The debate on the potential of STAT5 inhibition to block CML stem cells and lymphoid expansion will only be settled by the development of STAT5 inhibitors and their use in human patients.

Table 1

Similarities and differences in the experimental setup used to determine the in vivo effect of Stat5 deficiency in BCR-ABL1–induced leukemogenesis

Experimental setupWalz et alHoelbl et al
Mouse strain Balb/c C57/B6 
Time point of STATS deletion Disease induction Established disease 
Viral titer* Higher Lower 
Number of cells injected (intravenously) 5 × 105 1 × 106 
Induction of Mx1-Cre transgene plpC 250 μg (4×) pIpC 400μg (l×)/IFN-β (1000 U/mL) 
Age of donor and recipient mice 6 wk 6 wk 
5-FU treatment dose 150 mg/kg 150 mg/kg 
Retroviral vector pMSCV-p210-IRES-eGFP pMSCV-p210-IRES-eGFP 
Experimental setupWalz et alHoelbl et al
Mouse strain Balb/c C57/B6 
Time point of STATS deletion Disease induction Established disease 
Viral titer* Higher Lower 
Number of cells injected (intravenously) 5 × 105 1 × 106 
Induction of Mx1-Cre transgene plpC 250 μg (4×) pIpC 400μg (l×)/IFN-β (1000 U/mL) 
Age of donor and recipient mice 6 wk 6 wk 
5-FU treatment dose 150 mg/kg 150 mg/kg 
Retroviral vector pMSCV-p210-IRES-eGFP pMSCV-p210-IRES-eGFP 

IFN-β, interferon β; pIpC, polyinosinic polycytidylic acid; 5-FU, 5-fluorouracil.

*

V Sexl and R Van Etten, personal communication, 2010.

Further work has identified STAT5 not only as an integral player in CML pathogenesis but also as an important modulator in the response of BCR-ABL1–expressing cells to therapy with kinase inhibitors. Whereas low levels of STAT5 protein are associated with increased sensitivity of BCR-ABL1+ cells to imatinib in vitro, enhanced STAT5 expression leads to a reduction of imatinib-induced cytotoxicity.34  These results have been confirmed in vivo: mice injected with Abelson virus (v-ABL)–transformed cells acquired resistance to imatinib treatment if STAT5 was ectopically expressed. STAT5 mRNA expression and protein levels are consistently increased in more advanced phases of CML, as well as in samples from TKI-resistant patients. The emergence of imatinib resistance is strictly dependent on the transcriptional activity of STAT5 and may be mediated by increased expression of the antiapoptotic STAT5 downstream target genes BclXL and Bcl-2, possibly building up a barrier against apoptosis and cytotoxicity.34  A recent publication described a highly significant correlation between the level of STAT5A mRNA and the occurrence of BCR-ABL1 mutations in a cohort of 50 CML patients, possibly mediated by the enforced production of reactive oxygen intermediates.39  Further support for a link between STAT5 activity and TKI response is provided by a recent phosphoprotein-profiling study that found a significant correlation between the level of phosphorylated STAT5 and the response to TKI treatment.40  It will be of interest to study whether and how STAT3 induces TKI resistance as STAT3 can compensate for STAT5 under certain circumstances, and there is preliminary evidence to implicate STAT3 in drug resistance in CML.41,42 

Initial evidence for the involvement of JAK signaling downstream of the Abelson oncogene dates to 1995, when Danial et al43  reported a physical interaction of v-ABL with JAK1 and JAK3. Using a temperature-sensitive mutant of v-ABL, they showed a tight correlation of JAK activity with the presence of oncogenic Abelson tyrosine kinase. One year later, BCR-ABL1 was shown to phosphorylate JAK2 constitutively in cell lines expressing p210BCR-ABL1. These observations sparked considerable interest, and it was not long before JAK kinases were being discussed as potential therapeutic targets in hematological malignancies including CML.44,45  Subsequently, activation of JAK2 was verified in several human and murine cell lines expressing distinct forms of BCR-ABL1, as well as in leukemic cells derived from CML patients.46  Imatinib treatment of CML cell lines was able to reduce JAK2 tyrosine phosphorylation, substantiating the link between BCR-ABL1 and JAK2 activity.46  Over the past years, the “signallosome” surrounding BCR-ABL1 has been discovered, and coimmunoprecipitation experiments in murine and human BCR-ABL1+ cell lines have shown JAK2 to be 1 of the components.

JAK2 interacts physically with the C terminus of BCR-ABL1, whereas the SH2 domain of BCR-ABL1 is required for the efficient phosphorylation of JAK2 on tyrosine residue Y1007, a prerequisite for JAK2 activation.46  It has been proposed that the BCR-ABL1/JAK2 complex is essential for full-blown v-myc myelocytomatosis viral oncogene homolog (c-MYC) induction downstream of BCR-ABL1 by 3 independent mechanisms. First, JAK2 increases c-MYC mRNA levels by phosphorylating v-akt murine thymoma viral oncogene homolog, thereby causing deactivation of glycogen synthase kinase-3β, a negative regulator of c-MYC expression.47  This process may involve β-catenin as glycogen synthase kinase-3β–mediated phosphorylation of β-catenin causes its degradation, which leads to the down-regulation of target genes such as cyclin D1, c-JUN, and c-MYC.48-50  Second, JAK2 activation maintains a high level of c-MYC protein by inhibiting ubiquitin/26S proteasome-dependent degradation.51  Finally, JAK2 deactivates the phosphatase protein phosphatase 2A (PP2A) (see below), preventing c-MYC’s dephosphorylation and degradation.52,53  The phosphorylation of BCR-ABL1 and JAK2 is reciprocal. Besides v-Src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (SRC) kinase family members like v-yes-1 Yamaguchi sarcoma viral related oncogene homolog (LYN),54-58  JAK2 is able to phosphorylate BCR-ABL1 on tyrosine residue 177.52  This particular residue is critical for BCR-ABL1–induced disease maintenance as it allows binding of the SH2/SH3 domain-containing growth factor receptor-bound protein 2 (GRB2) protein and the rat sarcoma (RAS)-activating nucleotide exchange factor son-of-sevenless (SOS), critical components of the pathway by which tyrosine kinases induce RAS activation.59,60  Already 20 years ago, GRB2 and SOS were shown to link BCR-ABL1 activity to mitogen-activated protein kinase signaling.61,62  GRB2 directly binds BCR-ABL1 via its SH2 domain, resulting in a BCR-ABL1-GRB2-SOS complex that activates RAS. The GRB2 SH2 domain also allows binding to other phosphorylated proteins such as the receptor tyrosine kinase Src-homology collagen protein, which induces phosphatidylinositol 3-kinase signaling, providing a link of BCR-ABL1 to this essential survival pathway and allowing GRB2-independent RAS activation.62-66  Another protein found in a complex with BCR-ABL1/JAK2 is the proto-oncogene Abelson helper integration site 1 (Ahi-1). The enforced expression of Ahi-1 in hematopoietic cells suffices to induce a leukemic phenotype in vivo and collaborates with BCR-ABL1 to drive an aggressive form of leukemia.67  Ahi-1 not only enhances BCR-ABL1–dependent transformation but also reduces the TKI response of CML stem/progenitor cells, which can be overcome by combined treatment with JAK2 inhibitors.67,68 

Suppression of the phosphatase PP2A has a central role in the pathogenesis of CML. PP2A activity is substantially impaired in CML-CP and barely detectable in CML-BC.69  BCR-ABL1–mediated inhibition of PP2A is crucial for the leukemic cells because PP2A, if active, would counteract and block BCR-ABL1 signaling via the downstream tyrosine phosphatase SHP1. SHP1 is capable of dephosphorylating and thus deactivating BCR-ABL1.69  The kinase BCR-ABL1 and the phosphatase PP2A share common targets; they both regulate v-akt murine thymoma viral oncogene homolog, mitogen-activated protein kinase, LYN, c-MYC, RB, STAT5, and JAK2. In BCR-ABL1+ cells, PP2A inhibition is achieved by BCR-ABL1–dependent up-regulation of the inhibitor proteins cancerous inhibitor of protein phosphatase 2A70  and SET nuclear oncogene (SET), a nuclear/cytoplasmic phospho-protein overexpressed in solid and hematological malignancies.71,72  The upregulation and activation of these proteins support CML cells to circumvent apoptosis.69,73  Accordingly, SET knockdown and PP2A-activating drugs restore PP2A activity and decrease BCR-ABL1 expression and activity, leading to apoptosis in CML cells.69  Jak2 TKI treatment and knockdown of JAK2 reduced SET protein levels, leading to the concept that JAK2 directly regulates SET.74  These studies defined PP2A deactivation as a key signaling event downstream of the BCR-ABL1-JAK2 axis. In addition, the SRC kinase LYN was identified as a JAK2 target regulated by the SET-PP2A-SHP1 pathway (Figure 1).

Figure 1

The BCR-ABL1-JAK2-PP2A network. The scheme depicts how the BCR-ABL1-JAK2–mediated up-regulation of the phosphatase SET helps to maintain BCR-ABL1 activity, BCL2 expression, and MYC stability.

Figure 1

The BCR-ABL1-JAK2-PP2A network. The scheme depicts how the BCR-ABL1-JAK2–mediated up-regulation of the phosphatase SET helps to maintain BCR-ABL1 activity, BCL2 expression, and MYC stability.

Close modal

The key role of JAK2 downstream of BCR-ABL1 was underlined by monitoring proliferation, apoptosis, and tumorogenicity of CML cells after treatment with JAK2 TKIs. The JAK2 inhibitor AG490 induced cell death in a dose-dependent manner in 32Dp210 and K562 cells, as well as in imatinib-resistant BCR-ABL1+ Ba/F3 cells.47,51  This finding supported the idea that JAK2 inhibitors might represent a novel way to treat imatinib-resistant CML patients.52  Colony formation of 32Dp210 cells that were imatinib sensitive or resistant to imatinib was drastically reduced on AG490 treatment. Similar results were obtained with the JAK2 inhibitor HBC90.74  Importantly, leukemic cells derived from CML patients in CP, AP, and BC underwent apoptosis on treatment with 1 of these inhibitors, irrespective of whether they were sensitive to imatinib.74  Clinical studies have only recently become possible with the availability of more specific JAK2 inhibitors (Table 2).75-86  The IC50 values of TG101209 (a precursor of TG101348 currently in clinical studies for use in the treatment of JAK2V617F+ MPN) required to induce apoptosis in imatinib-sensitive and -resistant murine and human cell lines are within the low micromolar range, approaching values that may be reached in patients.74  Moreover, CD34+ cells derived from CP and BC imatinib-resistant CML patients proved sensitive to TG101209 treatment.52  Treatment of leukemic mice with JAK2 inhibitors induced a significant therapeutic response.52  It should be noted that all cell viability studies were undertaken with JAK2 TKIs: to date, no studies have used small interfering/short hairpin RNA–mediated knockdown of JAK2.

Table 2

JAK kinase specificity profile and clinical trials of distinct TKIs

Drug nameIC50 (nM)Clinical trialReference
JAK1JAK2JAK3TYK2
INCB18424 (Ruxolitinib) 3.3 2.8 428 19 FDA approved for MF; phase II for CML-BC; recruiting for phase I/II for CML under nilotinib treatment 75 
TQ101348 (SAR302503) 115 1002 405 Phase I/II in MPN 76 
CYT387 11 18 155 NA Phase I/II in MPN 77 
SB1518 (Pacritinib) 1280 23 520 50 Phase II in MPN 78 
CEP7O1 (Lestaurtinib) NA 0.9 NA Phase II in MPN 79 
LY2784544 NA 48 NA Phase I ongoing in MPN 80 
NS-018 33 39 22 Phase I/II ongoing in MPN 81 
AZD1480 1.3 0.3 3.9 NA Phase I/II ongoing in MPN 82 
BMS-911543 356 73 66 Phase I/II ongoing in MPN 83 
LY3009104 (Baricitinib) 5.9 5.7 560 53 Phase II ongoing in rheumatoid arthritis 84 
TG101209 NA 169 NA In vitro use only 85 
JAK inhibitor 1 15 In vitro use only 86 
Drug nameIC50 (nM)Clinical trialReference
JAK1JAK2JAK3TYK2
INCB18424 (Ruxolitinib) 3.3 2.8 428 19 FDA approved for MF; phase II for CML-BC; recruiting for phase I/II for CML under nilotinib treatment 75 
TQ101348 (SAR302503) 115 1002 405 Phase I/II in MPN 76 
CYT387 11 18 155 NA Phase I/II in MPN 77 
SB1518 (Pacritinib) 1280 23 520 50 Phase II in MPN 78 
CEP7O1 (Lestaurtinib) NA 0.9 NA Phase II in MPN 79 
LY2784544 NA 48 NA Phase I ongoing in MPN 80 
NS-018 33 39 22 Phase I/II ongoing in MPN 81 
AZD1480 1.3 0.3 3.9 NA Phase I/II ongoing in MPN 82 
BMS-911543 356 73 66 Phase I/II ongoing in MPN 83 
LY3009104 (Baricitinib) 5.9 5.7 560 53 Phase II ongoing in rheumatoid arthritis 84 
TG101209 NA 169 NA In vitro use only 85 
JAK inhibitor 1 15 In vitro use only 86 

NA, not available.

Interpretation of experiments with inhibitors is complicated by the fact that all of them hit >1 target,87,88  so dose-dependent effects on the off-targets need to be taken into consideration. To test the role of JAK2 in BCR-ABL1–induced leukemogenesis, we generated complete knockout and conditional Jak2 mice.89,90  Although JAK2 was essential for the initial transformation of lymphoid cells by v-ABL and p185BCR-ABL1, the initial transformation of myeloid cells by p210BCR-ABL1 was unaffected by the lack of JAK291  To investigate the role of JAK2 in the maintenance and survival of BCR-ABL1+ cells, we generated Jak2fl/fl mx1-Cre–positive cell lines. The CRE-mediated deletion of Jak2 in either lymphoid or myeloid BCR-ABL1+ cell lines had no impact on cell proliferation, cell cycle progression, or induction of apoptosis.91  In line with these in vitro findings, we observed no differences in disease latency on deletion of Jak2 in vivo.91  The experiment should be interpreted with caution: no long-term studies were performed, so we cannot exclude the possibility that JAK2 TKI inhibition provokes CML stem cell exhaustion; we cannot be certain that our transgenic mice represent a true model of the human disease or that retroviral infections faithfully mimic disease development; and the generation of gene-targeted mice might interfere with microRNAs that contribute to disease development. Despite these caveats, the results cast doubt on the significance of the proposed BCR-ABL1/JAK2 network for CML cell survival and proliferation.

All studies supporting the conclusion that JAK2 has a central role in CML cell survival relied on the use of JAK2 TKIs, whose off-target effects would provide an obvious explanation for the effects. We tested the effects of a panel of 5 distinct JAK2 TKIs (AG490,92  JAK inhibitor I, TG101209,85  TG101348,76  and INCB-01842475 ) on wild-type and Jak2-deficient BCR-ABL1+ cell lines. Three of the inhibitors (AG490, TG101209, and TG101348) induced cell death in BCR-ABL1+ cells, irrespective of whether JAK2 was expressed.91  As JAK inhibitor I in the concentration applied is a potent pan-JAK inhibitor86  and had no impact on CML cell survival, it is safe to conclude that JAKs are not involved in apoptosis induction by JAK inhibitors but that other off–targets induce cell death in BCR-ABL1–transformed cells, accounting for the discrepant observations. Remarkably, all 3 inhibitors that induced CML cell death were also able to inhibit BCR-ABL1 kinase activity. The choice of JAK2 TKI thus determines the outcome of the experiment. We consistently found that only inhibitors that target and inhibit BCR-ABL1 kinase were able to induce apoptosis in the low micromolar range (≤2 µM). JAK2 TKIs that do not target BCR-ABL1 failed to do so (JAK inhibitor I and INCB-018424).91 

Several investigations of the role of JAK2 in STAT5 activation in CML have reached similar conclusions: phosphorylation of tyrosine residue 694 and the resulting activation of STAT5 are independent of JAK2 in BCR-ABL1+ cells. An early study found STAT1 and STAT5 constitutively phosphorylated in BCR-ABL1–transformed cell lines but failed to detect a parallel increase in the steady-state tyrosine phosphorylation of JAK kinases, which is indicative of their activation.93  A similar mismatch between the levels of constitutive JAK2 and STAT5 tyrosine phosphorylation in BCR-ABL1–transformed Ba/F3 cells was observed by Ilaria and Van Etten; the extent of STAT5 activation was comparable to that in interleukin (IL)-3–stimulated maternal Ba/F3 cells, whereas JAK activation by BCR-ABL1 was considerably lower.94  Most importantly, the expression of dominant negative JAK2 mutants failed to interfere with the constitutive activation of STAT5 in BCR-ABL1+ cells but significantly decreased IL-3–dependent STAT5 activation.46,94  Evidence that SRC kinases are involved in the pathogenesis of BCR-ABL1–driven leukemia came from the finding of activated SRC kinases in a complex with BCR-ABL1 in myeloid cells.54,95  Although hemopoietic cell kinase (HCK) was shown to phosphorylate STAT5B on tyrosine 699 in murine 32D cells transfected with BCR-ABL1, the HCK STAT5B link could not be confirmed in human CML cell lines.91  As the SRC inhibitors PP1 and CGP76030 do not impact pSTAT5 levels in 32D cells expressing BCR-ABL1-T315I, the inhibition of pSTAT5 in wild-type BCR-ABL1–expressing cells is most likely caused by off-target effects.96  However, a role for SRC kinases in STAT5 activation cannot be entirely excluded; in murine Ph+ lymphoid leukemia, the SRC family kinases LYN, HCK, and Gardner-Rasheed feline sarcoma viral (V-Fgr) oncogene homolog are activated by p185BCR-ABL1.97  Disease latency was significantly enhanced in p185BCR-ABL1-triggered disease on deletion of ≥2 of these SRC kinases in genetically modified mice. In contrast, the induction of CML was not affected.

Our data support the concept that signaling in BCR-ABL1+ cells is “rewired” and that STAT5 activation becomes uncoupled from JAK2. The deletion of both Jak2 alleles did not affect the pSTAT5 level in BCR-ABL1–transformed cells. Moreover, treatment of BCR-ABL1–expressing Ba/F3 cells with the JAK2 TKIs INCB-018428, JAK inhibitor I, TG101209, and TG101348 (at a dosage that does not interfere with BCR-ABL1) did not alter the level of tyrosine-phosphorylated STAT5, despite abolishing IL-3– and JAK2-dependent STAT5 phosphorylation in the parental cells.91  siRNA-mediated knockdown of all 4 JAK kinases individually or in combination in Ku812 and K562 cells failed to change pSTAT5 levels. Finally, in vitro ABL kinase assays revealed a Km for STAT5 of ∼100 µM, within the range for the well-defined BCR-ABL1 target V-Crk sarcoma virus CT10 oncogene homolog (CRKL) under identical experimental conditions.91  The BCR-ABL1 target and adaptor protein CRKL physically interacts with BCR-ABL1 via its SH3 domain98  and is required for BCR-ABL1–induced STAT5 phosphorylation.99  It is attractive to speculate that BCR-ABL1 interacts with STAT5 via CRKL, although convincing experimental evidence is still lacking.

Our data show that JAK2 is not essential for CML induced by retrovirally expressed BCR-ABL1. Nevertheless, it is possible that the canonical JAK2-STAT5 pathway is important for more primitive CML stem/progenitor cells that may rely on cytokine-activated JAK-STAT signaling in addition to BCR-ABL1 signaling. It has been postulated that “sanctuaries” such as the bone marrow provide a protective environment, thereby accounting for CML stem cell resistance to TKIs. There is a general consensus that TKIs inhibit BCR-ABL1 activity in primitive lineage CD34+CD38 cells.100,101  In contrast to progenitor cells, CML stem cells are able to survive in vitro for prolonged periods of time despite complete oncogene inactivation.101  To cure a patient, it is necessary to eliminate cells that are either not or only partially dependent on BCR-ABL1 signaling. It is conceivable that the bone marrow microenvironment contains a distinct milieu of cytokines and growth factors that allow BCR-ABL1–independent survival and thus TKI drug resistance. In the presence of cytokines, short-term BCR-ABL1 kinase inhibition with 100 nM dasatinib fails to reduce CD34+-dependent colony formation. Remarkably, the inhibition of JAK activity using JAK inhibitor I re-established the sensitivity of CML progenitors to BCR-ABL1 inhibition despite the presence of cytokines.102  Similarly, K562 cells become resistant to BCR-ABL1 TKIs when cultured in bone marrow stroma–derived conditioned medium (CM), highlighting the importance of the microenvironment.41  Stroma-induced drug resistance correlates with increased pSTAT3 levels.41  Reducing the expression of JAK2 and TYK2 by siRNA or inhibiting JAK kinase activity by INCB-018424 blocked CM-mediated STAT3 activation and sensitized CML cells to nilotinib-mediated cell death.42  Accordingly, the combined treatment of Lin34+ patient-derived cells with INCB-018242 and nilotinib potentiated cell death and apoptosis when cocultured with bone marrow stromal cells.42 

A recent study highlights the influence of the microenvironment for the survival of CML cells.103  High concentrations of the JAK2-activating factors IL-6, granulocyte–colony-stimulating factor, and granulocyte-macrophage–colony-stimulating factor (GM-CSF) are present in CM and enhance survival of CML CD34+ cells under imatinib treatment. Only in the presence of CM does the combination of imatinib and the JAK2 inhibitor TG101209 or CYT387 increase apoptosis.103  This observation unequivocally links the CM-protective effect to JAK2 signaling and provides a rationale for combining imatinib and JAK2 TKIs in patient treatment. In a murine CML model, JAK2 inhibition with high doses of TG101209 (200 mg/kg per day) only moderately prolonged survival, suggesting that monotherapy merely delays disease progression. In contrast, combined treatment with nilotinib and high doses of TG101209 was more effective against BCR-ABL1+ cells than nilotinib alone, although the beneficial effect was nullified by toxic effects to nonleukemic cells.103  As combination treatments with low-dose TG101209 (75 mg/kg per day) did not demonstrate any advantage compared with treatment with nilotinib alone, the window for combining JAK2 and ABL inhibitor therapy is probably very narrow.103 

The microenvironment is not the only source of cytokines, and autocrine production of IL-3 and granulocyte–colony-stimulating factor by CD34+ CML cells has been reported. Both cytokines activate STAT5 in a JAK2-dependent manner, which provides an additional rationale for JAK2 TKI treatment in clinical settings.104  Secretion of GM-CSF has also been shown in CD34+ cells derived from imatinib-resistant patients: GM-CSF induces BCR-ABL1–independent activation of JAK2-STAT5 and thus counteracts TKI-based apoptosis. Cotreatment of the cells with a JAK2 inhibitor restores imatinib response.105 

In summary, the 2 opposing opinions may not be as contradictory as they initially appear. There is convincing genetic evidence that JAK2 is not absolutely required for the maintenance of BCR-ABL1+ leukemia; BCR-ABL1 itself appears capable to circumvent the requirement for JAK2 by directly activating the critical downstream transcription factor STAT5 (Figure 2). Nevertheless, it is possible that JAK2 inhibitors may be valuable in the treatment of BCR-ABL1–driven diseases, particularly those involving leukemic stem cells where BCR-ABL1 appears to have a subordinate role and JAK2 could be critical for cell survival (Figure 3). We are currently unable to predict the effects of JAK2 inhibition on stem and progenitor cells, although our limited information suggests a critical role for JAK2-dependent signaling in both cellular compartments.106  Identifying the JAK2 vulnerabilities in stem/progenitor cells and defining the potential therapeutic window remains a major challenge. Further studies are required to understand the combined effects of BCR-ABL1 and JAK2 TKIs in mice and humans. More detailed answers will only result from transplantation of bone marrow samples from treated individuals into NOD-scid IL2rγnull recipients. Two clinical trials are currently planned to recruit CML patients to test the efficacy of the JAK1/2 inhibitior ruxolitinib (INCB-018424) in combination with approved BCR-ABL1 TKIs (http://www.clinicaltrials.gov/ct2/results?term=ruxolitinib&pg=1). It is hoped that the results of the trials will show conclusively whether inhibition of JAK1/2 can benefit CML patients.

Figure 2

CML progenitor cell treatment with BCR-ABL1 TKIs leads to an abrogation of STAT5 signaling essential for survival and proliferation of the cell.

Figure 2

CML progenitor cell treatment with BCR-ABL1 TKIs leads to an abrogation of STAT5 signaling essential for survival and proliferation of the cell.

Close modal
Figure 3

Leukemic stem cell. The presence of a cytokine-enriched microenvironment leads to a BCR-ABL1–independent activation of STAT3 and STAT5 via JAK2. Targeting both pathways via BCR-ABL1 and JAK2 TKIs would interfere with these essential survival signals.

Figure 3

Leukemic stem cell. The presence of a cytokine-enriched microenvironment leads to a BCR-ABL1–independent activation of STAT3 and STAT5 via JAK2. Targeting both pathways via BCR-ABL1 and JAK2 TKIs would interfere with these essential survival signals.

Close modal

The authors thank Graham Tebb for scientific discussions and critical reading of the manuscript.

Work in the laboratory of V.S. was supported by the Austrian Science Fund (FWF).

Contribution: W.W., C.W., and V.S. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

The current affiliation for W.W. is Cambridge Institute for Medical Research and Department of Haematology, University of Cambridge, Cambridge, United Kingdom.

Correspondence: Veronika Sexl, Veterinaerplatz 1, A-1210 Vienna, Austria; e-mail: veronika.sexl@vetmeduni.ac.at.

1
Stark
 
GR
Darnell
 
JE
The JAK-STAT pathway at twenty.
Immunity
2012
, vol. 
36
 
4
(pg. 
503
-
514
)
2
Schindler
 
C
Plumlee
 
C
Inteferons pen the JAK-STAT pathway.
Semin Cell Dev Biol
2008
, vol. 
19
 
4
(pg. 
311
-
318
)
3
O’Shea
 
JJ
Murray
 
PJ
Cytokine signaling modules in inflammatory responses.
Immunity
2008
, vol. 
28
 
4
(pg. 
477
-
487
)
4
Giordanetto
 
F
Kroemer
 
RT
Prediction of the structure of human Janus kinase 2 (JAK2) comprising JAK homology domains 1 through 7.
Protein Eng
2002
, vol. 
15
 
9
(pg. 
727
-
737
)
5
Ihle
 
JN
Nosaka
 
T
Thierfelder
 
W
Quelle
 
FW
Shimoda
 
K
Jaks and Stats in cytokine signaling.
Stem Cells
1997
, vol. 
15
 
Suppl 1
(pg. 
105
-
111
)
6
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
)
7
Baxter
 
EJ
Scott
 
LM
Campbell
 
PJ
, et al. 
Cancer Genome Project
Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders.
Lancet
2005
, vol. 
365
 
9464
(pg. 
1054
-
1061
)
8
James
 
C
Ugo
 
V
Le Couédic
 
JP
, et al. 
A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera.
Nature
2005
, vol. 
434
 
7037
(pg. 
1144
-
1148
)
9
Kralovics
 
R
Passamonti
 
F
Buser
 
AS
, et al. 
A gain-of-function mutation of JAK2 in myeloproliferative disorders.
N Engl J Med
2005
, vol. 
352
 
17
(pg. 
1779
-
1790
)
10
Levine
 
RL
Wadleigh
 
M
Cools
 
J
, et al. 
Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis.
Cancer Cell
2005
, vol. 
7
 
4
(pg. 
387
-
397
)
11
Steensma
 
DP
Dewald
 
GW
Lasho
 
TL
, et al. 
The JAK2 V617F activating tyrosine kinase mutation is an infrequent event in both “atypical” myeloproliferative disorders and myelodysplastic syndromes.
Blood
2005
, vol. 
106
 
4
(pg. 
1207
-
1209
)
12
Walz
 
C
Cross
 
NC
Van Etten
 
RA
Reiter
 
A
Comparison of mutated ABL1 and JAK2 as oncogenes and drug targets in myeloproliferative disorders.
Leukemia
2008
, vol. 
22
 
7
(pg. 
1320
-
1334
)
13
Jatiani
 
SS
Baker
 
SJ
Silverman
 
LR
Reddy
 
EP
Jak/STAT pathways in cytokine signaling and myeloproliferative disorders: approaches for targeted therapies.
Genes Cancer
2010
, vol. 
1
 
10
(pg. 
979
-
993
)
14
Li
 
WX
Canonical and non-canonical JAK-STAT signaling.
Trends Cell Biol
2008
, vol. 
18
 
11
(pg. 
545
-
551
)
15
Rowley
 
JD
Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining.
Nature
1973
, vol. 
243
 
5405
(pg. 
290
-
293
)
16
Nowell
 
PC
The minute chromosome (Phl) in chronic granulocytic leukemia.
Blut
1962
, vol. 
8
 (pg. 
65
-
66
)
17
Kantarjian
 
HM
Keating
 
MJ
Talpaz
 
M
, et al. 
Chronic myelogenous leukemia in blast crisis. Analysis of 242 patients.
Am J Med
1987
, vol. 
83
 
3
(pg. 
445
-
454
)
18
Rousselot
 
P
Huguet
 
F
Rea
 
D
, et al. 
Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years.
Blood
2007
, vol. 
109
 
1
(pg. 
58
-
60
)
19
Mahon
 
FX
Réa
 
D
Guilhot
 
J
, et al. 
Intergroupe Français des Leucémies Myéloïdes Chroniques
Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial.
Lancet Oncol
2010
, vol. 
11
 
11
(pg. 
1029
-
1035
)
20
O’Hare
 
T
Zabriskie
 
MS
Eiring
 
AM
Deininger
 
MW
Pushing the limits of targeted therapy in chronic myeloid leukaemia.
Nat Rev Cancer
2012
, vol. 
12
 
8
(pg. 
513
-
526
)
21
Vannucchi
 
AM
Guglielmelli
 
P
Tefferi
 
A
Advances in understanding and management of myeloproliferative neoplasms.
CA Cancer J Clin
2009
, vol. 
59
 
3
(pg. 
171
-
191
)
22
Santos
 
FP
Verstovsek
 
S
Breakthroughs in myeloproliferative neoplasms.
Hematology
2012
, vol. 
17
 
Suppl 1
(pg. 
S55
-
S58
)
23
Santos
 
FP
Verstovsek
 
S
JAK2 inhibitors: what’s the true therapeutic potential?
Blood Rev
2011
, vol. 
25
 
2
(pg. 
53
-
63
)
24
Reddy
 
MM
Deshpande
 
A
Sattler
 
M
Targeting JAK2 in the therapy of myeloproliferative neoplasms.
Expert Opin Ther Targets
2012
, vol. 
16
 
3
(pg. 
313
-
324
)
25
Cui
 
Y
Riedlinger
 
G
Miyoshi
 
K
, et al. 
Inactivation of Stat5 in mouse mammary epithelium during pregnancy reveals distinct functions in cell proliferation, survival, and differentiation.
Mol Cell Biol
2004
, vol. 
24
 
18
(pg. 
8037
-
8047
)
26
Hoelbl
 
A
Kovacic
 
B
Kerenyi
 
MA
, et al. 
Clarifying the role of Stat5 in lymphoid development and Abelson-induced transformation.
Blood
2006
, vol. 
107
 
12
(pg. 
4898
-
4906
)
27
Moriggl
 
R
Sexl
 
V
Kenner
 
L
, et al. 
Stat5 tetramer formation is associated with leukemogenesis.
Cancer Cell
2005
, vol. 
7
 
1
(pg. 
87
-
99
)
28
Kovacic
 
B
Hoelbl
 
A
Litos
 
G
, et al. 
Diverging fates of cells of origin in acute and chronic leukaemia.
EMBO Mol Med
2012
, vol. 
4
 
4
(pg. 
283
-
297
)
29
Ilaria
 
RL
Hawley
 
RG
Van Etten
 
RA
Dominant negative mutants implicate STAT5 in myeloid cell proliferation and neutrophil differentiation.
Blood
1999
, vol. 
93
 
12
(pg. 
4154
-
4166
)
30
Sillaber
 
C
Gesbert
 
F
Frank
 
DA
Sattler
 
M
Griffin
 
JD
STAT5 activation contributes to growth and viability in Bcr/Abl-transformed cells.
Blood
2000
, vol. 
95
 
6
(pg. 
2118
-
2125
)
31
de Groot
 
RP
Raaijmakers
 
JA
Lammers
 
JW
Jove
 
R
Koenderman
 
L
STAT5 activation by BCR-Abl contributes to transformation of K562 leukemia cells.
Blood
1999
, vol. 
94
 
3
(pg. 
1108
-
1112
)
32
Huang
 
M
Dorsey
 
JF
Epling-Burnette
 
PK
, et al. 
Inhibition of Bcr-Abl kinase activity by PD180970 blocks constitutive activation of Stat5 and growth of CML cells.
Oncogene
2002
, vol. 
21
 
57
(pg. 
8804
-
8816
)
33
Scherr
 
M
Chaturvedi
 
A
Battmer
 
K
, et al. 
Enhanced sensitivity to inhibition of SHP2, STAT5, and Gab2 expression in chronic myeloid leukemia (CML).
Blood
2006
, vol. 
107
 
8
(pg. 
3279
-
3287
)
34
Warsch
 
W
Kollmann
 
K
Eckelhart
 
E
, et al. 
High STAT5 levels mediate imatinib resistance and indicate disease progression in chronic myeloid leukemia.
Blood
2011
, vol. 
117
 
12
(pg. 
3409
-
3420
)
35
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
)
36
Daley
 
GQ
Van Etten
 
RA
Baltimore
 
D
Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome.
Science
1990
, vol. 
247
 
4944
(pg. 
824
-
830
)
37
Walz
 
C
Ahmed
 
W
Lazarides
 
K
, et al. 
Essential role for Stat5a/b in myeloproliferative neoplasms induced by BCR-ABL1 and JAK2(V617F) in mice.
Blood
2012
, vol. 
119
 
15
(pg. 
3550
-
3560
)
38
Hoelbl
 
A
Schuster
 
C
Kovacic
 
B
, et al. 
Stat5 is indispensable for the maintenance of bcr/abl-positive leukaemia.
EMBO Mol Med
2010
, vol. 
2
 
3
(pg. 
98
-
110
)
39
Warsch
 
W
Grundschober
 
E
Berger
 
A
, et al. 
STAT5 triggers BCR-ABL1 mutation by mediating ROS production in chronic myeloid leukaemia.
Oncotarget
2012
, vol. 
3
 
12
(pg. 
1669
-
1687
)
40
Jalkanen
 
SE
Lahesmaa-Korpinen
 
AM
Heckman
 
CA
, et al. 
 
Phosphoprotein profiling predicts response to tyrosine kinase inhibitor therapy in chronic myeloid leukemia patients. Exp Hematol. 2012;40(9):705-714
41
Bewry
 
NN
Nair
 
RR
Emmons
 
MF
Boulware
 
D
Pinilla-Ibarz
 
J
Hazlehurst
 
LA
Stat3 contributes to resistance toward BCR-ABL inhibitors in a bone marrow microenvironment model of drug resistance.
Mol Cancer Ther
2008
, vol. 
7
 
10
(pg. 
3169
-
3175
)
42
Nair
 
RR
Tolentino
 
JH
Argilagos
 
RF
Zhang
 
L
Pinilla-Ibarz
 
J
Hazlehurst
 
LA
Potentiation of Nilotinib-mediated cell death in the context of the bone marrow microenvironment requires a promiscuous JAK inhibitor in CML.
Leuk Res
2012
, vol. 
36
 
6
(pg. 
756
-
763
)
43
Danial
 
NN
Pernis
 
A
Rothman
 
PB
Jak-STAT signaling induced by the v-abl oncogene.
Science
1995
, vol. 
269
 
5232
(pg. 
1875
-
1877
)
44
Wilson-Rawls
 
J
Xie
 
S
Liu
 
J
Laneuville
 
P
Arlinghaus
 
RB
P210 Bcr-Abl interacts with the interleukin 3 receptor beta(c) subunit and constitutively induces its tyrosine phosphorylation.
Cancer Res
1996
, vol. 
56
 
15
(pg. 
3426
-
3430
)
45
Wilson-Rawls
 
J
Liu
 
J
Laneuville
 
P
Arlinghaus
 
RB
P210 Bcr-Abl interacts with the interleukin-3 beta c subunit and constitutively activates Jak2.
Leukemia
1997
, vol. 
11
 
Suppl 3
(pg. 
428
-
431
)
46
Xie
 
S
Wang
 
Y
Liu
 
J
, et al. 
Involvement of Jak2 tyrosine phosphorylation in Bcr-Abl transformation.
Oncogene
2001
, vol. 
20
 
43
(pg. 
6188
-
6195
)
47
Samanta
 
AK
Lin
 
H
Sun
 
T
Kantarjian
 
H
Arlinghaus
 
RB
Janus kinase 2: a critical target in chronic myelogenous leukemia.
Cancer Res
2006
, vol. 
66
 
13
(pg. 
6468
-
6472
)
48
Rubinfeld
 
B
Albert
 
I
Porfiri
 
E
Fiol
 
C
Munemitsu
 
S
Polakis
 
P
Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly.
Science
1996
, vol. 
272
 
5264
(pg. 
1023
-
1026
)
49
Hülsken
 
J
Behrens
 
J
Birchmeier
 
W
Tumor-suppressor gene products in cell contacts: the cadherin-APC-armadillo connection.
Curr Opin Cell Biol
1994
, vol. 
6
 
5
(pg. 
711
-
716
)
50
Yost
 
C
Torres
 
M
Miller
 
JR
Huang
 
E
Kimelman
 
D
Moon
 
RT
The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3.
Genes Dev
1996
, vol. 
10
 
12
(pg. 
1443
-
1454
)
51
Xie
 
S
Lin
 
H
Sun
 
T
Arlinghaus
 
RB
Jak2 is involved in c-Myc induction by Bcr-Abl.
Oncogene
2002
, vol. 
21
 
47
(pg. 
7137
-
7146
)
52
Samanta
 
A
Perazzona
 
B
Chakraborty
 
S
, et al. 
Janus kinase 2 regulates Bcr-Abl signaling in chronic myeloid leukemia.
Leukemia
2011
, vol. 
25
 
3
(pg. 
463
-
472
)
53
Arnold
 
HK
Sears
 
RC
Protein phosphatase 2A regulatory subunit B56alpha associates with c-myc and negatively regulates c-myc accumulation.
Mol Cell Biol
2006
, vol. 
26
 
7
(pg. 
2832
-
2844
)
54
Warmuth
 
M
Bergmann
 
M
Priess
 
A
Häuslmann
 
K
Emmerich
 
B
Hallek
 
M
The Src family kinase Hck interacts with Bcr-Abl by a kinase-independent mechanism and phosphorylates the Grb2-binding site of Bcr.
J Biol Chem
1997
, vol. 
272
 
52
(pg. 
33260
-
33270
)
55
Stanglmaier
 
M
Warmuth
 
M
Kleinlein
 
I
Reis
 
S
Hallek
 
M
The interaction of the Bcr-Abl tyrosine kinase with the Src kinase Hck is mediated by multiple binding domains.
Leukemia
2003
, vol. 
17
 
2
(pg. 
283
-
289
)
56
Meyn
 
MA
Wilson
 
MB
Abdi
 
FA
, et al. 
Src family kinases phosphorylate the Bcr-Abl SH3-SH2 region and modulate Bcr-Abl transforming activity.
J Biol Chem
2006
, vol. 
281
 
41
(pg. 
30907
-
30916
)
57
Miething
 
C
Mugler
 
C
Grundler
 
R
Hoepfl
 
J
Bai
 
RY
Peschel
 
C
Duyster
 
J
Phosphorylation of tyrosine 393 in the kinase domain of Bcr-Abl influences the sensitivity towards imatinib in vivo.
Leukemia
2003
, vol. 
17
 
9
(pg. 
1695
-
1699
)
58
Wu
 
J
Meng
 
F
Lu
 
H
, et al. 
Lyn regulates BCR-ABL and Gab2 tyrosine phosphorylation and c-Cbl protein stability in imatinib-resistant chronic myelogenous leukemia cells.
Blood
2008
, vol. 
111
 
7
(pg. 
3821
-
3829
)
59
Feig
 
LA
Cooper
 
GM
Relationship among guanine nucleotide exchange, GTP hydrolysis, and transforming potential of mutated ras proteins.
Mol Cell Biol
1988
, vol. 
8
 
6
(pg. 
2472
-
2478
)
60
Schlessinger
 
J
How receptor tyrosine kinases activate Ras.
Trends Biochem Sci
1993
, vol. 
18
 
8
(pg. 
273
-
275
)
61
Pendergast
 
AM
Quilliam
 
LA
Cripe
 
LD
, et al. 
BCR-ABL-induced oncogenesis is mediated by direct interaction with the SH2 domain of the GRB-2 adaptor protein.
Cell
1993
, vol. 
75
 
1
(pg. 
175
-
185
)
62
Puil
 
L
Liu
 
J
Gish
 
G
, et al. 
Bcr-Abl oncoproteins bind directly to activators of the Ras signalling pathway.
EMBO J
1994
, vol. 
13
 
4
(pg. 
764
-
773
)
63
Pear
 
WS
Miller
 
JP
Xu
 
L
, et al. 
Efficient and rapid induction of a chronic myelogenous leukemia-like myeloproliferative disease in mice receiving P210 bcr/abl-transduced bone marrow.
Blood
1998
, vol. 
92
 
10
(pg. 
3780
-
3792
)
64
Zhang
 
X
Subrahmanyam
 
R
Wong
 
R
Gross
 
AW
Ren
 
R
The NH(2)-terminal coiled-coil domain and tyrosine 177 play important roles in induction of a myeloproliferative disease in mice by Bcr-Abl.
Mol Cell Biol
2001
, vol. 
21
 
3
(pg. 
840
-
853
)
65
Chu
 
S
Li
 
L
Singh
 
H
Bhatia
 
R
BCR-tyrosine 177 plays an essential role in Ras and Akt activation and in human hematopoietic progenitor transformation in chronic myelogenous leukemia.
Cancer Res
2007
, vol. 
67
 
14
(pg. 
7045
-
7053
)
66
Goga
 
A
McLaughlin
 
J
Afar
 
DE
Saffran
 
DC
Witte
 
ON
Alternative signals to RAS for hematopoietic transformation by the BCR-ABL oncogene.
Cell
1995
, vol. 
82
 
6
(pg. 
981
-
988
)
67
Zhou
 
LL
Zhao
 
Y
Ringrose
 
A
, et al. 
AHI-1 interacts with BCR-ABL and modulates BCR-ABL transforming activity and imatinib response of CML stem/progenitor cells.
J Exp Med
2008
, vol. 
205
 
11
(pg. 
2657
-
2671
)
68
Chen
 
M
Gallipoli
 
P
DeGeer
 
D
, et al. 
Targeting primitive chronic myeloid leukemia cells by effective inhibition of a new AHI-1-BCR-ABL-JAK2 complex.
J Natl Cancer Inst
2013
, vol. 
105
 
6
(pg. 
405
-
423
)
69
Neviani
 
P
Santhanam
 
R
Trotta
 
R
, et al. 
The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein.
Cancer Cell
2005
, vol. 
8
 
5
(pg. 
355
-
368
)
70
Lucas
 
CM
Harris
 
RJ
Giannoudis
 
A
Copland
 
M
Slupsky
 
JR
Clark
 
RE
Cancerous inhibitor of PP2A (CIP2A) at diagnosis of chronic myeloid leukemia is a critical determinant of disease progression.
Blood
2011
, vol. 
117
 
24
(pg. 
6660
-
6668
)
71
Carlson
 
SG
Eng
 
E
Kim
 
EG
Perlman
 
EJ
Copeland
 
TD
Ballermann
 
BJ
Expression of SET, an inhibitor of protein phosphatase 2A, in renal development and Wilms’ tumor.
J Am Soc Nephrol
1998
, vol. 
9
 
10
(pg. 
1873
-
1880
)
72
Fornerod
 
M
Boer
 
J
van Baal
 
S
, et al. 
Relocation of the carboxyterminal part of CAN from the nuclear envelope to the nucleus as a result of leukemia-specific chromosome rearrangements.
Oncogene
1995
, vol. 
10
 
9
(pg. 
1739
-
1748
)
73
Perrotti
 
D
Neviani
 
P
ReSETting PP2A tumour suppressor activity in blast crisis and imatinib-resistant chronic myelogenous leukaemia.
Br J Cancer
2006
, vol. 
95
 
7
(pg. 
775
-
781
)
74
Samanta
 
AK
Chakraborty
 
SN
Wang
 
Y
, et al. 
Jak2 inhibition deactivates Lyn kinase through the SET-PP2A-SHP1 pathway, causing apoptosis in drug-resistant cells from chronic myelogenous leukemia patients.
Oncogene
2009
, vol. 
28
 
14
(pg. 
1669
-
1681
)
75
Quintás-Cardama
 
A
Vaddi
 
K
Liu
 
P
, et al. 
Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms.
Blood
2010
, vol. 
115
 
15
(pg. 
3109
-
3117
)
76
Wernig
 
G
Kharas
 
MG
Okabe
 
R
, et al. 
Efficacy of TG101348, a selective JAK2 inhibitor, in treatment of a murine model of JAK2V617F-induced polycythemia vera.
Cancer Cell
2008
, vol. 
13
 
4
(pg. 
311
-
320
)
77
Pardanani
 
A
Lasho
 
T
Smith
 
G
Burns
 
CJ
Fantino
 
E
Tefferi
 
A
CYT387, a selective JAK1/JAK2 inhibitor: in vitro assessment of kinase selectivity and preclinical studies using cell lines and primary cells from polycythemia vera patients.
Leukemia
2009
, vol. 
23
 
8
(pg. 
1441
-
1445
)
78
Hart
 
S
Goh
 
KC
Novotny-Diermayr
 
V
, et al. 
SB1518, a novel macrocyclic pyrimidine-based JAK2 inhibitor for the treatment of myeloid and lymphoid malignancies.
Leukemia
2011
, vol. 
25
 
11
(pg. 
1751
-
1759
)
79
Hexner
 
EO
Serdikoff
 
C
Jan
 
M
, et al. 
Lestaurtinib (CEP701) is a JAK2 inhibitor that suppresses JAK2/STAT5 signaling and the proliferation of primary erythroid cells from patients with myeloproliferative disorders.
Blood
2008
, vol. 
111
 
12
(pg. 
5663
-
5671
)
80
Ma
 
L
Clayton
 
JR
Walgren
 
RA
, et al. 
Discovery and characterization of LY2784544, a small-molecule tyrosine kinase inhibitor of JAK2V617F.
Blood Cancer J
2013
, vol. 
3
 pg. 
e109
 
81
Nakaya
 
Y
Shide
 
K
Niwa
 
T
, et al. 
Efficacy of NS-018, a potent and selective JAK2/Src inhibitor, in primary cells and mouse models of myeloproliferative neoplasms.
Blood Cancer J
2011
, vol. 
1
 
7
pg. 
e29
 
82
Hedvat
 
M
Huszar
 
D
Herrmann
 
A
, et al. 
The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors.
Cancer Cell
2009
, vol. 
16
 
6
(pg. 
487
-
497
)
83
Purandare
 
AV
McDevitt
 
TM
Wan
 
H
, et al. 
Characterization of BMS-911543, a functionally selective small-molecule inhibitor of JAK2.
Leukemia
2012
, vol. 
26
 
2
(pg. 
280
-
288
)
84
van Vollenhoven
 
RF
Small molecular compounds in development for rheumatoid arthritis.
Curr Opin Rheumatol
2013
, vol. 
25
 
3
(pg. 
391
-
397
)
85
Pardanani
 
A
Hood
 
J
Lasho
 
T
, et al. 
TG101209, a small molecule JAK2-selective kinase inhibitor potently inhibits myeloproliferative disorder-associated JAK2V617F and MPLW515L/K mutations.
Leukemia
2007
, vol. 
21
 
8
(pg. 
1658
-
1668
)
86
Thompson
 
JE
Cubbon
 
RM
Cummings
 
RT
, et al. 
Photochemical preparation of a pyridone containing tetracycle: a Jak protein kinase inhibitor.
Bioorg Med Chem Lett
2002
, vol. 
12
 
8
(pg. 
1219
-
1223
)
87
Karaman
 
MW
Herrgard
 
S
Treiber
 
DK
, et al. 
A quantitative analysis of kinase inhibitor selectivity.
Nat Biotechnol
2008
, vol. 
26
 
1
(pg. 
127
-
132
)
88
Davis
 
MI
Hunt
 
JP
Herrgard
 
S
, et al. 
Comprehensive analysis of kinase inhibitor selectivity.
Nat Biotechnol
2011
, vol. 
29
 
11
(pg. 
1046
-
1051
)
89
Wagner
 
KU
Krempler
 
A
Triplett
 
AA
Qi
 
Y
George
 
NM
Zhu
 
J
Rui
 
H
Impaired alveologenesis and maintenance of secretory mammary epithelial cells in Jak2 conditional knockout mice.
Mol Cell Biol
2004
, vol. 
24
 
12
(pg. 
5510
-
5520
)
90
Neubauer
 
H
Cumano
 
A
Müller
 
M
Wu
 
H
Huffstadt
 
U
Pfeffer
 
K
Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis.
Cell
1998
, vol. 
93
 
3
(pg. 
397
-
409
)
91
Hantschel
 
O
Warsch
 
W
Eckelhart
 
E
, et al. 
BCR-ABL uncouples canonical JAK2-STAT5 signaling in chronic myeloid leukemia.
Nat Chem Biol
2012
, vol. 
8
 
3
(pg. 
285
-
293
)
92
Gazit
 
A
Yaish
 
P
Gilon
 
C
Levitzki
 
A
Tyrphostins I: synthesis and biological activity of protein tyrosine kinase inhibitors.
J Med Chem
1989
, vol. 
32
 
10
(pg. 
2344
-
2352
)
93
Carlesso
 
N
Frank
 
DA
Griffin
 
JD
Tyrosyl phosphorylation and DNA binding activity of signal transducers and activators of transcription (STAT) proteins in hematopoietic cell lines transformed by Bcr/Abl.
J Exp Med
1996
, vol. 
183
 
3
(pg. 
811
-
820
)
94
Ilaria
 
RL
Van Etten
 
RA
P210 and P190(BCR/ABL) induce the tyrosine phosphorylation and DNA binding activity of multiple specific STAT family members.
J Biol Chem
1996
, vol. 
271
 
49
(pg. 
31704
-
31710
)
95
Danhauser-Riedl
 
S
Warmuth
 
M
Druker
 
BJ
Emmerich
 
B
Hallek
 
M
Activation of Src kinases p53/56lyn and p59hck by p210bcr/abl in myeloid cells.
Cancer Res
1996
, vol. 
56
 
15
(pg. 
3589
-
3596
)
96
Warmuth
 
M
Simon
 
N
Mitina
 
O
, et al. 
Dual-specific Src and Abl kinase inhibitors, PP1 and CGP76030, inhibit growth and survival of cells expressing imatinib mesylate-resistant Bcr-Abl kinases.
Blood
2003
, vol. 
101
 
2
(pg. 
664
-
672
)
97
Hu
 
Y
Liu
 
Y
Pelletier
 
S
, et al. 
Requirement of Src kinases Lyn, Hck and Fgr for BCR-ABL1-induced B-lymphoblastic leukemia but not chronic myeloid leukemia.
Nat Genet
2004
, vol. 
36
 
5
(pg. 
453
-
461
)
98
Heaney
 
C
Kolibaba
 
K
Bhat
 
A
, et al. 
Direct binding of CRKL to BCR-ABL is not required for BCR-ABL transformation.
Blood
1997
, vol. 
89
 
1
(pg. 
297
-
306
)
99
Seo
 
JH
Wood
 
LJ
Agarwal
 
A
, et al. 
A specific need for CRKL in p210BCR-ABL-induced transformation of mouse hematopoietic progenitors.
Cancer Res
2010
, vol. 
70
 
18
(pg. 
7325
-
7335
)
100
Corbin
 
AS
Agarwal
 
A
Loriaux
 
M
Cortes
 
J
Deininger
 
MW
Druker
 
BJ
Human chronic myeloid leukemia stem cells are insensitive to imatinib despite inhibition of BCR-ABL activity.
J Clin Invest
2011
, vol. 
121
 
1
(pg. 
396
-
409
)
101
Hamilton
 
A
Helgason
 
GV
Schemionek
 
M
, et al. 
Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival.
Blood
2012
, vol. 
119
 
6
(pg. 
1501
-
1510
)
102
Hiwase
 
DK
White
 
DL
Powell
 
JA
, et al. 
Blocking cytokine signaling along with intense Bcr-Abl kinase inhibition induces apoptosis in primary CML progenitors.
Leukemia
2010
, vol. 
24
 
4
(pg. 
771
-
778
)
103
Traer
 
E
MacKenzie
 
R
Snead
 
J
, et al. 
Blockade of JAK2-mediated extrinsic survival signals restores sensitivity of CML cells to ABL inhibitors.
Leukemia
2012
, vol. 
26
 
5
(pg. 
1140
-
1143
)
104
Jiang
 
X
Lopez
 
A
Holyoake
 
T
Eaves
 
A
Eaves
 
C
Autocrine production and action of IL-3 and granulocyte colony-stimulating factor in chronic myeloid leukemia.
Proc Natl Acad Sci USA
1999
, vol. 
96
 
22
(pg. 
12804
-
12809
)
105
Wang
 
Y
Cai
 
D
Brendel
 
C
, et al. 
Adaptive secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF) mediates imatinib and nilotinib resistance in BCR/ABL+ progenitors via JAK-2/STAT-5 pathway activation.
Blood
2007
, vol. 
109
 
5
(pg. 
2147
-
2155
)
106
Park
 
SO
Wamsley
 
HL
Bae
 
K
, et al. 
Conditional deletion of Jak2 reveals an essential role in hematopoiesis throughout mouse ontogeny: implications for Jak2 inhibition in humans.
PLoS ONE
2013
, vol. 
8
 
3
pg. 
e59675
 
Sign in via your Institution