Abstract
This review focuses on topical issues in the biology and treatment of the myeloproliferative neoplasms (MPNs). Studies in transgenic mice suggest that BCR-ABL1 reduces the fraction of self-renewing ‘leukemic’ stem cells in the bone marrow but that some of these cells survive treatment with imatinib. This also seems to operate in humans. Data from models also strongly support the notion that JAK2V617F can initiate and sustain MPNs in mice; relevance to disease in humans is less clear. These data also support the hypothesis that level of JAK2V617F expression influences the MPN phenotype: higher levels favor erythrocytosis whereas lower levels favor thrombocytosis. Although TET2-mutations were thought to precede JAK2V617F in some persons with MPNs, it now appears that TET2 mutations may occur after JAK2V617F. Further understanding of signal-transduction pathways activated in chronic myeloid leukemia suggests various possible targets for new therapies including the WNT/beta catenin, notch and hedgehog pathways. Finally, the clinical role of the new JAK2- and BCR-ABL1-inhibitors is considered. Much further progress is likely in several of these areas soon.Introduction
The advent of tyrosine kinase inhibitors (TKI) has proved to be an extraordinarily important advance in the management of patients with early phase Ph-positive chronic myeloid leukemia and has also influenced the direction of research into the underlying biology of leukemia and other tumors. The identification of the JAK2 mutation in polycythemia vera and other Ph-negative myeloproliferative neoplasms (MPN) has made an extremely important contribution to our understanding of the basic biology of these disorders. There remain, however, many unanswered questions. This paper has three discrete but related themes. First, it reviews some of the murine systems that have been developed in recent years to model the JAK2-positive MPNs and BCR-ABL1-positive chronic myeloid leukemia (CML) with a view to defining those questions that might be answerable with appropriate model systems. Second, it reviews the very recent data relevant to the issue of whether TET2 mutations predispose to development of an MPN or are ‘merely’ secondary events. Third, it summarizes briefly some of the recent results of using new agents to treat the Ph-negative MPN and describes some molecular pathways that could be exploited for therapy in the future.
(a) Transgenic models of JAK2-mutant MPNs
The MPNs and related conditions, many of which are characterized by dysregulated tyrosine kinase (TK) signaling,1 are good candidates for mouse models. These models are typically established by expressing the relevant mutant signaling molecules, e.g. BCR-ABL1 in CML or mutant PTPN11 (in juvenile myelomonocytic leukemia) in mouse hematopoietic cells. There are two common strategies: (1) gene transfer into hematopoietic cells by retro- or lenti-viruses followed by transplantation; and (2) expression via a chromosomal trans-gene (for review see 2). Each method has advantages and disadvantages for modeling and for pre-clinical evaluation of molecularly targeted therapies.
Several previous publications reported data on the retro-viral strategy for modeling MPNs induced by JAK2,3–6 JAK2 exon-12 mutants7 and MPL W515L/K mutants.8 However, the transgenic mouse field is rapidly catching up. Three new transgenic models of JAK2-induced MPNs were presented at the 2009 ASH meeting.9–11 Here, we compare these new models with previously published models and consider what they can teach us about the pathophysiology of seemingly-corresponding MPNs (Table 1).
One of the major advantages of the transgene approach is the ability to express the mutant TK at near physiological levels. This contrasts with retroviral vector models in which relevant oncogenes are expressed at relatively high levels.3 This disparity might be particularly important for JAK2 as studies in cell lines indicate JAK2 must associate with a type-I cytokine receptor (such as EpoR or MPL) for signaling activity.17 Consequently, competition between the retrovirus-encoded mutant JAK2 and endogenous, wild-type JAK2 may influence disease phenotype.18 The first published transgenic model from Skoda and co-workers12 used a novel conditional inverted allele of human JAK2 under the control of the human JAK2–promoter. A constitutively expressed Vav-Cre transgene or the interferon-inducible Mx-Cre transgene was used to flip the transgene into the correct orientation. Depending on whether Cre was expressed at high/sustained levels (Vav-Cre) or at lower/transient levels (Mx-Cre), recombination took place predominantly within or between tandemly repeated transgenes yielding different copy numbers and expression of mutant versus wild-type JAK2 with mRNA ratios of ~0.6 for Vav-Cre and ~1.0 for Mx-Cre. The Vav-Cre;FF (flip-flop) mice had normal hemoglobin and WBC levels but increased platelets. In contrast, the Mx-Cre;FF mice had variable but significant increases in WBCs and platelets and increased hemoglobin (170–210 g/L) and low plasma Epo levels. Skoda and co-workers concluded that lower expression of JAK2 favoured an ET-like phenotype whereas higher expression favored a PV-like phenotype. Subsequently, reports of two transgenic JAK2 models were published wherein mouse or human JAK2 was expressed from an H-2Kb or Vav promoter.13;14 Here, there were significant differences in the phenotype and penetrance between founder mice (Table 1). However, in both models mice developed MPNs with variable degrees of polycythemia and thrombocytosis, extramedullary hematopoiesis, splenomegaly and Epo-independent erythroid colony (EEC) formation. Mice with lower relative levels of mutant JAK2 expression tended towards an ET-like phenotype with predominant thrombocytosis. These data support a correlation between level of JAK2 expression and MPN phenotype.
Founder transgenic mice with constitutive expression of dysregulated TKs (such as BCR-ABL1 and JAK2) show marked phenotype variability. This may be related to different transgene insertion sites and/or to deleterious effects of transgene expression during embryogenesis,19 the consequence of which is to select for decreased transgene expression in survivors.
To circumvent these problems four recent JAK2 models used “knock-in” approaches whereby the mutation was introduced into the normal JAK2 locus, so that the mutant JAK2 would be expressed physiologically. In two models, mutant JAK2 expression was further conditionally activated or regulated by Cre-lox recombination (Table 1). Mice in all four models developed MPN phenotypes. In the model from Golam Mohi and colleagues,9;15 a mouse JAK2 allele was expressed after Mx-Cremediated recombination. Heterozygous and homozygous transgenic mice developed an MPN with poly-cythemia and thrombocytosis with a more marked phenotype in homozygotes. In the model described by Tony Green and co-workers,10 mice expressing a conditional human JAK2 allele developed predominantly an ET-like phenotype with thrombocytosis and moderate polycythemia but not splenomegaly or myelofibrosis. The model reported by Jean-Luc Villeval and colleagues expressed a knock-in mouse JAK2 allele constitutively;11 these mice developed a severe MPN phenotype with polycythemia, thrombocytosis, splenomegaly and myelofibrosis.
Most recently, Ebert and colleagues reported the phenotype of a similar constitutively expressed murine Jak2 knock-in allele, where mice developed fatal MPN with polycythemia and splenomegaly but lacking thrombocytosis and myelofibrosis.16 The MPN was transferred to secondary recipients by transplantation of stem (Lin/Sca/Kit, LSK) cells, but not by committed progenitors.
What can we learn from these diverse mouse models of aberrant JAK2-expression? First, although considerable data suggest that one or more mutation(s) may antedate the JAK2 mutation in persons with MPNs,20 the high prevalence of one or more MPN phenotypes in the knock-in transgenic models coupled with the polyclonal MPN observed in the retroviral models5 strongly suggests that JAK2 can initiate and sustain MPN in mice. Whether this conclusion applies to humans is unknown.
Second, one of the most interesting questions is how one genetic lesion, JAK2, can cause diverse MPN phenotypes. Is dose the answer? For example, in humans homozygosity for JAK2 occurs exclusively in PV, and not in ET.21 In these models, there is support for the concept that expression of JAK2 at levels similar to or higher than endogenous JAK2 is associated with erythrocytosis whereas lower expression levels favor thrombocytosis. In the most recent and more “physiological” knock-in models, JAK2 expression is at levels equal to endogenous JAK2. In the models of Green and Villeval mice developed polycythemia, albeit to different extents.
However, the model reported by Mohi does not fit this paradigm: JAK2 expression was (unexpectedly) only about half that of JAK2 allele yet mice developed polycythemia. Homozygosity increased platelets further (Table 1).
There are various possible reasons for these phenotype differences including divergence between human and mouse JAK2 and variability in mouse strains (which influences the MPN phenotype in retroviral models)5. Lastly, we should recall that these mice differ from humans with MPNs in that the mice lack a normal population of hematopoietic stem cells and are wholly dependent on JAK2–associated hematopoiesis for blood cell production. As always, we can learn much from mouse models but they rarely replicate precisely the disease in humans. In fact, the diversity of MPNs in humans with a seemingly canonical JAK2 mutation far exceeds the aforementioned diversity of mouse models. It is well known that familial MPNs with JAK2 mutation have diverse phenotypes.22 This must mean factors other than JAK2 operate.
(b) Mouse models of Ph–positive MPNs
Mouse models of Ph-positive CML are critical for the understanding of functionally relevant cellular and molecular abnormalities and were developed using three approaches: (1) xenotransplants; (2) retroviral transplantation models; and (3) transgenic mice. Although xenotransplants of human CML cells to mice allow us to study and manipulate human cells in vivo, engraftment of human cells in NOD/SCID or similar mouse strains is generally less efficient than transplants using allogeneic or syngeneic murine cells. Moreover, although transplantation of human CML acute phase cells causes acute leukemia in NOD/SCID mice,23 chronic phase (CP) cells do not cause disease despite obvious engraftment.23;24 In contrast, retroviral expression of BCR-ABL1 in hematopoietic stem and progenitor cells results in versatile mouse models of CML facilitating functional analysis of BCR-ABL1 mutants, identification of critical target genes and cooperating oncogenes and assessment of new therapies.25–28 This topic was reviewed by van Etten.29 In addition, transgenic mouse models were generated that express BCR-ABL1 under the control of different promoters and enhancers. These models shed light on the nature of the CML-initiating cell and CML stem cell biology. Here we update only the transgenic mouse models (P210 BCR-ABL1).
Honda and co-workers crossed Tec promoter-driven BCR-ABL1 transgenic mice with BXH2 mice in a screen for common integration sites and identified a novel cooperating gene, Zfp423.
Coordinate overexpression of BCR-ABL1 and Zpf423 induced acute myeloid and lymphoid (B and T cell) leukemias in mice reminiscent of acute phase.30 These investigators reported a similar outcome by crossing Tec-BCR-ABL1 transgenic mice with Bcl11 or H2AX mice.31
Sánchez-García and co-workers generated transgenic mice expressing BCR-ABL1 under the control of a Sca-1 promoter32 to recapitulate CML as a stem cell disease in mice. Interestingly, embryonic lethality, commonly described previously in BCR-ABL1 transgenic mice,19 was not reported.32 Within 6–18 months, the transgenic mice developed a CML-like disease characterized by mildly increased WBC, increased neutrophils, splenomegaly and hepatomegaly and ultimately acute leukemia resembling the acute phase of CML in humans. The authors reported that cross-breeding of two founder lines shortened development of this phenotype to 1–2 months. BCR-ABL1 expression was confined to the Sca-1 cell population including hematopoietic stem cells (HSCs). However, the fact that a significant proportion of mice developed solid cancers (10% lung adenocarcinoma, 3% liver cancer, 2% gastrointestinal stromal cancers and others) shows that Sca-1 driven oncogene expression in non-hematopoietic cells may hamper some aspects of transgenic mouse modeling.
Koschmieder and co-workers generated BCR-ABL1 expressing mouse strains using the murine stem cell leukemia gene (SCL) 3′ enhancer.33 After inter-crossing SCLtTA and TRE-BCR-ABL1 mice to generate double-transgenic SCLtTA/BCR-ABL1 mice, BCR-ABL1 expression was induced by tetracycline withdrawal. These mice developed a CML-like disease within 30 to 120 days (increased WBC and neutrophils, splenomegaly and granulocyte infiltration of the liver, gut and lung); these features were reversible and re-inducible. These data suggest that developing a CML phenotype requires sustained BCR-ABL1 expression. Furthermore, Schemionek and coworkers from the same group showed that this CP-CML is transplantable into congenic mice.34 The LSK population, but not the Lin/Sca1/Kit progenitor or the granulocyte compartments, contains the CML-initiating cell. BCR-ABL1 reduces long-term repopulating (LT-) HSC numbers in the bone marrow and induces their differentiation. Spleen LT-HSC numbers are not reduced. Further experiments showed that the leukemia-inducing potential of BCR-ABL1 positive bone marrow is compromised by serial transplantation. These data suggest that BCR-ABL1 decreases the fraction of self-renewing stem cells in the bone marrow. Interestingly, this is very similar to a recently developed knock-in mouse model for JAK2 which showed reduced leukemia stem cell (LSK cell) numbers and impaired repopulation potential in the JAK2-positive mice.35 This parallel reinforces important similarities of the JAK2 and BCR-ABL1 oncogenes in vivo. The CML-like disease in transplant recipients is reversible by stopping BCR-ABL1-expression and by giving imatinib.
However, CML-initiating cells survive independently of BCR-ABL1 expression and give rise to recurrent CML when the BCR-ABL1 is re-expressed or imatinib is discontinued.
Zhang and colleagues36 were able to confirm these results. Using quantitative competitive repopulation assays they showed that the frequency of functional HSCs was reduced substantially in BCR-ABL1-positive mice compared to controls. Moreover, although one in 6 Flt3CD150CD48 LSK cells possessed repopulating activity, only one in 80 cells caused leukemia in transplanted mice. This may reflect the presence of BCR-ABL1-negative stem cells and thus allows the study of BCR-ABL1- positive-and –negative hematopoiesis in the same mouse.
This group provided evidence for significant in vivo activity of the combination of imatinib and the nuclear histone deacetylase (HDAC) inhibitors LAQ824 or LBH589.37 BCR-ABL1-transgenic mice treated with imatinib and LBH589 had prolonged leukemia-free survival after discontinuing treatment compared with mice treated with either drug alone. Expression of one of the putative targets, MCL-1, was strongly inhibited by combination therapy, suggesting a role for MCL-1-inhibition in controlling or killing CML stem cells.
Transgenic mice may be valuable to identify molecular target that may prove clinically useful. For example, Perrotti and co-workers showed that RNA binding proteins hnRNA-A1, -K, and –E2 are increased in BCR-ABL1 positive transgenic mice and that this results in SET activation and suppression of protein phosphatase 2A (PP2A).38;39 These authors reported that the PP2A activator, FTY-720, induces apoptosis of BCR-ABL1 positive LSK cells in SCLtTA/BCR-ABL1 mice.39 This work is described in greater detail later in this paper.
Finally, Sengupta and co-workers showed that BCR-ABL1-positive spleen cells can transplant CML-like disease. These cells have decreased adhesion and increased migration compared to BCR-ABL1 negative cells.40 By crossing BCR-ABL1 transgenic mice with Rac2 mice these investigators showed that Rac2 is critical for BCR-ABL1-induced disease and for the proliferation and survival of leukemia stem cells (LSC).40
Together with retroviral transplantation and as xenotransplant models, transgenic mouse models can be exploited in studies of the CML phenotype, of CML stem cell biology, and of new therapeutic strategies.
(c) Role of TET2 in the Ph-negative MPNs
Why is it difficult to identify pre-JAK2 events in MPN? It is quite likely that even if these events are important they have subtle or no clinical consequence by themselves. Shortly after the JAK2 mutation was described studies of clonality of hematopoietic cells suggested the existence of pre-JAK2 mutations, at least in some people.41–43
The first pre-JAK2 event described was deletion of the long arm of chromosome 20 (del[20q]). In some patients with MPNs del(20q) seemed to precede the JAK2 mutation. In others this sequence was reversed.20;44 Thus del(20q) appeared independent and was unlikely to predispose to JAK2mutation. Other data from persons with MPNs and JAK2 mutation whose disease transformed to blastic phase (BP) gave an intriguing picture: leukemia cells from one-half of the cases were JAK2-negative.4;43;45 These data argue for the existence of molecular lesions that might predispose to JAK2 mutations in MPN and/or to other JAK2 independent acute myeloid leukemias.
In 2009, mutations in TET2 and ASXL1 were identified in various myeloid neoplasms, including in around 10% of patients with MPNs.46–49 TET2, like other members of the TET family enzymes, presumably has an important function in DNA demethylation. It has recently been shown that the murine Tet proteins catalyze the conversion of 5 methylcytosine to 5 hydroxymethylcytosine on DNA.50 Thus TET2 mutations are thought to alter hematopoietic stem cell (HSC) functions and myeloid development via epigenetic modifications. These mutations were ideal pre-JAK2 candidate events. Indeed, studies of clonality in appropriate patients with typical MPNs showed that most cells had both mutations but that some cells had a TET2 mutation without a JAK2 mutation44;47 suggesting that the TET2 mutation was antecedent. In one case, the TET2 mutant clone acquired JAK2 and MPL mutations in distinct sub-clones.51 Is this the whole story? Unfortunately, no. Data from other patients with MPNs show a JAK2 mutation preceding the TET2 mutation.44;52;53 Other patients with MPNs had clones with either a JAK2 or TET2 mutation but not both.44 Similar studies of ASXL1 mutations have not been published but the same complexity is expected. What if people with these mutations develop transformation to blastic phase? In some instances, blast phase cells with TET2 defects had no TET2 mutation detected before transformation.51;54
In contrast, in 2 of 3 cases of MPN with ASXL1 mutations after transformation the mutation was also detected before transformation.51;54 This seeming discordance with TET2 should be tempered as TET2 mutations were detected in 10–20% of de novo cases of acute myelogenous leukemia (AML) and MPNs undergoing transformation to blastic phase.55
It is, therefore, possible that TET2 mutations, but also ASXL1 and del(20q), have different roles in initiating MPNs and causing AML alone or are associated with other molecular abnormalities.51;54
What of familial MPNs? The first studies of TET2 mutations in familial cases reported no inherited mutations.52 However, a recent report described sisters with a germline mutation of TET2.44 One presented with a JAK2-positive PV; the second was normal. This is the first evidence that TET2 mutations can be present in hematopoietic cells with no clinical consequence for decades. These data suggest no or only a very subtle effect on hematopoiesis of TET2 mutations. It could give a slight proliferative advantage to HSCs by causing a few “extra” mitoses, which could prepare the way for subsequent oncogenic mutations. This is reminiscent of the “fertile ground” hypothesis proposed for the JAK2 46/1 haplotype to explain the predisposition to acquire a JAK2 MPN.56–60 Intriguingly, this haplotype is the only marker that we can reasonably call a “pre-JAK2 condition” to date, although most individuals who carry it will never develop any JAK2 MPN.
(d) What molecular changes in the CML stem cells are critical for leukemia initiation or progression?
First and second generation TKI given to chronic phase (CP) CML patients substantially reduce the number of leukemia cells in the blood and bone marrow with improved survival. However, the story of the rationally-designed TKIs (imatinib, nilotinib, dasatinib and bosutinib) seems to have reached an ‘impasse’.61 For example, TKI-based therapies are largely ineffective in blast phase (BP) CML. More importantly, they are apparently unable to eradicate CML leukemia stem cells (LSCs). This raises several important questions; (1) Is BCR-ABL1 expression and/or activity essential to maintain the LSC reservoir in CP and BP? (2) Does BP result from genetic or epigenetic events that occur in LSCs or in more committed cells like granulocyte-macrophage progenitor (GMP) cells; and (3) what role, if any, does BCR-ABL1 play in blastic transformation?
Molecular events critical for the CML LSC
Considerable data indicate that TKI do not kill quiescent Ph-positive LSCs (defined as lineage-negative CD34/CD38/CD90 cells capable of self-renewal and of inducing transplantable leukemia in mice). Consequently, there is agreement that BCR-ABL1 expression and/or activity is not essential for self-renewal and survival of the CML LSC.62
Several observations in this context are of interest: (1) increased genomic instability correlating with levels of reactive oxygen species (ROS) in quiescent and proliferating CML LSCs and progenitors is associated with BCR-ABL1 expression;63 (2) enhanced activation of the β-catenin and hedgehog pathways64;65 is important for CML LSC survival and self-renewal;66–70 (3) treatment with the PP2A activator FTY720 decreases the number of Ph-positive but not of normal quiescent HSCs;39;65 (4) activity of FoxO transcription factors also appears important for maintaining CML LSC quiescence;71 (5) ALOX5-regulated MSR1 gene might be critical for maintaining CML LSCs; (6) BMI-1 expression decreases bone marrow homing and increases proliferation and maturation of leukemia Lin/Sca/Kit HSCs;72 (7) deficient RAC2 expression decreases survival of CML LSCs;73 (8) loss of BCL6 expression results in decreased self-renewal of BCR-ABL1-transduced bone marrow cells;74 and (9) oxygen levels in the bone marrow microenvironment may also be responsible for the quiescence and TKI-resistance of CML LSCs.75;76 These diverse data suggest that CML LSC behavior is controlled by different factors. The various pieces of this puzzle need to be molecularly and functionally inter-connected and could then form the basis for new therapeutic interventions.
Molecular events critical for the CML blastic phase granulocyte/macrophage progenitor
Lymphoblastic transformation may be characterized by non-random genetic aberrations (involving CDKN2A/B and IKZF1) and by expression of the activation induced deaminase (AID) mutator enzyme.77–79 In contrast, myeloid blastic transformation seems correlated with increased BCR-ABL1 expression and kinase activity in CD34 BP progenitors.61 BCR-ABL1 dose and kinase-dependent loss of PP2A activity and ROS induced genomic instability appear critical for the epigenetic and genetic heterogeneity characteristic of myeloid transformation.63;80 Increased BCR-ABL1 activity also appears essential for the differentiation-arrested phenotype of granulocyte/macrophage progenitors (GMP) in BP through alteration of the hnRNP-E2-CEBPα-miR-328 network.81 There is a concomitant decrease in PTEN that is BCR-ABL1-dependent which accelerates development of a BP phenotype in mice.82 Interestingly, it seems that higher levels of BCR-ABL1 are necessary for the non-hypoxic induction of the hypoxia-inducible factor-1α (HIF-1α) that, in turn, seems to be required by BP progenitors to tolerate enhanced BCR-ABL1 signaling and to exhibit TKI-resistance in vivo.83
The real role of BCR-ABL1 is controversial both in the regulation of survival and in the acquisition of a stem cell phenotype (e.g. self-renewal) by the GMPs. It has been reported that BP GMPs act as LSCs and that acquisition of self-renewal is dependent, at least in part, on BCR-ABL1-dependent induction of β-catenin expression/activity.84;85 However, some recent data suggest that BCR-ABL1 activity is actually dispensable for the β-catenin-dependent self-renewal of BP GMPs39;68;80;86 and that aberrant modulation of other pathways (e.g. sonic hedgehog, HES1) may also contribute but do not, per se, confer “stemness” to BCR-ABL1-positive GMPs.67;87 Likewise, recent reports indicate that survival of BP progenitors might occur in a BCR-ABL1-independent manner and involve aberrant expression of pro-survival factors (e.g. BCL2, BCL-XL, MCL1),88–91 despite the fact that a plethora of evidence accumulated during the past two decades showed a strict dependence between BCR-ABL1 activity and proliferation/survival signals in CD34 BP progenitors.61 It was recently reported that targeting BCR-ABL1 expression in the lineage-negative Sca cell compartment induces a CML-like MPD that in 70% of mice progresses to BP-like disease with lymphoid or myeloid features.32
These apparent incongruities might depend on the existence in BP of BCR-ABL1-positive progenitors which are capable of originating or maintaining a BP phenotype and that, for still unknown reasons, are not “oncogene addicted” and, therefore, intrinsically resistant both in vitro and in vivo to TKI treatment. Other data support this hypothesis and perhaps explain the paradoxical short- but not long-term BCR-ABL1-dependence of BP CD34 progenitors in TKI-treated BP patients. For example, in vitro clonogenic or survival assays performed with CD34 BP progenitors exposed to different TKIs never show complete inhibition of leukemic cell growth/survival, and other oncogenic tyrosine kinases (e.g. JAK2, LYN) may regulate and/or overcome BCR-ABL1 expression or activity.91;92 Interestingly, inhibition of the activity of the tumor suppressor PP2A plays a pivotal role in controlling BCR-ABL1-positive stem and progenitor cell survival because it impairs not only BCR-ABL1 but also JAK2, LYN and β-catenin activities when pharmacologically or molecularly reactivated.93
(f) What is the role of the second generation TKI in front-line management of patients with CML in first chronic phase?
For patients with CML in CP the introduction of imatinib mesylate in 1998 was an important therapeutic milestone. About 65% of patients treated with imatinib achieve and maintain for seven or more years a complete cytogenetic response (CCyR) and their survival is substantially longer than that achievable with any previous therapies. The adverse events attributable to imatinib are relatively mild and usually manageable.94–96
Conversely this means that one-third of CP patients cannot tolerate imatinib or have a leukemia that proves to be resistant to the drug.96 The best characterized mechanism of resistance is the acquisition of mutations in the BCR-ABL1 kinase domain (KD), of which the T315I mutation is the leading example,97 although the majority of CP patients resistant to imatinib have no identifiable KD mutation.
Efforts have, therefore, been made to improve the results achieved with the original TKI. Dasatinb and nilotinib may now replace imatinib as first-line therapy on the basis of their efficacy in patients with CML in CP who had failed imatinib. The third new TKI, bosutinib, is not yet licensed but is now in a phase III clinical trial, initial results of which will be reported shortly. In the short term, all three drugs seem valuable for patients who have failed imatinib; about 40–50% of these patients achieve a CCyR.98;99
The initial results of the ENESTnd trial in which nilotinib was compared prospectively with standard dose imatinib, and of the DASISION study comparing dasatinib with imatinib, were reported recently.100;101 Both studies were designed for patients with previously untreated CML in CP. Dasatinib and nilotinib both proved better than imatinib. The principal efficacy observations were a higher rate of incidence of CCyR at 12 months, which was achieved at a faster rate, and a reduced rate of progression to advanced phase following 18 months of follow up for the nilotinib study and 12 months for the dasatinib study.
All three drugs of the newer TKI have specific side effects. Thus dasatinib has been associated with pleural effusions, nilotinib with biochemical changes in liver function and bosutinib with gastrointestinal effects, particularly diarrhea. None of these drugs is active for patients in whom the Ph-positive clone consists predominantly of cells with a T315I mutation. Preliminary data suggest that a new multi-kinase inhibitor, ponatinib (previously AP24534), may be able to overcome resistance attributable to the T315I.102;103
There are still a number of unresolved issues in the management of CML in CP. For example, (a) it is not yet clear whether the clinician should leave treatment unchanged in a patient who achieves a complete cytogenetic response but no major molecular response; (b) it is not yet clear whether the second generation TKIs should be used for all newly diagnosed patients or whether it would be expedient to retain imatinib as initial therapy in some cases; (c) it would be very valuable if one could reliably predict responses in individual patients and design appropriate therapy for those who develop resistance to existing TKIs; and most importantly, (d) we need a therapeutic strategy (other than allogeneic stem cell transplantation) designed to ‘cure’ CML (see below).
(f) How can we target CML stem cells in responders to TKIs?
CML is recognized as a paradigm for cancer stem cells. Although TKIs have dramatically altered the history of chronic phase CML these drugs are expensive and unavailable to most people with CML who live outside North America or Western Europe. Side effects are substantial and drug-adherence studies suggest overall compliance levels of about 70 percent at two years, with levels of compliance strongly correlated to disease response.104;105
CML cells that originate from a CML stem cell follow a near normal differentiation hierarchy despite the fact that they originate from a leukemia stem cell (LSC). Therapy with TKIs targets the more mature BCR-ABL1 and results in and causes a rapid decline in the numbers of leukemia cells in the blood and bone marrow.106;107 This is followed by a much slower reduction in CML progenitor cell numbers over several years. Thereafter, it is unclear whether the slope continues very slowly downwards with cure predicted by 17–20 years107 or whether disease levels reach a plateau, presumably because at least some CML LSCs are resistant to killing by TKIs. Experimental evidence shows that CML LSCs survive despite continued exposure to TKIs was shown in vitro108–110 and in vivo.111
Major efforts are underway to apply systems biology approaches to compare normal HSC with CML LSC with the goal of finding novel targets that will allow selective eradication of LSCs without harming normal HSCs.112;113 Many investigators have already begun pre-clinical work to target LSCs (Table 2). These approaches can be broadly categorized in 3 ways (Table 3): (1) drugs/strategies that induce apoptosis of quiescent LSCs in a selective or non-selective manner; (2) drugs/strategies that address micro-environmental LSC interactions and affect quiescence, self-renewal and/or differentiation; and (3) drugs/strategies focused on immune therapy.
In this rapidly developing field it is difficult to see where the most important advances will occur. Whilst others are focusing on optimizing vaccinations designed to eliminate LSC, the groups of Tessa Holyoake and Ravi Bhatia have studied approaches that target quiescent LSCs directly or aim to drive LSCs into cell-cycle to sensitize them to TKIs. Some of these approaches have been or will be in clinical trials.67;137;138
Autophagy is a cellular lysosomal degradation pathway essential for the regulation of cell survival and death. One of the key regulators of autophagy is mTOR. In normal HSCs, nutrients and growth factors signal via PI3K/AKT/mTOR and keep autophagy suppressed. This pathway is mimicked by BCR-ABL1 signaling in CML LSCs. In normal HSCs, growth factor deprivation results in inhibition of mTOR and induces autophagy. Similarly, when CML LSCs are exposed to TKIs there is potent inhibition of BCR-ABL1 signaling, which induces autophagy. We recently reported that in CML LSCs this induced autophagy provides a survival mechanism that, at least in part, explains resistance to TKIs.137 Autophagy inhibitors are being developed but none is currently available for clinical trials. Chloroquine is a potent inhibitor of autophagy. We hypothesized that combining a TKI with chloroquine would shift the balance between apoptosis and induced autophagy in favor of enhanced apoptosis of CML LSCs. This was shown in vitro and in animal models and is the basis of a randomized clinical trial comparing imatinib with or without hydroxy-chloroquine in chronic phase CML patients achieving a major cytogenetic response.
(e) New therapies for Ph-negative MPNs?
The evolving understanding of the molecular pathogenesis of Ph-negative MPNs has led to an unprecedented variety of different therapies. JAK2-inhibitor testing began testing in 2007. These trials focused on persons with MPN-associated myelofibrosis (post-PV MF, post-ET MF and fibrotic PMF). These drugs are now being studied with persons with PV and ET unresponsive to other therapies. Alternative therapeutic strategies against different targets are also being developed.
MPN-associated myelofibrosis
There is no FDA-approved therapy for MPN-associated myelofibrosis. Current therapies are off-label indications palliative for anemia and/or reduction in splenomegaly.140 Allotransplants cure some persons with MPN-associated myelofibrosis but are used in fewer than 5% of affected persons.141
Patterns of efficacy and toxicity of JAK2-inhibitors are increasingly overlapping. Drugs in the most advanced phases of testing include INCB18424,114 TG101348,116 SB1518117 and CEP-701118 (Table 2). All rapidly reduce splenomegaly and improve myelofibrosis-associated constitutional symptoms, without significantly changing molecular or histological features of the disease.
Toxicities include anemia and thrombocytopenia. Gastrointestinal toxicity is common amongst drugs inhibiting FLT3.
The immune-modulatory drug pomalidomide produces durable anemia responses without bone marrow suppression or neuropathy.122 The nuclear histone deacetylase inhibitor LBH589 (Novartis) has promising activity in myelofibrosis with responses in splenomegaly and anemia.120 A phase II study is ongoing in the USA. The mTOR (mammalian target of rapamycin)-inhibitor RAD001 has activity similar to JAK2-inhibitors (decreased splenomegaly and improved constitutional symptoms).121
Polycythemia vera and essential thrombocythemia
Therapy of PV and ET relies on short-term anti-platelet drugs142 and selected cytoreduction for persons at high risk of vascular events (hydroxycarbamide, anagrelide and alkylating agents).143 In PV/ET we still need new drugs (1) for persons resistant or intolerant of current drugs to prevent vascular events; and (2) to delay progression to myelofibrosis and blastic transformation.
Data on therapy with INCB18424 were reported in 73 PV/ET patients failing hydroxycarbamide.115 There was a complete response in 94% of patients with PV and 61% of patients with ET. No patients had a vascular event on-study.
Splenomegaly and constitutional symptoms improved similar to data in MPN-associated myelofibrosis. Results from a phase II trial with CEP701 in 39 subjects were less impressive with several on-study vascular events and substantial gastrointestinal toxicity.119
Conventional interferon alpha has been used in persons with ET and PV but it is not well-tolerated. Two recent trials show better tolerance, efficacy in preventing vascular events and the potential to produce molecular and histological responses in persons with high-risk ET and PV.123;124 This might decrease the likelihood of progression to myelofibrosis and blastic transformation. An international phase III trial comparing hydroxyurea and pegylated interferon alpha-2a is planned.
Participants
The paper follows a small meeting in Natchez, Mississippi, USA immediately after the 2009 American Society of Hematology myeloproliferative neoplasms (MPNs). The organisers invited some discussants to briefly review selected topics addressed at the Natchez meeting and add relevant new data presented or published in 2010. The following persons participated in the meeting: Ralph Arlinghaus (USA), Tiziano Barbui (Italy), Francois Delhommeau (Paris), Connie Eaves (Canada), Alan Gewirtz (USA), John Goldman (UK), Anthony Green (UK), Oliver Hantschel (Austria), Rudiger Hehlmann (Germany), Tessa Holyoake (UK), Brian Huntly (UK), Catriona Jamieson (USA), Steffen Koschmieder (Germany), Robert Kralovics (Austria), Ruben Mesa (US), Tariq Mughal (UK), Vivian Oehler (USA), Heike Pahl (Germany), Animesh Pardanani (USA), Emmanuel Passegue (USA), Danilo Perrotti (USA), Josef Prchal (USA), Giuseppe Saglio (Italy), Richard Silver (USA), Tomasz Skorski (USA), Simona Soverini (Italy), Moshe Talpaz (USA), Richard van Etten (USA), Alessando Vannucchi (Italy), Jean Luc Villeval (France)
Footnotes
- Authorship and Disclosures The information provided by the authors about contributions from persons listed as authors and in acknowledgments is available with the full text of this paper at www.haematologica.org.
- Financial and other disclosures provided by the authors using the ICMJE (www.icmje.org) Uniform Format for Disclosure of Competing Interests are also available at www.haematologica.org.
- Funding: RvE is supported in part by NIH grant HL089747. SK is supported in part by DFG grant KO2155/2-2. DP is supported in part by grants from the National Cancer Institute CA095512, the US Army and the CML Research Program, W81XWH-07-1-0270, and The Leukemia and Lymphoma Society. FD is supported by the MPD Foundation, the Association L. Fugain, the Fondation de France and the Ligue Nationale Contre le Cancer.
- Received October 15, 2010.
- Revision received December 14, 2010.
- Accepted January 4, 2011.
References
- Van Etten RA, Shannon KM. Focus on myeloproliferative diseases and myelodysplastic syndromes. Cancer Cell. 2004; 6(6):547-52. PubMedhttps://doi.org/10.1016/j.ccr.2004.12.004Google Scholar
- Van Etten RA. Models of chronic myeloid leukemia. Curr Oncol Rep. 2001; 3(3):228-37. PubMedGoogle Scholar
- Lacout C, Pisani DF, Tulliez M, Gachelin FM, Vainchenker W, Villeval JL. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006; 108(5):1652-60. PubMedhttps://doi.org/10.1182/blood-2006-02-002030Google Scholar
- Wernig G, Mercher T, Okabe R, Levine RL, Lee BH, Gilliland DG. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006; 107(11):4274-81. PubMedhttps://doi.org/10.1182/blood-2005-12-4824Google Scholar
- Zaleskas VM, Krause DS, Lazarides K, Patel N, Hu Y, Li S, Van Etten RA. Molecular pathogenesis and therapy of polycythemia induced in mice by JAK2 V617F. PLoS One. 2006; 1:e18. PubMedhttps://doi.org/10.1371/journal.pone.0000018Google Scholar
- Bumm TG, Elsea C, Corbin AS, Loriaux M, Sherbenou D, Wood L. Characterization of murine JAK2V617F-positive myeloproliferative disease. Cancer Res. 2006; 66(23):11156-65. PubMedhttps://doi.org/10.1158/0008-5472.CAN-06-2210Google Scholar
- Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton MR. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007; 356(5):459-68. PubMedhttps://doi.org/10.1056/NEJMoa065202Google Scholar
- Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo M. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006; 3(7):e270. PubMedhttps://doi.org/10.1371/journal.pmed.0030270Google Scholar
- Akada H, Yan D, Zou H, Hutchison RE, Mohi MG. Conditional Expression of Heterozygous or Homozygous Jak2V617F From Its Endogenous Promoter Induces a Polycythemia Vera-Like Disease. ASH Annual Meeting Abstracts. 2009; 114(22):437. Google Scholar
- Ghevaert CJ, Li J, Severin S, Auger J, Watson S, Green AR. Physiological Levels of Jak2 V617F Result in Enhanced Megakaryocyte Differentiation, Proplatelet Formation and Platelet Reactivity. ASH Annual Meeting Abstracts. 2009; 114(22):226. Google Scholar
- Marty C, Lacout C, Fong L, Martin A, Vainchenker W, Villeval JL. Constitutive Heterozygous Expression of JAK2V617F Mutated Kinase Triggers Severe Myeloproliferative Disorder in Knock-in Mice. ASH Annual Meeting Abstracts. 2009; 114(22):2902. Google Scholar
- Tiedt R, Hao-Shen H, Sobas MA, Looser R, Dirnhofer S, Schwaller J, Skoda RC. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood. 2008; 111(8):3931-40. PubMedhttps://doi.org/10.1182/blood-2007-08-107748Google Scholar
- Shide K, Shimoda HK, Kumano T, Karube K, Kameda T, Takenaka K. Development of ET, primary myelofibrosis and PV in mice expressing JAK2 V617F. Leukemia. 2008; 22(1):87-95. PubMedhttps://doi.org/10.1038/sj.leu.2405043Google Scholar
- Xing S, Wanting TH, Zhao W, Ma J, Wang S, Xu X. Transgenic expression of JAK2V617F causes myeloproliferative disorders in mice. Blood. 2008; 111(10):5109-17. PubMedhttps://doi.org/10.1182/blood-2007-05-091579Google Scholar
- Akada H, Yan D, Zou H, Fiering S, Hutchison RE, Mohi MG. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood. 2010; 115(17):3589-97. PubMedhttps://doi.org/10.1182/blood-2009-04-215848Google Scholar
- Mullally A, Lane SW, Ball B, Megerdichian C, Okabe R, Al Shahrour F. Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. Cancer Cell. 2010; 17(6):584-96. PubMedhttps://doi.org/10.1016/j.ccr.2010.05.015Google Scholar
- Lu X, Levine R, Tong W, Wernig G, Pikman Y, Zarnegar S. Expression of a homodimeric type I cytokine receptor is required for JAK2V617F-mediated transformation. Proc Natl Acad Sci USA. 2005; 102(52):18962-7. PubMedhttps://doi.org/10.1073/pnas.0509714102Google Scholar
- James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, Lacout C. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005; 434(7037):1144-8. PubMedhttps://doi.org/10.1038/nature03546Google Scholar
- Heisterkamp N, Jenster G, Kioussis D, Pattengale PK, Groffen J. Human bcr-abl gene has a lethal effect on embryogenesis. Transgenic Res. 1991; 1(1):45-53. PubMedGoogle Scholar
- Kralovics R, Teo SS, Li S, Theocharides A, Buser AS, Tichelli A, Skoda RC. Acquisition of the V617F mutation of JAK2 is a late genetic event in a subset of patients with myeloproliferative disorders. Blood. 2006; 108(4):1377-80. PubMedhttps://doi.org/10.1182/blood-2005-11-009605Google Scholar
- Scott LM, Scott MA, Campbell PJ, Green AR. Progenitors homozygous for the V617F mutation occur in most patients with polycythemia vera, but not essential thrombocythemia. Blood. 2006; 108(7):2435-7. PubMedhttps://doi.org/10.1182/blood-2006-04-018259Google Scholar
- Bellanne-Chantelot C, Chaumarel I, Labopin M, Bellanger F, Barbu V, De Toma C. Genetic and clinical implications of the Val617Phe JAK2 mutation in 72 families with myeloproliferative disorders. Blood. 2006; 108(1):346-52. PubMedhttps://doi.org/10.1182/blood-2005-12-4852Google Scholar
- Dazzi F, Capelli D, Hasserjian R, Cotter F, Corbo M, Poletti A. The kinetics and extent of engraftment of chronic myelogenous leukemia cells in non-obese diabetic/severe combined immunodeficiency mice reflect the phase of the donor’s disease: an in vivo model of chronic myelogenous leukemia biology. Blood. 1998; 92(4):1390-6. PubMedGoogle Scholar
- Eisterer W, Jiang X, Christ O, Glimm H, Lee KH, Pang E. Different subsets of primary chronic myeloid leukemia stem cells engraft immunodeficient mice and produce a model of the human disease. Leukemia. 2005; 19(3):435-41. PubMedhttps://doi.org/10.1038/sj.leu.2403649Google Scholar
- 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; 247(4944):824-30. PubMedhttps://doi.org/10.1126/science.2406902Google Scholar
- Gishizky ML, Johnson-White J, Witte ON. Efficient transplantation of BCR-ABL-induced chronic myelogenous leukemia-like syndrome in mice. Proc Natl Acad Sci USA. 1993; 90(8):3755-9. PubMedhttps://doi.org/10.1073/pnas.90.8.3755Google Scholar
- Li S, Ilaria RL, Million RP, Daley GQ, Van Etten RA. The P190, P210, and P230 forms of the BCR/ABL oncogene induce a similar chronic myeloid leukemia-like syndrome in mice but have different lymphoid leukemogenic activity. J Exp Med. 1999; 189(9):1399-412. PubMedhttps://doi.org/10.1084/jem.189.9.1399Google Scholar
- Hu Y, Swerdlow S, Duffy TM, Weinmann R, Lee FY, Li S. Targeting multiple kinase pathways in leukemic progenitors and stem cells is essential for improved treatment of Ph+ leukemia in mice. Proc Natl Acad Sci USA. 2006; 103(45):16870-5. PubMedhttps://doi.org/10.1073/pnas.0606509103Google Scholar
- Van Etten RA. Studying the pathogenesis of BCR-ABL+ leukemia in mice. Oncogene. 2002; 21(56):8643-51. PubMedhttps://doi.org/10.1038/sj.onc.1206091Google Scholar
- Miyazaki K, Yamasaki N, Oda H, Kuwata T, Kanno Y, Miyazaki M. Enhanced expression of p210BCR/ABL and aberrant expression of Zfp423/ZNF423 induce blast crisis of chronic myelogenous leukemia. Blood. 2009; 113(19):4702-10. PubMedhttps://doi.org/10.1182/blood-2007-05-088724Google Scholar
- Nagamachi A, Yamasaki N, Miyazaki K, Oda H, Miyazaki M, Honda Z. Haploinsufficiency and acquired loss of Bcl11b and H2AX induces blast crisis of chronic myelogenous leukemia in a transgenic mouse model. Cancer Sci. 2009; 100(7):1219-26. PubMedhttps://doi.org/10.1111/j.1349-7006.2009.01172.xGoogle Scholar
- Perez-Caro M, Cobaleda C, Gonzalez-Herrero I, Vicente-Duenas C, Bermejo-Rodriguez C, Sanchez-Beato M. Cancer induction by restriction of oncogene expression to the stem cell compartment. EMBO J. 2009; 28(1):8-20. PubMedhttps://doi.org/10.1038/emboj.2008.253Google Scholar
- Koschmieder S, Gottgens B, Zhang P, Iwasaki-Arai J, Akashi K, Kutok JL. Inducible chronic phase of myeloid leukemia with expansion of hematopoietic stem cells in a transgenic model of BCR-ABL leukemogenesis. Blood. 2005; 105(1):324-34. PubMedhttps://doi.org/10.1182/blood-2003-12-4369Google Scholar
- Schemionek M, Elling C, Steidl U, Baumer N, Hamilton A, Spieker T. BCR-ABL enhances differentiation of long-term repopulating hematopoietic stem cells. Blood. 2010; 115(16):3185-95. PubMedhttps://doi.org/10.1182/blood-2009-04-215376Google Scholar
- Li J, Spensberger D, Ahn JS, Anand S, Beer PA, Ghevaert C. JAK2 V617F impairs hematopoietic stem cell function in a conditional knock-in mouse model of JAK2 V617F-positive essential thrombocythemia. Blood. 2010; 116(9):1528-38. PubMedhttps://doi.org/10.1182/blood-2009-12-259747Google Scholar
- Zhang B, Ho YW, Lee SU, Maeda T, Huettner C, Bhatia R. Characterization of Leukemia-Initiating Cells in a Transgenic Model of Chronic Phase Chronic Myelogenous Leukemia (CML). ASH Annual Meeting Abstracts. 2009; 114(22):858. Google Scholar
- Zhang B, Strauss AC, Chu S, Li M, Ho Y, Shiang KD. Effective Targeting of Quiescent Chronic Myelogenous Leukemia Stem Cells by Histone Deacetylase Inhibitors in Combination with Imatinib Mesylate. Cancer Cell. 2010; 17(5):427-42. PubMedhttps://doi.org/10.1016/j.ccr.2010.03.011Google Scholar
- Harb J, Neviani P, Huettner C, Marcucci G, Perrotti D. BCR/ABL Dosage Hierarchically and Temporally Influences hnRNP A1, hnRNP K and hnRNP E2 Expression in Hematopoietic Stem and Progenitor Cells. ASH Annual Meeting Abstracts. 2009; 114 (22):191. Google Scholar
- Oaks J, Neviani P, Mukhopadhyay A, Santhanam R, Ma Y, Mao C. FTY720 but Not Its Immunosuppressive Phosphorylated Form FTY720-P Exerts Anti-Leukemic Activity towards Ph(+) and Ph(−) Myeloproliferative Disorders through Reactivation of the PP2A Tumor Suppressor. ASH Annual Meeting Abstracts. 2009; 114(22):3259. Google Scholar
- Sengupta A, Arnett J, Dunn S, Williams DA, Cancelas JA. Rac2 GTPase deficiency depletes BCR−ABL+ leukemic stem cells and progenitors in vivo. Blood. 2010; 116(1):81-4. PubMedhttps://doi.org/10.1182/blood-2009-10-247437Google Scholar
- Nussenzveig RH, Swierczek SI, Jelinek J, Gaikwad A, Liu E, Verstovsek S. Polycythemia vera is not initiated by JAK2V617F mutation. Exp Hematol. 2007; 35(1):32-8. PubMedGoogle Scholar
- Levine RL, Belisle C, Wadleigh M, Zahrieh D, Lee S, Chagnon P. X-inactivation-based clonality analysis and quantitative JAK2V617F assessment reveal a strong association between clonality and JAK2V617F in PV but not ET/MMM, and identifies a subset of JAK2V617F-negative ET and MMM patients with clonal hematopoiesis. Blood. 2006; 107(10):4139-41. PubMedhttps://doi.org/10.1182/blood-2005-09-3900Google Scholar
- Campbell PJ, Baxter EJ, Beer PA, Scott LM, Bench AJ, Huntly BJ. Mutation of JAK2 in the myeloproliferative disorders: timing, clonality studies, cytogenetic associations, and role in leukemic transformation. Blood. 2006; 108(10):3548-55. PubMedhttps://doi.org/10.1182/blood-2005-12-013748Google Scholar
- Schaub FX, Jager R, Looser R, Hao-Shen H, Hermouet S, Girodon F. Clonal analysis of deletions on chromosome 20q and JAK2-V617F in MPD suggests that del20q acts independently and is not one of the predisposing mutations for JAK2-V617F. Blood. 2009; 113(9):2022-7. PubMedhttps://doi.org/10.1182/blood-2008-07-167056Google Scholar
- Theocharides A, Boissinot M, Girodon F, Garand R, Teo SS, Lippert E. Leukemic blasts in transformed JAK2-V617F-positive myeloproliferative disorders are frequently negative for the JAK2-V617F mutation. Blood. 2007; 110(1):375-9. PubMedhttps://doi.org/10.1182/blood-2006-12-062125Google Scholar
- Carbuccia N, Murati A, Trouplin V, Brecqueville M, Adelaide J, Rey J. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia. 2009; 23(11):2183-6. PubMedhttps://doi.org/10.1038/leu.2009.141Google Scholar
- Delhommeau F, Dupont S, Della Valle V, James C, Trannoy S, Massé A. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009; 360(22):2289-301. PubMedhttps://doi.org/10.1056/NEJMoa0810069Google Scholar
- Gelsi-Boyer V, Trouplin V, Adelaide J, Bonansea J, Cervera N, Carbuccia N. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009; 145(6):788-800. PubMedhttps://doi.org/10.1111/j.1365-2141.2009.07697.xGoogle Scholar
- Langemeijer SM, Kuiper RP, Berends M, Knops R, Aslanyan MG, Massop M. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat Genet. 2009; 41(7):838-42. PubMedhttps://doi.org/10.1038/ng.391Google Scholar
- Ito S, D’Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010; 466(7310):1129-33. PubMedhttps://doi.org/10.1038/nature09303Google Scholar
- Beer PA, Delhommeau F, LeCouedic JP, Dawson MA, Chen E, Bareford D. Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood. 2010; 115(14):2891-900. PubMedhttps://doi.org/10.1182/blood-2009-08-236596Google Scholar
- Saint-Martin C, Leroy G, Delhommeau F, Panelatti G, Dupont S, James C. Analysis of the ten-eleven translocation 2 (TET2) gene in familial myeloproliferative neoplasms. Blood. 2009; 114(8):1628-32. PubMedhttps://doi.org/10.1182/blood-2009-01-197525Google Scholar
- Swierczek S, Bellanne-Chantelot C, Yoon D, Saint-Martin C, Kim SJ, Najman A, Prchal JT. TET2 Mutations in Polycythemia Vera (PV) in Some Cases Follow Rather Than Precede JAK2 V617F Mutation, Are Not a Disease-Initiating Event, Affect Mainly Erythropoiesis, and Contribute to Increased Aggressivity of PV Clone. ASH Annual Meeting Abstracts. 2009; 114(22):3913. Google Scholar
- Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat C. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res. 2010; 70(2):447-52. PubMedhttps://doi.org/10.1158/0008-5472.CAN-09-3783Google Scholar
- Aslanyan MG, Langemeijer SMC, Cilloni D, Saglio G, Marie JP, Tang R. Incidence and Clinical Impact of TET2 Mutations in Acute Myeloid Leukemia Patients Treated within the EORTC/GIMEMA AML-12/06991 AML Trial. ASH Annual Meeting Abstracts. 2009; 114(22):2609. Google Scholar
- Campbell PJ. Somatic and germline genetics at the JAK2 locus. Nat Genet. 2009; 41(4):385-6. PubMedhttps://doi.org/10.1038/ng0409-385Google Scholar
- Jones AV, Chase A, Silver RT, Oscier D, Zoi K, Wang YL. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet. 2009; 41(4):446-9. PubMedhttps://doi.org/10.1038/ng.334Google Scholar
- Kilpivaara O, Mukherjee S, Schram AM, Wadleigh M, Mullally A, Ebert BL. A germline JAK2 SNP is associated with predisposition to the development of JAK2(V617F)-positive myeloproliferative neoplasms. Nat Genet. 2009; 41(4):455-9. PubMedhttps://doi.org/10.1038/ng.342Google Scholar
- Olcaydu D, Harutyunyan A, Jager R, Berg T, Gisslinger B, Pabinger I. A common JAK2 haplotype confers susceptibility to myeloproliferative neoplasms. Nat Genet. 2009; 41(4):450-4. PubMedhttps://doi.org/10.1038/ng.341Google Scholar
- Cross NCP, Campbell P, Beer PA, Schnittger S, Vannucchi AM, Zoi K. The JAK2 46/1 Haplotype Predisposes to Myeloproliferative Neoplasms Characterized by Diverse Mutations. ASH Annual Meeting Abstracts. 2009; 114(22):433. Google Scholar
- Perrotti D, Jamieson C, Goldman J, Skorski T. Chronic myeloid leukemia: mechanisms of blastic transformation. J Clin Invest. 2010; 120(7):2254-64. PubMedhttps://doi.org/10.1172/JCI41246Google Scholar
- Nicholson E, Holyoake T. The chronic myeloid leukemia stem cell. Clin Lymphoma Myeloma. 2009; 9(Suppl 4):S376-S381. PubMedhttps://doi.org/10.3816/CLM.2009.s.037Google Scholar
- Nieborowska-Skorska M, Koptyra M, Bolton E, Ray R, Ngaba D, Hoser G. ROS-Induced DNA Damage Causing Genomic Instability in CML Stem and/or Progenitor Cells and in Quiescent and/or Proliferating Cells: Role of Mitochondrial Respiratory Chain Complex III. Blood (ASH Annual Meeting Abstracts). 2009; 114(22):3268. Google Scholar
- Hu Y, Chen Y, Douglas L, Li S. beta-Catenin is essential for survival of leukemic stem cells insensitive to kinase inhibition in mice with BCR-ABL-induced chronic myeloid leukemia. Leukemia. 2009; 23(1):109-16. PubMedhttps://doi.org/10.1038/leu.2008.262Google Scholar
- Neviani P, Santhanam R, Ma Y, Marcucci G, Byrd JC, Chen CS. Activation of PP2A by FTY720 Inhibits Survival and Self-Renewal of the Ph(+) Chronic Myelogenous Leukemia (CML) CD34+/CD38− Stem Cell through the Simultaneous Suppression of BCR/ABL and BCR/ABL- independent Signals. Blood (ASH Annual Meeting Abstracts). 2008; 112(11):189. Google Scholar
- Zhao C, Chen A, Jamieson CH, Fereshteh M, Abrahamsson A, Blum J. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature. 2009; 458(7239):776-9. PubMedhttps://doi.org/10.1038/nature07737Google Scholar
- Dierks C, Beigi R, Guo GR, Zirlik K, Stegert MR, Manley P. Expansion of Bcr-Abl-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell. 2008; 14(3):238-49. PubMedhttps://doi.org/10.1016/j.ccr.2008.08.003Google Scholar
- Peterson LF, Turbiak AJ, Giannola DM, Donato N, Showalter HHD, Fearon ER, Talpaz M. Wnt-Pathway Directed Compound Targets Blast Crisis and Chronic Phase CML Leukemia Stem Progenitors. Blood (ASH Annual Meeting Abstracts). 2009; 114(22):2168. Google Scholar
- Irvine DA, Zhang B, Allan EK, Holyoake TL, Dorsch M, Manley PW. Combination of the Hedgehog Pathway Inhibitor LDE225 and Nilotinib Eliminates Chronic Myeloid Leukemia Stem and Progenitor Cells. Blood (ASH Annual Meeting Abstracts). 2009; 114(22):1428. Google Scholar
- Nagao R, Ashihara E, Kimura S, Yao H, Takeuchi M, Yokota A. Inhibition of Wnt/{beta}-Catenin Signaling by AV65 Treatment Caused Cell Cycle Arrest and Induced Caspase-Independent or -Dependent Apoptosis in CML Cells. Blood (ASH Annual Meeting Abstracts). 2009; 114 (22):2184. Google Scholar
- Cilloni D, Pellicano F, Helgason VG, Panuzzo C, Messa F, Huntly BJ. Foxo Transcription Factor Activity Is Retained in Quiescent Chronic Myeloid Leukaemia Stem Cells and Activated by Tyrosine Kinase Inhibitors to Mediate “induced-quiescence” in More Mature progenitors. Blood (ASH Annual Meeting Abstracts). 2009; 114 (22):187. Google Scholar
- Sengupta A, Arnett J, Dunn S, Cancelas J. Bmi-1 Overexpression Synergizes with p210-BCR-ABL to Induce Stem Cell and Progenitor Transformation. ASH Annual Meeting Abstracts. 2009; 114(22):3251. Google Scholar
- Sengupta A, Arnett J, Dunn S, Cancelas J. Deletion of Rac2 inhibits Proliferation of Chronic Myelogenous Leukemia (CML) Stems Cells and Progenitors (HSC/P) In Vivo and Promotes Survival of Scl/p210-BCR-ABL Mice. ASH Annual Meeting Abstracts. 2009; 114(22):3253. Google Scholar
- Hurtz C, Duy C, Cerchietti L, Park E, Ci W, Swaminathan S. BCL6 Is Required for Leukemia-Initiation and Self-Renewal Signaling in Chronic Myeloid Leukemia. ASH Annual Meeting Abstracts. 2009; 114(22):2167. Google Scholar
- Takeuchi M, Kimura S, Kuroda J, Ashihara E, Kawatani M, Osada H. Hypoxia-Adapted CML Cells Are More Primitive Population and Are Eradicated by Glyoxalase-1 Inhibitors. ASH Annual Meeting Abstracts. 2009; 114(22):2166. Google Scholar
- Ng KP, Poh TY, Sun WT, Chuah C, Ong ST. Physiologic Hypoxia Protects Chronic Myelogenous Leukemia Progenitors From Elimination by Imatinib Mesylate. ASH Annual Meeting Abstracts. 2009; 114(22):2181. Google Scholar
- Klemm L, Duy C, Iacobucci I, von Levetzow G, Feldhahn N, Kim Ym. The B Cell Mutator AID Promotes B Lymphoid Blast Crisis and Drug-Resistance in Chronic Myeloid Leukemia. ASH Annual Meeting Abstracts. 2009; 114(22):3274. Google Scholar
- Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma J. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008; 453(7191):110-4. PubMedhttps://doi.org/10.1038/nature06866Google Scholar
- Iacobucci I, Storlazzi CT, Cilloni D, Lonetti A, Ottaviani E, Soverini S. Identification and molecular characterization of recurrent genomic deletions on 7p12 in the IKZF1 gene in a large cohort of BCR-ABL1-positive acute lymphoblastic leukemia patients: on behalf of Gruppo Italiano Malattie Ematologiche dell’Adulto Acute Leukemia Working Party (GIMEMA AL WP). Blood. 2009; 114(10):2159-67. PubMedhttps://doi.org/10.1182/blood-2008-08-173963Google Scholar
- Neviani P, Santhanam R, Trotta R, Notari M, Blaser BW, Liu S. 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; 8(5):355-68. PubMedhttps://doi.org/10.1016/j.ccr.2005.10.015Google Scholar
- Eiring AM, Harb JG, Neviani P, Garton C, Oaks JJ, Spizzo R. miR-328 functions as an RNA decoy to modulate hnRNP E2 regulation of mRNA translation in leukemic blasts. Cell. 2010; 140(5):652-65. PubMedhttps://doi.org/10.1016/j.cell.2010.01.007Google Scholar
- Peng C, Chen Y, Yang Z, Zhang H, Osterby L, Rosmarin AG, Li S. PTEN is a tumor suppressor in CML stem cells and BCR-ABL-induced leukemias in mice. Blood. 2010; 115(3):626-35. PubMedhttps://doi.org/10.1182/blood-2009-06-228130Google Scholar
- Zhao F, Mancuso A, Bui TV, Tong X, Gruber JJ, Swider CR. Imatinib resistance associated with BCR-ABL upregulation is dependent on HIF-1alpha-induced metabolic reprograming. Oncogene. 2010; 29(20):2962-72. PubMedhttps://doi.org/10.1038/onc.2010.67Google Scholar
- Jamieson CH. Chronic myeloid leukemia stem cells. Hematology Am Soc Hematol Educ Program. 2008:436-42. Google Scholar
- Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, Zehnder JL. Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med. 2004; 351(7):657-67. PubMedhttps://doi.org/10.1056/NEJMoa040258Google Scholar
- Lim S, Saw TY, Chuah C, Ong ST. Overexpression and Phosphorylation of eIF4E Is Required for Beta-Catenin Activation in Blast Crisis Chronic Myelogenous Leukemia. ASH Annual Meeting Abstracts. 2009; 114(22):41. Google Scholar
- Nakahara F, Sakata-Yanagimoto M, Komeno Y, Kato N, Uchida T, Haraguchi K. Hes1 immortalizes committed progenitors and plays a role in blast crisis transition in chronic myelogenous leukemia. Blood. 2010; 115(14):2872-81. PubMedhttps://doi.org/10.1182/blood-2009-05-222836Google Scholar
- Goff D, Abrahamsson-Schairer A, Shih A, Black JM, Chuang R, Tesi RJ. The Broad Spectrum Bcl-2 Inhibitor Apogossypol Induces Apoptosis and Differentiation of Blast Crisis Chronic Myeloid Leukemia Stem Cells. ASH Annual Meeting Abstracts. 2009; 114(22):3275. Google Scholar
- Mak DH, Schober WD, Konopleva M, Cortes J, Kantarjian HM, Andreeff M, Carter BZ. Inhibition of Bcl-2/Bcl-XL Promotes Apoptosis in Blast Crisis CML Including Quiescent Primitive Progenitor Cells Regardless of Cellular Responses to Tyrosine Kinase Inhibitors. ASH Annual Meeting Abstracts. 2009; 114(22):646. Google Scholar
- Belloc F, Airiau K, Jeanneteau M, Mahon FX. ABT-737 Cooperates in a Strong Synergism with Tyrosine Kinase Inhibitors to Induce Apoptosis of Chronic Myeloid Leukemia Cells. ASH Annual Meeting Abstracts. 2009; 114(22):3246. Google Scholar
- Pasquet JM, Gioia R, Drullion C, Lagarde V, Leroy C, Roche S. Tyrosine Kinase Proteins profiling of Nilotinib Resistant Chronic Myelogenous Leukemia Cells Unravels a Tyrosine Kinase-Mediated Bypass. ASH Annual Meeting Abstracts. 2009; 114(22):2175. Google Scholar
- Samanta AK, Chakraborty SN, Sun X, Schlette E, Priebe W, Arlinghaus RB. Jak2 Phosphorylates Tyr 177 of Bcr-Abl Activating the Ras and PI-3 Kinase Pathways and Maintains Functional Levels of Bcr-Abl in Chronic Myelogenous Leukemia. ASH Annual Meeting Abstracts. 2009; 114(22):39. Google Scholar
- Perrotti D, Neviani P. Protein phosphatase 2A (PP2A), a drugable tumor suppressor in Ph1(+) leukemias. Cancer Metastasis Rev. 2008; 27(2):159-68. PubMedhttps://doi.org/10.1007/s10555-008-9119-xGoogle Scholar
- Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001; 344(14):1031-7. PubMedhttps://doi.org/10.1056/NEJM200104053441401Google Scholar
- O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2003; 348(11):994-1004. PubMedhttps://doi.org/10.1056/NEJMoa022457Google Scholar
- Mughal TI, Schrieber A. Principal long-term adverse effects of imatinib in patients with chronic myeloid leukemia in chronic phase. Biologics. 2010; 4:315-23. PubMedGoogle Scholar
- Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan J, Sawyers CL. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell. 2002; 2(2):117-25. PubMedhttps://doi.org/10.1016/S1535-6108(02)00096-XGoogle Scholar
- Talpaz M, Shah NP, Kantarjian H, Donato N, Nicoll J, Paquette R. Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med. 2006; 354(24):2531-41. PubMedhttps://doi.org/10.1056/NEJMoa055229Google Scholar
- Kantarjian HM, Giles F, Gattermann N, Bhalla K, Alimena G, Palandri F. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance. Blood. 2007; 110(10):3540-6. PubMedhttps://doi.org/10.1182/blood-2007-03-080689Google Scholar
- Kantarjian H, Shah NP, Hochhaus A, Cortes J, Shah S, Ayala M. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2010; 362(24):2260-70. PubMedhttps://doi.org/10.1056/NEJMoa1002315Google Scholar
- Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010; 362 (24):2251-9. PubMedhttps://doi.org/10.1056/NEJMoa0912614Google Scholar
- O’Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell. 2009; 16(5):401-12. PubMedhttps://doi.org/10.1016/j.ccr.2009.09.028Google Scholar
- Cortes J, Talpaz M, Deininger M, Shah N, Flinn IW, Mauro MJ. A Phase 1 Trial of Oral AP24534 in Patients with Refractory Chronic Myeloid Leukemia and Other Hematologic Malignancies: First Results of Safety and Clinical Activity against T315I and Resistant Mutations. ASH Annual Meeting Abstracts. 2009; 114(22):643. Google Scholar
- Noens L, van Lierde MA, De Bock R, Verhoef G, Zachee P, Berneman Z. Prevalence, determinants, and outcomes of nonadherence to imatinib therapy in patients with chronic myeloid leukemia: the ADAGIO study. Blood. 2009; 113(22):5401-11. PubMedhttps://doi.org/10.1182/blood-2008-12-196543Google Scholar
- Marin D, Bazeos A, Mahon FX, Eliasson L, Milojkovic D, Bua M. Adherence is the critical factor for achieving molecular responses in patients with chronic myeloid leukemia who achieve complete cytogenetic responses on imatinib. J Clin Oncol. 2010; 28(14):2381-8. PubMedhttps://doi.org/10.1200/JCO.2009.26.3087Google Scholar
- Michor F, Hughes TP, Iwasa Y, Branford S, Shah NP, Sawyers CL, Nowak MA. Dynamics of chronic myeloid leukaemia. Nature. 2005; 435(7046):1267-70. PubMedhttps://doi.org/10.1038/nature03669Google Scholar
- Roeder I, Horn M, Glauche I, Hochhaus A, Mueller MC, Loeffler M. Dynamic modeling of imatinib-treated chronic myeloid leukemia: functional insights and clinical implications. Nat Med. 2006; 12(10):1181-4. PubMedhttps://doi.org/10.1038/nm1487Google Scholar
- Graham SM, Jorgensen HG, Allan E, Pearson C, Alcorn MJ, Richmond L, Holyoake TL. Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood. 2002; 99(1):319-25. PubMedhttps://doi.org/10.1182/blood.V99.1.319Google Scholar
- Jorgensen HG, Allan EK, Jordanides NE, Mountford JC, Holyoake TL. Nilotinib exerts equipotent antiproliferative effects to imatinib and does not induce apoptosis in CD34+ CML cells. Blood. 2007; 109(9):4016-9. PubMedhttps://doi.org/10.1182/blood-2006-11-057521Google Scholar
- Copland M, Hamilton A, Elrick LJ, Baird JW, Allan EK, Jordanides N. Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction. Blood. 2006; 107(11):4532-9. PubMedhttps://doi.org/10.1182/blood-2005-07-2947Google Scholar
- Chu S, Lin A, McDonald T, Snyder DS, Forman SJ, Bhatia R. Persistence of Leukemia Stem Cells in Chronic Myelogenous Leukemia Patients in Complete Cytogenetic Remission on Imatinib Treatment for 5 Years. Blood (ASH Annual Meeting Abstracts). 2008; 112(11):194. Google Scholar
- Graham SM, Vass JK, Holyoake TL, Graham GJ. Transcriptional analysis of quiescent and proliferating CD34+ human hemopoietic cells from normal and chronic myeloid leukemia sources. Stem Cells. 2007; 25(12):3111-20. PubMedhttps://doi.org/10.1634/stemcells.2007-0250Google Scholar
- Griffiths SD, Burthem J, Unwin RD, Holyoake TL, Melo JV, Lucas GS, Whetton AD. The use of isobaric tag peptide labeling (iTRAQ) and mass spectrometry to examine rare, primitive hematopoietic cells from patients with chronic myeloid leukemia. Mol Biotechnol. 2007; 36(2):81-9. PubMedhttps://doi.org/10.1007/s12033-007-0005-5Google Scholar
- Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes-Franco J, Thomas DA. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med. 2010; 363(12):1117-27. PubMedhttps://doi.org/10.1056/NEJMoa1002028Google Scholar
- Verstovsek S, Passamonti F, Rambaldi A, Barosi G, Rosen P, Levy R. A Phase 2 Study of INCB018424, An Oral, Selective JAK1/JAK2 Inhibitor, in Patients with Advanced Polycythemia Vera (PV) and Essential Thrombocythemia (ET) Refractory to Hydroxyurea. Blood (ASH Annual Meeting Abstracts). 2009; 114(22):311. Google Scholar
- Pardanani AD, Gotlib JR, Jamieson C, Cortes J, Talpaz M, Stone R. A Phase I Evaluation of TG101348, a Selective JAK2 Inhibitor, in Myelofibrosis: Clinical Response Is Accompanied by Significant Reduction in JAK2V617F Allele Burden. Blood (ASH Annual Meeting Abstracts). 2009; 114(22):755. Google Scholar
- Verstovsek S, Odenike O, Scott B, Estrov Z, Cortes J, Thomas DA, Wood J, Ethirajulu K, Lowe A, Zhu HJ, Kantarjian H, Deeg HJ. Phase I Dose-Escalation Trial of SB1518, a Novel JAK2/FLT3 Inhibitor, in Acute and Chronic Myeloid Diseases, Including Primary or Post-Essential Thrombocythemia/Polycythemia Vera Myelofibrosis. Blood (ASH Annual Meeting Abstracts). 2009; 114(22):3905. Google Scholar
- Hexner E, Goldberg JD, Prchal JT, Demakos EP, Swierczek S, Weinberg RS. A Multicenter, Open Label Phase I/II Study of CEP701 (Lestaurtinib) in Adults with Myelofibrosis; a Report On Phase I: A Study of the Myeloproliferative Disorders Research Consortium (MPD-RC). Blood (ASH Annual Meeting Abstracts). 2009; 114 (22):754. Google Scholar
- Moliterno AR, Hexner E, Roboz GJ, Carroll M, Luger S, Mascarenhas J. An Open-Label Study of CEP-701 in Patients with JAK2 V617F-Positive PV and ET: Update of 39 Enrolled Patients. Blood (ASH Annual Meeting Abstracts). 2009; 114(22):753. Google Scholar
- Mascarenhas J, Wang X, Rodriguez A, Xu M, Gorman E, Zhang W. A Phase I Study of LBH589, a Novel Histone Deacetylase Inhibitor in Patients with Primary Myelofibrosis (PMF) and Post-Polycythemia/Essential Thrombocythemia Myelofibrosis (Post-PV/ET MF). Blood (ASH Annual Meeting Abstracts). 2009; 114 (22):308. Google Scholar
- Vannucchi AM, Guglielmelli P, Gattoni E, Antonioli E, Bogani C, Tozzi L. RAD001, An Inhibitor of mTOR, Shows Clinical Activity in a Phase I/II Study in Patients with Primary Myelofibrosis (PMF) and Post Polycythemia Vera/Essential Thrombocythemia Myelofibrosis (PPV/PET MF). Blood (ASH Annual Meeting Abstracts). 2009; 114(22):307. Google Scholar
- Mesa RA, Pardanani AD, Hussein K, Wu W, Schwager S, Litzow MR. Phase1/-2 study of Pomalidomide in myelofibrosis. Am J Hematol. 2010; 85(2):129-30. PubMedGoogle Scholar
- Kiladjian JJ, Cassinat B, Chevret S, Turlure P, Cambier N, Roussel M. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood. 2008; 112(8):3065-72. PubMedhttps://doi.org/10.1182/blood-2008-03-143537Google Scholar
- Quintas-Cardama A, Kantarjian H, Manshouri T, Luthra R, Estrov Z, Pierce S. Pegylated interferon alfa-2a yields high rates of hematologic and molecular response in patients with advanced essential thrombocythemia and polycythemia vera. J Clin Oncol. 2009; 27(32):5418-24. PubMedhttps://doi.org/10.1200/JCO.2009.23.6075Google Scholar
- Copland M, Pellicano F, Richmond L, Allan EK, Hamilton A, Lee FY. BMS-214662 potently induces apoptosis of chronic myeloid leukemia stem and progenitor cells and synergizes with tyrosine kinase inhibitors. Blood. 2008; 111(5):2843-53. PubMedhttps://doi.org/10.1182/blood-2007-09-112573Google Scholar
- Pellicano F, Copland M, Jorgensen HG, Mountford J, Leber B, Holyoake TL. BMS-214662 induces mitochondrial apoptosis in chronic myeloid leukemia (CML) stem/progenitor cells, including CD34+38-cells, through activation of protein kinase Cbeta. Blood. 2009; 114(19):4186-96. PubMedhttps://doi.org/10.1182/blood-2009-05-219550Google Scholar
- Heaney NB, Copland M, Stewart K, Godden J, Parker AN, McQuaker IG. Complete molecular responses are achieved after reduced intensity stem cell transplantation and donor lymphocyte infusion in chronic myeloid leukemia. Blood. 2008; 111(10):5252-5. PubMedhttps://doi.org/10.1182/blood-2007-10-118141Google Scholar
- Olavarria E, Siddique S, Griffiths MJ, Avery S, Byrne JL, Piper KP. Posttransplantation imatinib as a strategy to postpone the requirement for immunotherapy in patients undergoing reduced-intensity allografts for chronic myeloid leukemia. Blood. 2007; 110(13):4614-7. PubMedhttps://doi.org/10.1182/blood-2007-04-082990Google Scholar
- Bocchia M, Gentili S, Abruzzese E, Fanelli A, Iuliano F, Tabilio A. Effect of a p210 multipeptide vaccine associated with imatinib or interferon in patients with chronic myeloid leukaemia and persistent residual disease: a multicentre observational trial. Lancet. 2005; 365(9460):657-62. PubMedhttps://doi.org/10.1016/S0140-6736(05)17945-8Google Scholar
- Rojas JM, Knight K, Wang L, Clark RE. Clinical evaluation of BCR-ABL peptide immunisation in chronic myeloid leukaemia: results of the EPIC study. Leukemia. 2007; 21(11):2287-95. PubMedhttps://doi.org/10.1038/sj.leu.2404858Google Scholar
- Rezvani K, Yong AS, Tawab A, Jafarpour B, Eniafe R, Mielke S. Ex vivo characterization of polyclonal memory CD8+ T-cell responses to PRAME-specific peptides in patients with acute lymphoblastic leukemia and acute and chronic myeloid leukemia. Blood. 2009; 113(10):2245-55. PubMedhttps://doi.org/10.1182/blood-2008-03-144071Google Scholar
- Yong AS, Keyvanfar K, Hensel N, Eniafe R, Savani BN, Berg M. Primitive quiescent CD34+ cells in chronic myeloid leukemia are targeted by in vitro expanded natural killer cells, which are functionally enhanced by bortezomib. Blood. 2009; 113(4):875-82. PubMedhttps://doi.org/10.1182/blood-2008-05-158253Google Scholar
- Heaney NB, Pellicano F, Zhang B, Crawford L, Chu S, Kazmi SM. Bortezomib induces apoptosis in primitive chronic myeloid leukemia cells including LTC-IC and NOD/SCID repopulating cells. Blood. 2010; 115(11):2241-50. PubMedhttps://doi.org/10.1182/blood-2008-06-164582Google Scholar
- Jagani Z, Song K, Kutok JL, Dewar MR, Melet A, Santos T. Proteasome inhibition causes regression of leukemia and abrogates BCR-ABL-induced evasion of apoptosis in part through regulation of forkhead tumor suppressors. Cancer Res. 2009; 69(16):6546-55. PubMedhttps://doi.org/10.1158/0008-5472.CAN-09-0605Google Scholar
- Hu Z, Pan XF, Wu FQ, Ma LY, Liu DP, Liu Y. Synergy between proteasome inhibitors and imatinib mesylate in chronic myeloid leukemia. PLoS One. 2009; 4(7):e6257. PubMedhttps://doi.org/10.1371/journal.pone.0006257Google Scholar
- Bellodi C, Lidonnici MR, Hamilton A, Helgason GV, Soliera AR, Ronchetti M. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin Invest. 2009; 119(5):1109-23. PubMedhttps://doi.org/10.1172/JCI35660Google Scholar
- Jorgensen HG, Copland M, Allan EK, Jiang X, Eaves A, Eaves C, Holyoake TL. Intermittent exposure of primitive quiescent chronic myeloid leukemia cells to granulocyte-colony stimulating factor in vitro pro-motes their elimination by imatinib mesylate. Clin Cancer Res. 2006; 12(2):626-33. PubMedhttps://doi.org/10.1158/1078-0432.CCR-05-0429Google Scholar
- Foo J, Drummond MW, Clarkson B, Holyoake T, Michor F. Eradication of chronic myeloid leukemia stem cells: a novel mathematical model predicts no therapeutic benefit of adding G-CSF to imatinib. PLoS Comput Biol. 2009; 5(9):e1000503. PubMedhttps://doi.org/10.1371/journal.pcbi.1000503Google Scholar
- Drummond MW, Heaney N, Kaeda J, Nicolini FE, Clark RE, Wilson G. A pilot study of continuous imatinib vs pulsed imatinib with or without G-CSF in CML patients who have achieved a complete cytogenetic response. Leukemia. 2009; 23 (6):1199-201. PubMedhttps://doi.org/10.1038/leu.2009.43Google Scholar
- Mesa RA. How I treat symptomatic splenomegaly in patients with myelofibrosis. Blood. 2009; 113(22):5394-400. PubMedhttps://doi.org/10.1182/blood-2009-02-195974Google Scholar
- Kroger N, Holler E, Kobbe G, Bornhauser M, Schwerdtfeger R, Baurmann H. Allogeneic stem cell transplantation after reduced-intensity conditioning in patients with myelofibrosis: a prospective, multicenter study of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Blood. 2009; 114 (26):5264-70. PubMedhttps://doi.org/10.1182/blood-2009-07-234880Google Scholar
- Landolfi R, Marchioli R, Kutti J, Gisslinger H, Tognoni G, Patrono C, Barbui T. Efficacy and safety of low-dose aspirin in polycythemia vera. N Engl J Med. 2004; 350(2):114-24. PubMedhttps://doi.org/10.1056/NEJMoa035572Google Scholar
- Harrison CN, Campbell PJ, Buck G, Wheatley K, East CL, Bareford D. Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia. N Engl J Med. 2005; 353(1):33-45. PubMedhttps://doi.org/10.1056/NEJMoa043800Google Scholar