Critical Reviews in Oncology/Hematology 88 (2013) 667–679
The one-two punch: Combination treatment in chronic myeloid leukemia Kendra L. Sweet, Lori A. Hazlehurst, Javier Pinilla-Ibarz ∗ Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 13131 Magnolia Drive, 3 East, Rm 3056H, Tampa, FL 33612, United States Received 1 December 2012; received in revised form 31 May 2013; accepted 18 July 2013
Contents 1.
2.
3.
4.
5.
6.
7.
8.
∗
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.1. Are tyrosine kinase inhibitors curative for chronic myeloid leukemia? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.2. Need for a one–two punch . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . The JAK-STAT pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. JAK2 in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. STAT3 in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. STAT5 in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4. JAK/STAT pathway inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4.1. JAK2 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4.2. STAT3 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4.3. STAT5 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hedgehog signaling pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. The Hh pathway in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Hh inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wnt/β-catenin pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1. The Wnt pathway in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2. Wnt pathway inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Protein phosphatase 2A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1. Role of PP2A in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2. PP2A activators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . mTOR pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.1. mTOR complexes in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.2. Dual mTORC2/mTORC1 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Other drug classes/molecules with potential utility in CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.1. Interferon alfa . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.2. HDACis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.3. Alox5 (5-lipooxygenase) inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.4. HSP90 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conflict of interest statements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Corresponding author. Tel.: +1 813 745 1387; fax: +1 813 745 6817. E-mail address:
[email protected] (J. Pinilla-Ibarz).
1040-8428/$ – see front matter © 2013 Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.critrevonc.2013.07.017
668 668 668 669 669 669 669 670 671 671 671 671 671 671 672 672 672 672 673 673 673 674 674 675 675 675 675 675 675 675 676 676 676 679
668
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
Abstract Despite the success of tyrosine kinase inhibitor (TKI) therapy in patients with chronic myeloid leukemia (CML), minimal residual disease persists, requiring indefinite treatment. Accumulated evidence has shown that leukemic stem cells (LSCs) in the bone marrow can survive TKI treatment via downstream BCR-ABL1–independent signaling pathways that are activated by soluble growth factors and interactions with the extracellular matrix in the bone marrow microenvironment. Research efforts have therefore turned to the identification and development of agents that target LSCs, and together with TKIs, have the potential to eradicate CML. A number of such agents are now under clinical investigation, and others are soon to enter early-phase studies. This review examines the pathways, molecular targets, and potential new therapeutics that, with TKIs, may provide an effective “one-two punch” to cure CML. © 2013 Elsevier Ireland Ltd. All rights reserved. Keywords: Imatinib; Nilotinib; Dasatinib; Chronic myeloid leukemia; Tyrosine kinase; Leukemic stem cells
1. Introduction 1.1. Are tyrosine kinase inhibitors curative for chronic myeloid leukemia? Chronic myeloid leukemia (CML) is caused by the constitutive activity of an aberrant tyrosine kinase (BCR-ABL1) that is created by the translocation of the Abelson (ABL) 1 oncogene on chromosome 9 and the breakpoint cluster region (BCR) on chromosome 22 [1–3]. The BCR-ABL1 kinase, in turn, drives the activation of a series of pro-survival signaling pathways that eventually lead to malignant transformation [4]. Understanding of the molecular basis of CML enabled the development of targeted treatment to block BCR-ABL1 activity, a strategy that proved highly successful. Initial phase III data from the international randomized study of interferon and STI571 (IRIS) demonstrated overwhelmingly positive results for imatinib, the first available BCR-ABL1 tyrosine kinase inhibitor (TKI), versus the standard treatment at that time, interferon plus cytarabine [5]. Indeed, imatinib completely altered the treatment paradigm, making long-term survival a new reality for most patients. Eight-year results of the IRIS study showed estimated overall survival (OS) to be 85%; OS was 93% if only CML-related mortality and deaths prior to stem cell transplant were considered [6]. Yet it was evident from IRIS that some patients experienced disease progression while on treatment with imatinib. Most events occurred early, within the first 3 years of treatment [6]. Further, the stop imatinib (STIM) study, which probed the possibility of imatinib discontinuation in patients who had achieved a complete molecular remission (CMR; defined as undetectable residual disease on quantitative reverse transcriptase-polymerase chain reaction [qRT-PCR]) for at least 2 years, found that only select patients could cease treatment without relapse [7,8]. Even patients who do not relapse after discontinuation have evidence of disease when assessed by sophisticated methods such as qRT-PCR [9]. Indeed, some patients who are in prolonged remission with continued imatinib treatment may still harbor BCRABL1–positive stem cells [10]. Thus, despite its indisputable benefits, imatinib alone is unlikely to eradicate CML. Four more-potent TKIs, nilotinib, dasatinib, bosutinib, and ponatinib, were investigated and approved for the treatment of CML patients with intolerance of or resistance to
prior therapy including imatinib. Two of these agents, nilotinib and dasatinib, were evaluated in separate randomized, controlled studies as first-line treatment for CML in newly diagnosed patients. Nilotinib was compared with imatinib in the evaluating nilotinib efficacy and safety in clinical trials–newly diagnosed patients (ENESTnd) study [11], and dasatinib was compared with imatinib in the dasatinib versus imatinib study in treatment–naive cml patients (DASISION) [12]. Results of 4 years’ follow-up in ENESTnd and 3 years’ follow-up in DASISION are now available [13,14]. Nilotinib demonstrated a significantly higher rate of major molecular response (MMR) and CMR compared with imatinib, and dasatinib demonstrated a significantly higher MMR rate versus imatinib. The recent long-term data also show that fewer patients experienced disease progression with the newer agents; however, only the difference between nilotinib and imatinib was statistically significant [13,14]. Minimal residual disease remains evident with all available TKIs, however, making indefinite treatment necessary at this time. Although TKIs are generally well tolerated, long-term, potentially lifelong, treatment with TKIs increases the probability that patients will experience adverse events. The elimination of all residual disease would preclude the need for prolonged treatment and minimize the effect of TKI-related adverse events. It is clear that new strategies are needed in order to address these treatment gaps, with cure as the ultimate goal. 1.2. Need for a one–two punch In recent years, it has increasingly become appreciated that the bone marrow (BM) microenvironment provides a protective sanctuary in which leukemic stem cells (LSCs) can survive TKI treatment [15]. Normally, inhibition of BCR-ABL1 activity in CML cells leads to suppression of downstream pro-survival signaling pathways, tipping the balance toward pro-apoptotic mechanisms that lead to cell death. More-primitive progenitor cells that reside in the BM, however, are resistant to TKI-induced apoptosis, even though BCR-ABL1 activity is successfully inhibited [16–20]. It is believed that signaling pathways that are downstream of BCR-ABL1 remain intact in LSCs; these pathways may be activated by soluble growth factors and interactions with the extracellular matrix in the BM microenvironment, enabling
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
LSC survival [15]. Therefore, recent research efforts have focused on the identification of suitable targets and the development of agents that can block BCR-ABL1-independent pro-survival pathways in LSCs. In this review, a number of key pathways and molecules implicated in the survival of LSCs are examined. In addition, we review agents that show promise for providing the necessary “second punch” that may, in combination with TKI therapy, be curative for CML. Many such agents are currently being tested in various solid tumors and other hematologic disorders; investigation of some of these agents for the treatment of CML has also begun.
669
activation of downstream pathways, and ultimately interfered with oncogenic signaling [23]. 2.2. STAT3 in CML STAT3 is part of the signaling pathway downstream of JAK2 [23]. Data from in vitro CML models suggest a role for STAT3 in the development of TKI resistance in BM [24]. Bewry et al., used an in vitro coculture BM stroma model to determine the influence of BM soluble factors and extracellular matrices on imatinib sensitivity [24]. K562 cells (a transformed cell line derived from a CML patient), when cultured in BM stroma derived conditioned medium, were resistant to BCR-ABL1 inhibition. Drug resistance was associated with increased levels of STAT3. When STAT3 levels were reduced with small interfering RNA, K562 cells became sensitive to imatinib-induced cell death. Reducing STAT3 levels in regular growth conditions had no effect on imatinib sensitivity, suggesting that STAT3mediated drug resistance was due to factors present in the BM microenvironment. These data support a novel mechanism of BCR-ABL1-independent resistance and provide a rationale for the development of agents that can inhibit STAT3, thereby increasing the efficacy of TKIs in the BM.
2. The JAK-STAT pathway The janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway is the principal signaling mechanism for a multitude of cytokines and growth factors [21]. JAK activation stimulates cell proliferation, differentiation, migration, and apoptosis, which are critical to numerous processes including hematopoiesis, immune system development, and mammary gland development (Fig. 1) [22]. 2.1. JAK2 in CML
2.3. STAT5 in CML In vitro and in vivo preclinical studies suggest that the JAK2 signaling pathway is activated in CML; inhibition of the JAK2 pathway reduced phosphorylation of BCR-ABL1 and
STAT5, like STAT3, is a downstream component of the JAK/STAT pathway [25]. In vitro studies demonstrated that
Cytokine ligand JAK2
JAK2
JA
P
K2
P
JA
Stat
Stat
P
K JA
2
St
P
St
at
at
St
P
K2
?
JA P
at
St
Ras P
at
P Stat
SOC3
Stat
K2
? Raf
PI3K SOC1 P
MEK
Akt
SOC1 SOC3
P mTOR
ERK
FoxO P
P ERK
Activation of genes important in proliferation and survival
Fig. 1. JAK-STAT pathway. Adapted with permission from Levine et al. [22].
670
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
Table 1 Molecules in clinical development and their status in hematologic diseases. Molecule
Clinical development status for hematologic diseases
JAK2 inhibitors AZD1480
•Phase I and II study in PMF, PPV-MG, ET-MF [28]
CEP-701 (lestaurtinib)
•Phase I and II studies in MF [29,30] •Phase II study in FLT3 mutant AML [31] •Phase III study in infants with newly diagnosed ALL [32]
CYT387
•Phase I/II studies in PMF, PPV-MF, PET-MF [33,34]
LY2784544
•Phase I and II studies in MF, PV, and EF [35,36]
NS-018
•Phase I/II study in PMF, PPV-MF, or PET-MF [37]
Ruxolitinib (INCB018424)
•Indicated for intermediate or high-risk MF (PMF, PPV-MF, PET-MF] [38] •Phase I and II studies in PMF, PPV-MF and PET-MF [39–42] •Phase I and II study in refractory/relapsed AML or ALL [43] •Phase II study in AML, ALL, MDS, and CML [44,45] •Phase II study in relapsed/refractory DLBCL and PTCL [46]
SAR302503 (TG101348)
•Phase II and III studies in MF, PV-MF and ET-MF [47]
SB1518 (pacritinib)
•Phase I and II studies in AML, CML, CMML, MDS, MF [48,49] •Phase II studies in PMF, PPV-MF, PET-MF [50,51] •Phase I and II studies in HL, MCL, B-cell lymphoma [52,53]
STAT3 inhibitors ISIS 481464 (ISIS-STAT3Rx)
•Phase I study in advanced cancers [54]
OPB51602
•Phase I study in advanced hematologic malignancies (MM, NHL, AML, ALL, CML) [55]
Pyrimethamine
•Phase I/II study in CLL, SLL [56]
Hedgehog pathway agents BMS-833923 (XL139)
•Phase I study in multiple myeloma [57] •Phase II study in combination with dasatinib in CML [58]
GDC-0449 (vismodegib)
•Phase I study in multiple myeloma and plasma cell neoplasms [59]
IPI-926
•Phase II study in MF [60]
LDE225 (saridegib)
•Phase Ib study in combination with nilotinib in CML [61]
PF-04449913
•Phase I study in hematologic malignancies [62] •Phase Ib study in AML and MDS [63]
Wnt/-catenin pathway agents CWP232291
•Phase I study in acute myeloid leukemia [64]
PRI-724
•Phase I and II study in AML and CML [65]
Dual mTORC2/mTORC1 inhibitors CC-223
•Phase I/II study in advanced solid tumors, NHL and MM [66]
OSI-027
•Phase I study in patients with advanced solid tumors or lymphoma [67]
AML, acute myeloid leukemia; PMF, primary myelofibrosis; PPV-MF, post-polycythemia vera myelofibrosis; ET-MF, essential thrombocythemia myelofibrosis; PET-MF, post-essential thrombocythemia myelofibrosis; ALL, acute lymphoblastic leukemia; MDS, myelodysplastic syndrome; CML, chronic myelogenous leukemia; DLBL, diffuse large B-cell lymphoma; PTCL, peripheral T-cell lymphoma; CMML, chronic myelomonocytic leukemia; CLL, chronic lymphocytic leukemia; SLL, small lymphocytic leukemia; HL, Hodgkin lymphoma; MCL, mantle cell lymphoma; NHL, non-Hodgkin lymphoma; MM, multiple myeloma.
BCR-ABL1 can directly stimulate the tyrosine phosphorylation and dimerization of STAT5, bypassing JAK2, followed by the translocation of STAT5 dimers to the nucleus, where they promote activation of pro-survival genes [26]. More recently, high levels of STAT5 were shown to confer resistance to imatinib, nilotinib, and dasatinib, independently of JAK2. Whereas high levels of STAT5 were associated with reduced TKI-mediated cytotoxicity, reduction of STAT5 levels correlated with increased TKI-mediated killing. Further, STAT 5 levels from patient-derived cells correlated with TKI resistance and disease progression [27]. Although the authors
suggest that STAT5 inhibition may serve as an effective target for TKI resistance in patients with advanced disease [27], this strategy may also prove useful for eradicating CML cells residing in the protective BM microenvironment.
2.4. JAK/STAT pathway inhibitors The preclinical evidence to support the combination of JAK/STAT inhibitors with BCR-ABL TKIs is very promising.
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
2.4.1. JAK2 inhibitors A number of small-molecule JAK2 inhibitors are in various stages of clinical development in hematologic diseases (Table 1), and one, ruxolitinib, has received US Food and Drug Administration (FDA) approval for the treatment of other myeloproliferative disorders [38]. A phase II study of ruxolitinib monotherapy in 38 patients with various refractory or relapsed hematologic malignancies, including two patients with CML, demonstrated that ruxolitinib provided modest activity as a single agent. Three patients, all with acute myeloid leukemia (AML) after having had a myeloproliferative disorder, showed a significant response [45]. Notably, samples from five patients, all of whom had elevated STAT3 levels at baseline, showed significantly decreased STAT3 levels 2 h after ruxolitinib dosing. Recently, a series of preclinical studies has provided a rationale for the clinical study of the combination of ruxolitinib and nilotinib in CML [68]. Conditioned medium from BM-derived mesenchymal stromal cells was shown to activate STAT3 in blastic-phase CML-derived cell lines and in LSCs obtained from patients with CML; STAT activation protected cells against nilotinib-mediated cell death. Ruxolitinib was able to block STAT3 activation and, when combined with nilotinib, sensitized both cell lines and patient-derived LSCs to the cytotoxic effects of nilotinib [68]. Utilizing small interfering–RNA strategies, Nair et al., demonstrated that reduced expression of JAK2 and TYK2 was required to sensitize cells to nilotinib. These results suggest that the combination of a promiscuous JAK inhibitor and BCR-ABL1 inhibition may be able to overcome the protective effects of the BM microenvironment and eliminate LSCs that are responsible for minimal residual disease. In our institution and based on our previous investigations, we have recently started a phase I/II trial to evaluate the toxicity and efficacy in achieving CMR of the combination of nilotinib and ruxolitinib. The future approval of other JAK-2 inhibitors and their availability in clinical practice will require the investigation of other combinations in the near future. 2.4.2. STAT3 inhibitors Several agents that specifically target STAT3 are in early clinical development, including one being evaluated in CML (Table 1). A phase I study of OPB-51602 is ongoing in Japan in patients with relapsed or refractory hematologic malignancies, including CML [55]. ISIS 481464, an antisense oligonucleotide inhibitor, is being examined in a phase I trial in patients with advanced cancers [54]. Pyrimethamine is an older agent used to treat malaria and other parasitic infections. It recently was shown to inhibit STAT3, and a phase I/II study in relapsed chronic lymphocytic leukemia/small lymphocytic leukemia has been initiated, with completion estimated for February 2014 [56].
671
identified, by screening of compounds known to be safe in humans, to be a STAT5 inhibitor [69]. In vitro and ex vivo experiments demonstrated that pimozide decreases STAT5 target gene expression in KU812 and K562 CML cells and decreases colony-forming ability in patient-derived CD34+ BM cells from CML patients, but itself does not inhibit TK activity. Notably, the combination of pimozide with imatinib or nilotinib was synergistic for reducing STAT5 phosphorylation and decreasing the viability of KU812 cells, suggesting that such combinations may have clinical utility [69]. Indirubin is the major active antitumor component of a traditional Chinese herbal medicine that is reported to be useful in CML [70]. Preclinical studies have shown that compound E804, a potent derivative of indirubin, inhibits the STAT5 signaling pathway downstream of BCR-ABL1 and induces apoptosis of K562 CML cells, CML cells expressing the T315I mutation, and progenitor CD34+ CML cells from patient samples. 3. Hedgehog signaling pathway The Hedgehog (Hh) pathway, a signaling cascade that was initially identified in Drosophila melanogaster, is highly conserved among vertebrates [71]. It is essential for regulating the proliferation, migration, and differentiation of embryonic cells [72]. Aberrant reactivation of the pathway in adult tissue is associated with a wide variety of cancers, particularly basal cell carcinoma, medulloblastoma, and brain, gastrointestinal, breast, and prostate cancers. At a molecular level, in the absence of an Hh ligand, the patched (Ptch) receptor inhibits the action of smoothened (Smo), a G protein–coupled receptor. Binding of Hh ligands to Ptch relieves inhibition of Smo, which in turn activates zinc-finger like Gli proteins, leading to the transcription of tumor-promoting genes (Fig. 2) [71]. 3.1. The Hh pathway in CML Preclinical experiments have suggested that the Hh pathway is essential for the maintenance of LSCs in CML, and Smo, specifically, may be a rational therapeutic target. Smo is upregulated in LSCs relative to normal hematopoietic stem cells, and its inhibition in mice depletes LSCs, reduces the incidence of CML development, and, in combination with nilotinib, appeared to be more effective in preventing relapse than nilotinib alone [73]. In Smo knockout mice, hematopoietic stem cell renewal is impaired, and induction of CML by BCR-ABL1 is reduced [74]. A clinical strategy incorporating both Smo and BCR-ABL1 inhibition, therefore, may be successful in preventing the drug resistance and disease recurrence associated with TKI treatment alone [74]. 3.2. Hh inhibitors
2.4.3. STAT5 inhibitors The evaluation of agents that target STAT5 is at a preclinical stage. The psychotropic agent pimozide was recently
The preclinical data are convincing that the aberrant reactivation of the Hh pathway plays a significant role in
672
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
OFF
Hh ligand
ON
PTCH
PTCH SMO GLiR
SMO
Hh target genes ON
Hh target genes OFF
GLi
R
GLiA
GLiA
Fig. 2. Hedgehog signaling pathway. Adapted with permission from Mar et al. [71].
the preservation of LSCs. Among the Hh inhibitors currently being investigated for hematologic disorders (Table 1), two are being evaluated for CML. A phase Ib dose-finding study of the Smo antagonist LDE225 is being tested in combination with nilotinib in patients with CML in the chronic phase (CML-CP) who has failed prior treatment with other TKIs [61]. The other clinical trial is a large phase II randomized study evaluating the combination of the Smo inhibitor BMS833923 and dasatinib versus dasatinib alone in patients with CML-CP who did not achieve a major molecular response within 1 year of treatment with dasatinib alone. Some studies have shown that there are higher levels of Hh reactivation in the more advanced phases of CML than in CML-CP; if so, the combination of Hh or Smo inhibitors with TKIs may be more beneficial in advanced phases. The ongoing trials will better define the role of these compounds in different phases of CML.
displayed an enhanced capacity for self-renewal and leukemic potential [77]. The role of β-catenin in the maintenance of LSCs in CML was assessed in a mouse model [78]. Interestingly, deletion of β-catenin once CML was established did not increase survival in mice; however, β-catenin deletion plus imatinib synergized to eliminate CML stem cells that remained after imatinib treatment. This effect was also seen with pharmacologic inhibition of β-catenin with indomethacin [78]. Another research group has uncovered the involvement in CML of a little-known Wnt receptor, FZD-8, in a noncanonical Wnt/Ca2+ /nuclear factor of activated T-cell (NFAT) signaling pathway [79]. Antagonism of this pathway led to impaired NFAT activity, decreased cytokine production, and enhanced sensitivity to BCR-ABL1 inhibition in a mouse model. 4.2. Wnt pathway inhibitors
4. Wnt/β-catenin pathway The Wnt signaling pathway is a network of proteins involved in embryogenesis, in normal physiologic processes in adult animals, and in cancer [75]. Wnt signaling also appears to be a key pathway in stem cell regulation. In the canonical pathway, interaction of Wnt ligands with frizzled receptors and LDL receptor–related protein coreceptors blocks the degradation of β-catenin, an intracellular signaling molecule. The stabilized β-catenin then moves into the nucleus, where it induces the expression of target genes. In the case of mutations that block the degradation of β-catenin, target gene expression increases (Fig. 3) [76].
The demonstration that increased levels of β-catenin are associated with imatinib resistance and progression [77] makes use of a Wnt pathway inhibitor, or more specifically, a drug that directly inhibits β-catenin, a promising therapeutic avenue in combination with imatinib or other TKIs. Currently, there is one clinical trial investigating the safety and efficacy of Wnt pathway inhibition in patients with myeloid malignancies, including CML (Table 1). PRI-724, a specific inhibitor of cAMP response element-binding protein (CREB)-binding protein (CBP)/β-catenin, is being tested in a phase I/II dose-escalation study in patients with advanced malignancies. Patients with CML-CP who have failed two TKIs or patients with CML-AC/BC will be enrolled in the phase II portion of the study [65].
4.1. The Wnt pathway in CML Involvement of the Wnt pathway in CML was first described in 2004. Compared with normal stem cells from BM, progenitor CML cells from patients with accelerated phase/blast crisis (AP/BC) CML or patients who were resistant to imatinib demonstrated higher levels of βcatenin expression. Progenitor cells from these patients also
5. Protein phosphatase 2A Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase that has been identified as an important tumor suppressor that negatively regulates many pro-survival signaling pathways. Inhibition of PP2A, specifically by the
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
673
Normal HSC
Induced β-catenin expression in HSC
FZD
FZD LRP
LRP
Cytokine signals? β-cat
Proteasome
β-cat
β-cat
β-cat
β-cat
β-cat β-cat
Tcf-LEF
β-cat
Repression of Wnt target genes
β-cat
β-cat
Tcf–LEF
Expression of Wnt target genes
Multilineage progenitors
Production of mature blood cells
Lack of mature blood cells (anemia, immune deficiency)
Fig. 3. Wnt signaling pathway. Adapted with permission from Trowbridge et al. [76].
SET oncoprotein, is associated with transformation in a range of human cancers [80,81]. 5.1. Role of PP2A in CML Perrotti et al. [82] have shown that BCR-ABL1–positive cell lines and CML progenitor cells derived from patients with CML-CP and -BC have enhanced expression of the PP2A inhibitor SET (Fig. 4). SET expression is higher in progenitors derived from patients with CML-BC than those with CML-CP, and higher expression is associated with progressive loss of PP2A-mediated tumor suppression [82,83]. Reactivation of PP2A has been shown to suppress growth, increase apoptosis, and decrease leukemogenesis in model systems; this effect may occur through the PP2A-mediated activation of a second tumor suppressor, Src homology region 1–domain phosphatase 1 (SHP-1), which can inactivate BCRABL1 [82,83].
6. mTOR pathway Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that forms the core of two multi-protein signaling complexes, mTORC1 and mTORC2, which regulate cell growth, protein synthesis, and cell-cycle progression
PP2A inhibition p210-BCR-ABL1 hnRNP A1
SET
p190-BCR-ABL1 A C B PP2A
SHP-1
PP2A activation
CML-BP Ph(+) B-ALL
p210-BCR-ABL1 Forskolin
5.2. PP2A activators
FTY720
p190-BCR-ABL1
Interestingly, preclinical research in CML has identified FTY720, a drug approved for the treatment of multiple sclerosis, as an activator of PP2A [82]. Presently, there are no clinical trials evaluating the combination of TKI and FTY720 in CML. In the future we may see trials combining this drug with TKIs in the setting of advanced phases of CML or TKI resistance.
SHP-1
A C B PP2A Impairment of leukemogenesis
Fig. 4. Protein phosphatase 2A (PP2A). Adapted with permission from Perrotti et al. [82].
674
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
mTORC1 Growth Factors
Amino Acids
mTORC2
Energy
Growth Factors
Stress
De
PRR5/Protor
mTOR
pt
or
a
Ra p
FKB
P12
De
mTOR
TM-Site T450
P
P
HM-Site S473
P
HM-Site T389
P AKT
P
P SGK1
AGC Kinases
P
S6K1 dependent
mLST8/ GβL
hSIN1
PRAS40
Cap-
or
Rictor mLST8/ GβL
Raptor
4EBP1
pt
P PKCα
translation
Actin cytoskeleton organization Cell proliferation and survival
Cell growth and proliferation
Fig. 5. mTORC1 and mTORC2 signaling complexes. Adapted with permission from Foster and Fingar [85].
via complex interactions with numerous signaling pathways, including phosphatidylinositol 3-Kinase/Akt [84]. mTORC1 and mTORC2 are distinguished by their sensitivities to the mTOR inhibitor rapamycin and by their distinct partner proteins, substrates, and cellular functions. mTORC1, which is formed by mTOR, Raptor and mLST8, is allosterically inhibited by rapamycin and other analogs of rapamycin (e.g., temsirolimus, everolimus). mTORC2, which includes mTOR, Rictor, mLST8, and SIN1, is not inhibited by rapamycin and its analogs (Fig. 5) [85].
6.2. Dual mTORC2/mTORC1 inhibitors In vitro data show that the dual mTORC inhibitors induce apoptosis in both mutated and unmutated BCR-ABL–expressing cells [86,88]. Two dual mTORC2/mTORC1 inhibitors are in early clinical development (Table 1): OSI-027 and CC-223. Neither of these trials includes patients with CML. However, the combination of dual mTORC inhibitors with TKIs has good potential to induce apoptosis in LSCs that are resistant to TKIs.
A. 6.1. mTOR complexes in CML Recent preclinical research has demonstrated that rapamycin-insensitive (RI)–mTORC1 complexes are activated in BCR-ABL-expressing cells and are involved in the translation of genes that mediate mitogenic responses. mTORC2 complexes are also activated in, and are important for the growth and survival of, BCR-ABL1–expressing cells [86]. The mTORC1 inhibitor everolimus has shown promise in a phase I/II clinical trial of patients with relapsed or refractory hematologic malignancies; however, CML patients were not included in the trial [87]. Notably, a dual mTORC2/mTORC1 inhibitor, OSI-027, potently suppressed LSCs from CML patients, providing a rationale for incorporating such agents in combination therapy for CML (Fig. 6) [86].
B. LSCQ LSCP
LSCQ Self renewal
LSCP
EPC
EPC
LPC
LPC
LC
LC
Mobilize quiescent LSCs: • HDACinhibion • HSP90 inhibion • Interferon alfa • mTORC inhibion Target self renewal pathways: • Alox5 inhibion • Hh/Smo inhibion • JAK2 inhibion • PP2A acvaon • Wnt/ catenin inhibion
Fig. 6. Approaches to the eradication of leukemic stem cells (LSCs). A. LSCs cycle between quiescent (LSCQ ) and proliferative (LSCP ) states. LSCP have the capacity for self-renewal. Eventually, LSCP differentiate into early progenitor cells (EPC), which become late progenitor cells (LPC) that mature into leukemic cells (LC). B. Two main approaches to eradication of LSCs: mobilization of LSCQ and targeting of self-renewal pathways.
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
675
7. Other drug classes/molecules with potential utility in CML
ongoing phase I study (NCT00686218) is investigating the combination of imatinib and panobinostat.
A number of other molecules and drug classes are also being evaluated in hematologic disorders, including interferon alfa, histone deacetylase inhibitors (HDACis), an Alox5 inhibitor, and heat shock protein (HSP) 90 inhibitors.
7.3. Alox5 (5-lipooxygenase) inhibitor
7.1. Interferon alfa Interferon alfa has been an important treatment modality for CML for decades, although it has not been considered standard of care for CML-CP since the introduction of TKIs. Interest in interferon alfa for CML has shifted focus since observations were made that prior interferon alfa therapy may be correlated with sustained treatment-free remission after discontinuation of TKI therapy in clinical studies [89,90]. These clinical observations, in light of preclinical findings that exposure to interferon alfa can activate quiescent hematopoietic stem cells in a mouse model [91], suggest that prior or concomitant treatment of patients with interferon alfa can sensitize LSCs to eradication by TKI therapy. Several clinical studies have examined the combination of interferon alfa plus imatinib in patients with CML-CP [92–95]. Overall, the combination improves response rates or induces more-rapid responses over standard-dose imatinib alone, but the combination also increases the incidence of treatment-related adverse events. At present, others studies are ongoing that will evaluate the combination of interferon alfa plus nilotinib or imatinib (NCT00573378) and interferon alfa plus dasatinib (NCT01725204). Whether the combination of interferon alfa plus imatinib or other TKI results in LSC elimination remains to be determined and future trials with exhaustive correlative studies may answer this question. 7.2. HDACis Small-molecule HDACis have shown significant biological effects in preclinical models of oncogenesis; they induce growth arrest, differentiation, and apoptosis in vitro and in vivo, and can kill both actively proliferating and nonproliferating cancer cells [96,97]. In experimental models of CML, cotreatment with imatinib and the pan-HDACi LBH-589 induced apoptosis in nonproliferating, otherwise imatinib-resistant LSCs, significantly reduced LSC capacity to repopulate marrows of immunodeficient mice, reduced the number of putative CML LSCs in a transgenic mouse model, and reduced expression of genes related to primitive stem cell state [98]. Many such inhibitors are now in clinical trials, with several early-phase studies examining the combination of a TKI and HDACi in patients with CML. One phase I study (NCT00816283) has examined dasatinib in combination with vorinostat (no results have been published) and one
In a preclinical model of CML, Chen and colleagues have identified the arachidonate 5-lipoxygenase (5-LO) gene (Alox5) as a critical regulator of LSCs [99]. BCR-ABL1 does not induce CML in BCR-ABL1-transduced Alox5-/-mice, and Alox-deficient mice lack LSCs. These findings led to a phase I study of zileuton, a specific 5-lipoxygenase inhibitor approved for the treatment of asthma, in patients with CMLCP already receiving imatinib (NCT01130688). The study has a planned completion date of December 2014. 7.4. HSP90 inhibitors HSP90 is a molecular chaperone protein necessary for cellular survival [100]. HSPs monitor protein folding, prevent misfolding, and restore three-dimensional protein structure, activities that are essential during toxic cellular stress. Oncogenic cells depend on their ability to withstand endogenous (anoxia, nutrient deprivation, pH changes, and deranged signaling pathways) and exogenous (chemotherapy and radiation therapy) stressors for survival; inhibition of HSP90 in multiple cancer animal models has shown significant antitumor effects. Although the results of early efficacy studies of HSP90 inhibitors were encouraging, the ideal use of HSP90targeted therapeutics will require a deeper understanding of the functional differences in malignant neoplastic disease and normal cellular physiology.
8. Summary The BCR-ABL1 TKIs have been a groundbreaking success in treating CML. However, these agents may be unable to completely eradicate CML. Although long-term treatmentfree periods have been noted in some patients who have discontinued TKI treatment, evidence of minimal residual disease remains. Experimental studies suggest that quiescent LSCs survive TKI treatment because of microenvironmental factors in the BM. Novel pathways and molecules have been the focal point of drug development to target LSCs. It is anticipated that agents that inhibit critical pathways in LSCs, in combination with TKIs, may provide the one-two punch that is needed to eradicate CML.
Conflict of interest statements Javier Pinilla-Ibarz has received honoraria and research support from Bristol-Myers Squibb and Novartis. Kendra L. Sweet and Lori A. Hazlehurst have no conflicts of interest to report.
676
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
Reviewers Elias Jabbour, M.D., Associate Professor, University of Texas MD Anderson, Leukemia, 1515 Holcombe Blvd, Department of Leukemia, Unit 428, Houston, TX 77030, United States. Photis Beris, Associate Professor, Faculté de Médecine, Geneva University, Swiss Internal Medicine, 51 avenue Blanc, CH-1202 Genève, Switzerland. Michael J. Mauro, M.D., Professor, Oregon Health Science University, Division of Hematology and Medical Oncology, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, United States. Francis Giles, M.D., Professor, NUI Galway, HRB Clinical Research Facility, Geata an Eolais, University Road, Galway, Ireland. Dominik Heim, Head of Hematology Department, University Hospital, Basel, Spitalstrasse 21, Petersgraben 4, CH-4031 Basel, Switzerland.
[9]
[10]
[11]
[12]
[13]
[14]
Acknowledgements Financial support for medical editorial assistance was provided by Novartis Pharmaceuticals Corporation. We thank Mariana Ovnic, PhD, and Anna Lau, PhD, of Percolation Communications LLC for medical editorial assistance.
[15]
[16]
References [17] [1] Heisterkamp N, Stam K, Groffen J, de Klein A, Grosveld G. Structural organization of the bcr gene and its role in the Ph’ translocation. Nature 1985;315(6022):758–61. [2] Nowell PC, Hungerford DA. Chromosome studies on normal and leukemic human leukocytes. Journal of the National Cancer Institute 1960;25:85–109. [3] Rowley JD. Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 1973;243(5405):290–3. [4] Deininger MW, Goldman JM, Melo JV. The molecular biology of chronic myeloid leukemia. Blood 2000;96(10):3343–56. [5] O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. New England Journal of Medicine 2003;348(11):994–1004. [6] Deininger M, O’Brien SG, Guilhot F, Goldman JM, Hochhaus A, Hughes TP, et al. International randomized study of interferon vs STI571 (IRIS) 8-year follow up: sustained survival and low risk for progression or events in patients with newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP) treated with imatinib. Blood (ASH Annual Meeting Abstracts) 2009;114(22): 1126. [7] Mahon FX, Rea D, Guilhot J, Guilhot F, Huguet F, Nicolini F, et al. 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 Oncology 2010;11(11):1029–35. [8] Mahon F-X, Rea D, Guilhot J, Guilhot F, Huguet F, Nicolini FE, et al. Discontinuation of imatinib in patients with chronic myeloid leukemia who have maintained complete molecular response: update
[18]
[19]
[20]
[21] [22]
[23]
[24]
[25] [26]
results of the STIM study. Blood (ASH Annual Meeting Abstracts) 2011;118(21):603. Ross DM, Branford S, Seymour JF, Schwarer AP, Arthur C, Bartley PA, et al. Patients with chronic myeloid leukemia who maintain a complete molecular response after stopping imatinib treatment have evidence of persistent leukemia by DNA PCR. Leukemia 2010;24(10):1719–24. Chu S, McDonald T, Lin A, Chakraborty S, Huang Q, Snyder DS, et al. Persistence of leukemia stem cells in chronic myelogenous leukemia patients in prolonged remission with imatinib treatment. Blood 2011;118(20):5565–72. Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C, et al. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. New England Journal of Medicine 2010;362(24):2251–9. Kantarjian H, Shah NP, Hochhaus A, Cortes J, Shah S, Ayala M, et al. Dasatinib versus imatinib in newly diagnosed chronicphase chronic myeloid leukemia. New England Journal of Medicine 2010;362(24):2260–70. Hochhaus A, Shah NP, Cortes JE, Baccarani M, Bradley-Garelik MB, Dejardin D, et al. Dasatinib versus imatinib (IM) in newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP): DASISION 3-year follow-up. Journal of Clinical Oncology 2012;30(suppl): 6504. Kantarjian HM, Kim D-W, Issaragrisil S, Clark RE, Reiffers J, Nicolini FE, et al. Enestnd 4-year (y) update: continued superiority of nilotinib vs imatinib in patients (pts) with newly diagnosed Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia in chronic phase (CML-CP). Blood (ASH Annual Meeting Abstracts) 2012;120(21):1676. Nair RR, Tolentino J, Hazlehurst LA. The bone marrow microenvironment as a sanctuary for minimal residual disease in CML. Biochemical Pharmacology 2010;80(5):602–12. Copland M, Hamilton A, Elrick LJ, Baird JW, Allan EK, Jordanides N, et al. 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. 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. Journal of Clinical Investigation 2011;121(1):396–409. Graham SM, Jorgensen HG, Allan E, Pearson C, Alcorn MJ, Richmond L, et al. Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood 2002;99(1):319–25. Konig H, Holtz M, Modi H, Manley P, Holyoake TL, Forman SJ, et al. Enhanced BCR-ABL kinase inhibition does not result in increased inhibition of downstream signaling pathways or increased growth suppression in CML progenitors. Leukemia 2008;22(4):748–55. Konig H, Holyoake TL, Bhatia R. Effective and selective inhibition of chronic myeloid leukemia primitive hematopoietic progenitors by the dual Src/Abl kinase inhibitor SKI-606. Blood 2008;111(4):2329–38. Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. Journal of Cell Science 2004;117(Pt 8):1281–3. Levine RL, Pardanani A, Tefferi A, Gilliland DG. Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders. Nature Reviews Cancer 2007;7(9):673–83. Samanta A, Perazzona B, Chakraborty S, Sun X, Modi H, Bhatia R, et al. Janus kinase 2 regulates BCR-ABL signaling in chronic myeloid leukemia. Leukemia 2011;25(3):463–72. 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;7(10):3169–75. Turhan AG. STAT5 as a CML target: STATinib therapies? Blood 2011;117(12):3252–3. Carlesso N, Frank DA, Griffin JD. Tyrosyl phosphorylation and DNA binding activity of signal transducers and activators of
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679
[27]
[28]
[29]
[30]
[31]
[32]
[33]
[34]
[35]
[36]
[37]
[38] [39]
[40]
[41]
transcription (STAT) proteins in hematopoietic cell lines transformed by BCR/ABL. Journal of Experimental Medicine 1996;183(3):811–20. Warsch W, Kollmann K, Eckelhart E, Fajmann S, Cerny-Reiterer S, Holbl A, et al. High STAT5 levels mediate imatinib resistance and indicate disease progression in chronic myeloid leukemia. Blood 2011;117(12):3409–20. Verstovsek S. Study to Assess the Safety of Azd1480 in Patients with Myeloproliferative Diseases; 2012 http://clinicaltrials.gov/ ct2/show/NCT00910728 (accessed 18.07.12). Silverman L, Hoffman R. CEP-701 (Lestaurtinib) in 2012 http://clinicaltrials.gov/ct2/show/ Myelofibrosis; NCT00668421?term=Lestaurtinib&rank=1 (accessed 18.07.12). Santos FP, Kantarjian HM, Jain N, Manshouri T, Thomas DA, GarciaManero G, et al. Phase 2 study of CEP-701, an orally available JAK2 inhibitor, in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. Blood 2010;115(6):1131–6. Levis M, Ravandi F, Wang ES, Baer MR, Perl A, Coutre S, et al. Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood 2011;117(12):3294–301. Brown P. Combination Chemotherapy With or Without Lestaurtinib in Treating Infants With Newly Diagnosed Acute Lymphoblastic Leukemia; 2012 http://www.clinicaltrials.gov/ ct2/show/NCT00557193?term=NCT00557193&rank=1 (accessed 02.08.12). Sterling K. Safety Study Evaluating Twice-Daily Administration of Cyt387 in Primary Myelofibrosis or Post-Polycythemia Vera or Post-Essential Thrombocythemia Myelofibrosis; 2012 http://clinicaltrials.gov/ct2/show/NCT01423058?term=CYT387 &rank=2 (accessed 18.07.12). Tefferi A. Safety and Efficacy Study of CYT387 in Primary Myelofibrosis (PMF) or Post-Polycythemia Vera (PV) or Post-Essential Thrombocythemia (ET); 2012 http://clinicaltrials.gov/ct2/show/NCT00935987?term=CYT387 &rank=1 (accessed 18.07.12). Eli Lilly and Company. A Phase 1 Study of LY2784544 in Patients With Jak2 V617F-Positive Myeloproliferative Disorders; 2012 http://www.clinicaltrials.gov/ct2/show/ NCT01134120?term=NCT01134120&rank=1 (accessed 02.08.12). Eli Lilly and Company. A Phase 2 Study of LY278544 in Patients With Myeloproliferative Neoplasms; 2012 http://www.clinicaltrials. gov/ct2/show/NCT01594723?term=NCT01594723&rank=1 (accessed 02.08.12). Verstovsek S. A Phase 1/2, Open-Label, Dose-Escalation Multi-Center Study to Assess the Safety, Tolerability, PK and PD of Orally Administered NS-018 in Patients With Primary Myelofibrosis (MF), Post-Polycythemia Vera MF, or PostEssential Thrombocythemia MF; 2012 http://www.clinicaltrials. gov/ct2/show/NCT01423851?term=NCT01423851&rank=1 (accessed 02.08.12). Jakafi. Jakafi (Ruxolitinib) Prescribing Information. Wilmington, DE: Incyte Corporation; 2011. Talpaz M, Hamburg SL, Jamieson K. Preliminary safety and efficacy of ruxolitinib in patients with primary and secondary myelofibrosis with platelet counts of 50–100(10(9) (abstract 6630). In: Paper Presented At: Annual Meeting of the American Society of Clinical Oncology. 2012. Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes-Franco J, Thomas DA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. New England Journal of Medicine 2010;363(12):1117–27. Novartis Pharmaceuticals. A Multi-National Open-Label Phase II Study of the JAK Inhibitor INC424 in Patients with Primary Myelofibrosis, Post-Polycythemia Vera Myelofibrosis or Post-Essential Thrombocythemia Myelofibrosis; 2012
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53] [54]
[55]
[56]
[57]
677
http://www.clinicaltrials.gov/ct2/show/NCT01392443?term= NCT01392443&rank=1 (accessed 02.08.12). Verstovsek S. Evaluation of Ruxolitinib and Lenalidomide Combination as a Therapy for Patients with Myelofibrosis; 2012 http://www.clinicaltrials.gov/ct2/show/NCT01375140?term= NCT01375140&rank=1 (accessed 02.08.12). Verstovsek S. A Phase I/II Study to Determine the Safety and Efficacy of Ruxolitinib, a JAK1/JAK2 Inhibitor, in Subjects with Relapsed or Refractory Acute Leukemia; 2012 http://www.clinicaltrials.gov/ct2/show/NCT01251965?term= NCT01251965&rank=1 (accessed 02.08.12). Ravandi-Kashani F. INCB018424 in Patients with Advanced Hematologic Malignancies; 2012 http://clinicaltrials.gov/ct2/show/ NCT00674479?term=ruxolitinib&rank=18 (accessed 18.07.12). Eghtedar A, Verstovsek S, Estrov Z, Burger J, Cortes J, Bivins C, et al. Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood 2012;119(20):4614–8. Vose JM. A Phase 2 Multicenter, Investigator Initiated Study of Oral Ruxolitinib Phosphate for the Treatment of Relapsed or Refractory Diffuse Large B-Cell and Peripheral T-Cell NonHodgkin Lymphoma; 2012 http://www.clinicaltrials.gov/ct2/ show/NCT01431209?term=NCT01431209&rank=1 (accessed 02.08.12). Sanofi-Aventis. Study with SAR302503 in Patients with Polycythemia Vera or Essential Thrombocythemia; 2012 http:// clinicaltrials.gov/ct2/show/NCT00079482?term=Lestaurtinib &rank=3 (accessed 18.07.12). Garcia-Manero G. Phase II Study of SB1518 for Patients with Myelodysplastic Syndrome (MDS); 2012 http://www.clinicaltrials. gov/ct2/show/NCT01436084?term=NCT01436084&rank=1 (accessed 02.08.12). Verstovsek S. A Phase 1/2 Study of SB1518 for the Treatment of Advanced Myeloid Malignancies; 2012 http://clinicaltrials.gov/ ct2/show/NCT00719836?term=NCT00719836&rank=1 (accessed 18.09.12). Deeg HJ, Odenike O, Scott BL, Estrov Z, Cortes JE, Thomas DA, et al. Phase II study of SB1518, an orally available novel JAK2 inhibitor, in patients with myelofibrosis. Journal of Clinical Oncology 2011;29(5):6515. Komrokji RS, Wadleigh M, Seymour JF, Roberts AW, To LB, Zhu HJ, et al. Results of a phase 2 study of pacritinib (SB1518), a novel oral JAK2 inhibitor, in patients with primary, post-polycythemia vera, and post-essential thrombocythemia myelofibrosis. Blood (ASH Annual Meeting Abstracts) 2011;118(21):282. Younes A. A Phase 1 Study of SB1518 for the Treatment of Advanced Lymphoid Malignancies; 2012 http://www.clinicaltrials.gov/ ct2/show/NCT00741871?term=NCT00741871&rank=1 (accessed 02.08.12). Younes A. A Phase 2 Safety and Efficacy Study of SB1518 for the Treatment of Advanced Lymphoid Malignancies; 2012. Isis Pharmaceuticals. Phase I, Open-Label, Dose-Escalation Study of ISIS-STAT3Rx, Administered to Patients with Advanced Cancers; 2012 http://www.clinicalconnection.com/exp/EPVS.aspx? studyID=322467&slID=5291032 (accessed 18.07.12). Otsuka Pharmaceutical. A Dose-Escalation Trial to Investigate the Safety and Tolerability of OPB-51602 in Patients with Relapsed or Refractory Hematologic Malignancies (Phase 1); 2012 http://www.clinicaltrials.gov/ct2/show/NCT01344876?term= NCT01344876&rank=1 (accessed 02.08.12). Brown JR. Pyrimethamine for the Treatment of Relapsed Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma; 2012 http://clinicaltrials.gov/ct2/show/NCT01066663 (accessed 18.07.12). Bristol-Myers-Squibb. Multiple Ascending Dose (MAD) Combination in Subjects with Multiple Myeloma; 2012
678
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
[71]
[72]
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679 http://clinicaltrials.gov/ct2/show/NCT00884546?term=BMS833923+AND+myeloma&rank=1 (accessed 18.07.12). Bristol-Myers-Squibb. Dasatinib (BMS-354825) Combined with SMO Inhibitor (BMS-833923; XL139) in CML with Resistance or Suboptimal Response to a Prior TKI; 2012 http://www.clinicaltrials.gov/ct2/show/NCT01218477? term=NCT01218477&rank=1 (accessed 02.08.12). Huff CA. GDC-0449 in Treating Patients with High-risk First Remission or Relapsed Multiple Myeloma Who Received An Autologous Stem Cell Transplant; 2012 http://clinicaltrials.gov/ ct2/show/NCT01330173 (accessed 18.07.12). Verstovsek S. A Phase 2 Study With IPI-926 in Patients With Myelofibrosis; 2012 http://clinicaltrials.gov/ct2/show/ NCT01371617?term=NCT+01371617&rank=1 (accessed 18.09.12). Novartis Pharmaceuticals. A Single-Arm Dose-Finding Phase IB Multicenter Study of the Oral Smoothened Antagonist LDE225 in Combination With Nilotinib in Chronic Phase Chronic Myeloid Leukemia Patients Who Have Failed Prior Therapy With Other BCR-ABL TyrosineKinase Inhibitors; 2012 http://www.clinicaltrials.gov/ct2/show/ NCT01456676?term=NCT01456676&rank=1 (accessed 02.08.12). Pfizer Inc. A Study of PF-04449913 in Select Hematologic Malignancies; 2012 http://clinicaltrials.gov/ct2/show/NCT00953758 (accessed 18.07.12). Pfizer Inc. A Phase 1B Study to Evaluate the Safety and Preliminary Efficacy of PF-04449913, An Oral Hedgehog Inhibitor, in Combination With Intensive Chemotherapy, Low Dose Ara-C or Decitabine in Patients With Acute Myeloid Leukemia or High Risk Myelodysplastic Syndrome; 2012 http://www.clinicaltrials.gov/ct2/show/ NCT01546038?term=NCT01546038&rank=1 (accessed 02.08.12). de Haan H. Phase I Clinical Study of CWP232291 in Acute Myeloid Leukemia Patients; 2012 http://clinicaltrials.gov/ct2/ show/NCT01398462 (accessed 18.07.12). Cortes J. An Open-Label, Dose-Escalation Phase I/II Study of PRI-724 for Patients with Advanced Myeloid 2012 http://www.clinicaltrials.gov/ct2/show/ Malignancies; NCT01606579?term=NCT01606579&rank=1 (accessed 02.08.12). Hege K. Phase 1/2, Multi-Center, Open-Label, Dose Finding Study to Assess the Safety, Tolerability, Pharmacokinetics and Preliminary Efficacy of the mTOR Knase Inhibitor CC-223 Administered Orally to Subjects with Advanced Solid Tumors, Non-Hodgkin Lymphoma or Multiple Myeloma; 2012 http://www.clinicaltrials.gov/ ct2/show/NCT01177397?term=NCT01177397&rank=1 (accessed 02.08.12). Astellas Pharma Inc. A Phase 1 Dose-escalation Study of Intermittent, Once Weekly, and Continuous Daily Oral OSI-027 Dosing in Patients with Advanced Solid Tumors or Lymphoma; 2012. 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. Leukemia Research 2012;36(6): 756–63. Nelson EA, Walker SR, Weisberg E, Bar-Natan M, Barrett R, Gashin LB, et al. The STAT5 inhibitor pimozide decreases survival of chronic myelogenous leukemia cells resistant to kinase inhibitors. Blood 2011;117(12):3421–9. Nam S, Scuto A, Yang F, Chen W, Park S, Yoo HS, et al. Indirubin derivatives induce apoptosis of chronic myelogenous leukemia cells involving inhibition of Stat5 signaling. Molecular Oncology 2012;6(3):276–83. Mar BG, Amakye D, Aifantis I, Buonamici S. The controversial role of the Hedgehog pathway in normal and malignant hematopoiesis. Leukemia 2011;25(11):1665–73. Gupta S, Takebe N, Lorusso P. Targeting the Hedgehog pathway in cancer. Therapeutic Advances in Medical Oncology 2010;2(4):237–50.
[73] Dierks C, Beigi R, Guo GR, Zirlik K, Stegert MR, Manley P, et al. Expansion of BCR-ABL-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell 2008;14(3):238–49. [74] Zhao C, Chen A, Jamieson CH, Fereshteh M, Abrahamsson A, Blum J, et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 2009;458(7239):776–9. [75] Lie DC, Colamarino SA, Song HJ, Desire L, Mira H, Consiglio A, et al. Wnt signalling regulates adult hippocampal neurogenesis. Nature 2005;437(7063):1370–5. [76] Trowbridge JJ, Moon RT, Bhatia M. Hematopoietic stem cell biology: too much of a Wnt thing. Nature Immunology 2006;7(10):1021–3. [77] Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, Zehnder JL, et al. Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. New England Journal of Medicine 2004;351(7):657–67. [78] Heidel FH, Bullinger L, Feng Z, Wang Z, Neff TA, Stein L, et al. Genetic and pharmacologic inhibition of beta-catenin targets imatinib-resistant leukemia stem cells in CML. Cell Stem Cell 2012;10(4):412–24. [79] Gregory MA, Phang TL, Neviani P, Alvarez-Calderon F, Eide CA, O’Hare T, et al. Wnt/Ca2+/NFAT signaling maintains survival of Ph+ leukemia cells upon inhibition of BCR-ABL. Cancer Cell 2010;18(1):74–87. [80] Arroyo JD, Hahn WC. Involvement of PP2A in viral and cellular transformation. Oncogene 2005;24(52):7746–55. [81] Switzer CH, Cheng RY, Vitek TM, Christensen DJ, Wink DA, Vitek MP. Targeting SET/I(2)PP2A oncoprotein functions as a multipathway strategy for cancer therapy. Oncogene 2011;30(22):2504–13. [82] Perrotti D, Jamieson C, Goldman J, Skorski T. Chronic myeloid leukemia: mechanisms of blastic transformation. Journal of Clinical Investigation 2010;120(7):2254–64. [83] Neviani P, Santhanam R, Trotta R, Notari M, Blaser BW, Liu S, 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;8(5):355–68. [84] Teachey DT, Grupp SA, Brown VI. Mammalian target of rapamycin inhibitors and their potential role in therapy in leukaemia and other haematological malignancies. British Journal of Haematology 2009;145(5):569–80. [85] Foster KG, Fingar DC. Mammalian target of rapamycin (mTOR): conducting the cellular signaling symphony. Journal of Biological Chemistry 2010;285(19):14071–7. [86] Carayol N, Vakana E, Sassano A, Kaur S, Goussetis DJ, Glaser H, et al. Critical roles for mTORC2- and rapamycin-insensitive mTORC1complexes in growth and survival of BCR-ABL-expressing leukemic cells. Proceedings of the National Academy of Sciences of the United States of America 2010;107(28):12469–74. [87] Yee KW, Zeng Z, Konopleva M, Verstovsek S, Ravandi F, Ferrajoli A, et al. Phase I/II study of the mammalian target of rapamycin inhibitor everolimus (RAD001) in patients with relapsed or refractory hematologic malignancies. Clinical Cancer Research 2006;12(17):5165–73. [88] Janes MR, Limon JJ, So L, Chen J, Lim RJ, Chavez MA, et al. Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor. Nature Medicine 2010;16(2):205–13. [89] Rousselot P, Huguet F, Rea D, Legros L, Cayuela JM, Maarek O, et al. Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years. Blood 2007;109(1):58–60. [90] Rousselot P, Makhoul PC, Rea D, Agape P, Nicolini FE, Legros L, et al. Fluctuating values of molecular residual disease (MRD) without molecular progression after imatinib discontinuation in patients (pts) with chronic myeloid leukemia (CML) who have maintained complete molecular response: implications for re-treatment criteria and role of prior interferon therapy. A pilot study of the French CML Group (FILMC). Blood (ASH Annual Meeting Abstracts) 2011;118(21):3781.
K.L. Sweet et al. / Critical Reviews in Oncology/Hematology 88 (2013) 667–679 [91] Essers MA, Offner S, Blanco-Bose WE, Waibler Z, Kalinke U, Duchosal MA, et al. IFNalpha activates dormant haematopoietic stem cells in vivo. Nature 2009;458(7240):904–8. [92] Hehlmann R, Lauseker M, Jung-Munkwitz S, Leitner A, Muller MC, Pletsch N, et al. Tolerability-adapted imatinib 800 mg/d versus 400 mg/d versus 400 mg/d plus interferon-alpha in newly diagnosed chronic myeloid leukemia. Journal of Clinical Oncology 2011;29(12):1634–42. [93] Palandri F, Castagnetti F, Iacobucci I, Martinelli G, Amabile M, Gugliotta G, et al. The response to imatinib and interferon-alpha is more rapid than the response to imatinib alone: a retrospective analysis of 495 Philadelphia-positive chronic myeloid leukemia patients in early chronic phase. Haematologica 2010;95(8):1415–9. [94] Preudhomme C, Guilhot J, Nicolini FE, Guerci-Bresler A, RigalHuguet F, Maloisel F, et al. Imatinib plus peginterferon alfa-2a in chronic myeloid leukemia. New England Journal of Medicine 2010;363(26):2511–21. [95] Simonsson B, Gedde-Dahl T, Markevarn B, Remes K, Stentoft J, Almqvist A, et al. Combination of pegylated IFN-alpha2b with imatinib increases molecular response rates in patients with low- or intermediate-risk chronic myeloid leukemia. Blood 2011;118(12):3228–35. [96] Burgess A, Ruefli A, Beamish H, Warrener R, Saunders N, Johnstone R, et al. Histone deacetylase inhibitors specifically kill nonproliferating tumour cells. Oncogene 2004;23(40):6693–701. [97] Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nature Reviews Cancer 2006;6(1):38–51. [98] Zhang B, Strauss AC, Chu S, Li M, Ho Y, Shiang KD, et al. 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. [99] Chen Y, Li D, Li S. The Alox5 gene is a novel therapeutic target in cancer stem cells of chronic myeloid leukemia. Cell Cycle 2009;8(21):3488–92. [100] Den RB, Lu B. Heat shock protein 90 inhibition: rationale and clinical potential. Therapeutic Advances in Medical Oncology 2012;4(4):211–8.
Biographies Kendra L. Sweet, MD: Dr. Kendra Sweet is Clinical Instructor in Hematology and Medical Oncology at the H. Lee Moffitt Cancer Center and the University of South Florida in Tampa, Florida. Dr. Sweet is currently involved in a clinical
679
trial looking at combination therapy in CML and has done research on prognostic factors and outcomes in CML. She has presented her findings at annual meetings of the American Society of Clinical Oncology, the American Society of Hematology, and the European School of Hematology. Lori A. Hazlehurst, PhD: Dr. Hazlehurst is an associate member of the Molecular Oncology program and a member of the Malignant Hematology program at the H. Lee Moffitt Cancer Center in Tampa, Florida. Dr. Hazlehurst’s research is focused on the identification and targeting of mechanisms of drug resistance emanating from the bone marrow microenvironment. Her work has been published in journals including Blood, Cancer Research, Molecular Cancer Therapeutics, and Leukemia Research. Javier Pinilla-Ibarz, MD, PhD: Dr. Javier Pinilla-Ibarz is an associate member of the Malignant Hematology and Immunology program at the H. Lee Moffitt Cancer Center in Tampa, Florida, and is an Associate Professor in the Department of Oncologic Sciences, College of Medicine, University of South Florida, Tampa, Florida. Dr. Pinilla-Ibarz is leading the chronic leukemia program at Moffitt. The program’s principal clinical interest is the treatment of patients with myeloid and lymphoid chronic leukemia. In this capacity, Dr. Pinilla-Ibarz has participated in multiple clinical trials involving tyrosine kinase inhibitors (TKIs). Dr. Pinilla-Ibarz is also the Director of Immunotherapy for Malignant Hematology. He has been involved in developing new immunotherapeutic approaches to the treatment of chronic myeloid leukemia. He is currently leading several clinical trials of peptide and cellular vaccines in different hematologic disorders. Other research areas include the role of the bone marrow microenvironment in mediating resistance to TKIs and the interaction of TKIs with the immune system. Dr. Pinilla-Ibarz frequently presents his findings at annual meetings of the American Hematology Association and the American Society of Clinical Oncology, and other national/international meetings. His work has been published in journals including Blood, Nature Immunology, Seminars of Hematology, and Leukemia. He has also authored multiple book chapters.