Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jan 5.
Published in final edited form as: Expert Rev Hematol. 2014 Aug;7(4):439–464. doi: 10.1586/17474086.2014.932687

Small Molecule Inhibitors in Acute Myeloid Leukemia: From the Bench to the Clinic

Muneera Al-Hussaini 1, John F DiPersio 1,2
PMCID: PMC4283573  NIHMSID: NIHMS651420  PMID: 25025370

Abstract

Many patients with acute myeloid leukemia (AML) will eventually develop refractory or relapsed disease. In the absence of standard therapy for this population, there is currently an urgent unmet need for novel therapeutic agents. Targeted therapy with small molecule inhibitors (SMIs) represents a new therapeutic intervention that has been successful for the treatment of multiple tumors (e.g., gastrointestinal stromal tumors, chronic myelogenous leukemia). Hence, there has been great interest in generating selective small molecule inhibitors targeting critical pathways of proliferation and survival in AML. This review highlights a selective group of intriguing therapeutic agents and their presumed targets in both preclinical models and in early human clinical trials.

Keywords: acute myeloid leukemia, small molecule inhibitors, therapeutic agents

Introduction

Anthracyclin and cytosine arabinoside-based chemotherapy achieves complete remission (CR) in the majority of patients with acute myeloid leukemia (AML).1 Despite this fact, approximately 50% of patients will relapse within 1 to 2 years. The 5-year survival rate for patients who are less than 60 years old remains less than 50%.13 In a recent analysis by Wingard et al, residual and recurrent disease was the leading causes of death (43%–47%) in the first nine years following allogeneic bone marrow transplantation.4 The natural course of AML in patients who are greater than 60 years old is dismal with complete remission rates (CRR) of 40% to 65%, relapse rates of 60%–85% within 2–3 years of diagnosis, median overall survival (OS) of less than 6 months, and a 5-year overall survival (OS) of only 3%–8%.2,3 The poor prognosis for this older population is thought to arise from a higher rate of drug resistance, co-morbidities, poor tolerance to chemotherapy, overexpression of the multidrug resistance genes (MRD1 and other ATP Binding Cassette/ABC genes), unfavorable cytogenetics, and a high treatment-related mortality rate (≥25%).2,59 Accumulating data relating to the biology and initiating events of cancer have resulted in the identification of prognostic markers in AML and the development of novel targeted therapies in the hope of discovering a more efficient and less toxic alternative to conventional chemotherapy. The two small molecular inhibitors (SMIs), imatinib mesylate, a tyrosine kinase inhibitor that represses the function of BCR-ABL kinase (as well as other related tyrosine kinases), which has resulted in an 80% complete cytogenetic response rate in patients with chronic phase CML, and all-trans retinoic acid (ATRA), which can induce compete responses in patients with the APL (acute promyelocytic leukemia; M3 AML), have spawned great interest in the development of SMIs for the treatment of AML.1012 However, unlike in CML and APL, the identification of potential targets in AML has been limited by the heterogeneous clonal architecture of non-M3 AML and by the contribution of numerous driver mutations in its onset and progression. In this article, we will review SMIs for a number of biologically relevant targets in AML that are currently in clinical development, with a refrence to the ongoing clinical trials (Table.1), and the possible mechanisms of action and resistance to these reagents in AML.

Table.1.

Selected Ongoing Clinical Trials of Small Molecular Inhibitors in Patients with Acute Myeloid Leukemia

Molecular Target Small Molecule
Inhibitor
Phase of the
study
Study Design ClinicalTrials.gov
Identifier
Aurora kinase Alisertib
(Aurora A Kinase inhibitor)
Phase 1 alisertib in combination with 7+3 induction chemotherapy NCT01779843
AMG 900 Phase 1 Single agent NCT01380756

Farnesyl transferase Lestaurtinib Phase 1/ 2 lestaurtinib + cytarabine + idarubicin relapsed and refractory AML NCT00469859

Histone deacetylase Valproic acid Phase 1/ 2 ATRA+ Valproic acid+ low dose cytarabine NCT00995332
Entinostat
(MS275)
Phase 2 Entinostat + 5AZC in elderly NCT01305499
Vorinostat Phase 2 Temozolomide + vorinostat in patients more than 60 years old with newly diagnosed or relapsed or refractory AML NCT01550224
Vorinostat Phase 3 Cytarabine + daunorubicin hydrochloride or idarubicin+ cytarabine with or without vorinostat NCT01802333

Proteasome inhibitors Bortezomib Phase 2 Comparing decitabine with decitabine + subcutaneously injected bortezomib in untreated AML NCT01420926
Ixazomib Phase 2 Single agent for NPM1c relapsed/ refractory AML NCT02030405

CXCR4 Plerixafor Phase 1 Plerixafor+ decitabine for induction and post-remission therapy in elderly NCT01352650
Plerixafor Phase 1/2 Plerixafor+ GCSF + chemotherapy in refractory or relapsed AML NCT00906945
BL-8040 Phase 2A, multicenter, open-label Single agent therapy for relapsed and refractory AML NCT01838395
BMS-936564 (Anti-CXCR4) Phase 1, multicenter, open-label Single agent in relapsed AML, and B cell malignancies NCT01120457

HSP90 Ganetespib Phase 1/2 To combine either the tyrosine kinase inhibitor AC220, plerixafor, or ganetespib with chemotherapy in older patients with AML and high-risk myelodysplastic syndrome (AML18 Pilot) NCT01236144

FLT3 Crenolanib Phase 2 Single agent therapy in relapsed and refractory AML with FLT3- D835 activating mutation NCT01522469
Sorafenib Phase 1/2 Sorafenib + vorinostat, + bortezomib for AML with complex or poor-risk (monosomy 5/7) cytogenetics or FLT3-ITD–positive mutation NCT01534260
Sorafenib Phase 1 Single agent maintenance therapy after allogeneic stem cell transplantation. NCT01398501
Sorafenib Phase 3 Bortezomib+ sorafenib for de novo AML with high allelic ratio FLT3-ITD disease NCT01371981
Midostaurin
(PKC412)
Phase 2 Single agent therapy for c-kit or FLT3-ITD mutated t(8;21) AML NCT01830361

c-kit Nilotinib Phase 1/2 Nilotinib+ mitoxantrone, etoposide +high-dose cytarabine (NOVE-HiDAC) induction chemotherapy followed by consolidation therapy for poor-risk patients up to 65 years old with c-kit positive AML NCT01222143

Hedgehog pathway PF-04449913 Phase 2 Single agent therapy for high risk AML post-allogeneic stem cell transplantation relapse NCT01841333

WNT pathway PRI-724 Phase 1/2 Single agent tharpy for advanced malignancies. NCT01606579
CWP232291 Phase 1 Single agent therapy for relapsed and refractory AML, chronic myelomonocytic leukemia, myelodysplastic syndrome or high-risk myelofibrosis NCT01398462

IDH1/2 AG-120 Phase 1 Monotherapy in advanced hematological malignancies with IDH1 mutation NCT02074839
AG-221 Phase 1 Monotherapy in advanced hematological malignancies with IDH2 mutation NCT01915498

AML, Acute myeloid leukemia; 5AZC, 5-Azacytidine.

Nucleophosmin (NPM1)

NPM1, which encodes a nucleolar phosphoprotein, is mapped to the long arm of chromosome 5. Three isoforms of NPM1 are generated by alternative splicing. It has been implicated in genomic stability and cell cycle progression by acting as a histone chaperone and a nucleus-cytoplasmic shuttle. It participates in chromatin remodeling, ribosomal biogenesis, centrosome duplication, ribosomal RNA cleavage, DNA synthesis, RNA transcription, and DNA repair.13,14 Accumulation of NPM1 protein has been observed in cancerous cells, likely reflecting increased DNA replication.15,16 Approximately, 35% of AML patients harbor NPM1 mutations, most of which are structurally defined by an insertion in exon 12 with the duplication of a TCTG sequence at positions 956–959, leading to changes in the amino acid sequence of the C-terminal domain and loss of trp288 and trp290, thereby resulting in unfolding of the C-terminal region in the NPM1 protein and reduced nucleolar binding. A new nuclear export signal motif is also formed that increases NPM1-CRM1 heterodimerization and export to the cytoplasm.1719 NPM1 haploinsufficiency predisposes mice to tumor formation.20 This is thought to be related to the cytoplasmic dislocation of p19ARF (p14ARF ortholog) thus inhibiting its tumor suppressor effect by allowing mouse double minute 2 homolog (Mdm2) binding and inactivation of TP53, or by eliciting the post-translational sumoyl modification of the NPM1 protein in a TP53-independent mechanism.2123 Of note, the nuclear factor-κβ (NF-κβ) has an important role in the promotion of metastasis, angiogenesis, and the survival of cancer cells, and it is hyperactivated in the majority of AML patients.2426 The favorable outcome of NPM1-mutated AML has been attributed to NF-κβ binding to the mutated form of NPM1 (NPM1c) resulting in cytoplasmic sequestration and inactivation of NF-κβ, leading directly and indirectly to leukemic cell chemosensitization.17,27,28

Different strategies of NPM1c targeting have been proposed. Conceptually, transporting NPM1c from the cytoplasm back to the nucleus is an interesting approach, but it remains challenging. Leptomycin B is an Exportin-1/CRM1 inhibitor that exhibits in vitro tumoricidal activity by stabilizing TP53 through disrupting its CRM1-mediated nuclear export.2931 However, a phase 1 trial with Leptomycin B was discouraging, without objective responses and with significant toxicity manifesting as marked fatigue and anorexia.32 New, less toxic CRM1 inhibitors, such as CBS9106 and KPT 330, have been developed.3337 Additive effects of CRM1 inhibitors with cytarabine, FLT3 inhibitors, and histone deacetylase inhibitors in AML have been reported in recent preclinical studies.38,39

Inhibiting the interaction of NPM1 with other proteins has also been investigated. Cytoplasmic relocalization of HEXIM1 can be mediated by NPM1c, resulting in HEXIM1 inactivation and stimulation of the positive transcription elongation factor (P-TEFb), a cyclin-dependent kinase that regulates mRNA synthesis. A P-TEFb inhibitor, seliciclib was evaluated in a phase 1 trial by Bensen et al.40,41 Seliciclib was given for 7 days on a 3-week cycle, but tumor responses were not observed. Disease stabilization was observed in 8 out of 21 evaluable patients for up to 18 weeks, with dose-limiting toxicities (DLTs) of fatigue, hypokalemia, and urticarial rash. The investigators linked the lack of clinical responses to the significantly lower plasma concentrations in the study participants compared to the levels achieving tumor regression in human xenograft models.41 Given the heterozygosity of NPM1 mutations, it is conceivable that leukemic cells possess low nuclear levels of wild type NPM1, contributing to the cell growth. Wild type NPM1 levels are also expected to be lower in heterozygous mutant cells compared to normal cells because of dimerization with the NPM1c.42 Therefore, targeting the wild type NPM1 might also be an effective therapeutic approach via indirectly inhibiting the NPM1c-mutant and/or signaling pathways.23.13

MDM2

TP53 is a tumor suppressor that responds to stress signals and regulates cell cycle arrest, senescence, and apoptosis to maintain genomic stability.43 TP53 mutations are found in ~50% of tumors, leading to partial or complete loss of the TP53 function and consequently, to oncogenic transformation.4446 TP53 levels and activity are also downregulated by the oncoprotein MDM2 in an autoregulatory circuit, which was proposed as an alternative mechanism of TP53 inactivation in AML rather than TP53 mutation.47,48 Binding of the N-terminal domain of MDM2 with the N-terminus of the TP53 transactivation domain results in TP53 suppression, nuclear export, and ubiquitination followed by proteasomal degradation.49,50 This interaction explains the correlation of MDM2 overexpression with chemoresistance and poor outcomes in many tumors and provides a basis for the development of SMI against MDM2 in an attempt to enhance TP53 activity.5155 Nutlin-3 is an MDM2 antagonist that binds the TP53 pocket of MDM2. It increases TP53 levels and induces TP53-dependent apoptosis of AML blasts in vitro and an additive/synergistic anti-AML effect when combined with chemotherapy such as doxorubicin and cytosine arabinoside.56 Furthermore, Nutlin-3 induces a TP53-independent apoptotic effect by interfering with MDM2’s interaction with other proteins (e.g., p73, E2F-1, HIF-1α) that share the binding site of TP53.5759 Anti-angiogenic properties of Nutlin-3 were also observed in preclinical studies and were attributed to TP53 activation in endothelial cells, leading to the induction of anti-angiogenic proteins, such as TSP-1 and BAL1 and to downregulation of pro-angiogenic proteins such as VEGF, bFGF, FGF-BP, and COX-2.60,61 The initial results of a phase 1 trial conducted by Andreeff et al in patients with AML using RG7112, a new oral MDM2 antagonist, were promising. Of 31 evaluable patients, CR was reported in 5 after 1 cycle, with signs of hematological improvement in 12 other patients.62 RG7112 was further studied by Yee and colleagues in combination with cytarabine using 2 schedules. The DLT were diarrhea, rash and thrombocytopenia. The ORR was 52%. Of interest, CR was seen in an elderly patient with disease refractory to previous cytarabine therapy and that all patients with CR had TP53 wild type status. Bridging to transplant was also possible in 3 patients with relapsed/refractory disease.63

An analysis of 109 AML specimens after in vitro exposure to the MDM2 inhibitor MI-219 revealed resistance in all samples harboring TP53 mutations and in 30% of those with wild type TP53, with an enhanced response in association with high MDM2 expression.64 Drug resistance despite the presence of a wild type TP53 phenotype was assumed to be related to a post-translational modification of TP53 that reduces TP53 activity or to impaired TP53 pathways. Of interest, 84% of the FLT3-mutant (ITD) responded well to MI-219 (IC50 < 5 µM), suggesting a therapeutic potential of MDM2 inhibitors in FLT3-mutated AML. Various compounds that block MDM2–TP53 binding are currently being studied in clinical trials in both hematological and solid tumors. Initial studies, although limited, suggest reasonable safety and efficacy.65,66

Aurora kinases

Aurora kinases A, B, and C (AKA, AKB, and AKC) are serine/threonine kinases that regulate mitosis and cytokinesis and have been implicated in both tumor formation and progression.6771 The AKA gene maps to chromosome 20q13.2.72 In RNA interference studies, AKA inhibition disrupts spindle assembly, centrosome maturation and segregation, thereby leading to G2/M arrest and cell death.73,74 AKA is also a negative regulator of TP53 activity: the phosphorylation of TP53 at Ser315 by AKA induces MDM2-TP53 interaction, thereby causing TP53 inactivation.75,76 Thus, AKA inhibition increases TP53 levels and activity resulting in G2/M arrest followed by apoptosis. In addition, AKA overexpression in cancer has been linked to chromosomal instability and shortened survival.7780 Of note, CD34+/CD38 leukemic stem cells exhibit higher expression of AKA as compared with normal CD34+ progenitors, with a selective and potent anti-proliferative and apoptotic response to AKA inhibition.81

AKB is a chromosomal passenger protein encoded on chromosome 17p13.1.69 Inhibition of AKB’s function is associated with cytokinesis defects, polyploidy, and apoptosis.82 Its upregulation in patients with non–small-cell lung cancer, ovarian cancer, oral and laryngeal squamous cell carcinoma, and hepatocellular carcinoma correlates with the increased likelihood of metastasis, chemoresistance and poor survival.8387 Recent data suggests a supportive role of AKC to AKB function and that AKC overexpression can promote malignant transformation.88,89 Preclinical data support the antitumor activity of several of the aurora kinase inhibitors (AKIs). A phase 1 trial of patients with advanced hematological malignancies treated with AT9283, a small molecule inhibitor of AKA, AKB, JAK2/3, and ABL1, resulted in only a transient blast reduction of 30% in patients with AML despite reduced levels of the phosphorylated histone H3, a downstream target of AKB. Significant DLTs were reported, including pneumonia, myocardial infarction, and tumor lysis syndrome.90 Of note, cardiac toxicity manifesting as QTc prolongation was also reported with another AKI (MK-0457) and was attributed to hERG channel suppression.91

In contrast to patients with solid tumors, AML patients demonstrated a better response to the selective AKB inhibitor barasertib (AZD1152).92,93 Barasertib was given as a 7-day infusion on a 21-day cycle, with an overall hematological response of 25%. The majority of the patients enrolled in this study had relapsed AML. Recently, a randomized phase 2 study from MD Anderson Cancer Center reported higher CRR and CRp (CR with incomplete platelet recovery) in older patients with AML in response to a 7-day infusion of barasertib at a dose of 1200 mg/day as compared with those receiving a low dose of cytarabine of 20 mg/day subcutaneously for 10 days every 28 days (35% vs. 12%). The median duration of remission ranged from 28 to 321 days in the barasertib group and from 30 to 85 days in the cytarabine group.94 Responses were observed mostly after two cycles and included patients from all cytogenetic risk subsets in the barasertib-only arm while being limited to patients with good and intermediate risk cytogenetic profiles in the cytarabine arm. Treatment-related mortality was comparable between the two cohorts 30 days after the start of treatment, but it was lower in the barasertib arm at 90 days. However, this study was hampered by the small number and uneven distribution of patients in terms of age and performance status. Interestingly, some reports with AT9283 suggested TP53 mutation as a predictor of sensitivity.95 Alternatively, increased pHH3 marks a tumor response to the selective AKA inhibitor MLN8054.96 Because AKA stimulates the mTOR/Akt pathway and is implicated in taxane and carboplatin resistance, AKIs have been combined with taxane and carboplatin to overcome drug resistance, with encouraging results.9799 One interpretation of the limited clinical response to AKIs is the fact that they target the mitotic process, which might limit their activity to the proliferating cells.70 Several AKIs are in early development with promising results (e.g., TLK60404, CHR-3520 and Aki-100).70

Farnesyl transferase

RAS proteins regulate intracellular signal transduction and several critical pathways involved in cell growth, adhesion, migration, invasion, and apoptosis.100 Approximately 12% to 27% of patients with AML harbor activating mutations in the RAS proto-oncogene family predominantly at codon 12 of N-RAS and K-RAS.101108 RAS upregulation without an activating mutation has also been reported with AML.109 One mechanism of RAS inhibition is via the inhibition of farnesyl transferase, an essential enzyme for the post-translational modification of RAS. Farnesyl transferase adds a farnesyl moiety to the cysteine residue of the C-terminal CAAX motif, thereby allowing RAS to anchor itself to the inner leaflet of the plasma membrane. Data support the ability of farnesyl transferase inhibitors to induce apoptosis in multiple cancer cell lines (including AML lines), most likely by inhibiting the activity of the pro-survival proteins (e.g., AKT, mTOR, MAP kinases) and by activating pro-apoptotic proteins (e.g., Bax, PUMA, BAK).110116

A phase 1 trial of a farnesyl transferase inhibitor, lonafarnib, demonstrated limited clinical activity in patients with myelodysplastic syndrome (MDS) and secondary AML. Excessive toxicity was noted in the majority of participants, many of whom required dose reductions after the first treatment cycle or cessation of therapy.117 Tipifarnib is another farnesyl transferase inhibitor with a more favorable toxicity profile. The initial results of the CEP-20 trial demonstrated that tipifarnib induced CRR/CRp in 14% of poor-risk older AML patients (>70 years old) with a median survival of 18 months in responders irrespective of RAS mutational status. The dose used in this study was 600 mg twice a day for 3 weeks during every 4-week cycle.118 Of note, a larger trial comparing this regimen with best supportive care failed to replicate these results (CRR was only 8%, with no difference in survival).119 The AML16 trial that combined tipifarnib with low-dose cytarabine also failed to demonstrate significant differences in CRR and 12-month OS rates, leading to the premature closure of the study.120 Jabbour and colleagues reported similar negative results in a phase 1/2 trial of the addition of tipifarnib to idarubicin and cytarabine in patients with AML and poor-risk patients with myelodysplastic syndrome (MDS).121 However, subanalysis revealed a higher CRR among patients with unfavorable cytogenetic abnormalities involving chromosomes 5 and 7 as compared with historical controls receiving chemotherapy alone (58% vs. 19%). This observation was consistent with similar analysis performed for another study by Brenwin and colleagues, which suggested heightened sensitivity to tipifarnib-based therapy in the high-risk population.122

Geranylgeranylation of K-RAS and N-RAS is proposed as another way to circumvent farnesyl transferase inhibition and to restore RAS activation.123,124 In support of this is the fact that the mutation of H-RAS, which does not undergo geranylgeranylation, renders tumors more sensitive to farnesyl transferase inhibitors as compared with those tumors carrying N-RAS and K-RAS mutations.125,126 Dual targeting of the farnesyl and geranylgeranyl transferases effectively produced an anti-tumor response in vivo, but a high level of toxicity was initially encountered.127,128 L-778,123 is a dual inhibitor that was investigated in patients after being well tolerated in vivo.129 Despite successful inhibition of geranylgeranylation, L-778,123 did not suppress K-Ras prenylation in patient samples. Appels et al evaluated AZD3409, another dual inhibitor, in patients with solid tumor malignancies (n=29).130 The MTD was 750 mg twice a day, with DLTs of diarrhea and nausea. There were no objective responses. Interestingly, the pharmakodynamic data indicated two important points: 1) the maximal inhibition of farnesyl transferase activity was only 49% of pretreatment levels; 2) the maximal farnesyl transferase inhibition occurred at plasma levels considerably lower than those associated with drug-induced toxicity.

Histone deacetylase

Histones are alkaline proteins that associate with DNA to form small packaging units called nucleosomes. Histone deacetylases (HDACs) are involved in the post-translational modification of histones via removing ε-N-acetyl lysine residue, resulting in increased coiled DNA winding around the histone core, thereby reducing the accessibility of transcription factors and rendering functional genes inactive. Non-histone substrates to HDACs have also been identified, including TP53, heat shock protein 90 (HSP90), and PTEN.131133 The theoretical role of HDAC inhibitors (HDACIs) as anticancer agents is thought to be through the re-expression of epigenetically silenced tumor genes and tumor suppressor genes resulting in tumor regression. Trials of HDACIs in cutaneous T-cell lymphoma led to the approval of two HDACIs (vorinostat and romidepsin).134,135

The HDAC inhibitory activity of valproic acid (VPA) was tested in a phase 2 study of patients with MDS (n = 43) and AML (n = 32), which resulted in overall response rate (ORR) of 30% and 16%, respectively. The addition of ATRA did not increase VPA responses.136 Enhanced responses (52%) were noted in the MDS subgroup of patients with normal blast counts. By contrast, further studies with VPA, both as a single agent and with ATRA in high-risk patients with MDS and AML, demonstrated lower response, thereby suggesting that VPA may be more effective for low-risk patients with MDS.137,138 Treatment of 11 AML patients (70 years or older), unfit for chemotherapy with VPA, ATRA, and theophylline resulted in one CR, 2 CRi, and stable disease for 4–6 months in 3 patients.139 This combination was also evaluated in 20 AML patients with one CR, 2 PR and eradication of peripheral blood blasts (but not the bone marrow) in one patient.140 Fredly et al studied the addition of either hydroxyurea, 6-mercaptopurine or both to a combination of ATRA, VPA and theophylline in five patients with advanced AML. This was found to be an effective palliative strategy in reducing blast counts.141 Another study was conducted by Fredly et al in 36 AML patients unfit for intensive chemotherapy. The patients were treated with continuous VPA, and low-dose cytarabine (10mg/m2 for 10 days), and intermittent ATRA (for 14 days) on every 12-week cycle. Either hydroxyurea or 6-mercaptopurine was used to control non-responding hyperleukocytosis to low-dose cytarabine. Nine patients demonstrated hematological improvement, with a CRR of 6%, which is lower than the CRR of another study (18%) that used a higher dose of cytarabine (20mg/m2 twice daily for 10 days every 4–6 weeks).142,143 On the other hand, the CRR of Fredly et al study is similar to another trial (7.4%) that used cytarabine alone at 20 mg/m2 once a day for 10 days on monthly basis.144,145

Depsipeptide is another HDACI that demonstrated preclinical anti-leukemic activity with evidence of c-Myc downregulation. However, its clinical development was halted as a result of limited clinical responses and significant side effects, including profound fatigue and gastrointestinal toxicity.146 MGCD0103 (mocetinostat) is an HDACI with a prolonged half-life and selectivity to the HDAC isotypes 1, 2, 3, and 11. MGCD0103 was administered three times a week (20 to 80 mg/m2) to patients with refractory or relapsed AML (n = 22), CML (n = 1), ALL (n = 1), and MDS (n = 5). The MTD was 60 mg/m2. Three of the 23 evaluable patients achieved a CR after one to three cycles at higher dose levels of 60 to 80mg/m2. Two of these patients had refractory AML.147

Garcia-Manero and colleagues studied vorinostat in patients with hematological malignancies (31 of 41 had AML). The drug was given two to three times daily according to a 2-weeks-on/1-week-off schedule.137 The ORR was 17% (two patients had CR, two had CRp, and three had a >50% reduction in blasts), which continued for up to 53 weeks. All responders had AML, and bone marrow transplantation was successfully performed after treatment response in three patients. DNA hypermethylation and histone deacetylation could mutually reinforce epigenetic silencing of tumor suppressors in AML.148,149 The modest single-agent activity of HDACI and the evidence of a synergistic interaction with cytotoxic and DNA hypomethylating agents in preclinical work have prompted several combination clinical trials. E1905 study is a phase 2 randomized study that compared the hyopmethylating agent, azacitidine (AZA) alone (50 mg/m2 for 10 days) with the combination of AZA and etinostat (a class I HDACI, with a prolonged half-life of 4.5 days) in patients with AML (n=52) and MDS (n=97). Of interest, the median OS was unexpectedly worse in the combination group (13 months) compared to the AZA alone group (18 months), as well as the rate of infections and thrombocytopenia. Correlative studies showed less global hypomethylation in the combination arm which was attributed to etinostat-mediated inhibition of AZA-induced cell cycle arrest. It has therefore been suggested to administer the two reagents sequentially rather than simultaneously to mimic the successful in vitro model.150 On the basis of preclinical work indicating enhanced ATRA activity in association with VPA and 5-azacytidine (5-AZA), a total of 53 patients with AML and poor-risk patients with MDS were treated with VPA at escalating doses in combination with 5-AZA (75 mg/m2) and ATRA (45 mg/m2, started on day 3) during a 7-day course.151 Hematological responses of 42% including a CRR of 22% were observed. The CRR in this study is similar to another study using the same combination despite a prolonged exposure to ATRA (21 days).152 The combination of decitabine (20 mg/m2 for 10 days), another hypomethylating agent, with escalating doses of VPA (days 5 through 21) was also investigated, but the ORR (44%) of this small study did not differ from that of decitabine alone.153 In another study, neurotoxicity was encountered at higher doses of VPA (35 to 50 mg/m2) when it was used with decitabine (15 mg/m2 for 10 days). An ORR of 22% (50% among untreated elderly patients) was obtained.154 But, in contrast to the previous study, the response was not linked to plasma VPA levels, despite the significant increase in histone acetylation with higher VPA doses.

Idarubicin (12 mg/m2) and vorinostat were administered to 41 patients with refractory or relapsed AML. CR was noted in 17% of the evaluable patients. Garcia-Manero and colleagues administered vorinostat at a dose of 500 mg three times daily for 3 days followed by cytarabine and idarubicin infusion followed by consolidation therapy for those AML patients in remission.155 Vorinostat was continued as a maintenance therapy for up to 1 year in 17% of the participants. Vorinostat dose was reduced in 66% of the patients due to bone marrow suppression or grade 3 or 4 gastrointestinal toxicity. The ORR of 85% (100% in the presence of FLT3-ITD) was considered promising given the patients’ poor risk profile. Gogo and colleagues treated advanced and high-risk AML patients (n = 21) with vorinostat at escalating doses for 7 days followed by etoposide (100 mg/m2) and cytarabine (1 to 2 g/m2) on days 11 to 14.156 They documented an ORR of 33% and a median remission time of 7 months. The CR was achieved in 46% of those patients treated with the MTD of 200 mg twice daily.

Panobinostat (LBH589) is a hydroxamic acid analogue and a pan-HDACI that produced only transient reduction in AML blasts in a phase 1 study by Giles et al.157 In a phase 1b/2 trial conducted by Tan and colleagues, the administration of Azacytidine (75mg/m2 for 5 days) with escalating doses of panobinostat (3 times a week for 7 doses) on monthly basis in AML (n=29) and high risk MDS (n=10) patients resulted in ORR of 31% in AML and 50% in MDS patients. Fatigue was reported as the main DLT.158 The phase 2 trial of Panobinostat in relapsed/refractory acute lymphoblastic leukemia (ALL) and AML was recently completed, but the results have not yet been reported (NCT00723203).

Most recently, pracinostat gained Orphan Drug status by the FDA in 2014 for the future development in AML. It was shown to induce prolonged CRR of 14% in a phase 1 trial of older AML patients, lasting for 206 and 362 days (unpublished data). An ongoing phase II trial is currently evaluating pracinostat in combination with azacitidine in newly diagnosed AML (NCT01912274). Alternatively, the combination of pracinostat with the JAK2/FLT3 inhibitor pacritinib had significant synergetic anti-proliferative effects in vitro and in vivo, particularly in AML cell lines carrying JAK2V617F and activating FLT3 mutations.159 This synergistic effect is presumably due to the ability of this combination to counteract the rise in FLT3 and pSTAT5 (observed when using pacritinib alone), or to the suppression of stroma-mediated release of growth factors and cytokines.159 Of note, McCormack et al has also shown that combining the pan-HDACI, VPA with Nultin-3 (MDM2 antagonist) in vitro increases p53, acetylated p53, and p21 expression, and enhances the apoptotic response of AML cells, predominantly of those with higher CD34 expression.160

The exact mechanism by which HDACIs act as anti-neoplastic agents is not yet clear. Increased reactive oxygen species levels (ROS) and antioxidant gene expression following HDACI exposure were observed in vitro.133 Further, a correlation was found between antioxidant gene expression and vorinostat resistance, suggesting ROS-mediated apoptosis in HDACI-treated cells.137 Consistent with this hypothesis, the anti-leukemic activity of vorinostat was enhanced by resveratrol likely through inducing ROS production. Two additional pathways were proposed: 1) Sirt1 activation leading to the abrogation of HDACI-mediated p65 acetylation leading to NF-κβ inactivation and 2) S-phase cycle arrest, sensitizing cells to vorinostat-induced apoptosis.161

In addition, accumulating evidence suggests that HDACIs induce autophagy, presumably by increasing the expression of LC3B (a modulator of autophagy) or via the hyperacetylation and inhibition of HSP90 and subsequent degradation of AKT (a client protein of HSP90 and a negative regulator of autophagy).162166 This is supported by Wei and colleagues’ findings of increased autophagy and apoptosis when HDACIs are combined with GX15-070 (a BH3 mimetic) and the reversal of this effect by chloroquine (an autophagy inhibitor).167 The inhibition of Akt/mTOR pathway by HDACIs was shown in prostate and mantle cell lymphoma cell lines.168,169 Also, HDACI-mediated apoptosis and differentiation of AML cells was significantly increased by the addition of an mTOR inhibitor.170 Sandilya et al reported CR in two older AML patients, with high-risk features when treated with VPA (500 mg three times a day) and sirolimus, an mTOR inhibitor (1 mg per day). Of interest is that one of the patients failed 7+3 induction prior to enrollment in the study and that the responses lasted for over 9 months.171 A detailed review of the activity and toxicity of HDACIs was published by Bruserud and colleagues.172

Proteasome inhibitors

Proteasomes are intracellular organelles responsible for polyubiquitinated protein degradation including oxidized, damaged, and misfolded proteins through trypsin-like, chemotrypsin-like, or peptidyl-glutamyl peptide hydrolyzing activities.173,174 In addition, proteasomes regulate the levels of proteins involved in the cell cycle (e.g., TP53, cyclins, p27), apoptosis, and intracellular signaling pathways.175177 Proteasome inhibitors have successfully induced the in vitro killing of multiple cancers, including hematological malignancies. 178,179,178,179,178,179,178,179,178,179,178,179,178,179,178,179,178,179,178,179

Bortezomib is a reversible inhibitor of chemotrypsin-like activity in the proteasome that has been approved for the treatment of multiple myeloma and mantle cell lymphoma.180,181 Guzman and colleagues demonstrated that bortezomib induced an apoptotic response of leukemic stem cells by suppressing the degradation of (and thus increasing the levels of Iκβ), a negative regulator of NF-κβ.182 Bortezomib also improves the response of myelomonocytic leukemic cells to tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) in vitro, which is also related to bortezomib-mediated abrogation of NF-κβ activation, downregulation of the anti-apoptotic proteins C-FLIP and XIAP and expression of the TRAIL receptors DR4 and DR5.183,184

Another proposed mechanism of bortezomib is the induction of G2/M cell cycle arrest via the stabilization of cyclin and the cyclin-dependent kinase inhibitors p21 and p27.182,185 In a study by Cortes et al, bortezomib was administered to 15 patients with relapsed/refractory AML at escalating doses (0.75 to 1.5 mg/m2) twice weekly for 4 weeks every 6 weeks. Only a transient response was observed in five of the six patients who received the MTD dose of 1.25 mg/m2, with DLTs at 1.5 mg/m2 including postural hypotension, fluid retention, nausea, and diarrhea. Four patients experienced a greater than 50% reduction in peripheral or bone marrow blast count, and one demonstrated an increasing neutrophil count.186

Sarlo and colleagues treated 14 high-risk older patients with previously untreated and refractory or relapsed AML with bortezomib at a dose of 1.5 mg/m2 twice weekly for 2 weeks of a 3-week cycle. Treatment response was limited to reduced blast counts in peripheral blood and bone marrow in 61% of the participants. Neurological toxicity developed in 54% of patients, resulting in bortezomib discontinuation in 30% of cases.187 Alternative more limited bortezomib exposure was well tolerated in 31 patients with newly diagnosed AML and older patients with relapsed AML, with higher doses of up to 1.5 mg/m2 used in combination with cytarabine and idarubicin as induction therapy. The CR achieved among patients with relapsed AML (67%) exceeded the previously reported CR in literature (20–40%), with median disease-free survival of 15.3 months among responders.188 A similar response was achieved in a phase 2 CALGB study.189

Blum and colleagues treated 19 high-risk and refractory patients with decitabine and escalating doses of bortezomib (up to 1.3 mg/m2), with an ORR of 37%.190 This response was similar to a previous study of decitabine alone. Correlative studies suggested that bortezomib could potentiate decitabine cytotoxicity by increasing the expression of miR-293, thereby leading to the inhibition of the SP1/NF-κβ complex and ultimately to FLT3 downregulation. Therefore, an alternative sequential therapy of bortezomib followed by decitabine was proposed to increase the clinical benefit. Extrapolating evidence from multiple myeloma studies, the authors proposed subcutaneous and once-weekly dosing to minimize bortezomib-related neurotoxicity while maintaining its therapeutic efficacy.191,192 A synergistic anti-leukemic interaction was found between bortezomib and the HDACI belinostat in vitro.193,194 This combination was investigated in a phase I trial conducted in hematological malignancies, including AML (n=19). Results suggest safety of this regimen, without DLT. One CR (despite prior resistance to 7+3 regimen) and 2 PR were observed in AML patients which allowed these patients to proceed to allotransplantation.195

SDF1/CXCR4

The chemokine receptor CXCR4 (CD184) is an adhesion molecule widely expressed on many cell types. The interaction of CXCR4 with its ligand SDF1 (CXCL12) is implicated in the migration, homing, and quiescence of hematopoietic and leukemic stem cells in the bone marrow.196199 The heterodimerization of SDF1 with CXCR4 also activates the intracellular signaling of several survival pathways (e.g., PI3K/AKT, MAPK, PKC, JAK/STAT).200 In AML, CXCR4 overexpression has been linked to poor clinical outcomes.201,202 Plerixafor (AMD3100) is a bicyclam SMI that blocks SDF1 binding to CXCR4 and it was approved as a peripheral stem cell mobilizer for the treatment of multiple myeloma and non-Hodgkin’s disease. Plerixafor has been used successfully in preclinical mouse models of AML to sensitize AML to chemotherapy in vivo presumably by “mobilizing” leukemic stem cells and detaching them from the anti-apoptotic effects of the hematopoietic niche.203

Uy and colleagues published the results of 46 patients with refractory or relapsed AML who were treated with plerixafor (0.24 mg/kg/day) and a regimen of mitoxantrone, etoposide, and cytarabine.204 The authors described a twofold increase in circulating leukemic blasts, which peaked at 6 to 12 hours after plerixafor administration. The CRR/CRp was 46% representing modest improvements as compared to chemotherapy alone in previous reports. Additional preclinical data from MD Anderson demonstrated anti-leukemic synergy between CXCR4 blockers and FLT3 inhibitors.205 A preliminary report on the response to plerixafor, granulocyte colony-stimulating factor (GCSF), and sorafenib in relapsed/refractory FLT3-ITD+ AML demonstrated a CRR/CRp of 77%. This was associated with increased peripheral blood counts of blasts by 41-fold and of leukemic stem cells by 148-fold (NCT00943943).206 The degree of leukemic stem cell mobilization was noted to be related to baseline blast counts and VLA-4 expression. Of note is that a response was observed in three patients for whom prior treatment with FLT3 inhibitors was not effective.

HSP90

HSP90 belongs to the heat shock protein family and is comprised of two main isoforms: the inducible HSP90α and the constitutively expressed HSP90β.207 It is a pleotropic chaperone that is upregulated in response to cellular stress and is required for folding, transport, stabilization, and proteasomal degradation of proteins involved in the cell cycle, apoptosis, steroid receptor function, and signal transduction.208212 The engagement of a client protein with the HSP70/HSP40/HSP90 complex and p60HOP forms the HSP90 multichaperone complex, in which functional conformation and activation of the client protein require the binding of ATP to the N-terminal domain of HSP90.208,212214 Several oncogenic proteins are regulated by HSP90, including c-kit, HER-2, FLT3, BCR-ABL, ZAP-70, Akt, NPM-ALK, Raf-1, and mutated TP53.209,215222 High HSP90 levels were identified in many solid tumors.223 HSP90 is also highly expressed in patients with AML, and it confers chemoresistance and poor survival.224226 Furthermore, HSP90 exists predominantly in its active multiprotein complex state in cancerous cells as compared with normal tissues, which suggests that there is malignant cell dependence on this protein.227 Reikvam et al reported increased HSP protein content (including HSP90) in FLT3-ITD+ AML, and that the apoptotic response to HSP90 inhibition is more prominent in FLT3-ITD+ AML samples than in wild-type AML.228

HSP90 inhibitors act mostly by blocking HSP90 ATP binding, presumably leading to the trapping of the client oncogenic protein in a misfolded form, which is then followed by proteasomal degradation. The in vitro exposure of AML cells to the HSP90 inhibitor 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) produced the downregulation of Akt, Raf-1, N-RAS, K-RAS, and mutant p53; increased p21 levels; the activation of apoptotic protein Bax; and G2/M arrest.229,230 17-AAG is a potent and tumor-specific agent as a result of its high affinity for the HSP90 heterocomplex.227 The preclinical activity of 17-AAG positively correlated with the percentage of leukemic cells expressing HSP90, which may define a subset of potentially 17-AAG sensitive AML patients.226 Eleven patients with refractory or relapsed AML were treated with 17-AAG followed by bortezomib. Hepatotoxicity related to the benzoquinone moiety of 17-AAG was noted as the primary DLT, without hematological response apart from a transient reduction in blast count from 79% to 9% in one patient.231 QTc prolongation was also reported, and it was attributed to previous anthracyclin therapy and the 17-AAG formulation since it was not encountered in two other studies of multiple myeloma despite higher doses of 17-AAG.232,233

Alvespimycin (17-DMAG) is a 17-AAG analog with more potent effects and a longer-half life.234 It was given intravenously to 24 patients with refractory AML at escalating doses (8 to 32 mg/m2) twice a week for 2 out of 3 weeks, with a CRR of 17% and blast reduction of more than 50% in one patient. However, there were no further plans for clinical development as a result of the significant toxicity associated with this drug, including neurotopenia, diarrhea, fatigue, and myocardial ischemia.235 Patients who were co-treated with cytarabine exhibited synergetic anti-myeloid effects.236,237 HDACIs were also able to synergistically enhance 17-AAG activity against leukemic cells, including those expressing FLT3 mutation.238,239 The mechanisms of resistance to HSP90 inhibition were extensively reviewed by Piper and colleagues.240 One of the proposed mecahinsms is upregulation of another molecular chaperone HSP70. Combining 17-DMAG with the HSP70 inhibitor VER-155008 has shown a synergestic activity against primary AML cells in vitro.241 In addition to CXC4, studies are ongoing to explore the therapeuti potential of other prtective factors in the tumor niche.(table.2).

Table.2.

Potential therapeutic targets in the tumor microenvirment

Molecular target Biological Significance and related Studies Refrences
VLA-4/VCAM-1
  • BIO5192, is a VLA-4 antagonist that causes disruption of the intearction of VLA-4 adhesion molecule on progenotors with its ligand VCAM-1 on mesenchymal stroma cells, and results in significant progenitor cell mobilization.

  • VCAM-1/VLA-4 intraction stimulates NFκB signaling and is linked to chemoresistance in AML.

[368, 369]
Angiogenesis
  • The addition of Bevcizumab to chemotherapy had no additive effect in a phase II tria of older AML patients with newly diagnosed AML.

  • Inhibition of the PI3K/Akt/mTOR signaling reduced angioregulatory cytokine release from the tumor and the stroma cells (fibroblasts, osteoblasrts and endothelial cells), with an anti-proliferative effect in vitro.

  • The interaction between AML cells and endothelial cells enhances angiogenesis through upreglation of VEGF and Notch/Dll4 pathway.

[370372]
HIF-1α
  • Upreglates VEGF and induces angiogenesis.

  • Upregulates CXCR4 and CXCL12 leading to increased CXCR4-CXCL12-mediated chemotaxis, and migration of leukemic stem cell to the protective bone marrow niche.

  • The cross-talk between HIF-1α and Notch pathway promotes leukemic stem cell survival.

  • Echinomycin, an HIF inhibitor showed an anti-leukemic activity in vitro and in vivo.

[373376]
Arginase II
  • Released by AML blasts.

  • Associated with suppressed proliferation of T cells and CD34+ heamatopoietic progenitors, polarization of monocytes to immunosuppressive M2-like phenotype, and increased AML engrafment.

  • The immunosuppresive effects could be revrersed with the use of the Arginase inhibitor NOHA, and the NO synthase inhibitor L-NMMA.

  • Clinical development was delayed due to concens of drug toxicity.

[377, 378]
CD44
  • Mediates cell-cell and cell-extracelluar matrix adhesion.

  • Binding to hyaluronan ligand induces intracellur signaling.

  • CD44 antibody eliminated leukemic stem cells in vivo, as demonstrated by serial transplantation experiment.

[373, 379]

VLA-4 , veruy late antigen-4; VCAM-1, vascular cell adhesion molecule; VEGF, vascular endothelial growth factor; HIF-α, hypoxia induced factor-1 alpha; AML, acute myeloid leukemia.

Kinase Inhibitors

FLT3

The FMS-like tyrosine kinase-3 (FLT3) is a class III tyrosine kinase receptor that is predominantly expressed in hematopoietic progenitors. FLT3 homodimerizes with itself upon binding to its ligand (FLT3L), thereby leading to the activation of the cytoplasmic kinase domain and the trans-phosphorylation of the intracytoplasmic tyrosine residues, which trigger the phosphorylation of critical signal transduction proteins that drive cell differentiation, proliferation, and survival (e.g., STAT5, ERK1/2).242,243 FLT3 mutations that result in constitutively activated signaling have been described in AML, the most notable one being the internal tandem duplication (ITD) that results from a frameshift mutation involving insertions in the juxtamembrane domain of FLT3. FLT3-ITD is recognized as the most frequent mutation among individuals with AML. It is found in 24% of adult patients, and it represents a poor prognostic feature of AML with normal cytogenetics.244 The adverse effect of FLT3-ITD also overrides the favorable prognosis of NPM1 mutation, especially with high allelic burden.245 The size of FLT3-ITD (>70bp) was investigated as a prognostic indicator of survival, with conflicting reports.246,247 Point mutations in the activation loop of the tyrosine kinase domain (FLT3-TKD) were also identified in 7% of patients with AML; these were most commonly a missense mutation of (D835Y) that changes tyrosine to aspartate.248250

Semaxinibis (SU5416) is a SMI against FLT3, c-kit, and VEGFR (1–2). It was tested in a phase II trial of 43 patients with c-kit positive AML (>30%) who had refractory disease or who were deemed unfit for induction chemotherapy.251 The twice-weekly administration of 145 mg/m2 was associated with a partial response of more than 50% blast reduction in 7 patients, but this effect lasted only 1 to 5 months, whereas CRp was achieved in only 1 patient, who relapsed after 2 months. The main adverse events were nausea, headache, and severe bone pain. Both high VEGF expression and low c-kit expression were favorable predictors of response. FLT3 phosphorylation was minimally inhibited by SU5416 among the patients who were examined; however, a reduction in bone marrow vessel density was noted in all responders (100%) as compared with 72% of the non-responders. Thus, it was postulated that the clinical benefit of SU5416 is mainly the result of its anti-angiogenic properties and that an additional anti-leukemic effect could be established by targeting c-kit–mediated and FLT3-mediated proliferation.

Sorafenib is an oral multikinase inhibitor approved for the use in patients with renal cell carcinoma, with activity against serine/ threonine kinase Raf-1, c-Kit, FLT-3, PDGFR and VEGFR. A recent German randomized, placebo controlled trial in older AML patients (n=201) revealed that the use of sorafenib (400 mg twice a day) in between induction (cytarabine and daunorubicin) and consolidation chemotherapy followed by maintenance duration after CR for 1 year from the beginning of therapy was associated with higher toxicity and mortality (17 vs. 7%), lower CRR (46 vs. 60%) and ORR (57 vs.64%) after induction, and lack of improvement in event free-survival (EFS) (7 vs. 5 months) and OS (15 vs. 13 months), including those patients with FLT3-ITD.252 In the SORAML trial, younger AML patients (n=276) received either sorafeninb (800mg a day) or placebo concomitantly with induction and consolidation therapy and for 1 year as maintenance therapy following consolidation. Preliminary results were suggestive of improvement in EFS, but not in the 2-year OS in the sorafenib arm, particularly in patients carrying FLT3-ITD. There was no significant difference in CRR between the placebo and sorafenib arms (56% and 60%, respectively). In a retrospective analysis of refractory and relapsed FLT3-ITD AML patients (n=69), Metzelder et al reported CRR on sorafenib monotherapy in 23% of the cases, with more frequent and durable response (median of 197 days) in patients who relapsed post- allogeneic stem cell transplant compared to those who relapsed after chemotherapy (median of 136 days).253 This raised the possibility of an enhanced sorafenib activity by graft-versus-leukemia (GVL) effect. Elevated FLT3 ligand levels following chemotherapy resulting in FLT3 receptor activation was postulated as a mechanism of resistance to FLT-3 inhibitors.254 Ravandi et al studied the combination of azacytidine and sorfenib in 43 patients with refractory or relapsed FLT-3 ITD AML. The CRR was 43% (ORR 46%) but with a median duration of only 2.3 months.255

Sunitinib (SU11248) is another oral inhibitor of FLT3, c-kit, PDGFR, and VEGFR (1–2). It was evaluated in 15 patients with AML, who were given two doses of 50 mg/day and 75 mg/day for a 4-week cycle followed by 1 or 2 weeks of rest. The responses were limited to partial remissions for short durations (4 to 16 weeks) and morphological remission in 1 patient. These effects were mainly observed in patients with FLT3 mutations. The higher dose of 75 mg was associated with DLTs of cardiac failure, hypertension, and profound fatigue.256

Midostaurin (PKC412) is another FLT3 inhibitor that was examined in patients with relapsed or refractory AML and high-risk MDS (n = 95). They were given doses of 50 mg and 100 mg twice daily. The drug was well tolerated and induced hematological improvement in patients with wild-type and FLT3 mutations (42% and 71%, respectively).257 A subsequent phase 3 CALGB study was launched in 2008, in which patients aged 60 years or less with newly diagnosed AML with FLT3 mutations were randomized to either midostaurin with standard induction and consolidation therapy or standard chemotherapy and consolidation without midostaurin. The results are pending (NCT00651261). Finally, patients with activating mutations of FLT3 who are in first remission will be randomized (phase II) to either observation or mitostaurin maintenance after allogeneic stem cell transplantation (NCT01883362). This trial is just beginning and the results will be expected in the next 3–5 years.

Quizartinib (AC220) is a new selective FLT3 inhibitor with improved efficacy and a better pharmacokinetic profile. A phase 1 study of 76 patients with relapsed or refractory AML and who were treated with quizartinib (12 to 450 mg/day) revealed an ORR of 53% in the FLT3-ITD+ group (CR noted in 24%) and 14% in the FLT3-ITD group.258 Quizartinib was further studied in a phase 2 trial of AML, in which an ORR of 68% was achieved in patients with refractory or relapsed AML, including those who relapsed after allogeneic bone marrow transplantation.259 Allogeneic transplantation after quizartinib response was performed in 35% of both FLT3-ITD+ and FLT3-ITD cases. The OS at 1 year for FLT3-ITD+ patients post-allogeneic stem cell transplant was 39% among those achieving complete and partial remission. Alternatively, the OS at 1 year without allogeneic stem cells transplantation after complete and partial remission were 25% and 5%, respectively.260 A phase 1 clinical trial is currently examining the role of Quizartinib as maintenance in AML post-allogeneic stem cell transplantation (NCT01468467).

On the basis of in vitro analyses of primary AML samples, it was concluded that increased mutant allelic burden (>50%) and disease relapse confer increased sensitivity to FLT3 inhibitors.261 The type and location of FLT3 mutations have also influenced the therapeutic response.262264 Parmar et al demonstrated resistance of CD34+FLT3-ITD+ leukemic progenitors in vitro to FLT3 inhibitors when cultured with stromal niche cells.265 Alternatively, recent publications by Weisberg and colleagues demonstrated the benefits of using multikinase targeting agents (e.g., AKT, JAK2, Abl) to overcome stromal-cell–derived protective effects and to sensitize AML blasts to FLT3 inhibitors.266,267

C-KIT

The stem cell factor receptor (c-kit) is a type III tyrosine kinase receptor encoded by the proto-oncogene kit. The binding of c-kit to its ligand, the stem cell factor (SCF), results in dimerization and autophosphorylation of the receptor and subsequent activation of multiple pro-survival signaling pathways, including PI3 kinase, RAS-RAF-MAP kinase, and JAK/STAT.268,269 C-kit is expressed in normal hematopoietic cells and in more than 70% of patients with AML, and it plays important roles in cell differentiation, expansion, and survival.268,270,271 Constitutive activation of c-kit is most frequently due to point mutations in the receptor tyrosine kinase or to ITDs, and these were associated with an increased risk of relapse, drug resistance, and shorter survival (particularly with t(8;21) AML).272,273276 Imatinib is a SMI that blocks the ATP binding site of the tyrosine kinases c-kit, PDGFRα and PDGFRβ, and BCR-ABL fusion protein.277 The published data regarding the efficacy of imatinib for the treatment of refractory AML are inconsistent. This might be explained by variations in patient selection, c-kit mutational status and level of expression and/or dependency of leukemic cell survival on other genetic alterations.278282 The addition of imatinib to the reinduction regimen of mitoxantrone, etoposide and cytarabine in a phase I/II trial (n=33) have shown a good response, particularly in patients with relapsed disease, and responses were correlated with AKT inhibition.283 Imatinib resistance due to the D816 c-kit and to other secondary mutations in c-kit that develop after exposure to imatinib has been described in patients with GIST and CML.282 In preclinical experiments, Santos et al showed an apoptotic/anti-proliferative response of AML progenitor cells to dasatinib, a new c-kit and src inhibitor, with an additive effect when administered in combination with chemotherapy.284 A recent CALGB/Alliance and other clinical trials are testing the role of dasatinib in patients with t(8;21) and inv16 AML, both AMLs that can be negatively impacted by activating mutations of c-kit (NCT00850382, NCT02013648, NCT01876953).

JAK2

Janus kinases (JAKs) are non-receptor tyrosine kinases that include JAK1, JAK2, and tyrosine kinase 2 (TYK2).285 JAKs are associated with the intracellular domain of the class 1 and 2 cytokine receptors for the transmission of cytokine-induced growth and inflammatory signaling to the nucleus through the phosphorylation of the transcription factors STATs.286,287 Hyperactivated JAK/STAT signaling is frequently observed in patients with AML, and a negative impact of elevated phosphorylated JAK2 levels on treatment outcome was recently demonstrated.288291 The activating mutation JAK2V617F was reported in 0.5% to 8% of de novo AML, with a higher frequency seen in patients with t(8;21) leukemia, in whom it is considered a cooperating mutation.290,292295 Alterations in the negative regulators of JAK/STAT signaling have also been identified, including the downregulation of CD45 in patients with t(8;21) AML, and hypermethylation of the suppressor of cytokine signaling-1, and the protein inhibitor of activated stats-2 in 70% of AML population.296298

The treatment of refractory leukemia with ruxolitinib (Jakafi, a JAK1/2 inhibitor FDA approved for the treatment of advanced myelofibrosis) in a phase 2 trial resulted in CR in only 3 of the 38 participants.299 All responders had post-myeloproliferative AML, and the JAK2V617F mutation did not influence the treatment response. The toxicity was limited, and it manifested mainly as thrombocytopenia and increased transaminases. Hyperactivated JAK/STAT signaling was postulated as a mechanism of resistance to selective FLT3 inhibitors.300 The simultaneous targeting of FLT3 and JAK/STAT pathways has shown synergistic effects against AML cell lines.300,301 Of note, lestaurtinib, a JAK2/FLT3 inhibitor, failed to improve patient responses or survival in a phase 2 study when given 2 days after salvage chemotherapy in patients with FLT3-mutant AML (n = 224). These results were attributed to unfavorable pharmacokinetics resulting in inadequate suppression of FLT3 or to post-chemotherapy elevation in FLT3 ligand levels (thus minimizing FLT3 inhibition) and α1-acid glycoprotein (thus decreasing the free serum levels of the drug).302 Another FLT3/JAK2 inhibitor, pacritinib (SB1518), demonstrated impressive preclinical activity in FLT3-TKI resistant AML, which has led to a phase 1/2 study in patients with advanced AML (NCT00719836).301

Polo-Like Kinases (Plk)

The Polo-like Kinases (Plks) from the family of serine/threonine kinases (Plk1-4) have emerged as important regulators of cytokinesis and cell cycle progression. But Plk1 (STPK13) is the only isoform that has been shown to have a critical role in activation of Cdc2, centrosomal maturation and normal spindle formation.303 These family members, especially Plk1, are overexpressed in many tumors and have been thought to be logical therapeutic targets, especially in light of in vitro data suggesting that genetic deletion of Plk1 or knockdown of Plks in cell lines results in either loss of viability or decrease in proliferation. Of note is that this occurred predominantly in those cell lines harboring K-RAS mutationa.304 Although multiple Plk inhibitors are now in early phase clinical trials for non-hematologic malignancies, no studies have thus far been opened for the treatment on AML.

Hedgehog Pathway

Hedgehog signaling is critical for hematopoietic stem cell differentiation and survival. It is positively regulated by the transmembrane molecule, Smoothened (SMO), and the Glioma family of transcription factors (GLI1, GLI2 and GLI3). The transmembrane proteins, Patched (PTCH1 and PTCH2), have inhibitory effects through the suppression of SMO. When GLI2 and GLI3 are phosphorylated on serine and threonine by glycogen synthase kinase (GSK3β), protein kinase A, and casein kinase, they undergo C-terminal ubiquitination and proteasomal proteolysis, yielding truncated proteins that inhibit the transcription of downstream targets. Ligand binding results in PTCH and SMO dissociation followed by SMO accumulation and stabilization of GLI2 and GLI3, which translocate to the nucleus and induce transcription of GLI1, PTCH1, and PTCH2, which then mediate cell cycle progression, apoptosis, and stem cell renewal.305 Aberrant Hedgehog signaling can contribute to tumor initiation, angiogenesis, and cancer cell survival via gain-of-function SMO mutations, loss-of-function PTCH mutations, aberrant GLI2 and GLI3 expression, upregulation of drug resistance genes (e.g., p-glycoprotein, MDR1, BCRP, and ABCG2) or by influencing the interaction of cancer stem cells with the stroma. 306312 The expression of SHH, GLI1, and GLI2 has been demonstrated in AML blasts, and an anti-leukemic effect was observed in vitro in response to cyclopamine, an SMI of Hedgehog/SMO signaling.309

Minami and colleagues demonstrated that the SMO inhibitor PF-04449913 could inhibit leukemia-initiating cells as assessed by the serial xenotransplantation of primary AML blasts.313 The initial results from a phase 1 trial evaluating PF-04449913 in multiple hematological malignancies are suggestive of good safety profile and treatment response.314 PF-04449913 could also reduce the resistance to Ara-C mediated by co-culturing of AML cell lines with bone marrow stromal cells in vitro.315 A clinical trial examining the effect of PF-04449913 with chemotherapy in patients with AML or MDS is currently underway (NCT01546038). In addition, a GLI2 inhibitor (GANT61) has recently shown promising anti-leukemic activity in vitro.316

WNT/β-Catenin Pathway

WNT is a glycoprotein that plays an important role in hematopoietic stem cell growth and renewal and in organogenesis during embryonic development.317 WNT is normally in an inactive state in adults, but it is overexpressed and activated in patients with multiple cancers.318 It is also implicated in epithelial-mesenchymal transition (EMT) that increases the metastatic potential of tumors.319 Growing evidence supports the role of the WNT/β-catenin (canonical) pathway in AML. The activation of this pathway is induced by the engagement of WNT with the Frizzled receptor and its co-receptors, the lipoprotein-receptor–related proteins (LRP5 and LRP6). This leads to the relocation of the multiprotein degradation complex (MDC), which consists of glycogen synthase kinase (GSK3β), adnomatous polyposis coli (APC) and axin proteins to the cell membrane resulting in suppression of MDC function. MDC is normally responsible for the phosphorylation of β-catenin which is then targeted by the E3–ubiquitin–ligase complex for proteasomal degradation. Thus, the inactivation of MDC leads to the intracellular accumulation of β-catenin, which translocates to the nucleus to be associated with the T-cell receptor factor and the lymphoid enhancer-binding factor 1 (LEF1) leading to upregulation of survivin, cyclin-D1, C-MYC, and CD44 among others.320323

β-catenin expression in primary AML specimens has been associated with WNT activation and reduced patient survival.324,325 Multiple strategies are being investigated to abrogate WNT/β-Catenin signaling; 1)disrupting ligand interaction with WNT and its co-receptors (a soluble ligand binding domain of Fzd8, Fzd8-CRD-FC); 2)blocking β-catenin accumulation via the inactivation of Dishevelled, a protein that interacts with Frizzled and that is involved in MDC inactivation (e.g., niclosamide); or by the upregulation of axin (e.g., XAV939, IWR-1); 3) blocking β-catenin nuclear localization by NSAID; 4)the inhibition of HGF-mediated degradation of E-cadherin, a protein that normally complexes with β-catenin leading to β-catenin intracellular sequestration (e.g., PHA665752); 5)the inactivation of p38 MAPK, a kinase that normally phosphorylates and inactivates GSK3 resulting in upregulation of Snail and the downregulation of E-cadherin (e.g., PH-797804); and 6)blocking the formation and transactivation of β-catenin/LEF/TCF complex (e,g., ICG-001, CGP049090, ZTM000990, PFK115-584).326337 Several drugs targeting WNT/β-catenin signaling are undergoing clinical evaluation in AML (Table 1).

IDH1 and IDH2

Mutations in isocitate dehydrogenase (IDH1 and IDH2) are detected in around 6–16% and 8–19% of AML patients, respectively.338 The incidence is higher in patients with normal cytogenetics and NPM1 mutations.338340 But the data are conflicting regarding the prognostic significance of such mutations.338,341 However, recent studies suggested that an elevated serum level of 2-hydroxyglutarate protein (which is produced by IDH1 and IDH-2 mutant cells, and is thought to induce DNA hypermethylation and accelerate leukemogenesis) is a marker of IDH mutation, and disease activity and a predictor of poor outcome.340,342345 Cell line studies by Chaturvedi et al demonstrated suppression of growth and MAPK signaling in IDH1- mutant leukemic cells when treated with HMS-101 (a SMI of IDH1 mutation).346 AG-6780 is a SMI of IDH2/RI40Q mutation that has been shown to stimulate the expression of maturation markers in leukemic cells in vitro, reflecting induction of differentiation.347 AG-221, is another IDH2 inhibitor that is being evaluated as a single agent in patients with MDS, relapsed/refractory AML, and newly diagnosed, older AML patients carrying IDH2 mutation. The initial report of the phase I trial of AG-221 supports safety of the drug with early signs of efficacy. Responses were observed in 6 of 10 evaluable patients; with 2 CR (one of whom proceeded to allogeneic stem cell transplantation).348

Immunotherapy: Monoclonal and Bispecific Antibodies

Gemtuzumab ozogamicin (GO) is a humanized monoclonal antibody- drug conjugate directed against CD33, a transmembrane receptor expressed by the majority of AML blasts.349 An ORR of 26–30% in several Phase II trials of GO as monotherapy in relapsed AML led to the initial FDA approval in 2000. GO was withdrawn in 2010 due to increased death rate and lack of an additional benefit in a SWOG study. However, emerging data from subsequent phase III trials studying the incorporation of GO to standard chemotherapy indicated an improved disease-free and overall survival, especially in the low risk groups.350352 It was presumed that such controversy is due to using lower doses of daunorubicin in the GO arm of the SWOG trial.353 Further, GO was associated with high response and prolonged remission in APL when used as monotherapy (substituting chemotherapy in older patients or in molecular relapse) and as an alternative to anthracyclines in combination with ATRA and arsenic trioxide (for induction therapy of high-risk APL).354356

New technologies in protein engineering have led to the generation of promising immune-based therapeutic agents that could recruit immune effector cells to tumor targets. Such molecules are called bispecific antibodies. They combine two antigen specificities for the simultaneous binding to specific markers expressed by effector cells and tumor cells, thereby leading to immune response activation for the selective eradication of target cells in an antigen-specific and MHC-unrestricted manner.357359 Thus, bispecific antibodies could bypass some of the protective barriers of AML blasts against immune responses such as MHC-downregulation, impaired immunological synapse formation between T cells and blasts, and expression of negative co-stimulatory ligands (such as PD-1L) on target AML and tumor cells. Currently, the most developed platforms of bispecific antibodies are the bispecific T-cell engagers (BiTEs) and the dual affinity re-targeting molecules (DARTs). A CD3 × CD33 BiTE has shown promising tumoricidal effects in vitro and in vivo preclinical models.360 Our group has investigated a CD3 × CD123 DART in AML and shown near-complete eradication of the CD123+ human AML in the peripheral blood in a primary human AML xenotransplantation model.361 However, non-specific T cell activation and cytokine storm are potential risks with the use of such bi-specific agents. Blinatumomab (CD3 × C19 BiTE) has been shown to induce high rates of remissions in patients with B-precursor acute lymphoblastic leukemia (B-ALL), but with prolonged B lymphocyte depletion and hypogammaglobulinemia.181183 Cytokine release syndrome was observed, manifested with fever, rigors, and hypoxia, which could be ameliorated by steroid prophylaxis.184 Approximately 15% of patients experienced CNS complications, including convulsions and encephalopathy, which were reversible after drug withdrawal.362364 Wong and colleagues demonstrated recently the activation of T cell cytotoxicity against B-precursor CLL (B-CLL) following exposure to Blinatumomab in vitro.365 Blinatumomab is currently being evaluated in patients with ALL, B-NHL and B-CLL (NCT01466179).

Expert Commentary

Aberrant signal transduction has been considered the driving force of oncogenesis, with a high degree of complexity and cross-talk among the different pathways. The use of SMIs allows for the specific targeting of intracellular kinases and protein–protein interactions, which has been proven effective for improving the survival of GIST, APL and CML and which has prompted substantial interest in AML. However, in contrast with the preclinical results, there has been only a modest clinical benefit of SMIs in patients with AML, especially when used in the absence of chemotherapy. These limited clinical responses may involve the upregulation of the targeted gene, the selective expansion of a resistant clone (as a result of epigenetic alterations or primary or acquired mutations), a protective tumor microenvironment, the presence of other molecular aberrations that maintain leukemic cell proliferation and survival, or the properties of the cell lines or primary AML samples examined in the preclinical work.185 This argues for the importance of the combination with other targeting and cytotoxic agents while considering the potential toxicity that may result from the off-target effects on normal cells.

It is important to note that most of the data available are from early-phase studies. This preludes accurate interpretation in the light of the heterogenousity and small number of the patients, the short duration of follow up, selection bias, and the presence of multiple confounding factors that may influence treatment outcomes (e.g., drug exposure, clinical status, cytogenetics, interval of CR, and number of treatment cycles administered). In another aspect, the high remission rate of FLT3 inhibitors, although it is mostly short-lasting, can be considered as a bridging strategy for transplantation. Combining SMIs with conventional chemotherapy is an interesting investigational approach in AML to reduce disease burden and thereby improve transplantation outcome. Studies have shown the immunomodulatory properties of some SMIs.366 For example, sunitinib down regulates the expression of PDL-1 (by myeloid-derived suppressor cells {MDSCs}), and CTLA-4 and PD-1 (by tumor cells), reduces T-regulatory cells and MDSCs, and increases cytotoxic T cell expansion.367 It may therefore be reasonable to study such SMIs 1) as maintenance therapy post-transplantation to enhance GVL effect, prolong disease remission and eliminate minimal residual disease, and 2) to enhance the GVL effect of donor lymphocyte infusion for relapsed AML after transplantation. We also agree with previous reports that durable partial response and disease-stabilization of well tolerated SMIs are potential markers of clinical benefit, particularly in older patients, unfit for chemotherapy and transplantation, and in relapsed AML patients with limited therapeutic options.145

Five-year view

Compelling evidence supports the heterogenousity of AML, the importance of cytogenetics, and the use of gene expression profiling to stratify the risk and design a treatment plan that is based on the biological characteristics of the disease. It is intuitively appealing to incorporate molecular markers of prognosis and biochemical response on the basis of correlative scientific analyses of the relevant patient specimens for more personalized treatment decisions and clinical trial design. Understanding the redundancy of pro-survival pathways, tumor evolution, and cancerous cell interaction with protective niches is an ongoing challenge to the discovery and validation of new drug targets and combination therapies. Additional effort is needed to exploit the pharmacokinetics/dynamics and mechanisms of action and resistance of the current SMIs for enhanced therapeutic strategies in the future.

Key issues.

  • Small molecule inhibitors have been developed to modulate the activity of proteins encoded by mutated or overexpressed genes in patients with acute myeloid leukemia.

  • Clonal evolution, dependency on alternative signaling pathways, and pharmacodynamics are potential obstacles to the clinical development of small molecule inhibitors for the treatment of acute myeloid leukemia.

  • Multi-targeting and the combination of small molecule inhibitor treatment with chemotherapy may improve treatment efficacy.

  • The analysis of patient samples is an important tool to investigate resistance mechanisms and to discover and validate biological markers that could be used for the prediction and assessment of treatment response.

Acknowledgments

Funding Support: This work was supported by grants from the National Institutes of Health/National Cancer Institute (grants NIH/NCI P01 CA101937 [JFD] and NIH/NCI R01 CA152329 [JFD]).

Footnotes

Financial and competing interest: None.

References

  • 1.Litzow MR. Progress and strategies for patients with relapsed and refractory acute myeloid leukemia. Curr Opin Hematol. 2007;14:130–137. doi: 10.1097/MOH.0b013e32801684ef. [DOI] [PubMed] [Google Scholar]
  • 2.Luger SM. Treating the elderly patient with acute myelogenous leukemia. Hematology Am Soc Hematol Educ Program. 2010;2010:62–69. doi: 10.1182/asheducation-2010.1.62. [DOI] [PubMed] [Google Scholar]
  • 3.Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid leukemia. J Clin Oncol. 2011;29:487–494. doi: 10.1200/JCO.2010.30.1820. [DOI] [PubMed] [Google Scholar]
  • 4.Wingard JR, Majhail NS, Brazauskas R, et al. Long-term survival and late deaths after allogeneic hematopoietic cell transplantation. J Clin Oncol. 2011;29:2230–2239. doi: 10.1200/JCO.2010.33.7212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ungewickell A, Medeiros BC. Novel agents in acute myeloid leukemia. Int J Hematol. 2012;96:178–185. doi: 10.1007/s12185-012-1151-5. [DOI] [PubMed] [Google Scholar]
  • 6.Sekeres MA. Treatment of older adults with acute myeloid leukemia: state of the art and current perspectives. Haematologica. 2008;93:1769–1772. doi: 10.3324/haematol.2008.000497. [DOI] [PubMed] [Google Scholar]
  • 7.Popplewell LL, Forman SJ. Is there an upper age limit for bone marrow transplantation? Bone Marrow Transplant. 2002;29:277–284. doi: 10.1038/sj.bmt.1703382. [DOI] [PubMed] [Google Scholar]
  • 8.Leith CP, Kopecky KJ, Godwin J, et al. Acute myeloid leukemia in the elderly: assessment of multidrug resistance (MDR1) and cytogenetics distinguishes biologic subgroups with remarkably distinct responses to standard chemotherapy. A Southwest Oncology Group study. Blood. 1997;89:3323–3329. [PubMed] [Google Scholar]
  • 9.Mrozek K, Bloomfield CD. Chromosome aberrations, gene mutations and expression changes, and prognosis in adult acute myeloid leukemia. Hematology Am Soc Hematol Educ Program. 2006:169–177. doi: 10.1182/asheducation-2006.1.169. [DOI] [PubMed] [Google Scholar]
  • 10.O'Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2003;348:994–1004. doi: 10.1056/NEJMoa022457. [DOI] [PubMed] [Google Scholar]
  • 11.Huang ME, Ye YC, Chen SR, et al. Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Blood. 1988;72:567–572. doi: 10.1182/blood-2016-11-750182. [DOI] [PubMed] [Google Scholar]
  • 12.Castaigne S, Chomienne C, Daniel MT, et al. All-trans retinoic acid as a differentiation therapy for acute promyelocytic leukemia. I. Clinical results. Blood. 1990;76:1704–1709. [PubMed] [Google Scholar]
  • 13.Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer. Nat Rev Cancer. 2006;6:493–505. doi: 10.1038/nrc1885. [DOI] [PubMed] [Google Scholar]
  • 14.Lindstrom MS. NPM1/B23: A Multifunctional Chaperone in Ribosome Biogenesis and Chromatin Remodeling. Biochem Res Int. 2011;2011:195209. doi: 10.1155/2011/195209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ruggero D, Pandolfi PP. Does the ribosome translate cancer? Nat Rev Cancer. 2003;3:179–192. doi: 10.1038/nrc1015. [DOI] [PubMed] [Google Scholar]
  • 16.Nozawa Y, Van Belzen N, Van der Made AC, Dinjens WN, Bosman FT. Expression of nucleophosmin/B23 in normal and neoplastic colorectal mucosa. J Pathol. 1996;178:48–52. doi: 10.1002/(SICI)1096-9896(199601)178:1<48::AID-PATH432>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  • 17.Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352:254–266. doi: 10.1056/NEJMoa041974. [DOI] [PubMed] [Google Scholar]
  • 18.Falini B, Nicoletti I, Martelli MF, Mecucci C. Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features. Blood. 2007;109:874–885. doi: 10.1182/blood-2006-07-012252. [DOI] [PubMed] [Google Scholar]
  • 19.Grummitt CG, Townsley FM, Johnson CM, Warren AJ, Bycroft M. Structural consequences of nucleophosmin mutations in acute myeloid leukemia. J Biol Chem. 2008;283:23326–23332. doi: 10.1074/jbc.M801706200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sportoletti P, Grisendi S, Majid SM, et al. Npm1 is a haploinsufficient suppressor of myeloid and lymphoid malignancies in the mouse. Blood. 2008;111:3859–3862. doi: 10.1182/blood-2007-06-098251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.den Besten W, Kuo ML, Williams RT, Sherr CJ. Myeloid leukemia-associated nucleophosmin mutants perturb p53-dependent and independent activities of the Arf tumor suppressor protein. Cell Cycle. 2005;4:1593–1598. doi: 10.4161/cc.4.11.2174. [DOI] [PubMed] [Google Scholar]
  • 22.Colombo E, Martinelli P, Zamponi R, et al. Delocalization and destabilization of the Arf tumor suppressor by the leukemia-associated NPM mutant. Cancer Res. 2006;66:3044–3050. doi: 10.1158/0008-5472.CAN-05-2378. [DOI] [PubMed] [Google Scholar]
  • 23.Falini B, Bolli N, Liso A, et al. Altered nucleophosmin transport in acute myeloid leukaemia with mutated NPM1: molecular basis and clinical implications. Leukemia. 2009;23:1731–1743. doi: 10.1038/leu.2009.124. [DOI] [PubMed] [Google Scholar]
  • 24.Huang S, DeGuzman A, Bucana CD, Fidler IJ. Nuclear factor-kappaB activity correlates with growth, angiogenesis, and metastasis of human melanoma cells in nude mice. Clin Cancer Res. 2000;6:2573–2581. [PubMed] [Google Scholar]
  • 25.Li B, Li YY, Tsao SW, Cheung AL. Targeting NF-kappaB signaling pathway suppresses tumor growth, angiogenesis, and metastasis of human esophageal cancer. Mol Cancer Ther. 2009;8:2635–2644. doi: 10.1158/1535-7163.MCT-09-0162. [DOI] [PubMed] [Google Scholar]
  • 26.Guzman ML, Neering SJ, Upchurch D, et al. Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood. 2001;98:2301–2307. doi: 10.1182/blood.v98.8.2301. [DOI] [PubMed] [Google Scholar]
  • 27.Grandage VL, Gale RE, Linch DC, Khwaja A. PI3-kinase/Akt is constitutively active in primary acute myeloid leukaemia cells and regulates survival and chemoresistance via NF-kappaB, Mapkinase and p53 pathways. Leukemia. 2005;19:586–594. doi: 10.1038/sj.leu.2403653. [DOI] [PubMed] [Google Scholar]
  • 28.Cilloni D, Messa F, Rosso V, et al. Increase sensitivity to chemotherapeutical agents and cytoplasmatic interaction between NPM leukemic mutant and NF-kappaB in AML carrying NPM1 mutations. Leukemia. 2008;22:1234–1240. doi: 10.1038/leu.2008.68. [DOI] [PubMed] [Google Scholar]
  • 29.Freedman DA, Levine AJ. Nuclear export is required for degradation of endogenous p53 by MDM2 and human papillomavirus E6. Mol Cell Biol. 1998;18:7288–7293. doi: 10.1128/mcb.18.12.7288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hietanen S, Lain S, Krausz E, Blattner C, Lane DP. Activation of p53 in cervical carcinoma cells by small molecules. Proc Natl Acad Sci U S A. 2000;97:8501–8506. doi: 10.1073/pnas.97.15.8501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pathria G, Wagner C, Wagner SN. Inhibition of CRM1-mediated nucleocytoplasmic transport: triggering human melanoma cell apoptosis by perturbing multiple cellular pathways. J Invest Dermatol. 2012;132:2780–2790. doi: 10.1038/jid.2012.233. [DOI] [PubMed] [Google Scholar]
  • 32.Newlands ES, Rustin GJ, Brampton MH. Phase I trial of elactocin. Br J Cancer. 1996;74:648–649. doi: 10.1038/bjc.1996.415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Mutka SC, Yang WQ, Dong SD, et al. Identification of nuclear export inhibitors with potent anticancer activity in vivo. Cancer Res. 2009;69:510–517. doi: 10.1158/0008-5472.CAN-08-0858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sakakibara K, Saito N, Sato T, et al. CBS9106 is a novel reversible oral CRM1 inhibitor with CRM1 degrading activity. Blood. 2011;118:3922–3931. doi: 10.1182/blood-2011-01-333138. [DOI] [PubMed] [Google Scholar]
  • 35.Ranganathan P, Yu X, Na C, et al. Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia. Blood. 2012;120:1765–1773. doi: 10.1182/blood-2012-04-423160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kojima K, Kornblau SM, Ruvolo V, et al. Prognostic impact and targeting of CRM1 in acute myeloid leukemia. Blood. 2013;121:4166–4174. doi: 10.1182/blood-2012-08-447581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Savona MGR, Brown DP, et al. 55th ASH; 2013. 2013. Phase I trial of selinexor (KPT-330), a first-In-class oral selective inhibitor of nuclear export (SINE) in patients (pts) with advanced acute myelogenous leukemia (AML) [Google Scholar]
  • 38.Rettig MHM, Prior J, et al. Inhibition of nuclear transport modulator Xpo1/CRM1 for the treatment of acute myeloid leukemia (AML); 55th ASH annual meeting; 2013. [Google Scholar]
  • 39.Fiskus WBR, Karissa P, et al. Co-treatment with NPM1 antagonist and FLT3 inhibitor or pan-histone deacetylase inhibitor exerts superior efficacy against cultured and primary human AML cells co-expressing mutant NPM1 and FLT3-ITD; AACR 104th annual meeting; 2013; 2013. [Google Scholar]
  • 40.Gurumurthy M, Tan CH, Ng R, et al. Nucleophosmin interacts with HEXIM1 and regulates RNA polymerase II transcription. J Mol Biol. 2008;378:302–317. doi: 10.1016/j.jmb.2008.02.055. [DOI] [PubMed] [Google Scholar]
  • 41.Benson C, White J, De Bono J, et al. A phase I trial of the selective oral cyclin-dependent kinase inhibitor seliciclib (CYC202; R-Roscovitine), administered twice daily for 7 days every 21 days. Br J Cancer. 2007;96:29–37. doi: 10.1038/sj.bjc.6603509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Falini B, Bolli N, Shan J, et al. Both carboxy-terminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+ AML. Blood. 2006;107:4514–4523. doi: 10.1182/blood-2005-11-4745. [DOI] [PubMed] [Google Scholar]
  • 43.Vousden KH, Prives C. Blinded by the Light: The Growing Complexity of p53. Cell. 2009;137:413–431. doi: 10.1016/j.cell.2009.04.037. [DOI] [PubMed] [Google Scholar]
  • 44.Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 mutations in human cancers. Science. 1991;253:49–53. doi: 10.1126/science.1905840. [DOI] [PubMed] [Google Scholar]
  • 45.Muller PA, Vousden KH. p53 mutations in cancer. Nat Cell Biol. 2013;15:2–8. doi: 10.1038/ncb2641. [DOI] [PubMed] [Google Scholar]
  • 46.Levine AJ. p53, the cellular gatekeeper for growth and division. Cell. 1997;88:323–331. doi: 10.1016/s0092-8674(00)81871-1. [DOI] [PubMed] [Google Scholar]
  • 47.Schottelius A, Brennscheidt U, Ludwig WD, Mertelsmann RH, Herrmann F, Lubbert M. Mechanisms of p53 alteration in acute leukemias. Leukemia. 1994;8:1673–1681. [PubMed] [Google Scholar]
  • 48.Bueso-Ramos CE, Yang Y, deLeon E, McCown P, Stass SA, Albitar M. The human MDM-2 oncogene is overexpressed in leukemias. Blood. 1993;82:2617–2623. [PubMed] [Google Scholar]
  • 49.Wu X, Bayle JH, Olson D, Levine AJ. The p53-mdm-2 autoregulatory feedback loop. Genes Dev. 1993;7:1126–1132. doi: 10.1101/gad.7.7a.1126. [DOI] [PubMed] [Google Scholar]
  • 50.Freedman DA, Wu L, Levine AJ. Functions of the MDM2 oncoprotein. Cell Mol Life Sci. 1999;55:96–107. doi: 10.1007/s000180050273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zhou M, Gu L, Abshire TC, et al. Incidence and prognostic significance of MDM2 oncoprotein overexpression in relapsed childhood acute lymphoblastic leukemia. Leukemia. 2000;14:61–67. doi: 10.1038/sj.leu.2401619. [DOI] [PubMed] [Google Scholar]
  • 52.Rayburn E, Zhang R, He J, Wang H. MDM2 and human malignancies: expression, clinical pathology, prognostic markers, and implications for chemotherapy. Curr Cancer Drug Targets. 2005;5:27–41. doi: 10.2174/1568009053332636. [DOI] [PubMed] [Google Scholar]
  • 53.Ventura A, Kirsch DG, McLaughlin ME, et al. Restoration of p53 function leads to tumour regression in vivo. Nature. 2007;445:661–665. doi: 10.1038/nature05541. [DOI] [PubMed] [Google Scholar]
  • 54.Xue W, Zender L, Miething C, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–660. doi: 10.1038/nature05529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tovar C, Graves B, Packman K, et al. MDM2 small-molecule antagonist RG7112 activates p53 signaling and regresses human tumors in preclinical cancer models. Cancer Res. 2013;73:2587–2597. doi: 10.1158/0008-5472.CAN-12-2807. [DOI] [PubMed] [Google Scholar]
  • 56.Kojima K, Konopleva M, Samudio IJ, et al. MDM2 antagonists induce p53-dependent apoptosis in AML: implications for leukemia therapy. Blood. 2005;106:3150–3159. doi: 10.1182/blood-2005-02-0553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lau LM, Nugent JK, Zhao X, Irwin MS. HDM2 antagonist Nutlin-3 disrupts p73-HDM2 binding and enhances p73 function. Oncogene. 2008;27:997–1003. doi: 10.1038/sj.onc.1210707. [DOI] [PubMed] [Google Scholar]
  • 58.Ambrosini G, Sambol EB, Carvajal D, Vassilev LT, Singer S, Schwartz GK. Mouse double minute antagonist Nutlin-3a enhances chemotherapy-induced apoptosis in cancer cells with mutant p53 by activating E2F1. Oncogene. 2007;26:3473–3481. doi: 10.1038/sj.onc.1210136. [DOI] [PubMed] [Google Scholar]
  • 59.LaRusch GA, Jackson MW, Dunbar JD, Warren RS, Donner DB, Mayo LD. Nutlin3 blocks vascular endothelial growth factor induction by preventing the interaction between hypoxia inducible factor 1alpha and Hdm2. Cancer Res. 2007;67:450–454. doi: 10.1158/0008-5472.CAN-06-2710. [DOI] [PubMed] [Google Scholar]
  • 60.Secchiero P, Corallini F, Gonelli A, et al. Antiangiogenic activity of the MDM2 antagonist nutlin-3. Circ Res. 2007;100:61–69. doi: 10.1161/01.RES.0000253975.76198.ff. [DOI] [PubMed] [Google Scholar]
  • 61.Teodoro JG, Evans SK, Green MR. Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome. J Mol Med (Berl) 2007;85:1175–1186. doi: 10.1007/s00109-007-0221-2. [DOI] [PubMed] [Google Scholar]
  • 62.Andreeff MKK, Yee K, et al. Results of the Phase 1 Trial of RG7112, a Small-Molecule MDM2 Antagonist, in Acute Leukemia; 54th ASH annual meeting; 2012; 2012. [Google Scholar]
  • 63.Yee KMG, Assouline S, et al. Phase 1b study of the MDM2 antagonist RG7112 in combination with 2 doses/schedules of cytarabine; 55th ASH annual meeting; 2013; 2013. [Google Scholar]
  • 64.Long J, Parkin B, Ouillette P, et al. Multiple distinct molecular mechanisms influence sensitivity and resistance to MDM2 inhibitors in adult acute myelogenous leukemia. Blood. 2010;116:71–80. doi: 10.1182/blood-2010-01-261628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yee KMG, Assouline S, et al. Phase 1b Study Of The MDM2 Antagonist RG7112 In Combination With 2 Doses/Schedules Of Cytarabine; 55th ASH annual meeting; 2013; 2013. [Google Scholar]
  • 66.Ray-Coquard I, Blay JY, Italiano A, et al. Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: an exploratory proof-of-mechanism study. Lancet Oncol. 2012;13:1133–1140. doi: 10.1016/S1470-2045(12)70474-6. [DOI] [PubMed] [Google Scholar]
  • 67.Carmena M, Earnshaw WC. The cellular geography of aurora kinases. Nat Rev Mol Cell Biol. 2003;4:842–854. doi: 10.1038/nrm1245. [DOI] [PubMed] [Google Scholar]
  • 68.Fu J, Bian M, Jiang Q, Zhang C. Roles of Aurora kinases in mitosis and tumorigenesis. Mol Cancer Res. 2007;5:1–10. doi: 10.1158/1541-7786.MCR-06-0208. [DOI] [PubMed] [Google Scholar]
  • 69.Boss DS, Beijnen JH, Schellens JH. Clinical experience with aurora kinase inhibitors: a review. Oncologist. 2009;14:780–793. doi: 10.1634/theoncologist.2009-0019. [DOI] [PubMed] [Google Scholar]
  • 70.Kollareddy M, Zheleva D, Dzubak P, Brahmkshatriya PS, Lepsik M, Hajduch M. Aurora kinase inhibitors: progress towards the clinic. Invest New Drugs. 2012;30:2411–2432. doi: 10.1007/s10637-012-9798-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Katsha A, Soutto M, Sehdev V, et al. Aurora kinase A promotes inflammation and tumorigenesis in mice and human gastric neoplasia. Gastroenterology. 2013;145:1312–1322. e1–e8. doi: 10.1053/j.gastro.2013.08.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sen S, Zhou H, White RA. A putative serine/threonine kinase encoding gene BTAK on chromosome 20q13 is amplified and overexpressed in human breast cancer cell lines. Oncogene. 1997;14:2195–2200. doi: 10.1038/sj.onc.1201065. [DOI] [PubMed] [Google Scholar]
  • 73.Marumoto T, Honda S, Hara T, et al. Aurora-A kinase maintains the fidelity of early and late mitotic events in HeLa cells. J Biol Chem. 2003;278:51786–51795. doi: 10.1074/jbc.M306275200. [DOI] [PubMed] [Google Scholar]
  • 74.Carmena M, Ruchaud S, Earnshaw WC. Making the Auroras glow: regulation of Aurora A and B kinase function by interacting proteins. Curr Opin Cell Biol. 2009;21:796–805. doi: 10.1016/j.ceb.2009.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Katayama H, Sasai K, Kawai H, et al. Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53. Nat Genet. 2004;36:55–62. doi: 10.1038/ng1279. [DOI] [PubMed] [Google Scholar]
  • 76.Liu Q, Kaneko S, Yang L, et al. Aurora-A abrogation of p53 DNA binding and transactivation activity by phosphorylation of serine 215. J Biol Chem. 2004;279:52175–52182. doi: 10.1074/jbc.M406802200. [DOI] [PubMed] [Google Scholar]
  • 77.Nishida N, Nagasaka T, Kashiwagi K, Boland CR, Goel A. High copy amplification of the Aurora-A gene is associated with chromosomal instability phenotype in human colorectal cancers. Cancer Biol Ther. 2007;6:525–533. doi: 10.4161/cbt.6.4.3817. [DOI] [PubMed] [Google Scholar]
  • 78.Reiter R, Gais P, Jutting U, et al. Aurora kinase A messenger RNA overexpression is correlated with tumor progression and shortened survival in head and neck squamous cell carcinoma. Clin Cancer Res. 2006;12:5136–5141. doi: 10.1158/1078-0432.CCR-05-1650. [DOI] [PubMed] [Google Scholar]
  • 79.Nadler Y, Camp RL, Schwartz C, Rimm DL, Kluger HM, Kluger Y. Expression of Aurora A (but not Aurora B) is predictive of survival in breast cancer. Clin Cancer Res. 2008;14:4455–4462. doi: 10.1158/1078-0432.CCR-07-5268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Goos JA, Coupe VM, Diosdado B, et al. Aurora kinase A (AURKA) expression in colorectal cancer liver metastasis is associated with poor prognosis. Br J Cancer. 2013;109:2445–2452. doi: 10.1038/bjc.2013.608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Yang J, Ikezoe T, Nishioka C, Nobumoto A, Udaka K, Yokoyama A. CD34(+)/CD38(−) acute myelogenous leukemia cells aberrantly express Aurora kinase A. Int J Cancer. 2013;133:2706–2719. doi: 10.1002/ijc.28277. [DOI] [PubMed] [Google Scholar]
  • 82.Lens SM, Voest EE, Medema RH. Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat Rev Cancer. 2010;10:825–841. doi: 10.1038/nrc2964. [DOI] [PubMed] [Google Scholar]
  • 83.Takeshita M, Koga T, Takayama K, et al. Aurora-B overexpression is correlated with aneuploidy and poor prognosis in non-small cell lung cancer. Lung Cancer. 2013;80:85–90. doi: 10.1016/j.lungcan.2012.12.018. [DOI] [PubMed] [Google Scholar]
  • 84.Hetland TE, Nymoen DA, Holth A, et al. Aurora B expression in metastatic effusions from advanced-stage ovarian serous carcinoma is predictive of intrinsic chemotherapy resistance. Hum Pathol. 2013;44:777–785. doi: 10.1016/j.humpath.2012.08.002. [DOI] [PubMed] [Google Scholar]
  • 85.Erpolat OP, Gocun PU, Akmansu M, Karakus E, Akyol G. High expression of nuclear survivin and Aurora B predicts poor overall survival in patients with head and neck squamous cell cancer. Strahlenther Onkol. 2012;188:248–254. doi: 10.1007/s00066-011-0042-7. [DOI] [PubMed] [Google Scholar]
  • 86.Pannone G, Hindi SA, Santoro A, et al. Aurora B expression as a prognostic indicator and possible therapeutic target in oral squamous cell carcinoma. Int J Immunopathol Pharmacol. 2011;24:79–88. doi: 10.1177/039463201102400110. [DOI] [PubMed] [Google Scholar]
  • 87.Garcia-Fernandez E, De Diego JI, Collantes-Bellido E, et al. Aurora B kinase expression in laryngeal squamous cell carcinoma and its prognostic implications. Histopathology. 2011;58:368–376. doi: 10.1111/j.1365-2559.2011.03757.x. [DOI] [PubMed] [Google Scholar]
  • 88.Vader G, Lens SM. The Aurora kinase family in cell division and cancer. Biochim Biophys Acta. 2008;1786:60–72. doi: 10.1016/j.bbcan.2008.07.003. [DOI] [PubMed] [Google Scholar]
  • 89.Khan J, Ezan F, Cremet JY, et al. Overexpression of active Aurora-C kinase results in cell transformation and tumour formation. PLoS One. 2011;6:e26512. doi: 10.1371/journal.pone.0026512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Foran J, Ravandi F, Wierda W, et al. A Phase I and Pharmacodynamic Study of AT9283, a Small-Molecule Inhibitor of Aurora Kinases in Patients With Relapsed/Refractory Leukemia or Myelofibrosis. Clin Lymphoma Myeloma Leuk. 2013 doi: 10.1016/j.clml.2013.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Moore AS, Blagg J, Linardopoulos S, Pearson AD. Aurora kinase inhibitors: novel small molecules with promising activity in acute myeloid and Philadelphia-positive leukemias. Leukemia. 2010;24:671–678. doi: 10.1038/leu.2010.15. [DOI] [PubMed] [Google Scholar]
  • 92.Schwartz GK, Carvajal RD, Midgley R, et al. Phase I study of barasertib (AZD1152), a selective inhibitor of Aurora B kinase, in patients with advanced solid tumors. Invest New Drugs. 2013;31:370–380. doi: 10.1007/s10637-012-9825-7. [DOI] [PubMed] [Google Scholar]
  • 93.Lowenberg B, Muus P, Ossenkoppele G, et al. Phase 1/2 study to assess the safety, efficacy, and pharmacokinetics of barasertib (AZD1152) in patients with advanced acute myeloid leukemia. Blood. 2011;118:6030–6036. doi: 10.1182/blood-2011-07-366930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kantarjian HM, Martinelli G, Jabbour EJ, et al. Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia. Cancer. 2013;119:2611–2619. doi: 10.1002/cncr.28113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Curry J, Angove H, Fazal L, et al. Aurora B kinase inhibition in mitosis: strategies for optimising the use of aurora kinase inhibitors such as AT9283. Cell Cycle. 2009;8:1921–1929. doi: 10.4161/cc.8.12.8741. [DOI] [PubMed] [Google Scholar]
  • 96.Manfredi MG, Ecsedy JA, Meetze KA, et al. Antitumor activity of MLN8054, an orally active small-molecule inhibitor of Aurora A kinase. Proc Natl Acad Sci U S A. 2007;104:4106–4111. doi: 10.1073/pnas.0608798104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Taga M, Hirooka E, Ouchi T. Essential roles of mTOR/Akt pathway in Aurora-A cell transformation. Int J Biol Sci. 2009;5:444–450. doi: 10.7150/ijbs.5.444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Scharer CD, Laycock N, Osunkoya AO, et al. Aurora kinase inhibitors synergize with paclitaxel to induce apoptosis in ovarian cancer cells. J Transl Med. 2008;6:79. doi: 10.1186/1479-5876-6-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.VanderPorten EC, Taverna P, Hogan JN, Ballinger MD, Flanagan WM, Fucini RV. The Aurora kinase inhibitor SNS-314 shows broad therapeutic potential with chemotherapeutics and synergy with microtubule-targeted agents in a colon carcinoma model. Mol Cancer Ther. 2009;8:930–939. doi: 10.1158/1535-7163.MCT-08-0754. [DOI] [PubMed] [Google Scholar]
  • 100.Chang F, Steelman LS, Lee JT, et al. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia. 2003;17:1263–1293. doi: 10.1038/sj.leu.2402945. [DOI] [PubMed] [Google Scholar]
  • 101.Radich JP, Kopecky KJ, Willman CL, et al. N-ras mutations in adult de novo acute myelogenous leukemia: prevalence and clinical significance. Blood. 1990;76:801–807. [PubMed] [Google Scholar]
  • 102.Coghlan DW, Morley AA, Matthews JP, Bishop JF. The incidence and prognostic significance of mutations in codon 13 of the N-ras gene in acute myeloid leukemia. Leukemia. 1994;8:1682–1687. [PubMed] [Google Scholar]
  • 103.Neubauer A, Dodge RK, George SL, et al. Prognostic importance of mutations in the ras proto-oncogenes in de novo acute myeloid leukemia. Blood. 1994;83:1603–1611. [PubMed] [Google Scholar]
  • 104.De Melo MB, Lorand-Metze I, Lima CS, Saad ST, Costa FF. N-ras gene point mutations in Brazilian acute myelogenous leukemia patients correlate with a poor prognosis. Leuk Lymphoma. 1997;24:309–317. doi: 10.3109/10428199709039018. [DOI] [PubMed] [Google Scholar]
  • 105.Kiyoi H, Naoe T, Nakano Y, et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood. 1999;93:3074–3080. [PubMed] [Google Scholar]
  • 106.Meshinchi S, Stirewalt DL, Alonzo TA, et al. Activating mutations of RTK/ras signal transduction pathway in pediatric acute myeloid leukemia. Blood. 2003;102:1474–1479. doi: 10.1182/blood-2003-01-0137. [DOI] [PubMed] [Google Scholar]
  • 107.Ritter M, Kim TD, Lisske P, Thiede C, Schaich M, Neubauer A. Prognostic significance of N-RAS and K-RAS mutations in 232 patients with acute myeloid leukemia. Haematologica. 2004;89:1397–1399. [PubMed] [Google Scholar]
  • 108.Illmer T, Thiede C, Fredersdorf A, et al. Activation of the RAS pathway is predictive for a chemosensitive phenotype of acute myelogenous leukemia blasts. Clin Cancer Res. 2005;11:3217–3224. doi: 10.1158/1078-0432.CCR-04-2232. [DOI] [PubMed] [Google Scholar]
  • 109.Gougopoulou DM, Kiaris H, Ergazaki M, Anagnostopoulos NI, Grigoraki V, Spandidos DA. Mutations and expression of the ras family genes in leukemias. Stem Cells. 1996;14:725–729. doi: 10.1002/stem.140725. [DOI] [PubMed] [Google Scholar]
  • 110.Jiang K, Coppola D, Crespo NC, et al. The phosphoinositide 3-OH kinase/AKT2 pathway as a critical target for farnesyltransferase inhibitor-induced apoptosis. Mol Cell Biol. 2000;20:139–148. doi: 10.1128/mcb.20.1.139-148.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Niessner H, Beck D, Sinnberg T, et al. The farnesyl transferase inhibitor lonafarnib inhibits mTOR signaling and enforces sorafenib-induced apoptosis in melanoma cells. J Invest Dermatol. 2011;131:468–479. doi: 10.1038/jid.2010.297. [DOI] [PubMed] [Google Scholar]
  • 112.Ding H, Hackbarth J, Schneider PA, et al. Cytotoxicity of farnesyltransferase inhibitors in lymphoid cells mediated by MAPK pathway inhibition and Bim up-regulation. Blood. 2011;118:4872–4881. doi: 10.1182/blood-2011-02-334870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Chun KH, Lee HY, Hassan K, Khuri F, Hong WK, Lotan R. Implication of protein kinase B/Akt and Bcl-2/Bcl-XL suppression by the farnesyl transferase inhibitor SCH66336 in apoptosis induction in squamous carcinoma cells. Cancer Res. 2003;63:4796–4800. [PubMed] [Google Scholar]
  • 114.Sepp-Lorenzino L, Ma Z, Rands E, et al. A peptidomimetic inhibitor of farnesyl:protein transferase blocks the anchorage-dependent and -independent growth of human tumor cell lines. Cancer Res. 1995;55:5302–5309. [PubMed] [Google Scholar]
  • 115.Feldkamp MM, Lau N, Guha A. Growth inhibition of astrocytoma cells by farnesyl transferase inhibitors is mediated by a combination of anti-proliferative, pro-apoptotic and anti-angiogenic effects. Oncogene. 1999;18:7514–7526. doi: 10.1038/sj.onc.1203105. [DOI] [PubMed] [Google Scholar]
  • 116.Gomez-Benito M, Marzo I, Anel A, Naval J. Farnesyltransferase inhibitor BMS-214662 induces apoptosis in myeloma cells through PUMA up-regulation, Bax and Bak activation, and Mcl-1 elimination. Mol Pharmacol. 2005;67:1991–1998. doi: 10.1124/mol.104.007021. [DOI] [PubMed] [Google Scholar]
  • 117.Ravoet C, Mineur P, Robin V, et al. Farnesyl transferase inhibitor (lonafarnib) in patients with myelodysplastic syndrome or secondary acute myeloid leukaemia: a phase II study. Ann Hematol. 2008;87:881–885. doi: 10.1007/s00277-008-0536-2. [DOI] [PubMed] [Google Scholar]
  • 118.Lancet JE, Gojo I, Gotlib J, et al. A phase 2 study of the farnesyltransferase inhibitor tipifarnib in poor-risk and elderly patients with previously untreated acute myelogenous leukemia. Blood. 2007;109:1387–1394. doi: 10.1182/blood-2006-04-014357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Harousseau JL, Martinelli G, Jedrzejczak WW, et al. A randomized phase 3 study of tipifarnib compared with best supportive care, including hydroxyurea, in the treatment of newly diagnosed acute myeloid leukemia in patients 70 years or older. Blood. 2009;114:1166–1173. doi: 10.1182/blood-2009-01-198093. [DOI] [PubMed] [Google Scholar]
  • 120.Burnett AK, Russell NH, Culligan D, et al. The addition of the farnesyl transferase inhibitor, tipifarnib, to low dose cytarabine does not improve outcome for older patients with AML. Br J Haematol. 2012;158:519–522. doi: 10.1111/j.1365-2141.2012.09165.x. [DOI] [PubMed] [Google Scholar]
  • 121.Jabbour E, Kantarjian H, Ravandi F, et al. A phase 1–2 study of a farnesyltransferase inhibitor, tipifarnib, combined with idarubicin and cytarabine for patients with newly diagnosed acute myeloid leukemia and high-risk myelodysplastic syndrome. Cancer. 2011;117:1236–1244. doi: 10.1002/cncr.25575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Brandwein JM, Leber BF, Howson-Jan K, et al. A phase I study of tipifarnib combined with conventional induction and consolidation therapy for previously untreated patients with acute myeloid leukemia aged 60 years and over. Leukemia. 2009;23:631–634. doi: 10.1038/leu.2008.341. [DOI] [PubMed] [Google Scholar]
  • 123.Whyte DB, Kirschmeier P, Hockenberry TN, et al. K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem. 1997;272:14459–14464. doi: 10.1074/jbc.272.22.14459. [DOI] [PubMed] [Google Scholar]
  • 124.Morgan MA, Wegner J, Aydilek E, Ganser A, Reuter CW. Synergistic cytotoxic effects in myeloid leukemia cells upon cotreatment with farnesyltransferase and geranylgeranyl transferase-I inhibitors. Leukemia. 2003;17:1508–1520. doi: 10.1038/sj.leu.2403022. [DOI] [PubMed] [Google Scholar]
  • 125.End DW, Smets G, Todd AV, et al. Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Res. 2001;61:131–137. [PubMed] [Google Scholar]
  • 126.Lubet RA, Christov K, You M, et al. Effects of the farnesyl transferase inhibitor R115777 (Zarnestra) on mammary carcinogenesis: prevention, therapy, and role of HaRas mutations. Mol Cancer Ther. 2006;5:1073–1078. doi: 10.1158/1535-7163.MCT-05-0398. [DOI] [PubMed] [Google Scholar]
  • 127.Sun J, Blaskovich MA, Knowles D, et al. Antitumor efficacy of a novel class of non-thiol-containing peptidomimetic inhibitors of farnesyltransferase and geranylgeranyltransferase I: combination therapy with the cytotoxic agents cisplatin, Taxol, and gemcitabine. Cancer Res. 1999;59:4919–4926. [PubMed] [Google Scholar]
  • 128.Lobell RB, Omer CA, Abrams MT, et al. Evaluation of farnesyl:protein transferase and geranylgeranyl:protein transferase inhibitor combinations in preclinical models. Cancer Res. 2001;61:8758–8768. [PubMed] [Google Scholar]
  • 129.Lobell RB, Liu D, Buser CA, et al. Preclinical and clinical pharmacodynamic assessment of L-778,123, a dual inhibitor of farnesyl:protein transferase and geranylgeranyl:protein transferase type-I. Mol Cancer Ther. 2002;1:747–758. [PubMed] [Google Scholar]
  • 130.Appels NM, Bolijn MJ, Chan K, et al. Phase I pharmacokinetic and pharmacodynamic study of the prenyl transferase inhibitor AZD3409 in patients with advanced cancer. Br J Cancer. 2008;98:1951–1958. doi: 10.1038/sj.bjc.6604402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Felsenfeld G, Groudine M. Controlling the double helix. Nature. 2003;421:448–453. doi: 10.1038/nature01411. [DOI] [PubMed] [Google Scholar]
  • 132.Holdenrieder S, Nagel D, Schalhorn A, et al. Clinical relevance of circulating nucleosomes in cancer. Ann N Y Acad Sci. 2008;1137:180–189. doi: 10.1196/annals.1448.012. [DOI] [PubMed] [Google Scholar]
  • 133.Quintas-Cardama A, Santos FP, Garcia-Manero G. Histone deacetylase inhibitors for the treatment of myelodysplastic syndrome and acute myeloid leukemia. Leukemia. 2011;25:226–235. doi: 10.1038/leu.2010.276. [DOI] [PubMed] [Google Scholar]
  • 134.Olsen EA, Kim YH, Kuzel TM, et al. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J Clin Oncol. 2007;25:3109–3115. doi: 10.1200/JCO.2006.10.2434. [DOI] [PubMed] [Google Scholar]
  • 135.Whittaker SJ, Demierre MF, Kim EJ, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol. 2010;28:4485–4491. doi: 10.1200/JCO.2010.28.9066. [DOI] [PubMed] [Google Scholar]
  • 136.Kuendgen A, Knipp S, Fox F, et al. Results of a phase 2 study of valproic acid alone or in combination with all-trans retinoic acid in 75 patients with myelodysplastic syndrome and relapsed or refractory acute myeloid leukemia. Ann Hematol. 2005;84(Suppl 1):61–66. doi: 10.1007/s00277-005-0026-8. [DOI] [PubMed] [Google Scholar]
  • 137.Garcia-Manero G, Yang H, Bueso-Ramos C, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 2008;111:1060–1066. doi: 10.1182/blood-2007-06-098061. [DOI] [PubMed] [Google Scholar]
  • 138.Bug G, Ritter M, Wassmann B, et al. Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia. Cancer. 2005;104:2717–2725. doi: 10.1002/cncr.21589. [DOI] [PubMed] [Google Scholar]
  • 139.Raffoux E, Chaibi P, Dombret H, Degos L. Valproic acid and all-trans retinoic acid for the treatment of elderly patients with acute myeloid leukemia. Haematologica. 2005;90:986–988. [PubMed] [Google Scholar]
  • 140.Khanim FL, Bradbury CA, Arrazi J, et al. Elevated FOSB-expression; a potential marker of valproate sensitivity in AML. Br J Haematol. 2009;144:332–341. doi: 10.1111/j.1365-2141.2008.07449.x. [DOI] [PubMed] [Google Scholar]
  • 141.Fredly H, Stapnes Bjornsen C, Gjertsen BT, Bruserud O. Combination of the histone deacetylase inhibitor valproic acid with oral hydroxyurea or 6-mercaptopurin can be safe and effective in patients with advanced acute myeloid leukaemia--a report of five cases. Hematology. 2010;15:338–343. doi: 10.1179/102453310X12647083620967. [DOI] [PubMed] [Google Scholar]
  • 142.Fredly H, Ersvaer E, Kittang AO, Tsykunova G, Gjertsen BT, Bruserud O. The combination of valproic acid, all-trans retinoic acid and low-dose cytarabine as disease-stabilizing treatment in acute myeloid leukemia. Clin Epigenetics. 2013;5:13. doi: 10.1186/1868-7083-5-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109:1114–1124. doi: 10.1002/cncr.22496. [DOI] [PubMed] [Google Scholar]
  • 144.Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30:2670–2677. doi: 10.1200/JCO.2011.38.9429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Fredly H, Gjertsen BT, Bruserud O. Histone deacetylase inhibition in the treatment of acute myeloid leukemia: the effects of valproic acid on leukemic cells, and the clinical and experimental evidence for combining valproic acid with other antileukemic agents. Clin Epigenetics. 2013;5:12. doi: 10.1186/1868-7083-5-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Byrd JC, Marcucci G, Parthun MR, et al. A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia. Blood. 2005;105:959–967. doi: 10.1182/blood-2004-05-1693. [DOI] [PubMed] [Google Scholar]
  • 147.Garcia-Manero G, Assouline S, Cortes J, et al. Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia. Blood. 2008;112:981–989. doi: 10.1182/blood-2007-10-115873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683–692. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bhaumik SR, Smith E, Shilatifard A. Covalent modifications of histones during development and disease pathogenesis. Nat Struct Mol Biol. 2007;14:1008–1016. doi: 10.1038/nsmb1337. [DOI] [PubMed] [Google Scholar]
  • 150.Prebet T, Sun Z, Figueroa ME, et al. Prolonged Administration of Azacitidine With or Without Entinostat for Myelodysplastic Syndrome and Acute Myeloid Leukemia With Myelodysplasia-Related Changes: Results of the US Leukemia Intergroup Trial E1905. J Clin Oncol. 2014;32:1242–1248. doi: 10.1200/JCO.2013.50.3102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Soriano AO, Yang H, Faderl S, et al. Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome. Blood. 2007;110:2302–2308. doi: 10.1182/blood-2007-03-078576. [DOI] [PubMed] [Google Scholar]
  • 152.Raffoux E, Cras A, Recher C, et al. Phase 2 clinical trial of 5-azacitidine, valproic acid, and all-trans retinoic acid in patients with high-risk acute myeloid leukemia or myelodysplastic syndrome. Oncotarget. 2010;1:34–42. doi: 10.18632/oncotarget.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Blum W, Klisovic RB, Hackanson B, et al. Phase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemia. J Clin Oncol. 2007;25:3884–3891. doi: 10.1200/JCO.2006.09.4169. [DOI] [PubMed] [Google Scholar]
  • 154.Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B, et al. Phase 1/2 study of the combination of 5-aza-2'-deoxycytidine with valproic acid in patients with leukemia. Blood. 2006;108:3271–3279. doi: 10.1182/blood-2006-03-009142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Garcia-Manero G, Tambaro FP, Bekele NB, et al. Phase II trial of vorinostat with idarubicin and cytarabine for patients with newly diagnosed acute myelogenous leukemia or myelodysplastic syndrome. J Clin Oncol. 2012;30:2204–2210. doi: 10.1200/JCO.2011.38.3265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Gojo I, Tan M, Fang HB, et al. Translational phase I trial of vorinostat (suberoylanilide hydroxamic acid) combined with cytarabine and etoposide in patients with relapsed, refractory, or high-risk acute myeloid leukemia. Clin Cancer Res. 2013;19:1838–1851. doi: 10.1158/1078-0432.CCR-12-3165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Giles F, Fischer T, Cortes J, et al. A phase I study of intravenous LBH589, a novel cinnamic hydroxamic acid analogue histone deacetylase inhibitor, in patients with refractory hematologic malignancies. Clin Cancer Res. 2006;12:4628–4635. doi: 10.1158/1078-0432.CCR-06-0511. [DOI] [PubMed] [Google Scholar]
  • 158.Tan P, Wei A, Mithraprabhu S, et al. Dual epigenetic targeting with panobinostat and azacitidine in acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood Cancer J. 2014;4:e170. doi: 10.1038/bcj.2013.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Novotny-Diermayr V, Hart S, Goh KC, et al. The oral HDAC inhibitor pracinostat (SB939) is efficacious and synergistic with the JAK2 inhibitor pacritinib (SB1518) in preclinical models of AML. Blood Cancer J. 2012;2:e69. doi: 10.1038/bcj.2012.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.McCormack E, Haaland I, Venas G, et al. Synergistic induction of p53 mediated apoptosis by valproic acid and nutlin-3 in acute myeloid leukemia. Leukemia. 2012;26:910–917. doi: 10.1038/leu.2011.315. [DOI] [PubMed] [Google Scholar]
  • 161.Yaseen A, Chen S, Hock S, et al. Resveratrol sensitizes acute myelogenous leukemia cells to histone deacetylase inhibitors through reactive oxygen species-mediated activation of the extrinsic apoptotic pathway. Mol Pharmacol. 2012;82:1030–1041. doi: 10.1124/mol.112.079624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Shao Y, Gao Z, Marks PA, Jiang X. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci U S A. 2004;101:18030–18035. doi: 10.1073/pnas.0408345102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Cai D, Wang Y, Ottmann OG, Barth PJ, Neubauer A, Burchert A. FLT3-ITD-, but not BCR/ABL-transformed cells require concurrent Akt/mTor blockage to undergo apoptosis after histone deacetylase inhibitor treatment. Blood. 2006;107:2094–2097. doi: 10.1182/blood-2005-08-3317. [DOI] [PubMed] [Google Scholar]
  • 164.Gammoh N, Lam D, Puente C, Ganley I, Marks PA, Jiang X. Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc Natl Acad Sci U S A. 2012;109:6561–6565. doi: 10.1073/pnas.1204429109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Degtyarev M, De Maziere A, Orr C, et al. Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol. 2008;183:101–116. doi: 10.1083/jcb.200801099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Koren I, Kimchi A. Cell biology. Promoting tumorigenesis by suppressing autophagy. Science. 2012;338:889–890. doi: 10.1126/science.1230577. [DOI] [PubMed] [Google Scholar]
  • 167.Wei Y, Kadia T, Tong W, et al. The combination of a histone deacetylase inhibitor with the Bcl-2 homology domain-3 mimetic GX15-070 has synergistic antileukemia activity by activating both apoptosis and autophagy. Clin Cancer Res. 2010;16:3923–3932. doi: 10.1158/1078-0432.CCR-10-0032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Chen CS, Weng SC, Tseng PH, Lin HP. Histone acetylation-independent effect of histone deacetylase inhibitors on Akt through the reshuffling of protein phosphatase 1 complexes. J Biol Chem. 2005;280:38879–38887. doi: 10.1074/jbc.M505733200. [DOI] [PubMed] [Google Scholar]
  • 169.Kawamata N, Chen J, Koeffler HP. Suberoylanilide hydroxamic acid (SAHA; vorinostat) suppresses translation of cyclin D1 in mantle cell lymphoma cells. Blood. 2007;110:2667–2673. doi: 10.1182/blood-2005-11-026344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Nishioka C, Ikezoe T, Yang J, Koeffler HP, Yokoyama A. Blockade of mTOR signaling potentiates the ability of histone deacetylase inhibitor to induce growth arrest and differentiation of acute myelogenous leukemia cells. Leukemia. 2008;22:2159–2168. doi: 10.1038/leu.2008.243. [DOI] [PubMed] [Google Scholar]
  • 171.Sandilya VGV, Sharma K, et al. Comination HDACi and mTOR inhibitor therapy in poor-risk de novo and refractory patients with AML; ASCO annual meeting; 2013; 2013. [Google Scholar]
  • 172.Bruserud O, Stapnes C, Ersvaer E, Gjertsen BT, Ryningen A. Histone deacetylase inhibitors in cancer treatment: a review of the clinical toxicity and the modulation of gene expression in cancer cell. Curr Pharm Biotechnol. 2007;8:388–400. doi: 10.2174/138920107783018417. [DOI] [PubMed] [Google Scholar]
  • 173.Laney JD, Hochstrasser M. Substrate targeting in the ubiquitin system. Cell. 1999;97:427–430. doi: 10.1016/s0092-8674(00)80752-7. [DOI] [PubMed] [Google Scholar]
  • 174.Orlowski M, Wilk S. Catalytic activities of the 20 S proteasome, a multicatalytic proteinase complex. Arch Biochem Biophys. 2000;383:1–16. doi: 10.1006/abbi.2000.2036. [DOI] [PubMed] [Google Scholar]
  • 175.Pagano M, Tam SW, Theodoras AM, et al. Role of the ubiquitin-proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science. 1995;269:682–685. doi: 10.1126/science.7624798. [DOI] [PubMed] [Google Scholar]
  • 176.Maki CG, Huibregtse JM, Howley PM. In vivo ubiquitination and proteasome-mediated degradation of p53(1) Cancer Res. 1996;56:2649–2654. [PubMed] [Google Scholar]
  • 177.Mani A, Gelmann EP. The ubiquitin-proteasome pathway and its role in cancer. J Clin Oncol. 2005;23:4776–4789. doi: 10.1200/JCO.2005.05.081. [DOI] [PubMed] [Google Scholar]
  • 178.Adams J. The development of proteasome inhibitors as anticancer drugs. Cancer Cell. 2004;5:417–421. doi: 10.1016/s1535-6108(04)00120-5. [DOI] [PubMed] [Google Scholar]
  • 179.Mitsiades CS, Mitsiades N, Hideshima T, Richardson PG, Anderson KC. Proteasome inhibition as a new therapeutic principle in hematological malignancies. Curr Drug Targets. 2006;7:1341–1347. doi: 10.2174/138945006778559247. [DOI] [PubMed] [Google Scholar]
  • 180.Kane RC, Farrell AT, Sridhara R, Pazdur R. United States Food and Drug Administration approval summary: bortezomib for the treatment of progressive multiple myeloma after one prior therapy. Clin Cancer Res. 2006;12:2955–2960. doi: 10.1158/1078-0432.CCR-06-0170. [DOI] [PubMed] [Google Scholar]
  • 181.Kane RC, Dagher R, Farrell A, et al. Bortezomib for the treatment of mantle cell lymphoma. Clin Cancer Res. 2007;13:5291–5294. doi: 10.1158/1078-0432.CCR-07-0871. [DOI] [PubMed] [Google Scholar]
  • 182.Guzman ML, Swiderski CF, Howard DS, et al. Preferential induction of apoptosis for primary human leukemic stem cells. Proc Natl Acad Sci U S A. 2002;99:16220–16225. doi: 10.1073/pnas.252462599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Koschny R, Holland H, Sykora J, et al. Bortezomib sensitizes primary human astrocytoma cells of WHO grades I to IV for tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Clin Cancer Res. 2007;13:3403–3412. doi: 10.1158/1078-0432.CCR-07-0251. [DOI] [PubMed] [Google Scholar]
  • 184.Dijk M, Murphy E, Morrell R, et al. The Proteasome Inhibitor Bortezomib Sensitizes AML with Myelomonocytic Differentiation to TRAIL Mediated Apoptosis. Cancers (Basel) 2011;3:1329–1350. doi: 10.3390/cancers3011329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lu G, Punj V, Chaudhary PM. Proteasome inhibitor Bortezomib induces cell cycle arrest and apoptosis in cell lines derived from Ewing's sarcoma family of tumors and synergizes with TRAIL. Cancer Biol Ther. 2008;7:603–608. doi: 10.4161/cbt.7.4.5564. [DOI] [PubMed] [Google Scholar]
  • 186.Cortes J, Thomas D, Koller C, et al. Phase I study of bortezomib in refractory or relapsed acute leukemias. Clin Cancer Res. 2004;10:3371–3376. doi: 10.1158/1078-0432.CCR-03-0508. [DOI] [PubMed] [Google Scholar]
  • 187.Sarlo C, Buccisano F, Maurillo L, et al. Phase II Study of Bortezomib as a Single Agent in Patients with Previously Untreated or Relapsed/Refractory Acute Myeloid Leukemia Ineligible for Intensive Therapy. Leuk Res Treatment. 2013;2013:705714. doi: 10.1155/2013/705714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Attar EC, De Angelo DJ, Supko JG, et al. Phase I and pharmacokinetic study of bortezomib in combination with idarubicin and cytarabine in patients with acute myelogenous leukemia. Clin Cancer Res. 2008;14:1446–1454. doi: 10.1158/1078-0432.CCR-07-4626. [DOI] [PubMed] [Google Scholar]
  • 189.Attar EC, Johnson JL, Amrein PC, et al. Bortezomib added to daunorubicin and cytarabine during induction therapy and to intermediate-dose cytarabine for consolidation in patients with previously untreated acute myeloid leukemia age 60 to 75 years: CALGB (Alliance) study 10502. J Clin Oncol. 2013;31:923–929. doi: 10.1200/JCO.2012.45.2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Blum W, Schwind S, Tarighat SS, et al. Clinical and pharmacodynamic activity of bortezomib and decitabine in acute myeloid leukemia. Blood. 2012;119:6025–6031. doi: 10.1182/blood-2012-03-413898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Hainsworth JD, Spigel DR, Barton J, et al. Weekly treatment with bortezomib for patients with recurrent or refractory multiple myeloma: a phase 2 trial of the Minnie Pearl Cancer Research Network. Cancer. 2008;113:765–771. doi: 10.1002/cncr.23606. [DOI] [PubMed] [Google Scholar]
  • 192.Moreau P, Pylypenko H, Grosicki S, et al. Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study. Lancet Oncol. 2011;12:431–440. doi: 10.1016/S1470-2045(11)70081-X. [DOI] [PubMed] [Google Scholar]
  • 193.Dai Y, Chen S, Wang L, et al. Bortezomib interacts synergistically with belinostat in human acute myeloid leukaemia and acute lymphoblastic leukaemia cells in association with perturbations in NF-kappaB and Bim. Br J Haematol. 2011;153:222–235. doi: 10.1111/j.1365-2141.2011.08591.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Nie D, Huang K, Yin S, et al. Synergistic/additive interaction of valproic acid with bortezomib on proliferation and apoptosis of acute myeloid leukemia cells. Leuk Lymphoma. 2012;53:2487–2495. doi: 10.3109/10428194.2012.698273. [DOI] [PubMed] [Google Scholar]
  • 195.Holkova BBp, Tombes M, et al. Phase I trial of Belinostat and Bortezomib in patients with relapsed or refractory acute leukemia, myelodysplastic syndrome, or chronic myelogenous leukemia in blast crisis - one year update; 54th ASH anuual meeting; 2012; 2012. [Google Scholar]
  • 196.Peled A, Petit I, Kollet O, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845–848. doi: 10.1126/science.283.5403.845. [DOI] [PubMed] [Google Scholar]
  • 197.Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005;201:1307–1318. doi: 10.1084/jem.20041385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25:977–988. doi: 10.1016/j.immuni.2006.10.016. [DOI] [PubMed] [Google Scholar]
  • 199.Tavor S, Petit I, Porozov S, et al. CXCR4 regulates migration and development of human acute myelogenous leukemia stem cells in transplanted NOD/SCID mice. Cancer Res. 2004;64:2817–2824. doi: 10.1158/0008-5472.can-03-3693. [DOI] [PubMed] [Google Scholar]
  • 200.Teicher BA, Fricker SP. CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin Cancer Res. 2010;16:2927–2931. doi: 10.1158/1078-0432.CCR-09-2329. [DOI] [PubMed] [Google Scholar]
  • 201.Spoo AC, Lubbert M, Wierda WG, Burger JA. CXCR4 is a prognostic marker in acute myelogenous leukemia. Blood. 2007;109:786–791. doi: 10.1182/blood-2006-05-024844. [DOI] [PubMed] [Google Scholar]
  • 202.Rombouts EJ, Pavic B, Lowenberg B, Ploemacher RE. Relation between CXCR-4 expression, Flt3 mutations, and unfavorable prognosis of adult acute myeloid leukemia. Blood. 2004;104:550–557. doi: 10.1182/blood-2004-02-0566. [DOI] [PubMed] [Google Scholar]
  • 203.Nervi B, Ramirez P, Rettig MP, et al. Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100. Blood. 2009;113:6206–6214. doi: 10.1182/blood-2008-06-162123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Uy GL, Rettig MP, Motabi IH, et al. A phase 1/2 study of chemosensitization with the CXCR4 antagonist plerixafor in relapsed or refractory acute myeloid leukemia. Blood. 2012;119:3917–3924. doi: 10.1182/blood-2011-10-383406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Zeng Z, Shi YX, Samudio IJ, et al. Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood. 2009;113:6215–6224. doi: 10.1182/blood-2008-05-158311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Andreeff MZZ, Kelly AM, et al. Mobilization and elmination of FLT3-ITD+ acute myelogenous leukemia (AML) stem/progenitor cells by plerixafor/G-CSF/Sorafinib: results from a phase I trial in relapsed/refractory AML patients; 54th ASH annual meeting; 2012; 2012. [Google Scholar]
  • 207.Sreedhar AS, Kalmar E, Csermely P, Shen YF. Hsp90 isoforms: functions, expression and clinical importance. FEBS Lett. 2004;562:11–15. doi: 10.1016/s0014-5793(04)00229-7. [DOI] [PubMed] [Google Scholar]
  • 208.Stancato LF, Chow YH, Hutchison KA, Perdew GH, Jove R, Pratt WB. Raf exists in a native heterocomplex with hsp90 and p50 that can be reconstituted in a cell-free system. J Biol Chem. 1993;268:21711–21716. [PubMed] [Google Scholar]
  • 209.Sato S, Fujita N, Tsuruo T. Modulation of Akt kinase activity by binding to Hsp90. Proc Natl Acad Sci U S A. 2000;97:10832–10837. doi: 10.1073/pnas.170276797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Pratt WB, Toft DO. Regulation of signaling protein function and trafficking by the hsp90/hsp70-based chaperone machinery. Exp Biol Med (Maywood) 2003;228:111–133. doi: 10.1177/153537020322800201. [DOI] [PubMed] [Google Scholar]
  • 211.Fujita N, Sato S, Ishida A, Tsuruo T. Involvement of Hsp90 in signaling and stability of 3-phosphoinositide-dependent kinase-1. J Biol Chem. 2002;277:10346–10353. doi: 10.1074/jbc.M106736200. [DOI] [PubMed] [Google Scholar]
  • 212.Whitesell L, Bagatell R, Falsey R. The stress response: implications for the clinical development of hsp90 inhibitors. Curr Cancer Drug Targets. 2003;3:349–358. doi: 10.2174/1568009033481787. [DOI] [PubMed] [Google Scholar]
  • 213.Workman P. Overview: translating Hsp90 biology into Hsp90 drugs. Curr Cancer Drug Targets. 2003;3:297–300. doi: 10.2174/1568009033481868. [DOI] [PubMed] [Google Scholar]
  • 214.Lauria A, Ippolito M, Almerico AM. Inside the Hsp90 inhibitors binding mode through induced fit docking. J Mol Graph Model. 2009;27:712–722. doi: 10.1016/j.jmgm.2008.11.004. [DOI] [PubMed] [Google Scholar]
  • 215.Fumo G, Akin C, Metcalfe DD, Neckers L. 17-Allylamino-17-demethoxygeldanamycin (17-AAG) is effective in down-regulating mutated, constitutively activated KIT protein in human mast cells. Blood. 2004;103:1078–1084. doi: 10.1182/blood-2003-07-2477. [DOI] [PubMed] [Google Scholar]
  • 216.Xu W, Mimnaugh E, Rosser MF, et al. Sensitivity of mature Erbb2 to geldanamycin is conferred by its kinase domain and is mediated by the chaperone protein Hsp90. J Biol Chem. 2001;276:3702–3708. doi: 10.1074/jbc.M006864200. [DOI] [PubMed] [Google Scholar]
  • 217.Minami Y, Kiyoi H, Yamamoto Y, et al. Selective apoptosis of tandemly duplicated FLT3-transformed leukemia cells by Hsp90 inhibitors. Leukemia. 2002;16:1535–1540. doi: 10.1038/sj.leu.2402558. [DOI] [PubMed] [Google Scholar]
  • 218.An WG, Schulte TW, Neckers LM. The heat shock protein 90 antagonist geldanamycin alters chaperone association with p210bcr-abl and v-src proteins before their degradation by the proteasome. Cell Growth Differ. 2000;11:355–360. [PubMed] [Google Scholar]
  • 219.Castro JE, Prada CE, Loria O, et al. ZAP-70 is a novel conditional heat shock protein 90 (Hsp90) client: inhibition of Hsp90 leads to ZAP-70 degradation, apoptosis, and impaired signaling in chronic lymphocytic leukemia. Blood. 2005;106:2506–2512. doi: 10.1182/blood-2005-03-1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Bonvini P, Gastaldi T, Falini B, Rosolen A. Nucleophosmin-anaplastic lymphoma kinase (NPM-ALK), a novel Hsp90-client tyrosine kinase: down-regulation of NPM-ALK expression and tyrosine phosphorylation in ALK(+) CD30(+) lymphoma cells by the Hsp90 antagonist 17-allylamino,17-demethoxygeldanamycin. Cancer Res. 2002;62:1559–1566. [PubMed] [Google Scholar]
  • 221.Schulte TW, Blagosklonny MV, Ingui C, Neckers L. Disruption of the Raf-1-Hsp90 molecular complex results in destabilization of Raf-1 and loss of Raf-1-Ras association. J Biol Chem. 1995;270:24585–24588. doi: 10.1074/jbc.270.41.24585. [DOI] [PubMed] [Google Scholar]
  • 222.Blagosklonny MV, Toretsky J, Neckers L. Geldanamycin selectively destabilizes and conformationally alters mutated p53. Oncogene. 1995;11:933–939. [PubMed] [Google Scholar]
  • 223.Ochel HJ, Gademann G. Heat-shock protein 90: potential involvement in the pathogenesis of malignancy and pharmacological intervention. Onkologie. 2002;25:466–473. doi: 10.1159/000067442. [DOI] [PubMed] [Google Scholar]
  • 224.Yufu Y, Nishimura J, Nawata H. High constitutive expression of heat shock protein 90 alpha in human acute leukemia cells. Leuk Res. 1992;16:597–605. doi: 10.1016/0145-2126(92)90008-u. [DOI] [PubMed] [Google Scholar]
  • 225.Thomas X, Campos L, Mounier C, et al. Expression of heat-shock proteins is associated with major adverse prognostic factors in acute myeloid leukemia. Leuk Res. 2005;29:1049–1058. doi: 10.1016/j.leukres.2005.02.010. [DOI] [PubMed] [Google Scholar]
  • 226.Flandrin P, Guyotat D, Duval A, et al. Significance of heat-shock protein (HSP) 90 expression in acute myeloid leukemia cells. Cell Stress Chaperones. 2008;13:357–364. doi: 10.1007/s12192-008-0035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Kamal A, Thao L, Sensintaffar J, et al. A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature. 2003;425:407–410. doi: 10.1038/nature01913. [DOI] [PubMed] [Google Scholar]
  • 228.Reikvam H, Hatfield KJ, Ersvaer E, et al. Expression profile of heat shock proteins in acute myeloid leukaemia patients reveals a distinct signature strongly associated with FLT3 mutation status--consequences and potentials for pharmacological intervention. Br J Haematol. 2012;156:468–480. doi: 10.1111/j.1365-2141.2011.08960.x. [DOI] [PubMed] [Google Scholar]
  • 229.Nimmanapalli R, O'Bryan E, Kuhn D, Yamaguchi H, Wang HG, Bhalla KN. Regulation of 17-AAG-induced apoptosis: role of Bcl-2, Bcl-XL, and Bax downstream of 17-AAG-mediated down-regulation of Akt, Raf-1, and Src kinases. Blood. 2003;102:269–275. doi: 10.1182/blood-2002-12-3718. [DOI] [PubMed] [Google Scholar]
  • 230.Napper JM, Sollars VE. 17-N-Allylamino-17-demethoxygeldanamycin induces a diverse response in human acute myelogenous cells. Leuk Res. 2010;34:1493–1500. doi: 10.1016/j.leukres.2010.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Walker AR, Klisovic R, Johnston JS, et al. Pharmacokinetics and dose escalation of the heat shock protein inhibitor 17-allyamino-17-demethoxygeldanamycin in combination with bortezomib in relapsed or refractory acute myeloid leukemia. Leuk Lymphoma. 2013;54:1996–2002. doi: 10.3109/10428194.2012.760733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Richardson PG, Chanan-Khan AA, Lonial S, et al. Tanespimycin and bortezomib combination treatment in patients with relapsed or relapsed and refractory multiple myeloma: results of a phase 1/2 study. Br J Haematol. 2011;153:729–740. doi: 10.1111/j.1365-2141.2011.08664.x. [DOI] [PubMed] [Google Scholar]
  • 233.Richardson PG, Badros AZ, Jagannath S, et al. Tanespimycin with bortezomib: activity in relapsed/refractory patients with multiple myeloma. Br J Haematol. 2010;150:428–437. doi: 10.1111/j.1365-2141.2010.08264.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Lancet JE, Gojo I, Burton M, et al. Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia. Leukemia. 2010;24:699–705. doi: 10.1038/leu.2009.292. [DOI] [PubMed] [Google Scholar]
  • 235.Taldone T, Gozman A, Maharaj R, Chiosis G. Targeting Hsp90: small-molecule inhibitors and their clinical development. Curr Opin Pharmacol. 2008;8:370–374. doi: 10.1016/j.coph.2008.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Mesa RA, Loegering D, Powell HL, et al. Heat shock protein 90 inhibition sensitizes acute myelogenous leukemia cells to cytarabine. Blood. 2005;106:318–327. doi: 10.1182/blood-2004-09-3523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Walsby EJ, Lazenby M, Pepper CJ, Knapper S, Burnett AK. The HSP90 inhibitor NVP-AUY922-AG inhibits the PI3K and IKK signalling pathways and synergizes with cytarabine in acute myeloid leukaemia cells. Br J Haematol. 2013;161:57–67. doi: 10.1111/bjh.12215. [DOI] [PubMed] [Google Scholar]
  • 238.Rao R, Fiskus W, Yang Y, et al. HDAC6 inhibition enhances 17-AAG--mediated abrogation of hsp90 chaperone function in human leukemia cells. Blood. 2008;112:1886–1893. doi: 10.1182/blood-2008-03-143644. [DOI] [PubMed] [Google Scholar]
  • 239.George P, Bali P, Annavarapu S, et al. Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3. Blood. 2005;105:1768–1776. doi: 10.1182/blood-2004-09-3413. [DOI] [PubMed] [Google Scholar]
  • 240.Piper WPMH. Mechanism of resistance to Hsp90 inhibitor drugs: a complex mosiac emerges. Pharmaceuticals. 2011;4:1400–1422. doi: 10.3390/ph4111400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Reikvam H, Nepstad I, Sulen A, Gjertsen BT, Hatfield KJ, Bruserud O. Increased antileukemic effects in human acute myeloid leukemia by combining HSP70 and HSP90 inhibitors. Expert Opin Investig Drugs. 2013;22:551–563. doi: 10.1517/13543784.2013.791280. [DOI] [PubMed] [Google Scholar]
  • 242.Rosnet O, Buhring HJ, deLapeyriere O, et al. Expression and signal transduction of the FLT3 tyrosine kinase receptor. Acta Haematol. 1996;95:218–223. doi: 10.1159/000203881. [DOI] [PubMed] [Google Scholar]
  • 243.Zhang S, Fukuda S, Lee Y, et al. Essential role of signal transducer and activator of transcription (Stat)5a but not Stat5b for Flt3-dependent signaling. J Exp Med. 2000;192:719–728. doi: 10.1084/jem.192.5.719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Frohling S, Schlenk RF, Breitruck J, et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm. Blood. 2002;100:4372–4380. doi: 10.1182/blood-2002-05-1440. [DOI] [PubMed] [Google Scholar]
  • 245.Pratcorona M, Brunet S, Nomdedeu J, et al. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood. 2013;121:2734–2738. doi: 10.1182/blood-2012-06-431122. [DOI] [PubMed] [Google Scholar]
  • 246.Stirewalt DL, Kopecky KJ, Meshinchi S, et al. Size of FLT3 internal tandem duplication has prognostic significance in patients with acute myeloid leukemia. Blood. 2006;107:3724–3726. doi: 10.1182/blood-2005-08-3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Kusec R, Jaksic O, Ostojic S, Kardum-Skelin I, Vrhovac R, Jaksic B. More on prognostic significance of FLT3/ITD size in acute myeloid leukemia (AML) Blood. 2006;108:405–406. doi: 10.1182/blood-2005-12-5128. author reply 6. [DOI] [PubMed] [Google Scholar]
  • 248.Abu-Duhier FM, Goodeve AC, Wilson GA, Care RS, Peake IR, Reilly JT. Identification of novel FLT-3 Asp835 mutations in adult acute myeloid leukaemia. Br J Haematol. 2001;113:983–988. doi: 10.1046/j.1365-2141.2001.02850.x. [DOI] [PubMed] [Google Scholar]
  • 249.Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97:2434–2439. doi: 10.1182/blood.v97.8.2434. [DOI] [PubMed] [Google Scholar]
  • 250.Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99:4326–4335. doi: 10.1182/blood.v99.12.4326. [DOI] [PubMed] [Google Scholar]
  • 251.Fiedler W, Mesters R, Tinnefeld H, et al. A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia. Blood. 2003;102:2763–2767. doi: 10.1182/blood-2002-10-2998. [DOI] [PubMed] [Google Scholar]
  • 252.Serve H, Krug U, Wagner R, et al. Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. J Clin Oncol. 2013;31:3110–3118. doi: 10.1200/JCO.2012.46.4990. [DOI] [PubMed] [Google Scholar]
  • 253.Metzelder SK, Schroeder T, Finck A, et al. High activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained responses. Leukemia. 2012;26:2353–2359. doi: 10.1038/leu.2012.105. [DOI] [PubMed] [Google Scholar]
  • 254.Sato T, Yang X, Knapper S, et al. FLT3 ligand impedes the efficacy of FLT3 inhibitors in vitro and in vivo. Blood. 2011;117:3286–3293. doi: 10.1182/blood-2010-01-266742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Ravandi F, Alattar ML, Grunwald MR, et al. Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood. 2013;121:4655–4662. doi: 10.1182/blood-2013-01-480228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Fiedler W, Serve H, Dohner H, et al. A phase 1 study of SU11248 in the treatment of patients with refractory or resistant acute myeloid leukemia (AML) or not amenable to conventional therapy for the disease. Blood. 2005;105:986–993. doi: 10.1182/blood-2004-05-1846. [DOI] [PubMed] [Google Scholar]
  • 257.Fischer T, Stone RM, Deangelo DJ, et al. Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin Oncol. 2010;28:4339–4345. doi: 10.1200/JCO.2010.28.9678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Cortes JE, Kantarjian H, Foran JM, et al. Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal tandem duplication status. J Clin Oncol. 2013;31:3681–3687. doi: 10.1200/JCO.2013.48.8783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Levis JMAE, Dombret H, et al. Final results of a phase 2 open-label, monotherapy efficacy and safety study of Quizartinib (AC220) in patients with FLT3-ITD positive or negative relapsed/refractory acute myeloid leukemia after second-line chemotherapy or hematopoietic stem cell transplantation; 54th ASH annual meeting; 2012; 2012. [Google Scholar]
  • 260.Cortes JPA, Dombret H, et al. Response rate and bridging to hematopoietic stem cell transplantation (HSCT) with quizartinib (AC220) in patients with FLT3-ITD positive or negative relapsed/refractory AML after second-line chemotherapy or previous bone marrow transplant. ASCO Annual Meeting; 2013. 2013 [Google Scholar]
  • 261.Pratz KW, Sato T, Murphy KM, Stine A, Rajkhowa T, Levis M. FLT3-mutant allelic burden and clinical status are predictive of response to FLT3 inhibitors in AML. Blood. 2010;115:1425–1432. doi: 10.1182/blood-2009-09-242859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Grundler R, Thiede C, Miething C, Steudel C, Peschel C, Duyster J. Sensitivity toward tyrosine kinase inhibitors varies between different activating mutations of the FLT3 receptor. Blood. 2003;102:646–651. doi: 10.1182/blood-2002-11-3441. [DOI] [PubMed] [Google Scholar]
  • 263.Clark JJ, Cools J, Curley DP, et al. Variable sensitivity of FLT3 activation loop mutations to the small molecule tyrosine kinase inhibitor MLN518. Blood. 2004;104:2867–2872. doi: 10.1182/blood-2003-12-4446. [DOI] [PubMed] [Google Scholar]
  • 264.Smith CC, Wang Q, Chin CS, et al. Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature. 2012;485:260–263. doi: 10.1038/nature11016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Parmar A, Marz S, Rushton S, et al. Stromal niche cells protect early leukemic FLT3-ITD+ progenitor cells against first-generation FLT3 tyrosine kinase inhibitors. Cancer Res. 2011;71:4696–4706. doi: 10.1158/0008-5472.CAN-10-4136. [DOI] [PubMed] [Google Scholar]
  • 266.Weisberg E, Liu Q, Zhang X, et al. Selective Akt inhibitors synergize with tyrosine kinase inhibitors and effectively override stroma-associated cytoprotection of mutant FLT3-positive AML cells. PLoS One. 2013;8:e56473. doi: 10.1371/journal.pone.0056473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Weisberg E, Liu Q, Nelson E, et al. Using combination therapy to override stromal-mediated chemoresistance in mutant FLT3-positive AML: synergism between FLT3 inhibitors, dasatinib/multi-targeted inhibitors and JAK inhibitors. Leukemia. 2012;26:2233–2244. doi: 10.1038/leu.2012.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Linnekin D. Early signaling pathways activated by c-Kit in hematopoietic cells. Int J Biochem Cell Biol. 1999;31:1053–1074. doi: 10.1016/s1357-2725(99)00078-3. [DOI] [PubMed] [Google Scholar]
  • 269.Lennartsson J, Ronnstrand L. The stem cell factor receptor/c-Kit as a drug target in cancer. Curr Cancer Drug Targets. 2006;6:65–75. doi: 10.2174/156800906775471725. [DOI] [PubMed] [Google Scholar]
  • 270.Broudy VC. Stem cell factor and hematopoiesis. Blood. 1997;90:1345–1364. [PubMed] [Google Scholar]
  • 271.Ikeda H, Kanakura Y, Tamaki T, et al. Expression and functional role of the proto-oncogene c-kit in acute myeloblastic leukemia cells. Blood. 1991;78:2962–2968. [PubMed] [Google Scholar]
  • 272.Nanri T, Matsuno N, Kawakita T, et al. Mutations in the receptor tyrosine kinase pathway are associated with clinical outcome in patients with acute myeloblastic leukemia harboring t(8;21)(q22;q22) Leukemia. 2005;19:1361–1366. doi: 10.1038/sj.leu.2403803. [DOI] [PubMed] [Google Scholar]
  • 273.Park SH, Chi HS, Min SK, Park BG, Jang S, Park CJ. Prognostic impact of c-KIT mutations in core binding factor acute myeloid leukemia. Leuk Res. 2011;35:1376–1383. doi: 10.1016/j.leukres.2011.06.003. [DOI] [PubMed] [Google Scholar]
  • 274.Ashman LK, Ferrao P, Cole SR, Cambareri AC. Effects of mutant c-Kit in early myeloid cells. Leuk Lymphoma. 1999;34:451–461. doi: 10.3109/10428199909058472. [DOI] [PubMed] [Google Scholar]
  • 275.Ning ZQ, Li J, Arceci RJ. Activating mutations of c-kit at codon 816 confer drug resistance in human leukemia cells. Leuk Lymphoma. 2001;41:513–522. doi: 10.3109/10428190109060342. [DOI] [PubMed] [Google Scholar]
  • 276.Schnittger S, Kohl TM, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107:1791–1799. doi: 10.1182/blood-2005-04-1466. [DOI] [PubMed] [Google Scholar]
  • 277.Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med. 1996;2:561–566. doi: 10.1038/nm0596-561. [DOI] [PubMed] [Google Scholar]
  • 278.Kindler T, Breitenbuecher F, Marx A, et al. Efficacy and safety of imatinib in adult patients with c-kit-positive acute myeloid leukemia. Blood. 2004;103:3644–3654. doi: 10.1182/blood-2003-06-2071. [DOI] [PubMed] [Google Scholar]
  • 279.Cortes J, Giles F, O'Brien S, et al. Results of imatinib mesylate therapy in patients with refractory or recurrent acute myeloid leukemia, high-risk myelodysplastic syndrome, and myeloproliferative disorders. Cancer. 2003;97:2760–2766. doi: 10.1002/cncr.11416. [DOI] [PubMed] [Google Scholar]
  • 280.Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott KA, Zigler AJ. Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor. Blood. 2000;96:925–932. [PubMed] [Google Scholar]
  • 281.Malagola M, Martinelli G, Rondoni M, et al. Imatinib mesylate in the treatment of c-kit-positive acute myeloid leukemia: is this the real target? Blood. 2005;105:904. doi: 10.1182/blood-2004-08-3088. author reply 5. [DOI] [PubMed] [Google Scholar]
  • 282.Frost MJ, Ferrao PT, Hughes TP, Ashman LK. Juxtamembrane mutant V560GKit is more sensitive to Imatinib (STI571) compared with wild-type c-kit whereas the kinase domain mutant D816VKit is resistant. Mol Cancer Ther. 2002;1:1115–1124. [PubMed] [Google Scholar]
  • 283.Brandwein JM, Hedley DW, Chow S, et al. A phase I/II study of imatinib plus reinduction therapy for c-kit-positive relapsed/refractory acute myeloid leukemia: inhibition of Akt activation correlates with complete response. Leukemia. 2011;25:945–952. doi: 10.1038/leu.2011.34. [DOI] [PubMed] [Google Scholar]
  • 284.Dos Santos C, McDonald T, Ho YW, et al. The Src and c-Kit kinase inhibitor dasatinib enhances p53-mediated targeting of human acute myeloid leukemia stem cells by chemotherapeutic agents. Blood. 2013;122:1900–1913. doi: 10.1182/blood-2012-11-466425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Leonard WJ, O'Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. doi: 10.1146/annurev.immunol.16.1.293. [DOI] [PubMed] [Google Scholar]
  • 286.Darnell JE, Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264:1415–1421. doi: 10.1126/science.8197455. [DOI] [PubMed] [Google Scholar]
  • 287.Levy DE, Darnell JE., Jr Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol. 2002;3:651–662. doi: 10.1038/nrm909. [DOI] [PubMed] [Google Scholar]
  • 288.Gouilleux-Gruart V, Gouilleux F, Desaint C, et al. STAT-related transcription factors are constitutively activated in peripheral blood cells from acute leukemia patients. Blood. 1996;87:1692–1697. [PubMed] [Google Scholar]
  • 289.Spiekermann K, Biethahn S, Wilde S, Hiddemann W, Alves F. Constitutive activation of STAT transcription factors in acute myelogenous leukemia. Eur J Haematol. 2001;67:63–71. [PubMed] [Google Scholar]
  • 290.Steensma DP, McClure RF, Karp JE, et al. JAK2 V617F is a rare finding in de novo acute myeloid leukemia, but STAT3 activation is common and remains unexplained. Leukemia. 2006;20:971–978. doi: 10.1038/sj.leu.2404206. [DOI] [PubMed] [Google Scholar]
  • 291.Ikezoe T, Kojima S, Furihata M, et al. Expression of p-JAK2 predicts clinical outcome and is a potential molecular target of acute myelogenous leukemia. Int J Cancer. 2011;129:2512–2521. doi: 10.1002/ijc.25910. [DOI] [PubMed] [Google Scholar]
  • 292.Lee JW, Kim YG, Soung YH, et al. The JAK2 V617F mutation in de novo acute myelogenous leukemias. Oncogene. 2006;25:1434–1436. doi: 10.1038/sj.onc.1209163. [DOI] [PubMed] [Google Scholar]
  • 293.Schnittger S, Bacher U, Kern W, Haferlach T, Haferlach C. JAK2V617F as progression marker in CMPD and as cooperative mutation in AML with trisomy 8 and t(8;21): a comparative study on 1103 CMPD and 269 AML cases. Leukemia. 2007;21:1843–1845. doi: 10.1038/sj.leu.2404707. [DOI] [PubMed] [Google Scholar]
  • 294.Levine RL, Loriaux M, Huntly BJ, et al. The JAK2V617F activating mutation occurs in chronic myelomonocytic leukemia and acute myeloid leukemia, but not in acute lymphoblastic leukemia or chronic lymphocytic leukemia. Blood. 2005;106:3377–3379. doi: 10.1182/blood-2005-05-1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Iwanaga E, Nanri T, Matsuno N, Kawakita T, Mitsuya H, Asou N. A JAK2-V617F activating mutation in addition to KIT and FLT3 mutations is associated with clinical outcome in patients with t(8;21)(q22;q22) acute myeloid leukemia. Haematologica. 2009;94:433–435. doi: 10.3324/haematol.13283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Lo MC, Peterson LF, Yan M, et al. Combined gene expression and DNA occupancy profiling identifies potential therapeutic targets of t(8;21) AML. Blood. 2012;120:1473–1484. doi: 10.1182/blood-2011-12-395335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Watanabe D, Ezoe S, Fujimoto M, et al. Suppressor of cytokine signaling-1 gene silencing in acute myeloid leukaemia and human haematopoietic cell lines. Br J Haematol. 2004;126:726–735. doi: 10.1111/j.1365-2141.2004.05107.x. [DOI] [PubMed] [Google Scholar]
  • 298.Figueroa ME, Lugthart S, Li Y, et al. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell. 2010;17:13–27. doi: 10.1016/j.ccr.2009.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Eghtedar A, Verstovsek S, Estrov Z, et al. Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood. 2012;119:4614–4618. doi: 10.1182/blood-2011-12-400051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Zhou J, Bi C, Janakakumara JV, et al. Enhanced activation of STAT pathways and overexpression of survivin confer resistance to FLT3 inhibitors and could be therapeutic targets in AML. Blood. 2009;113:4052–4062. doi: 10.1182/blood-2008-05-156422. [DOI] [PubMed] [Google Scholar]
  • 301.Hart S, Goh KC, Novotny-Diermayr V, et al. Pacritinib (SB1518), a JAK2/FLT3 inhibitor for the treatment of acute myeloid leukemia. Blood Cancer J. 2011;1:e44. doi: 10.1038/bcj.2011.43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Levis M, Ravandi F, Wang ES, et al. Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood. 2011;117:3294–3301. doi: 10.1182/blood-2010-08-301796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Chopra P, Sethi G, Dastidar SG, Ray A. Polo-like kinase inhibitors: an emerging opportunity for cancer therapeutics. Expert Opin Investig Drugs. 2010;19:27–43. doi: 10.1517/13543780903483191. [DOI] [PubMed] [Google Scholar]
  • 304.Luo J, Emanuele MJ, Li D, et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell. 2009;137:835–848. doi: 10.1016/j.cell.2009.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Irvine DA, Copland M. Targeting hedgehog in hematologic malignancy. Blood. 2012;119:2196–2204. doi: 10.1182/blood-2011-10-383752. [DOI] [PubMed] [Google Scholar]
  • 306.Lee SW, Moskowitz MA, Sims JR. Sonic hedgehog inversely regulates the expression of angiopoietin-1 and angiopoietin-2 in fibroblasts. Int J Mol Med. 2007;19:445–451. [PubMed] [Google Scholar]
  • 307.Adolphe C, Hetherington R, Ellis T, Wainwright B. Patched1 functions as a gatekeeper by promoting cell cycle progression. Cancer Res. 2006;66:2081–2088. doi: 10.1158/0008-5472.CAN-05-2146. [DOI] [PubMed] [Google Scholar]
  • 308.Athar M, Li C, Tang X, et al. Inhibition of smoothened signaling prevents ultraviolet B-induced basal cell carcinomas through regulation of Fas expression and apoptosis. Cancer Res. 2004;64:7545–7552. doi: 10.1158/0008-5472.CAN-04-1393. [DOI] [PubMed] [Google Scholar]
  • 309.Kobune M, Takimoto R, Murase K, et al. Drug resistance is dramatically restored by hedgehog inhibitors in CD34+ leukemic cells. Cancer Sci. 2009;100:948–955. doi: 10.1111/j.1349-7006.2009.01111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Sims-Mourtada J, Izzo JG, Ajani J, Chao KS. Sonic Hedgehog promotes multiple drug resistance by regulation of drug transport. Oncogene. 2007;26:5674–5679. doi: 10.1038/sj.onc.1210356. [DOI] [PubMed] [Google Scholar]
  • 311.Kobune M, Iyama S, Kikuchi S, et al. Stromal cells expressing hedgehog-interacting protein regulate the proliferation of myeloid neoplasms. Blood Cancer J. 2012;2:e87. doi: 10.1038/bcj.2012.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Sheikh A, Alvi AA, Aslam HM, Haseeb A. Hedgehog pathway inhibitors - current status and future prospects. Infect Agent Cancer. 2012;7:29. doi: 10.1186/1750-9378-7-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Minami YFN, Naoe T, et al. Effect of treatment with hedgehog inhibitor PF-04449913, on leukemia-initiation potential in acute myeloid leukemia cells. ASCO annual meeting; 2013. 2013 [Google Scholar]
  • 314.Jamieson CCE, Oehler V, et al. Phase 1 Dose-Escalation Study of PF-04449913, An Oral Hedgehog (Hh) Inhibitor, in Patients with Select Hematologic Malignancies; 55th ASH annual meeting; 2013; 2013. [Google Scholar]
  • 315.Fukushima NmY, Hayakawa F, et al. Treatment With Hedgehog Inhibitor, PF-04449913, Attenuates Leukemia-Initiation Potential In Acute Myeloid Leukemia Cells; 55th ASH annual meeting; 2013; 2013. [Google Scholar]
  • 316.Wellbrock JKJ, Wagner W, et al. Expression of hedgehog pathway mediator Gli2 represents a clinically negative prognostic marker in acute myeloid leukemia and its inhibitor GANT61 exerts anti-leukemic effects in vitro; 55th ASH annual meeting; 2013; 2013. [Google Scholar]
  • 317.Reya T, Duncan AW, Ailles L, et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature. 2003;423:409–414. doi: 10.1038/nature01593. [DOI] [PubMed] [Google Scholar]
  • 318.Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434:843–850. doi: 10.1038/nature03319. [DOI] [PubMed] [Google Scholar]
  • 319.Muller T, Bain G, Wang X, Papkoff J. Regulation of epithelial cell migration and tumor formation by beta-catenin signaling. Exp Cell Res. 2002;280:119–133. doi: 10.1006/excr.2002.5630. [DOI] [PubMed] [Google Scholar]
  • 320.Zhang T, Otevrel T, Gao Z, Ehrlich SM, Fields JZ, Boman BM. Evidence that APC regulates survivin expression: a possible mechanism contributing to the stem cell origin of colon cancer. Cancer Res. 2001;61:8664–8667. [PubMed] [Google Scholar]
  • 321.Shtutman M, Zhurinsky J, Simcha I, et al. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci U S A. 1999;96:5522–5527. doi: 10.1073/pnas.96.10.5522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.He TC, Sparks AB, Rago C, et al. Identification of c-MYC as a target of the APC pathway. Science. 1998;281:1509–1512. doi: 10.1126/science.281.5382.1509. [DOI] [PubMed] [Google Scholar]
  • 323.Wielenga VJ, Smits R, Korinek V, et al. Expression of CD44 in Apc and Tcf mutant mice implies regulation by the WNT pathway. Am J Pathol. 1999;154:515–523. doi: 10.1016/S0002-9440(10)65297-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Ysebaert L, Chicanne G, Demur C, et al. Expression of beta-catenin by acute myeloid leukemia cells predicts enhanced clonogenic capacities and poor prognosis. Leukemia. 2006;20:1211–1216. doi: 10.1038/sj.leu.2404239. [DOI] [PubMed] [Google Scholar]
  • 325.Xu J, Suzuki M, Niwa Y, et al. Clinical significance of nuclear non-phosphorylated beta-catenin in acute myeloid leukaemia and myelodysplastic syndrome. Br J Haematol. 2008;140:394–401. doi: 10.1111/j.1365-2141.2007.06914.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Curtin JC, Lorenzi MV. Drug discovery approaches to target Wnt signaling in cancer stem cells. Oncotarget. 2010;1:552–566. doi: 10.18632/oncotarget.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.DeAlmeida VI, Miao L, Ernst JA, Koeppen H, Polakis P, Rubinfeld B. The soluble wnt receptor Frizzled8CRD-hFc inhibits the growth of teratocarcinomas in vivo. Cancer Res. 2007;67:5371–5379. doi: 10.1158/0008-5472.CAN-07-0266. [DOI] [PubMed] [Google Scholar]
  • 328.Mook RA, Jr, Chen M, Lu J, Barak LS, Lyerly HK, Chen W. Small molecule modulators of Wnt/beta-catenin signaling. Bioorg Med Chem Lett. 2013;23:2187–2191. doi: 10.1016/j.bmcl.2013.01.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Hsiao SJ, Smith S. Tankyrase function at telomeres, spindle poles, and beyond. Biochimie. 2008;90:83–92. doi: 10.1016/j.biochi.2007.07.012. [DOI] [PubMed] [Google Scholar]
  • 330.Chen B, Dodge ME, Tang W, et al. Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nat Chem Biol. 2009;5:100–107. doi: 10.1038/nchembio.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Christensen JG, Schreck R, Burrows J, et al. A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Res. 2003;63:7345–7355. [PubMed] [Google Scholar]
  • 332.Grosch S, Tegeder I, Niederberger E, Brautigam L, Geisslinger G. COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib. FASEB J. 2001;15:2742–2744. doi: 10.1096/fj.01-0299fje. [DOI] [PubMed] [Google Scholar]
  • 333.Xing L, Devadas B, Devraj RV, et al. Discovery and characterization of atropisomer PH-797804, a p38 MAP kinase inhibitor, as a clinical drug candidate. ChemMedChem. 2012;7:273–280. doi: 10.1002/cmdc.201100439. [DOI] [PubMed] [Google Scholar]
  • 334.Emami KH, Nguyen C, Ma H, et al. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription [corrected] Proc Natl Acad Sci U S A. 2004;101:12682–12687. doi: 10.1073/pnas.0404875101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Lepourcelet M, Chen YN, France DS, et al. Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex. Cancer Cell. 2004;5:91–102. doi: 10.1016/s1535-6108(03)00334-9. [DOI] [PubMed] [Google Scholar]
  • 336.Barker N, Clevers H. Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov. 2006;5:997–1014. doi: 10.1038/nrd2154. [DOI] [PubMed] [Google Scholar]
  • 337.Wei W, Chua MS, Grepper S, So S. Small molecule antagonists of Tcf4/beta-catenin complex inhibit the growth of HCC cells in vitro and in vivo. Int J Cancer. 2010;126:2426–2436. doi: 10.1002/ijc.24810. [DOI] [PubMed] [Google Scholar]
  • 338.Im AP, Sehgal AR, Carroll MP, et al. DNMT3A and IDH mutations in acute myeloid leukemia and other myeloid malignancies: associations with prognosis and potential treatment strategies. Leukemia. 2014 doi: 10.1038/leu.2014.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Patel KP, Ravandi F, Ma D, et al. Acute myeloid leukemia with IDH1 or IDH2 mutation: frequency and clinicopathologic features. Am J Clin Pathol. 2011;135:35–45. doi: 10.1309/AJCPD7NR2RMNQDVF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Janin M, Mylonas E, Saada V, et al. Serum 2-hydroxyglutarate production in IDH1- and IDH2-mutated de novo acute myeloid leukemia: a study by the Acute Leukemia French Association group. J Clin Oncol. 2014;32:297–305. doi: 10.1200/JCO.2013.50.2047. [DOI] [PubMed] [Google Scholar]
  • 341.Ravandi F, Patel K, Luthra R, et al. Prognostic significance of alterations in IDH enzyme isoforms in patients with AML treated with high-dose cytarabine and idarubicin. Cancer. 2012;118:2665–2673. doi: 10.1002/cncr.26580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Fathi AT, Sadrzadeh H, Borger DR, et al. Prospective serial evaluation of 2-hydroxyglutarate, during treatment of newly diagnosed acute myeloid leukemia, to assess disease activity and therapeutic response. Blood. 2012;120:4649–4652. doi: 10.1182/blood-2012-06-438267. [DOI] [PubMed] [Google Scholar]
  • 343.Losman JA, Looper RE, Koivunen P, et al. (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science. 2013;339:1621–1625. doi: 10.1126/science.1231677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Wang JH, Chen WL, Li JM, et al. Prognostic significance of 2-hydroxyglutarate levels in acute myeloid leukemia in China. Proc Natl Acad Sci U S A. 2013;110:17017–17022. doi: 10.1073/pnas.1315558110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Pollyea DA, Kohrt HE, Zhang B, et al. 2-Hydroxyglutarate in IDH mutant acute myeloid leukemia: predicting patient responses, minimal residual disease and correlations with methylcytosine and hydroxymethylcytosine levels. Leuk Lymphoma. 2013;54:408–410. doi: 10.3109/10428194.2012.701009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Chaturvedi A, Araujo Cruz MM, Jyotsana N, et al. Mutant IDH1 promotes leukemogenesis in vivo and can be specifically targeted in human AML. Blood. 2013;122:2877–2887. doi: 10.1182/blood-2013-03-491571. [DOI] [PubMed] [Google Scholar]
  • 347.Wang F, Travins J, DeLaBarre B, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science. 2013;340:622–626. doi: 10.1126/science.1234769. [DOI] [PubMed] [Google Scholar]
  • 348.Stein ETM, et al. Clinical safety and activity in a phase I trial of AG-221, a first in class, potent inhibitor of the IDH2-mutant protein, in patients with IDH2 mutant positive advanced hematologic malignancies. AACR annual meeting; 2014. 2014 [Google Scholar]
  • 349.Cowan AJ, Laszlo GS, Estey EH, Walter RB. Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin. Front Biosci (Landmark Ed) 2013;18:1311–1334. doi: 10.2741/4181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Burnett AK, Hills RK, Milligan D, et al. Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial. J Clin Oncol. 2011;29:369–377. doi: 10.1200/JCO.2010.31.4310. [DOI] [PubMed] [Google Scholar]
  • 351.Recher CPA, Witz F, et al. Addition of Gemtuzumab Ozogamycin to chemotherapy improves event-free survival but not overall survival of AML patients with intermediate cytogenetics not eligible for allogeneic transplantation. Results of the GOELAMS AML 2006 IR study; ASHH Annual meeting; 2011; 2011. [Google Scholar]
  • 352.Castaigne S, Pautas C, Terre C, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet. 2012;379:1508–1516. doi: 10.1016/S0140-6736(12)60485-1. [DOI] [PubMed] [Google Scholar]
  • 353.Rowe JM, Lowenberg B. Gemtuzumab ozogamicin in acute myeloid leukemia: a remarkable saga about an active drug. Blood. 2013;121:4838–4841. doi: 10.1182/blood-2013-03-490482. [DOI] [PubMed] [Google Scholar]
  • 354.Breccia M, Cimino G, Diverio D, Gentilini F, Mandelli F, Lo Coco F. Sustained molecular remission after low dose gemtuzumab-ozogamicin in elderly patients with advanced acute promyelocytic leukemia. Haematologica. 2007;92:1273–1274. doi: 10.3324/haematol.11329. [DOI] [PubMed] [Google Scholar]
  • 355.Lo-Coco F, Cimino G, Breccia M, et al. Gemtuzumab ozogamicin (Mylotarg) as a single agent for molecularly relapsed acute promyelocytic leukemia. Blood. 2004;104:1995–1999. doi: 10.1182/blood-2004-04-1550. [DOI] [PubMed] [Google Scholar]
  • 356.Ravandi F, Estey E, Jones D, et al. Effective treatment of acute promyelocytic leukemia with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab ozogamicin. J Clin Oncol. 2009;27:504–510. doi: 10.1200/JCO.2008.18.6130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Chames P, Baty D. Bispecific antibodies for cancer therapy: the light at the end of the tunnel? MAbs. 2009;1:539–547. doi: 10.4161/mabs.1.6.10015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Frankel SR, Baeuerle PA. Targeting T cells to tumor cells using bispecific antibodies. Curr Opin Chem Biol. 2013;17:385–392. doi: 10.1016/j.cbpa.2013.03.029. [DOI] [PubMed] [Google Scholar]
  • 359.Satta A, Mezzanzanica D, Turatti F, Canevari S, Figini M. Redirection of T-cell effector functions for cancer therapy: bispecific antibodies and chimeric antigen receptors. Future Oncol. 2013;9:527–539. doi: 10.2217/fon.12.203. [DOI] [PubMed] [Google Scholar]
  • 360.Aigner M, Feulner J, Schaffer S, et al. T lymphocytes can be effectively recruited for ex vivo and in vivo lysis of AML blasts by a novel CD33/CD3-bispecific BiTE antibody construct. Leukemia. 2013;27:1107–1115. doi: 10.1038/leu.2012.341. [DOI] [PubMed] [Google Scholar]
  • 361.AL Hussaini MRJ, Rettig M, et al. Targeting CD123 In Leukemic Stem Cells Using Dual Affinity Re-Targeting Molecules (DARTs®); 55thh ASH annual meeting; 2013; 2013. [Google Scholar]
  • 362.Bargou R, Leo E, Zugmaier G, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321:974–977. doi: 10.1126/science.1158545. [DOI] [PubMed] [Google Scholar]
  • 363.Topp MS, Kufer P, Gokbuget N, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol. 2011;29:2493–2498. doi: 10.1200/JCO.2010.32.7270. [DOI] [PubMed] [Google Scholar]
  • 364.Klinger M, Brandl C, Zugmaier G, et al. Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood. 2012;119:6226–6233. doi: 10.1182/blood-2012-01-400515. [DOI] [PubMed] [Google Scholar]
  • 365.Wong R, Pepper C, Brennan P, Nagorsen D, Man S, Fegan C. Blinatumomab induces autologous T-cell killing of chronic lymphocytic leukemia cells. Haematologica. 2013;98:1930–1938. doi: 10.3324/haematol.2012.082248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Reikvam H, Kittang AO, Melve G, et al. Targeted anti-leukemic therapy as disease-stabilizing treatment for acute myeloid leukemia relapse after allogeneic stem cell transplantation: Will it be possible to combine these strategies with retransplantation or donor lymphocyte infusions? Curr Cancer Drug Targets. 2013;13:30–47. [PubMed] [Google Scholar]
  • 367.Alfaro C, Suarez N, Gonzalez A, et al. Influence of bevacizumab, sunitinib and sorafenib as single agents or in combination on the inhibitory effects of VEGF on human dendritic cell differentiation from monocytes. Br J Cancer. 2009;100:1111–1119. doi: 10.1038/sj.bjc.6604965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Ramirez P, Rettig MP, Uy GL, et al. BIO5192, a small molecule inhibitor of VLA-4, mobilizes hematopoietic stem and progenitor cells. Blood. 2009;114:1340–1343. doi: 10.1182/blood-2008-10-184721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Jacamo R, Chen Y, Wang Z, et al. Reciprocal leukemia-stroma VCAM-1/VLA-4-dependent activation of NF-kappaB mediates chemoresistance. Blood. 2014;123:2691–2702. doi: 10.1182/blood-2013-06-511527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Ossenkoppele GJ, Stussi G, Maertens J, et al. Addition of bevacizumab to chemotherapy in acute myeloid leukemia at older age: a randomized phase 2 trial of the Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON) and the Swiss Group for Clinical Cancer Research (SAKK) Blood. 2012;120:4706–4711. doi: 10.1182/blood-2012-04-420596. [DOI] [PubMed] [Google Scholar]
  • 371.Reikvam H, Nepstad I, Bruserud O, Hatfield KJ. Pharmacological targeting of the PI3K/mTOR pathway alters the release of angioregulatory mediators both from primary human acute myeloid leukemia cells and their neighboring stromal cells. Oncotarget. 2013;4:830–843. doi: 10.18632/oncotarget.971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Zhang J, Ye J, Ma D, et al. Cross-talk between leukemic and endothelial cells promotes angiogenesis by VEGF activation of the Notch/Dll4 pathway. Carcinogenesis. 2013;34:667–677. doi: 10.1093/carcin/bgs386. [DOI] [PubMed] [Google Scholar]
  • 373.Konopleva MY, Jordan CT. Leukemia stem cells and microenvironment: biology and therapeutic targeting. J Clin Oncol. 2011;29:591–599. doi: 10.1200/JCO.2010.31.0904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley EJ, Krek W. Chemokine receptor CXCR4 downregulated by von Hippel-Lindau tumour suppressor pVHL. Nature. 2003;425:307–311. doi: 10.1038/nature01874. [DOI] [PubMed] [Google Scholar]
  • 375.Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–864. doi: 10.1038/nm1075. [DOI] [PubMed] [Google Scholar]
  • 376.Wang Y, Liu Y, Malek SN, Zheng P. Targeting HIF1alpha eliminates cancer stem cells in hematological malignancies. Cell Stem Cell. 2011;8:399–411. doi: 10.1016/j.stem.2011.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Mussai F, De Santo C, Abu-Dayyeh I, et al. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood. 2013;122:749–758. doi: 10.1182/blood-2013-01-480129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol. 2003;24:302–306. doi: 10.1016/s1471-4906(03)00132-7. [DOI] [PubMed] [Google Scholar]
  • 379.Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med. 2006;12:1167–1174. doi: 10.1038/nm1483. [DOI] [PubMed] [Google Scholar]

RESOURCES