Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2017

Open Access 01.12.2017 | Review

Emerging therapies for acute myeloid leukemia

verfasst von: Caner Saygin, Hetty E. Carraway

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2017

Abstract

Acute myeloid leukemia (AML) is characterized by clinical and biological heterogeneity. Despite the advances in our understanding of its pathobiology, the chemotherapy-directed management has remained largely unchanged in the past 40 years. However, various novel agents have demonstrated clinical activity, either as single agents (e.g., isocitrate dehydrogenase (IDH) inhibitors, vadastuximab) or in combination with standard induction/consolidation at diagnosis and with salvage regimens at relapse. The classes of agents described in this review include novel cytotoxic chemotherapies (CPX-351 and vosaroxin), epigenetic modifiers (guadecitabine, IDH inhibitors, histone deacetylase (HDAC) inhibitors, bromodomain and extraterminal (BET) inhibitors), FMS-like tyrosine kinase receptor 3 (FLT3) inhibitors, and antibody-drug conjugates (vadastuximab), as well as cell cycle inhibitors (volasertib), B-cell lymphoma 2 (BCL-2) inhibitors, and aminopeptidase inhibitors. These agents are actively undergoing clinical investigation alone or in combination with available chemotherapy.
Abkürzungen
ATRA
All-trans retinoic acid
NPM
Nucleophosmin

Background

Acute myeloid leukemia (AML) is a clonal disorder of myeloid progenitors characterized by clinical and biological heterogeneity. With decades of research, our understanding of the pathobiology, classification, and genomic landscape of AML has improved substantially [1, 2]. Concurrently, various promising agents have been evaluated in clinical trials, but the classical upfront treatment of AML (intensive induction with 7 days of cytarabine plus 3 days of an anthracycline (7 + 3), followed by consolidation chemotherapy or hematopoietic cell transplant (HCT)) has remained steadfast over the last 40 years. With traditional intensive chemotherapy regimens, only 40% of AML patients <60 years of age survive more than 5 years, and even patients with favorable-risk core-binding factor leukemia have a mortality rate of 56% at 10 years [3]. In older adults unfit for standard induction chemotherapy, outcomes of lower-intensity treatment (low-dose cytarabine, azacitidine, or decitabine) are not curative and median overall survival (OS) is often <1 year [4, 5]. This discordance between the explosive growth in trials investigating novel therapies in AML and the minimal progress made in current standard of care might be attributed to suboptimal preclinical models, exclusive criteria that limit patients eligible for enrollment into clinical trials, and single-agent clinical approach to early drug development as well as limited ability to eliminate the remnant leukemic clone due to ineffective novel agents or emergence of leukemic clonal promiscuity [6].
Despite the paucity of new drug approvals for AML treatment, identification of potential driver mutations through next-generation sequencing has revealed biologic intricacies of AML and led to new investigational drugs. Furthermore, antigen-specific immunotherapies hold promise to expand the armamentarium for treatment of AML. In this review, we highlight some of the promising novel approaches and agents that are currently in clinical trials and have published or presented data. This article is not meant to be an exhaustive review of all emerging agents. Instead, we summarize evolving treatment strategies with promising results in AML trials, which hence will likely emerge as new therapeutics in the near future (Table 1).
Table 1
Selected emerging therapies for the management of AML
Drug class/mechanism
Agent
Suggested patient population
Single/combination
Phase of developmenta
Ref.
Cytotoxic chemotherapy
 Liposomal formulation of 7 + 3
CPX-351
≥60 years, sAML, fit for induction therapy
Single agent
3
[1015]
 Topoisomerase II inhibitor
Vosaroxin
≥60 years, R/R
With cytarabine
3
[19]
Epigenetic modifiers
 DNMT inhibitor
Guadecitabine
Unfit for intensive therapy or R/R
Single agent
3
[24, 25]
 IDH1 inhibitor
AG-120, IDH305, FT-2102
IDH1 mutated
Single agent/with AZA or induction and consolidation
1b/2
[31, 33]
 IDH2 inhibitor
Enasidenib (AG-221)
IDH2 mutated
Single agent/with AZA or induction and consolidation
3
[34]
 HDAC inhibitors
Panobinostat
Ongoing investigation
With HMAs or induction and consolidation
1b/2
[38, 39]
Vorinostat
3
[4043]
Entinostat
2
[45]
Pracinostat
2
[46]
 BET inhibitor
OTX015
Ongoing investigation
Single agent
1
[47]
 DOT1L inhibitor
Pinometostat
MLL-rearranged
Single agent
1
[49]
 LSD1 inhibitor
Tranylcypromine, GSK2879552, ORY-1001
Ongoing investigation
Single agent/with ATRA
1/2
FLT3 inhibitors
Sorafenib
FLT3-ITD-mutated R/R
With AZA
2
[60]
Midostaurin
FLT3-ITD or FLT3-TKD, ≤60 years
With induction and consolidation
3
[63]
Quizartinib
FLT3-ITD-mutated R/R
Single agent
3
[6668]
Crenolanib
FLT3-ITD or FLT3-TKD
Single agent/with induction and consolidation
2
[7173]
Gilteritinib
FLT3-ITD or FLT3-TKD R/R
Single agent
3
[74]
Antibody-drug conjugates
 Anti-CD33
Vadastuximab (SGN-CD33A)
CD33+
Single agent/with HMAs or induction and consolidation
3
[7577]
Cell cycle inhibitors
 Polo-like kinase inhibitor
Volasertib
Unfit for intensive therapy
With LDAC or decitabine
3
[78]
Other agents
 BCL-2 inhibitor
Venetoclax (ABT-199)
R/R, or older (≥65 years) and unfit for intensive therapy
Single agent/with HMAs or LDAC
2
[82, 83]
 Aminopeptidase inhibitor
Tosedostat
≥60 year, R/R or unfit for intensive therapy
Single agent/with cytarabine or HMAs
2
[8487]
aDenotes the furthest phase in development
7 + 3 7 days of cytarabine and 3 days of daunorubicin, AML acute myeloid leukemia, AZA azacitidine, R/R relapsed or refractory, HMA hypomethylating agent, LDAC low-dose cytarabine, sAML secondary acute myeloid leukemia

Novel cytotoxic chemotherapy agents

The backbone of AML induction therapy has been anthracycline-cytarabine combination for decades, and the addition of other cytotoxics, including thioguanine, fludarabine, or etoposide, have offered no additional survival benefit [7, 8]. However, certain modifications in traditional 7 + 3, including manipulation of treatment intensity and duration of treatment, translated into improved OS for selected populations of adult patients [9]. Therefore, new formulations of cytotoxic chemotherapy that have similar mechanisms of action might continue to improve outcomes.

CPX-351

CPX-351 is the liposomal formulation of cytarabine and daunorubicin packaged at a 5:1 molar ratio within liposomes, which in animal models demonstrated higher efficacy compared with the same drugs administered conventionally [10]. The ratio is shown to be maximally synergistic and minimally antagonistic in vitro [11]. In a randomized, open-label, phase 2 trial, CPX-351 (100 U/m2, equivalent to 100 mg/m2 cytarabine and 44 mg/m2 daunorubicin) was compared with conventional 7 + 3 (100 mg/m2 cytarabine and 60 mg/m2 daunorubicin) as an induction therapy, with enrollment of 126 AML patients at age 60–75 years, who were fit for intensive chemotherapy [10]. The primary efficacy endpoint was composite complete response (CRc), combining morphologic complete response (CR) and morphologic CR with incomplete count recovery (CRi). Overall, CRc rates were higher (66.7 vs 51.2%, p = 0.07) in the CPX-351 arm, which met the predefined criteria for success (p < 0.1). There were no differences in true CR rate (48.8% in both arms), event-free survival (EFS), or OS when all patients were analyzed. However, preplanned subgroup analysis of patients with secondary AML (sAML; therapy-related AML or AML with a history of antecedent hematologic disorder) demonstrated an improved response rate (57.6 vs 31.6%, p = 0.06), with prolonged EFS (p = 0.08) and OS (p = 0.01). In a separate phase 2, randomized, open-label study, 125 AML patients at first relapse, who were between the ages of 18 and 65 years, were assigned to CPX-351 or investigators’ choice of first salvage treatment [12]. Overall, the CR rate was slightly higher in the CPX-351 arm (37 vs 31.8%), but the study did not achieve the predefined goal of survival improvement at 1 year.
Based on the promising results in patients with sAML, a randomized, multicenter, open-label, phase 3 study of first-line CPX-351 (100 U/m2) vs daunorubicin (60 mg/m2) plus cytarabine (100 mg/m2) in high-risk sAML patients was initiated [13]. A total of 309 patients between the ages of 60 and 75 years were randomized 1:1 to treatment arms. CPX-351 treatment resulted in better OS (median, 9.56 vs 5.95 months, p = 0.005), EFS (p = 0.021), and CRc rates (47.7 vs 33.3%, p = 0.016). Moreover, 60-day mortality was lower in CPX-351 (13.7 vs 21.2%), and grade 3–5 adverse events (AEs) were similar in frequency and severity in both arms. In a subgroup analysis of this large trial, patients aged 60–69 and 70–75 years were analyzed separately and both groups were found to have greater median OS (9.63 vs 6.87 months in patients aged 60–69 years and 8.87 vs 5.62 months in patients aged 70–75 years) and CRc rate (50 vs 36.3% in patients aged 60–69 years and 43.9 vs 27.8% in patients aged 70–75 years) with the CPX-351 treatment [14]. In another subgroup analysis, 91 transplanted patients were landmarked at the time of hematopoetic stem cell transplant (HSCT) (i.e., time of origin) and patients treated with CPX-351 were found to have better OS as compared to patients treated with standard 7 + 3 (p = 0.004) [15]. These results support the use of CPX-351 as first-line induction treatment for fit older patients with sAML, and this formulation may provide an effective bridge to successful HCT in this subset of patients.
In addition, since the drug has shown efficacy (i.e., CRc) at doses of 50 and 75 U/m2 in phase 1 and 2 trials, a current phase 2 study compares outcomes of newly diagnosed AML patients at these lower doses, who are otherwise at high risk of induction mortality [16]. Interim results demonstrated significantly better OS (HR, 0.2; p = 0.005) and EFS (HR, 0.25; p = 0.019) with 75 U/m2, and the study continues accrual with the third arm of 100 U/m2 (NCT02286726). Moreover, comparison of the outcomes of CPX-351 (100 U/m2) vs daunorubicin (90 mg/m2) + cytarabine treatment would be valuable to assess the superiority (or non-inferiority) of this formulation.

Vosaroxin

Vosaroxin is a first-in-class, non-anthracycline quinolone derivative that induces replication-dependent DNA damage by intercalating DNA and inhibiting topoisomerase II, thereby inducing G2 cell cycle arrest and apoptosis [3]. It is minimally metabolized, without production of free radicals that are implicated in the cardiotoxicity observed with other topoisomerase II inhibitors. Based on the encouraging results in early phase 1 and phase 1b/2 studies [17, 18], the randomized, placebo-controlled, double-blind phase 3 VALOR trial of cytarabine (1 g/m2 days 1–5) with or without vosaroxin (90 mg/m2 days 1–4) was conducted in adult patients with primary refractory AML or AML in first relapse [19]. A total of 711 patients were randomized 1:1 to treatment arms, and the study did not meet the primary endpoint of median OS difference between groups (7.5 months in vosaroxin arm vs 6.1 months in placebo arm, p = 0.06). However, the overall CR rate was nearly doubled in the vosaroxin arm compared with the placebo arm (30.1 vs 16.3%, p < 0.0001), and the responses were durable as shown by the leukemia-free survival data. Additionally, in the predefined analysis censoring at the time of HSCT, OS was better in the vosaroxin plus cytarabine group than in the placebo plus cytarabine group (6.7 vs 5.3 months, p = 0.02). In further preplanned analyses (based on age and time to relapse), vosaroxin-treated patients ≥60 years had significantly better OS (7.1 vs 5 months, p = 0.003) and those who relapsed <12 months and received vosaroxin had 1.5 months (6.7 vs 5.2 months, p = 0.03) of median OS benefit as compared to the placebo arm. There was no difference in 30- and 60-day all-cause mortality between treatment groups, but 15% of patients on the vosaroxin arm had grade 3 or 4 stomatitis. The recently presented updated survival data was consistent with the subgroup analysis of the primary report, and after a median of 39.9 months of follow-up, the survival benefit observed in patients ≥60 years was durable through 48 months [20].
Vosaroxin has also been investigated in the first-line setting for older patients with previously untreated poor-risk AML. In the single-agent, phase 2 REVEAL-1 trial, vosaroxin monotherapy was evaluated at doses of 72 and 90 mg/m2 and the 72 mg/m2 dose demonstrated a CRc rate of 35%, with an acceptable 30- and 60-day mortality of 7 and 17%, respectively [21]. Therefore, this dose was used in a subsequent open-label, randomized phase 2 study, which was designed with the “pick a winner” strategy, comparing in a 1:1 randomization of low-dose cytarabine (LDAC) vs vosaroxin monotherapy and LDAC vs LDAC + vosaroxin combination in older unfit AML patients [22]. The study demonstrated no CR or survival benefit for vosaroxin, and the trial was prematurely closed at its first interim analysis.
Vosaroxin at the dose of 90 mg/m2 demonstrated clinical activity in combination with cytarabine for relapsed or refractory (R/R) AML patients ≥60 years. However, the OS benefit was 2.1 months. Its utility is limited to the R/R setting, as the drug did not provide benefit when compared to LDAC in the first-line treatment for unfit older patients. A phase 2 study of vosaroxin and decitabine in older patients with newly diagnosed AML or high-risk myelodysplastic syndrome (MDS) is currently ongoing (NCT01893320).

Epigenetic modifiers

A great number of comprehensive whole genome sequencing, exome sequencing, and targeted sequencing studies have been performed in AML and myeloid neoplasms in the last decade. Many of the newly identified recurrently mutated genes are involved in the epigenetic regulation of transcription [1]. Epigenetic modifiers include proteins involved in modifications of DNA cytosine residues (e.g., methylation) or post-translational modifications of histones (acetylation, ubiquitination). Mutations in these genes often lead directly to aberrant gene expression in AML [23]. Currently, these mutations represent a major focus of interest, and several novel epigenetic therapies are in preclinical testing phases or have entered clinical trials (Fig. 1).

DNMT inhibitors

DNA methyltransferase 3A (DNMT3A) catalyzes de novo methylation of cytosine residues in DNA, and its gene is frequently mutated in AML, which leads to loss of function and confers adverse risk. The hypomethylating agents (HMAs), azacitidine and decitabine, are nucleoside analogs that integrate into DNA and inhibit DNMTs [23]. Along with LDAC, they represent a reasonable treatment option for low blast count AML patients who are unfit for intensive induction chemotherapy with CRc rates around 20–30% [4]. Guadecitabine (SGI-110) is a second-generation HMA formulated as a dinucleotide of decitabine and deoxyguanosine, which increases the half-life of decitabine by protecting it from deamination [3]. In a multicenter, phase 2, dose-response study, 51 previously untreated elderly (≥65 years) AML patients, who were ineligible for intensive chemotherapy, were randomized 1:1 to 60 or 90 mg/m2 guadecitabine [24]. There were no significant differences in CR rates between treatment groups, and in combined analysis, overall CR and CRc rates were 37 and 57%, respectively, and the median OS was 10.5 months. Based on these results, a randomized phase 3 trial has been initiated to compare guadecitabine vs treatment choice (i.e., azacitidine, decitabine, or LDAC) in patients with previously untreated AML, who are ineligible for intensive chemotherapy (NCT02348489).
Guadecitabine has also been investigated in R/R AML patients, and a recently reported long-term follow-up of phase 2 studies demonstrated that in 103 patients, 23% achieved CRc and median OS was 6.6 months with 1- and 2-year survival rates of 28 and 19%, respectively [25]. On the basis of these data, a phase 3 randomized, open-label study of guadecitabine vs treatment choice in R/R AML has been initiated (NCT02920008).
DNA methylation profiling identified biologically distinct subtypes of AML, and certain methylation profiles were associated with adverse outcome [26, 27]. Furthermore, differentially methylated regions of DNA at baseline distinguished patients who responded to HMA from non-responders in different myeloid malignancies [28, 29]. Specific methylation signatures may predict responsiveness to treatment with guadecitabine and offer an opportunity to improve management of elderly or R/R AML patients.
Guadecitabine has demonstrated clinical activity in first-line and R/R settings, and two ongoing phase 3 studies for these patient populations may provide evidence to justify its use over LDAC and first-generation HMAs. However, a substantial difference in cost with marginal difference in OS benefit might limit its use in the clinical setting.

IDH inhibitors

Isocitrate dehydrogenases 1 and 2 (IDH1 (cytoplasmic) and IDH2 (mitochondrial), respectively) catalyze the conversion of isocitrate to α-ketoglutarate (α-KG). In adults, IDH1 and IDH2 mutations occur with a frequency of 5–10 and 10–15% in adult AML, respectively, and are more common in patients with cytogenetically normal AML (10.4 and 15–20%, respectively) [23]. All IDH1 and IDH2 mutations are novel gain-of-function mutations, and the mutant IDH proteins possess a neomorphic enzyme activity catalyzing the conversion of α-KG to the oncometabolite 2-hydroxyglutarate (2-HG) (Fig. 2). This leads the competitive inhibition of α-KG-dependent enzymes, including TET2, hypermethylation of target genes, and impaired hematopoietic differentiation. The prognostic impact of IDH mutations in myeloid neoplasms remains controversial; however, one hypothesis is that inhibition of mutant IDH may decrease the levels of 2-HG and reverse the block in cellular differentiation [30]. In the short time since the IDH mutations were first discovered in 2009, several IDH inhibitors have been tested in clinical trials and early results demonstrated encouraging responses with durability and minimal toxicity.
AG-120, IDH305, and FT-2102 are oral inhibitors of mutant IDH1 that are currently in clinical development. In a phase 1, open-label, dose escalation and expansion study, AG-120 monotherapy was evaluated in patients with IDH1 mutant advanced hematologic malignancies (NCT02074839) [31]. Overall response rate (ORR) was 36% and CR rate was 18% in a cohort in which majority of patients (78%) had R/R AML. The drug was well tolerated, but three patients developed differentiation syndrome within the first 60 days of treatment, and they were successfully managed with hydroxyurea and steroids [32]. Dose expansion arms are currently enrolling patients with R/R and untreated AML. In another phase 1 dose-escalation study, IDH305, a mutant-selective allosteric IDH1 inhibitor, was evaluated in R/R AML and MDS (NCT02381886) [33]. In the interim analysis of 24 AML patients, ORR and CR rates were 33 and 9.5%, respectively. The most commonly reported AEs were raised bilirubin and lipase. A phase 1/1b study of FT-2102 as a single agent and in combination with azacitidine is currently ongoing (NCT02719574).
Enasidenib (AG-221) is an oral inhibitor of IDH2, which also reduces 2-HG levels in patients with IDH2-mutated AML to levels detected in healthy subjects [30]. Interim results of a phase 1/2 dose-escalation study of AG-221 demonstrated an ORR of 41% in R/R AML patients, regardless of the number of prior treatments [34]. Rates of CR, CRc, and partial response (PR) were 18, 26, and 15%, respectively. An additional 45% of patients had stable disease. Of interest, the mutant IDH2 variant allelic frequency (VAF) did not change from baseline in the majority of patients who attained CRc, suggesting that eradication of the clone was not necessary for response and gives insight into the putative mechanism of enasidenib as a differentiation agent. The drug was well tolerated, and the most common AEs were indirect hyperbilirubinemia (19%) and nausea (18%). Based on these results, the phase 3, randomized, open-label IDHENTIFY trial is currently recruiting AML patients ≥60 years who are R/R after two or three prior regimens (NCT02577406). Patients are randomized 1:1 to enasidenib or conventional care regimens, and enrollment will continue through 2019.
In early phase 1/2 trials, IDH inhibitors have demonstrated impressive single-agent activity in R/R AML patients. Additionally, both AG-120 and enasidenib are being investigated in patients with newly diagnosed AML with IDH1 and/or IDH2 mutations, in combination with induction and consolidation for patients eligible for intensive chemotherapy (NCT02632708), as well as with azacitidine in unfit patients (NCT02677922). A caveat is the lack of OS data, and it is unclear whether patients with “stable disease,” who represent 45% of patients in the initial phase 1/2 trial of enasidenib, will have a meaningful survival benefit. Despite this, these drugs offer a significant possibility of improving current standard of care in IDH mutant AML patients.

HDAC inhibitors

Histone acetylation and deacetylation are essential processes for the regulation of gene expression. Acetylation of histones relaxes the condensed chromatin and exposes the promoter regions of genes to transcription factors. On the other hand, deacetylation catalyzed by histone deacetylases (HDACs) results in gene silencing [23]. In leukemic cells, this balance is disrupted by several mechanisms, and therefore, HDAC inhibitors emerged as an attractive therapeutic approach to modulate disease (Fig. 2). Unlike IDH inhibitors, clinical activity of monotherapy with an HDAC inhibitor was low, with ORR of 17% for vorinostat [35] and 13% for mocetinostat [36], and no clinical response was achieved with entinostat monotherapy [37]. Therefore, current studies focus on combination regimens of HDAC inhibitors with other epigenetic agents like HMAs or alternatively directly intensive chemotherapy.
In a phase 1b/2 study, oral pan-HDAC inhibitor panobinostat was administered sequentially with azacitidine (75 mg/m2) to previously untreated patients with AML or high-risk MDS [38]. In 29 patients with AML, CRc and PR rates were 10 and 21%, respectively, and median OS was 8 months. Another phase 1b/2 study, Panobidara, combined panobinostat with induction chemotherapy consisting of idarubicin + cytarabine (IA), followed by panobinostat maintenance in elderly patients with newly diagnosed AML [39]. In 38 evaluable patients, CR was 64% and median OS of the whole cohort was 17 months. Even though results from early clinical studies are encouraging, larger randomized cohorts are necessary to make any statement about the benefit of adding panobinostat to HMAs or intensive chemotherapy.
Despite its modest activity as a single agent, a phase 2 study of vorinostat combined with IA for newly diagnosed young (≤65 years) AML or high-risk MDS patients showed an ORR of 85%, including 76% true CR, and a median OS of 82 weeks [40]. ORR was 93% in diploid patients and 100% in FLT3 internal tandem duplication (FLT3-ITD) patients. Based on these results, the randomized, phase 3 SWOG S1203 study compared 7 + 3 vs IA vs IA plus vorinostat in young (≤60 years) untreated AML patients [41]. There were no significant differences in CR rates, EFS, or OS among all three arms; thus, the vorinostat arm was stopped due to futility. Vorinostat has also been studied in combination with HMAs. In a phase 1 dose-escalation study of vorinostat with decitabine in 29 R/R and 31 untreated AML patients, ORRs with concurrent treatment were 15 and 46%, respectively [42]. Additionally, a recently presented phase 1 study of vorinostat with decitabine in young patients (≤60 years) with R/R AML who had mixed lineage leukemic (MLL) partial tandem duplication and received a median of two prior regimens, demonstrated modest toxicity with 35% CRc [43]. The future of vorinostat in patients eligible for intensive chemotherapy is in doubt. However, it has proven clinical activity in combination with HMAs for unfit patients and may find itself an indication for rare biological subsets of AML. Of note, data from the SWOG S1117 study, which randomized 277 high-risk MDS patients to azacitidine vs azacitidine plus vorinostat vs azacitidine plus lenalidomide, demonstrated no significant differences in ORR [44]. The study was not powered to evaluate OS, but a longer follow-up is needed to better evaluate effect on duration of response in patients treated with vorinostat.
Entinostat and pracinostat are two other oral HDAC inhibitors, which are in early phases of development. An open-label phase 2 trial (E1905) randomized 149 patients with AML/MDS to azacitidine (50 mg/m2 for 10 days) with or without overlapping/concurrent delivery schedule of entinostat [45]. The addition of entinostat did not increase ORR but was associated with decreased reversal of methylation compared to HMA monotherapy. Therefore, another phase 2 randomized study has opened in order to investigate the activity of prolonged azacitidine combined with two different entinostat schedules (concurrent vs sequential) in elderly (≥60 years) patients with AML (NCT01305499). This study hopes to determine if sequential therapy might improve clinical responses as concurrent administration may cause decreased HMA incorporation due to cell cycle arrest from the overlapping schedule of HDACi. Finally, a randomized phase 2 study that investigated pracinostat combined with azacitidine for elderly (≥65 years) AML patients who were unfit for intensive induction therapy was completed [46]. In 50 evaluable patients, the combination was well tolerated with CR and CRi rates of 32 and 14%, respectively. Most clinical responses occurred within the first 2 cycles and continued to improve with ongoing therapy. Pracinostat was granted Orphan Drug Status by the FDA in 2014, and the follow-up of patients treated on this study continues in order to calculate a median OS.

BET inhibitors

Bromodomain and extraterminal (BET) proteins play a major role in the epigenetic regulation of gene transcription by binding to acetylated histone tails and recruiting the transcriptional machinery to the promoter regions of genes (Fig. 1). BET inhibitors demonstrated remarkable anti-leukemic activity in vitro and in vivo in various AML models and are currently being tested in multiple early-phase trials [23]. In a dose-escalation, open-label, phase 1 study, orally active BET inhibitor OTX015 was given to 41 older (≥60 years) patients with R/R acute leukemia (36 AML, 1 high-risk MDS) [47]. Two patients achieved CR, one had CRp, and two patients had partial blast clearance. Common AEs were diarrhea and hyperbilirubinemia. The study did not identify any biomarkers to predict response. Various other BET inhibitors have entered early clinical trials in patients with R/R AML, including TEN-010 (NCT02308761), GSK525762 (NCT01943851), and CPI-0610 (NCT02158858).
BET inhibitors have raised great interest as a novel treatment approach, and ongoing phase 1 trials are investigating their single-agent activities. These drugs are also being investigated in combination with standard therapies and other novel agents. Furthermore, a vigorous search for potential biomarkers of response may identify patients with higher likelihood of BET inhibitor response.

DOT1L inhibitors

Rearrangements of the mixed lineage leukemic (MLL) gene at the 11q23 chromosome locus are present in 5–10% of AML cases and portend poor prognosis [48]. Most of the MLL fusion partners bind to disruptor of telomeric silencing 1-like (DOT1L), which is postulated to be the oncogenic driver of MLL-rearranged (MLL-r) AML via its histone methyltransferase activity (Fig. 1). Pinometostat (EPZ-5676) is a DOT1L inhibitor and had robust preclinical activity in MLL-r xenograft models [23]. Early results from an open-label phase 1 trial, which enrolled adult R/R acute leukemias, demonstrated an ORR of 12.2% (6 out of 49 patients) with an acceptable safety profile [49]. However, another phase 1 trial conducted in children with R/R MLL-r acute leukemia reported no ORR in 18 patients enrolled [50]. Both studies showed evidence of target inhibition, and next steps in development should explore pinometostat combinations with other anti-leukemic agents. Interesting preclinical work demonstrates that NPM1-mutated leukemogenesis is dependent on HOX and MEIS1 expression, which is controlled by specific chromatin regulatory complexes. Inhibition by DOT1L and the menin-MLL pathways can release the block on NPM1-mutated leukemia and result in differentiation. Furthermore, inhibition of the menin-MLL pathway led to profound down regulation of MEIS1 and subsequent suppression of the FLT3 expression [51]. These data suggest that the further development of these agents may ultimately play a large role in NPM1/FLT3-ITD-mutated leukemias.

LSD1 inhibitors

Lysine-specific demethylase 1 (LSD1) is a histone demethylase expressed in leukemic cells and regulates the differentiation block in AML [23, 52, 53]. The enzyme can be targeted by tranylcypromine (TCP), and the combination of ATRA and TCP is currently being investigated in multiple phase 1 and phase 1/2 studies for adults with R/R AML or MDS (NCT02273102, NCT02261779, NCT02717884). Selective TCP derivative LSD1 inhibitors, GSK2879552 and ORY-1001, have also entered early-phase trials for patients with R/R acute leukemia (NCT02177812, EudraCT number 2013-002447-29). Similarly, an open-label phase 1 trial of IMG-7289 with or without ATRA is recruiting participants (NCT02842827). At the time of this writing, no data are available from these studies.
In summary, studies of the mutational landscape of AML highlighted many epigenetic modifiers as attractive targets for personalized therapy. Some of these novel agents showed encouraging clinical activity in early-phase trials. Several other agents are in early preclinical development and will likely be tested in AML trials in the near future (e.g., EZH2 inhibitors). Utilization of accurate biomarkers of clinical response and rational combinations of targeted agents with other anti-leukemic therapy might revolutionize the management of AML in the near future.

FLT3 inhibitors

FMS-like tyrosine kinase receptor 3 (FLT3) is the most frequently mutated gene in AML. FLT3 internal tandem duplications (FLT3-ITDs) are seen in approximately 25–30% of patients with de novo AML, while an additional 5–10% of patients harbor point mutations in tyrosine kinase domain (TKD) that results in constitutive tyrosine kinase signaling (Fig. 2) [54, 55]. The high frequency of these mutations and the poor prognosis associated with FLT3-ITD compel the development of FLT3 inhibitors. Despite the disappointing results with early FLT3 inhibitors and low single-agent activity, newer agents and combination regimens have yielded encouraging results.

Sorafenib

Sorafenib is an oral multi-kinase inhibitor with activity against FLT3, KIT, vascular endothelial growth factor receptor (VEGFR), and platelet-derived growth factor receptor (PDGFR). It has been investigated in combination with standard 7 + 3 induction and cytarabine consolidation in elderly patients with AML [56]. The combination did not improve OS or EFS (even in the subgroup analysis of patients with FLT3-ITD) but resulted in higher treatment-related mortality. Similarly, combination of sorafenib with azacitidine (for FLT3-ITD-mutated AML) or LDAC (for high-risk MDS or AML) did not translate into meaningful survival benefit in untreated elderly patients [57, 58]. However, in a phase 2 trial of intensive chemotherapy (7 + 3 followed by consolidation) with or without sorafenib in younger (≤60 years) untreated AML patients, median EFS was significantly longer in the sorafenib arm vs the placebo arm (21 vs 9 months, p = 0.01) [59]. CR rates were similar (60 vs 59%), and the sorafenib group had a higher rate of AEs. In another phase 2 study of azacitidine with sorafenib in 43 patients with R/R AML, 93% had FLT3-ITD mutation and the combination resulted in an ORR of 46% [60]. Moreover, sorafenib maintenance after allogeneic HCT was shown to be safe and promising to prevent relapse in early-phase studies of patients with FLT3-ITD AML [61].
In first-line management of elderly AML patients, sorafenib demonstrated no benefit. The encouraging results in younger patients need to be supported with long-term OS data. Even though the drug might be useful for treatment of R/R AML, its future is limited due to the era of newer FLT3 inhibitors.

Midostaurin

Midostaurin is another oral multi-kinase inhibitor of FLT3, KIT, VEGFR, PDGFR, and protein kinase C. In early single-agent studies, activity of midostaurin was limited but promising; hence, it was further investigated in combination with standard intensive chemotherapy. In a phase 1b study of newly diagnosed young (≤60 years) AML patients, a midostaurin 50-mg twice-daily dose schedule in combination with 7 + 3 and high-dose cytarabine (HDAC) post-remission resulted in CR rates of 92 and 74% in patients with FLT3-mutant and FLT3-wild-type AML, respectively [62]. This led to the development of the international, randomized, placebo-controlled, phase 3 RATIFY trial of midostaurin (50 mg twice daily) in combination with 7 + 3 in young untreated AML patients with FLT3 mutation (ITD or TKD) [63]. A total of 717 patients were randomized 1:1 to either midostaurin or placebo, and the midostaurin arm had significantly longer median OS (74.7 vs 26 months, p = 0.007) and EFS (8 vs 3 months, p = 0.004). There were no significant differences in CR (59 vs 54%) and AE rates between the groups. In another phase 1/2 study of midostaurin in combination with azacitidine in R/R AML and MDS, ORR was 26%, while patients with FLT3-ITD AML who were previously unexposed to FLT3 inhibitors had an ORR of 33% [64]. Two phase 2 studies are currently investigating midostaurin with azacitidine (NCT01093573) and decitabine (NCT01846624) in FLT3-ITD AML.
Addition of midostaurin to first-line treatment has shown remarkable survival benefit in FLT3-mutated young AML patients and is expected to reshape their standard of care.

Quizartinib

Quizartinib is a highly selective FLT3 inhibitor with 10-fold lower affinity for other kinases and the first of its class to achieve meaningful single-agent activity. In a phase 1 study, ORR in R/R AML patients was 17% in the entire cohort and 53% in patients with FLT3-ITD mutation [65]. In subsequent phase 2 studies of FLT3-ITD-mutated R/R AML, quizartinib monotherapy resulted in CRc and ORR rates of 44–54 and 61–72%, respectively [6668]. Median duration of response was 3 months. Moreover, a recently presented phase 1/2 study of quizartinib with azacitidine or LDAC in R/R myeloid leukemias demonstrated an ORR of 73% among patients with FLT3-ITD, and the median OS was 14.8 months for the entire cohort [69]. A randomized, open-label, phase 3 study of quizartinib monotherapy vs salvage chemotherapy in patients with FLT3-ITD-mutated R/R AML is currently recruiting participants (NCT02039726).
Response rates achieved with single-agent quizartinib are highly encouraging, but the short duration of response is of concern. Resistance to quizartinib is attributed to mutations in the TKD of the FLT3 gene; thus, agents that can overcome this resistance and offer durable remissions may substitute it during drug development and approval.

Crenolanib

Crenolanib is a selective pan-FLT3 inhibitor (ITD and TKD mutants) and can overcome quizartinib resistance with its activity against the D835 mutant FLT3. Up to 22% of patients develop a TKD mutation during FLT3 inhibitor therapy, but the activity of aforementioned inhibitors against secondary point mutations is limited [70]. In an open-label, single-center, phase 2 study of crenolanib in 38 R/R AML patients with FLT3-ITD or FLT3-TKD, ORR was 62% in FLT3 inhibitor-naïve patients and 38% in patients who had prior FLT3 inhibitor therapy [71]. Median OS was significantly longer in the former (55 vs 13 weeks, p = 0.03). This impressive clinical activity led to another phase 2 trial in which crenolanib in combination with standard 7 + 3 induction and HDAC consolidation was given to newly diagnosed AML patients with FLT3 mutations [72]. In the interim analysis of 25 patients, the combination was well tolerated and 96% achieved CRc, with 88% true CR. At a median follow-up of 6 months, only three patients (all >60 years) relapsed. OS was not reached, and the trial is ongoing. Finally, interim results of a phase 2 trial of crenolanib in combination with salvage idarubicin and HDAC in multiply R/R FLT3-mutated AML showed a CRc rate of 67% from a total of six patients who received ≤2 prior AML therapies, while no one with >2 prior treatments achieved CR [73]. Median OS was longer in the former (259 vs 53 days). This trial is now expanded to allow combination of crenolanib with other salvage regimens.

Gilteritinib

Gilteritinib is a potent, selective FLT3/AXL inhibitor with activity against both FLT3-ITD and FLT3-TKD mutations. In a large open-label phase 1/2 study of gilteritinib in R/R AML patients, 252 patients were enrolled and 70% had ≥2 prior AML therapies [74]. While patients with wild-type FLT3 had minimal benefit, ORR in 169 FLT3-mutated patients receiving ≥80 mg was 52%, median duration of response was 20 weeks, and median OS was 31 weeks. With these encouraging results, a randomized open-label phase 3 trial of gilteritinib vs salvage chemotherapy in R/R FLT3-mutated AML is ongoing (NCT02421939). In addition, a phase 1 study investigating the safety and efficacy of gilteritinib in combination with standard induction and consolidation in newly diagnosed adult patients with AML is ongoing (NCT02236013).
Clinical development of FLT3 inhibitors has been a vigorous effort during the past decade. Newer agents have demonstrated remarkable activity in FLT3-mutated R/R AML. However, short durations of response with quizartinib (3 months) and gilteritinib (5 months) can be a limiting factor for patients in whom HCT is not considered. Crenolanib will be an interesting agent to follow due to its reported single-agent activity and is currently also being investigated in combination with other salvage regimens. The RATIFY study has demonstrated benefit of combining midostaurin with standard induction and consolidation therapy of FLT3-mutated AML in patients less than 60 years of age, and we await results if this agent will become the first FDA-approved inhibitor in the upfront setting for younger AML patients. In addition, crenolanib was shown to be safe and effective with intensive chemotherapy, and if gilteritinib improves the outcomes of standard therapy in ongoing studies, these two newer agents may help continue the personalized approach to therapy with subsequent incremental improvements in clinical outcome.

Monoclonal antibodies

Antigen-specific immunotherapies targeting various cell surface proteins on leukemic myeloblasts and leukemic stem cells are in clinical development. Most of these trials, including anti-CD47 (NCT02678338), anti-CD25 (NCT02588092), anti-CD56 (NCT02420873), ipilimumab (i.e., anti-CTLA4) (NCT01757639), and nivolumab (i.e., anti-PD1) (NCT02464657, NCT02397720), have not yet reported any in-depth results; hence, they will not be discussed in this review. Among others, there is considerable interest in the novel antibody-drug conjugate (ADC) vadastuximab talirine (SGN-CD33A).
Vadastuximab is a CD33-directed antibody conjugated to pyrrolobenzodiazepine dimer, and upon binding, the compound is internalized and the dimer is released via proteolytic cleavage in lysosomes, which leads to DNA cross-linking and cell death (Fig. 2). In a phase 1 study of vadastuximab monotherapy in older patients with treatment-naïve CD33+ AML, 54% achieved CRc and ORR was 73% [75]. Another phase 1 study of vadastuximab (10 μg/kg) plus azacitidine or decitabine in untreated older AML patients reported CRc rate of 73%, median RFS of 9.1 months, and 60-day mortality rate of 8% [76]. In both of these studies, grade 4 myelosuppression was the most common AE, but the drug had no off-target AEs. These encouraging results have led to the phase 3 CASCADE trial investigating HMA with or without vadastuximab in older AML patients, which is currently accruing with a goal target of 500 patients (NCT02785900). Moreover, a phase 1b study of vadastuximab in combination with 7 + 3 induction therapy enrolled 42 newly diagnosed young (≤65 years) AML patients and showed acceptable toxicity profile with 78% CRc rate and 88% ORR [77].
Vadastuximab has remarkable clinical activity with no off-target toxicity as a single agent and with HMAs in elderly patients. If the results of CASCADE trial demonstrate survival benefit, it may improve the current standard of care for older adults who are ineligible for induction chemotherapy.

Cell cycle inhibitors

Inhibitors of cyclin-dependent kinases, Wee1, MDM2, aurora kinases, and polo-like kinases (PLKs) have been investigated in an effort to block the proliferation of leukemic myeloblasts. However, most of these agents showed little to no additional benefit to standard of care, and the most encouraging results have come from the PLK-inhibitor volasertib.
PLKs play an important role in many cellular processes including entry into mitosis, DNA replication, and stress response to DNA damage. PLK1 is overexpressed in AML cells, and its inhibition with volasertib leads to disrupted spindle formation and cell cycle arrest. In a phase 2 trial, 89 previously untreated AML patients who were unfit for intensive chemotherapy were randomized 1:1 to LDAC with or without volasertib [78]. The LDAC + volasertib arm had a higher CRc rate (31 vs 13.3%, p = 0.05), longer median EFS (5.6 vs 2.3 months, p = 0.02), and OS (8 vs 5.2 months, p = 0.04). A randomized, placebo-controlled, double-blind, phase 3 trial is ongoing to validate the efficacy and safety of this combination (NCT01721876). Combination of volasertib with decitabine in older AML patients is also under investigation (NCT02003573).

Other agents

BCL-2 inhibitors

Overexpression of the anti-apoptotic protein B-cell lymphoma 2 (BCL-2) has been implicated in AML cell survival and chemotherapy resistance [79]. Venetoclax is an orally bioavailable, selective BCL-2 inhibitor and showed promising preclinical activity in AML xenograft models [80]. In a single-arm phase 2 study, venetoclax monotherapy resulted in one CR and four CRs out of 28 patients evaluable, majority of whom had R/R AML [81]. Of note, 33% of patients with IDH1/2 mutations achieved CRc, which might suggest BCL-2 dependence in this subgroup. Despite its modest single-agent activity, venetoclax combined with LDAC or HMAs showed encouraging results in newly diagnosed older (≥65 years) AML patients who were ineligible for intensive therapy. In a phase 1b/2 study of 18 patients, venetoclax + LDAC demonstrated an ORR of 44% with acceptable tolerability [82]. Another phase 1b study evaluated 39 patients treated with venetoclax plus decitabine or azacitidine and showed an ORR of 76% (82% in IDH1/2-mutated patients) [83]. These initial findings demonstrate acceptable toxicity and promising clinical activity with venetoclax combinations in older untreated AML patients and warrant future randomized studies to further investigate the efficacy and biomarkers of response.

Aminopeptidase inhibitors

Aminopeptidases regulate protein turnover by hydrolyzing the terminal amino acids from peptides, which is thought to be an important source of amino acids for malignant cells. Tosedostat is an oral aminopeptidase inhibitor, which showed significant anti-leukemic activity as a single agent, with an ORR of 27% in 51 AML patients, majority of whom were R/R [84]. In the open-label phase 2 OPAL study, 76 older patients (≥60 years) with R/R AML were randomized 1:1 to two different doses of tosedostat (120 mg once daily for 6 months or 240 mg once daily for 2 months followed by 120 mg for 4 months) and the study reported an overall CRc rate of 10% [85]. Efficacy of tosedostat in combination with cytarabine or azacitidine was investigated in a phase 1/2 study of R/R AML or high-risk MDS patients and showed an ORR of 33% [86]. Finally, a phase 2 study of tosedostat with cytarabine or decitabine in newly diagnosed older patients with AML or high-risk MDS demonstrated a CRc rate of 53% and true CR rate of 41% [87]. The combination was well tolerated, and median OS was 11.5 months.

JAK/STAT inhibitors

JAK2 mutations or fusion proteins leading to constitutive activation of JAK2 have been implicated in myeloproliferative neoplasms and AML. An oral JAK2 inhibitor, pacritinib (SB1518), was shown to have a synergistic effect with pracinostat in preclinical AML models [88]. A phase 2 study of pacritinib in combination with decitabine or cytarabine in older AML patients is currently ongoing (NCT02532010).

Conclusions

AML is a complex heterogeneous disease with multiple recurrently mutated genes. With increased understanding of AML pathogenesis, there is a strong impetus for development of novel agents against various actionable targets. Some of these agents, like IDH inhibitors, demonstrated impressive single-agent activity, but rational combinations can offer the greatest benefit as far as the diverse genetic landscape is concerned. A decade from now, it is possible that CPX-351 may be added to the standard first-line induction therapy for older adults with secondary AML depending on the cost/benefit ratio. Additionally, novel agents like IDH inhibitors, FLT3 inhibitors, and vadastuximab (SGN-CD33A) may routinely be combined with traditional induction and consolidation therapy in select patients. Improvements in outcomes in R/R AML patients may be realized by combining standard salvage regimens with vosaroxin, guadecitabine, venetoclax, and perhaps even with tosedostat. Furthermore, efforts to boost the host immune system for improved tumor surveillance and killing by the addition of checkpoint inhibitors (ipilimumab, nivolumab and others) may change the delivery of therapy at several phases including intensive chemotherapy, and at the time of relapse, and in the post-HCT setting. Timely application of targeted therapies (e.g., FLT3 inhibitors) in the setting of post-transplant maintenance may also drive improvements for leukemic patients. Development of drugs for AML is a challenging task with a long history of multiple failures and minimal gains. However, in this genomic age with various promising agents, one should have no hesitation to expect a revolution.

Acknowledgements

We would like to thank Amanda Mendelsohn from the Cleveland Clinic Center for Medical Art and Photography for her exceptional help with the figures of this manuscript.

Funding

The authors declare no sources of funding.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Authors’ contributions

CS and HC reviewed the literature and wrote and approved the manuscript.

Competing interests

The authors declare that they have no competing interests.
Does not apply for this review article.
Does not apply for this review article.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Network CGAR. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Eng J Med. 2013;368(22):2059–74.CrossRef Network CGAR. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Eng J Med. 2013;368(22):2059–74.CrossRef
2.
Zurück zum Zitat Papaemmanuil EGM, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem G, Van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S, O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves MF, Ganser A, Döhner K, Schlenk RF, Döhner H, Campbell PJ. Genomic classification and prognosis in acute myeloid leukemia. N Eng J Med. 2016;374(23):2209–21.CrossRef Papaemmanuil EGM, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem G, Van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S, O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves MF, Ganser A, Döhner K, Schlenk RF, Döhner H, Campbell PJ. Genomic classification and prognosis in acute myeloid leukemia. N Eng J Med. 2016;374(23):2209–21.CrossRef
4.
Zurück zum Zitat Podoltsev NA SM, Zeidan AM, Gore SD. Selecting initial treatment of acute myeloid leukaemia in older adults. Blood Rev. 2016. In press. Podoltsev NA SM, Zeidan AM, Gore SD. Selecting initial treatment of acute myeloid leukaemia in older adults. Blood Rev. 2016. In press.
5.
Zurück zum Zitat Pleyer LBS, Stauder R, Girschikofsky M, Sill H, Schlick K, Thaler J, Halter B, Machherndl-Spandl S, Zebisch A, Pichler A, Pfeilstöcker M, Autzinger E-M, Lang A, Geissler K, Voskova D, Geissler D, Sperr WR, Hojas S, Rogulj IM, Andel J, Greil R. Azacitidine front-line in 339 patients with myelodysplastic syndromes and acute myeloid leukaemia: comparison of French-American-British and World Health Organization classifications. J Hematol Oncol. 2016;9(1):39.CrossRefPubMedPubMedCentral Pleyer LBS, Stauder R, Girschikofsky M, Sill H, Schlick K, Thaler J, Halter B, Machherndl-Spandl S, Zebisch A, Pichler A, Pfeilstöcker M, Autzinger E-M, Lang A, Geissler K, Voskova D, Geissler D, Sperr WR, Hojas S, Rogulj IM, Andel J, Greil R. Azacitidine front-line in 339 patients with myelodysplastic syndromes and acute myeloid leukaemia: comparison of French-American-British and World Health Organization classifications. J Hematol Oncol. 2016;9(1):39.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat LE Estey E, Lowenberg B. Current challenges in clinical development of “targeted therapies”: the case of acute myeloid leukemia. Blood. 2016;125(16):2461–6.CrossRef LE Estey E, Lowenberg B. Current challenges in clinical development of “targeted therapies”: the case of acute myeloid leukemia. Blood. 2016;125(16):2461–6.CrossRef
7.
Zurück zum Zitat Holowiecki JGS, Giebel S, Robak T, Kyrcz-Krzemien S, Kuliczkowski K, Skotnicki AB, Hellmann A, Sulek K, Dmoszynska A, Kloczko J, Jedrzejczak WW, Zdziarska B, Warzocha K, Zawilska K, Komarnicki M, Kielbinski M, Piatkowska-Jakubas B, Wierzbowska A, Wach M, Haus O. Cladribine, but not fludarabine, added to daunorubicin and cytarabine during induction prolongs survival of patients with acute myeloid leukemia: a multicenter, randomized phase III study. J Clin Oncol. 2012;30(20):2441–8.CrossRefPubMed Holowiecki JGS, Giebel S, Robak T, Kyrcz-Krzemien S, Kuliczkowski K, Skotnicki AB, Hellmann A, Sulek K, Dmoszynska A, Kloczko J, Jedrzejczak WW, Zdziarska B, Warzocha K, Zawilska K, Komarnicki M, Kielbinski M, Piatkowska-Jakubas B, Wierzbowska A, Wach M, Haus O. Cladribine, but not fludarabine, added to daunorubicin and cytarabine during induction prolongs survival of patients with acute myeloid leukemia: a multicenter, randomized phase III study. J Clin Oncol. 2012;30(20):2441–8.CrossRefPubMed
8.
Zurück zum Zitat Bishop JFLR, Joshua D. Etoposide in acute nonlymphocytic leukemia: Australian Leukemia Study Group. Blood. 1990;75:27–32.PubMed Bishop JFLR, Joshua D. Etoposide in acute nonlymphocytic leukemia: Australian Leukemia Study Group. Blood. 1990;75:27–32.PubMed
9.
Zurück zum Zitat Fernandez HFSZ, Yao X, Litzow MR, Luger SM, Paietta EM, Racevskis J, Dewald GW, Ketterling RP, Bennett JM, Rowe JM, Lazarus HM, Tallman MS. Anthracycline dose intensification in acute myeloid leukemia. N Eng J Med. 2009;361(13):1249–59.CrossRef Fernandez HFSZ, Yao X, Litzow MR, Luger SM, Paietta EM, Racevskis J, Dewald GW, Ketterling RP, Bennett JM, Rowe JM, Lazarus HM, Tallman MS. Anthracycline dose intensification in acute myeloid leukemia. N Eng J Med. 2009;361(13):1249–59.CrossRef
10.
Zurück zum Zitat Lancet JECJ, Hogge DE, Tallman MS, Kovacsovics TJ, Damon LE, Komrokji R, Solomon SR, Kolitz JE, Cooper M, Yeager AM, Louie AC, Feldman EJ. Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood. 2014;123(21):3239–46.CrossRefPubMedPubMedCentral Lancet JECJ, Hogge DE, Tallman MS, Kovacsovics TJ, Damon LE, Komrokji R, Solomon SR, Kolitz JE, Cooper M, Yeager AM, Louie AC, Feldman EJ. Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood. 2014;123(21):3239–46.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Mayer LDHT, Tardi PG, Harasym NL, Shew CR, Johnstone SA, Ramsay EC, Bally MB, Janoff AS. Ratiometric dosing of anticancer drug combinations: controlling drug ratios after systemic administration regulates therapeutic activity in tumor-bearing mice. Mol Cancer Ther. 2006;5(7):1854–63.CrossRefPubMed Mayer LDHT, Tardi PG, Harasym NL, Shew CR, Johnstone SA, Ramsay EC, Bally MB, Janoff AS. Ratiometric dosing of anticancer drug combinations: controlling drug ratios after systemic administration regulates therapeutic activity in tumor-bearing mice. Mol Cancer Ther. 2006;5(7):1854–63.CrossRefPubMed
12.
Zurück zum Zitat Cortes JEGS, Feldman EJ, Rizzeri DA, Hogge DE, Larson M, Pigneux A, Recher C, Schiller G, Warzocha K, Kantarjian H, Louie AC, Kolitz JE. Phase II, multicenter, randomized trial of CPX-351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML. Cancer. 2015;121(2):234–42.CrossRefPubMed Cortes JEGS, Feldman EJ, Rizzeri DA, Hogge DE, Larson M, Pigneux A, Recher C, Schiller G, Warzocha K, Kantarjian H, Louie AC, Kolitz JE. Phase II, multicenter, randomized trial of CPX-351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML. Cancer. 2015;121(2):234–42.CrossRefPubMed
13.
Zurück zum Zitat Lancet JE UG, Cortes JE, Newell LF, Lin TL, Ritchie EK, Stuart RK, Strickland SA, Hogge D, Solomon SR, Stone RM, Bixby DL, Kolitz JE, Schiller GJ, Wieduwilt MJ, Ryan DH, Hoering A, Chiarella M, Louie AC, Medeiros BC. Final results of a phase III randomized trial of CPX-351 versus 7+3 in older patients with newly diagnosed high risk (secondary) AML. J Clin Oncol. 2016;34(suppl; abstr 7000). Lancet JE UG, Cortes JE, Newell LF, Lin TL, Ritchie EK, Stuart RK, Strickland SA, Hogge D, Solomon SR, Stone RM, Bixby DL, Kolitz JE, Schiller GJ, Wieduwilt MJ, Ryan DH, Hoering A, Chiarella M, Louie AC, Medeiros BC. Final results of a phase III randomized trial of CPX-351 versus 7+3 in older patients with newly diagnosed high risk (secondary) AML. J Clin Oncol. 2016;34(suppl; abstr 7000).
14.
Zurück zum Zitat Medeiros BCLJ, Cortes JE, Newell LF, Lin TL, Ritchie EK. Analysis of efficacy by age for patients aged 60–75 with untreated secondary acute myeloid leukemia (AML) treated with CPX-351 liposome injection versus conventional cytarabine and daunorubicin in a phase III trial. Blood. 2016;128(22):2780. Medeiros BCLJ, Cortes JE, Newell LF, Lin TL, Ritchie EK. Analysis of efficacy by age for patients aged 60–75 with untreated secondary acute myeloid leukemia (AML) treated with CPX-351 liposome injection versus conventional cytarabine and daunorubicin in a phase III trial. Blood. 2016;128(22):2780.
15.
Zurück zum Zitat Lancet JEHA, Uy GL, Cortes JE, Newell LF, Lin TL, Ritchie EK, Stuart RK, Strickland SA, Hogge D, Solomon SR, Stone RM, Bixby DL, Kolitz JE, Schiller GJ, Wieduwilt MJ, Ryan DH, Chiarella MT, Louie AC, Medeiros BC. Survival following allogeneic hematopoietic cell transplantation in older high-risk acute myeloid leukemia patients initially treated with CPX-351 liposome injection versus standard cytarabine and daunorubicin: subgroup analysis of a large phase III trial. Blood. 2016;128(22):906. Lancet JEHA, Uy GL, Cortes JE, Newell LF, Lin TL, Ritchie EK, Stuart RK, Strickland SA, Hogge D, Solomon SR, Stone RM, Bixby DL, Kolitz JE, Schiller GJ, Wieduwilt MJ, Ryan DH, Chiarella MT, Louie AC, Medeiros BC. Survival following allogeneic hematopoietic cell transplantation in older high-risk acute myeloid leukemia patients initially treated with CPX-351 liposome injection versus standard cytarabine and daunorubicin: subgroup analysis of a large phase III trial. Blood. 2016;128(22):906.
16.
Zurück zum Zitat Assi RKH, Verstovsek S, Garcia-Manero G, Konopleva M, Ravandi F. CPX-351 for the treatment of high-risk patients with acute myeloid leukemia. Blood. 2016;128(22):4047. Assi RKH, Verstovsek S, Garcia-Manero G, Konopleva M, Ravandi F. CPX-351 for the treatment of high-risk patients with acute myeloid leukemia. Blood. 2016;128(22):4047.
17.
Zurück zum Zitat Lancet JERF, Ricklis RM, Cripe LD, Kantarjian HM, Giles FJ, List AF, Chen T, Allen RS, Fox JA, Michelson GC, Karp JE. A phase Ib study of vosaroxin, an anticancer quinolone derivative, in patients with relapsed or refractory acute leukemia. Leukemia. 2011;25(12):1808–14.CrossRefPubMedPubMedCentral Lancet JERF, Ricklis RM, Cripe LD, Kantarjian HM, Giles FJ, List AF, Chen T, Allen RS, Fox JA, Michelson GC, Karp JE. A phase Ib study of vosaroxin, an anticancer quinolone derivative, in patients with relapsed or refractory acute leukemia. Leukemia. 2011;25(12):1808–14.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Lancet JERG, Cripe LD, Michelson GC, Fox JA, Leavitt RD, Chen T, Hawtin R, Craig AR, Ravandi F, Maris MB, Stuart RK, Karp JE. A phase 1b/2 study of vosaroxin in combination with cytarabine in patients with relapsed or refractory acute myeloid leukemia. Haematologica. 2015;100(2):231–7.CrossRefPubMedPubMedCentral Lancet JERG, Cripe LD, Michelson GC, Fox JA, Leavitt RD, Chen T, Hawtin R, Craig AR, Ravandi F, Maris MB, Stuart RK, Karp JE. A phase 1b/2 study of vosaroxin in combination with cytarabine in patients with relapsed or refractory acute myeloid leukemia. Haematologica. 2015;100(2):231–7.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Ravandi FRE, Sayar H, Lancet JE, Craig MD, Vey N, Strickland SA, Schiller GJ, Jabbour E, Erba HP, Pigneux A, Horst HA, Recher C, Klimek VM, Cortes J, Roboz GJ, Odenike O, Thomas X, Havelange V, Maertens J, Derigs HG, Heuser M, Damon L, Powell BL, Gaidano G, Carella AM, Wei A, Hogge D, Craig AR, Fox JA, Ward R, Smith JA, Acton G, Mehta C, Stuart RK, Kantarjian HM. Vosaroxin plus cytarabine versus placebo plus cytarabine in patients with first relapsed or refractory acute myeloid leukaemia (VALOR): a randomised, controlled, double-blind, multinational, phase 3 study. Lancet Oncol. 2015;16(9):1025–36.CrossRefPubMedPubMedCentral Ravandi FRE, Sayar H, Lancet JE, Craig MD, Vey N, Strickland SA, Schiller GJ, Jabbour E, Erba HP, Pigneux A, Horst HA, Recher C, Klimek VM, Cortes J, Roboz GJ, Odenike O, Thomas X, Havelange V, Maertens J, Derigs HG, Heuser M, Damon L, Powell BL, Gaidano G, Carella AM, Wei A, Hogge D, Craig AR, Fox JA, Ward R, Smith JA, Acton G, Mehta C, Stuart RK, Kantarjian HM. Vosaroxin plus cytarabine versus placebo plus cytarabine in patients with first relapsed or refractory acute myeloid leukaemia (VALOR): a randomised, controlled, double-blind, multinational, phase 3 study. Lancet Oncol. 2015;16(9):1025–36.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Ravandi FRE, Sayar H, Lancet JE, Craig MD, Vey N, Strickland SA, Schiller GJ, Jabbour E, Erba HP, Pigneux A, Horst HA, Recher C, Klimek VM, Cortes J, Roboz GJ, Craig A, Ward R, Smith J, Kantarjian H, Stuart RK. Durable overall survival benefit in patients ≥ 60 years with relapsed or refractory AML treated with vosaroxin/cytarabine vs placebo/cytarabine: updated results from the VALOR trial. Blood. 2016;128(22):903. Ravandi FRE, Sayar H, Lancet JE, Craig MD, Vey N, Strickland SA, Schiller GJ, Jabbour E, Erba HP, Pigneux A, Horst HA, Recher C, Klimek VM, Cortes J, Roboz GJ, Craig A, Ward R, Smith J, Kantarjian H, Stuart RK. Durable overall survival benefit in patients ≥ 60 years with relapsed or refractory AML treated with vosaroxin/cytarabine vs placebo/cytarabine: updated results from the VALOR trial. Blood. 2016;128(22):903.
21.
Zurück zum Zitat Stuart RKCL, Maris MB, Cooper MA, Stone RM, Dakhil SR, Turturro F, Stock W, Mason J, Shami PJ, Strickland SA, Costa LJ, Borthakur G, Michelson GC, Fox JA, Leavitt RD, Ravandi F. REVEAL-1, a phase 2 dose regimen optimization study of vosaroxin in older poor-risk patients with previously untreated acute myeloid leukaemia. Br J Haematol. 2015;168(6):796–805.CrossRefPubMed Stuart RKCL, Maris MB, Cooper MA, Stone RM, Dakhil SR, Turturro F, Stock W, Mason J, Shami PJ, Strickland SA, Costa LJ, Borthakur G, Michelson GC, Fox JA, Leavitt RD, Ravandi F. REVEAL-1, a phase 2 dose regimen optimization study of vosaroxin in older poor-risk patients with previously untreated acute myeloid leukaemia. Br J Haematol. 2015;168(6):796–805.CrossRefPubMed
22.
Zurück zum Zitat Dennis MRN, Hills RK, Hemmaway C, Panoskaltsis N, McMullin MF, Kjeldsen L, Dignum H, Thomas IF, Clark RE, Milligan D, Burnett AK. Vosaroxin and vosaroxin plus low-dose Ara-C (LDAC) vs low-dose Ara-C alone in older patients with acute myeloid leukemia. Blood. 2015;125(19):2923–32.CrossRefPubMedPubMedCentral Dennis MRN, Hills RK, Hemmaway C, Panoskaltsis N, McMullin MF, Kjeldsen L, Dignum H, Thomas IF, Clark RE, Milligan D, Burnett AK. Vosaroxin and vosaroxin plus low-dose Ara-C (LDAC) vs low-dose Ara-C alone in older patients with acute myeloid leukemia. Blood. 2015;125(19):2923–32.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Kropf P JE, Yee K, O’Connell C, Tibes R, Roboz GJ, Walsh K, Podoltsev NA, Savona M, Issa JP, Hao Y, Naim S, Azab M, Kantarjian H. Late responses and overall survival (OS) from long term follow up of a randomized phase 2 study of SGI-110 (guadecitabine) 5-day regimen in elderly AML who are not eligible for intensive chemotherapy. In: 20th Congress of the European Hematology Association [Abstract P571]. Vienna: European Hematology Association; 2015. Kropf P JE, Yee K, O’Connell C, Tibes R, Roboz GJ, Walsh K, Podoltsev NA, Savona M, Issa JP, Hao Y, Naim S, Azab M, Kantarjian H. Late responses and overall survival (OS) from long term follow up of a randomized phase 2 study of SGI-110 (guadecitabine) 5-day regimen in elderly AML who are not eligible for intensive chemotherapy. In: 20th Congress of the European Hematology Association [Abstract P571]. Vienna: European Hematology Association; 2015.
25.
Zurück zum Zitat Daver N KH, Roboz GJ, Kropf PL, Yee KW, O’Connell CL, Griffiths EA, Jabbour EJ, Stock W, Walsh KJ, Rizzieri DA, Berdeja JG, Su XY, Azab M, Issa PJ. Long term survival and clinical complete responses of various prognostic subgroups in 103 relapsed/refractory acute myeloid leukemia (r/r AML) patients treated with guadecitabine (SGI-110) in phase 2 studies. Blood. 2016;(suppl; abstract 904). Daver N KH, Roboz GJ, Kropf PL, Yee KW, O’Connell CL, Griffiths EA, Jabbour EJ, Stock W, Walsh KJ, Rizzieri DA, Berdeja JG, Su XY, Azab M, Issa PJ. Long term survival and clinical complete responses of various prognostic subgroups in 103 relapsed/refractory acute myeloid leukemia (r/r AML) patients treated with guadecitabine (SGI-110) in phase 2 studies. Blood. 2016;(suppl; abstract 904).
26.
Zurück zum Zitat Figueroa ME, Lugthart S, Li Y, Erpelinck-Verschueren C, Deng X, Christos PJ, Schifano E, Booth J, van Putten W, Skrabanek L, Campagne F, Mazumdar M, Greally JM, Valk PJ, Löwenberg B, Delwel R, Melnick A. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell. 2010;17(1):13–27.CrossRefPubMedPubMedCentral Figueroa ME, Lugthart S, Li Y, Erpelinck-Verschueren C, Deng X, Christos PJ, Schifano E, Booth J, van Putten W, Skrabanek L, Campagne F, Mazumdar M, Greally JM, Valk PJ, Löwenberg B, Delwel R, Melnick A. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell. 2010;17(1):13–27.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, Tallman MS, Sun Z, Wolniak K, Peeters JK, Liu W, Choe SE, Fantin VR, Paietta E, Lowenberg B, Licht JD, Godley LA, Delwel R, Valk PJ, Thompson CB, Levine RL, Melnick A. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–67.CrossRefPubMedPubMedCentral Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, Tallman MS, Sun Z, Wolniak K, Peeters JK, Liu W, Choe SE, Fantin VR, Paietta E, Lowenberg B, Licht JD, Godley LA, Delwel R, Valk PJ, Thompson CB, Levine RL, Melnick A. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–67.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Meldi K, Qin T, Buchi F, Droin N, Sotzen J, Micol JB, Selimoglu-Buet D, Masala E, Allione B, Gioia D, Poloni A, Lunghi M, Solary E, Abdel-Wahab O, Santini V, Figueroa ME. Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia. J Clin Invest. 2015;125(5):1857–72.CrossRefPubMedPubMedCentral Meldi K, Qin T, Buchi F, Droin N, Sotzen J, Micol JB, Selimoglu-Buet D, Masala E, Allione B, Gioia D, Poloni A, Lunghi M, Solary E, Abdel-Wahab O, Santini V, Figueroa ME. Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia. J Clin Invest. 2015;125(5):1857–72.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Traina F, Visconte V, Elson P, Tabarroki A, Jankowska AM, Hasrouni E, Sugimoto Y, Szpurka H, Makishima H, O’Keefe CL, Sekeres MA, Advani AS, Kalaycio M, Copelan EA, Saunthararajah Y, Olalla Saad ST, Maciejewski JP, Tiu RV. Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia. 2014;28(1):78–87.CrossRefPubMed Traina F, Visconte V, Elson P, Tabarroki A, Jankowska AM, Hasrouni E, Sugimoto Y, Szpurka H, Makishima H, O’Keefe CL, Sekeres MA, Advani AS, Kalaycio M, Copelan EA, Saunthararajah Y, Olalla Saad ST, Maciejewski JP, Tiu RV. Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia. 2014;28(1):78–87.CrossRefPubMed
30.
Zurück zum Zitat Medeiros BC FA, DiNardo CD, Pollyea DA, Chan SM, Swords R. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia. 2016. In press. Medeiros BC FA, DiNardo CD, Pollyea DA, Chan SM, Swords R. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia. 2016. In press.
31.
Zurück zum Zitat DiNardo CD dBS, Pollyea DA, Stein EM, Fathi AT, Roboz GJ, Collins R, Swords RT, Flinn IW, Altman JK, Tallman MS, Kantarjian HM, Derti A, Goldwasser M, Prahl M, Wu B, Yen K, Agresta S, Stone RM. Molecular profiling and relationship with clinical response in patients with IDH1 mutation-positive hematologic malignancies receiving AG-120, a first-in-class potent inhibitor of mutant IDH1, in addition to data from the completed dose escalation portion of the phase 1 study. Blood. 2015;126:1306.CrossRef DiNardo CD dBS, Pollyea DA, Stein EM, Fathi AT, Roboz GJ, Collins R, Swords RT, Flinn IW, Altman JK, Tallman MS, Kantarjian HM, Derti A, Goldwasser M, Prahl M, Wu B, Yen K, Agresta S, Stone RM. Molecular profiling and relationship with clinical response in patients with IDH1 mutation-positive hematologic malignancies receiving AG-120, a first-in-class potent inhibitor of mutant IDH1, in addition to data from the completed dose escalation portion of the phase 1 study. Blood. 2015;126:1306.CrossRef
32.
Zurück zum Zitat Birendra KC, DiNardo CD. Evidence for clinical differentiation and differentiation syndrome in patients with acute myeloid leukemia and IDH1 mutations treated with the targeted mutant IDH1 inhibitor, AG-120. Clin Lymphoma Myeloma Leuk. 2016;16(8):460–5.CrossRefPubMed Birendra KC, DiNardo CD. Evidence for clinical differentiation and differentiation syndrome in patients with acute myeloid leukemia and IDH1 mutations treated with the targeted mutant IDH1 inhibitor, AG-120. Clin Lymphoma Myeloma Leuk. 2016;16(8):460–5.CrossRefPubMed
33.
Zurück zum Zitat DiNardo CDSA, Yee KW, Hochhaus A, Kraemer A, Carvajal RD, Janku F, Bedard P, Carpio C, Wick A, Schwartz GK, Schöffski P, Wen P, van den Bent MJ, Rosenthal M, O’Keeffe M, Chen X, Pagliarini R, Schuck V, Myers A, Wei A. A phase I study of IDH305 in patients with advanced malignancies including relapsed/refractory AML and MDS that harbor IDH1R132 mutations. Blood. 2016;128(22):1073. DiNardo CDSA, Yee KW, Hochhaus A, Kraemer A, Carvajal RD, Janku F, Bedard P, Carpio C, Wick A, Schwartz GK, Schöffski P, Wen P, van den Bent MJ, Rosenthal M, O’Keeffe M, Chen X, Pagliarini R, Schuck V, Myers A, Wei A. A phase I study of IDH305 in patients with advanced malignancies including relapsed/refractory AML and MDS that harbor IDH1R132 mutations. Blood. 2016;128(22):1073.
34.
Zurück zum Zitat Stein EMDC, Altman JK, Collins R, DeAngelo DJ, Kantarjian HM, Sekeres MA, Fathi AT, Flinn IW, Frankel AE, Levine RL, Medeiros BC, Patel MR, Pollyea D, Roboz GJ, Stone RM, Swords RT, Tallman MS, Yen K, Attar EC, Xu Q, Tosolini A, Mei JM, Thakurta A, Knight RD, De Botton S. Safety and efficacy of AG-221, a potent inhibitor of mutant IDH2 that promotes differentiation of myeloid cells in patients with advanced hematologic malignancies: results of a phase 1/2 trial. Blood. 2015;126:323. Stein EMDC, Altman JK, Collins R, DeAngelo DJ, Kantarjian HM, Sekeres MA, Fathi AT, Flinn IW, Frankel AE, Levine RL, Medeiros BC, Patel MR, Pollyea D, Roboz GJ, Stone RM, Swords RT, Tallman MS, Yen K, Attar EC, Xu Q, Tosolini A, Mei JM, Thakurta A, Knight RD, De Botton S. Safety and efficacy of AG-221, a potent inhibitor of mutant IDH2 that promotes differentiation of myeloid cells in patients with advanced hematologic malignancies: results of a phase 1/2 trial. Blood. 2015;126:323.
35.
Zurück zum Zitat Garcia-Manero GYH, Bueso-Ramos C, Ferrajoli A, Cortes J, Wierda WG, Faderl S, Koller C, Morris G, Rosner G, Loboda A, Fantin VR, Randolph SS, Hardwick JS, Reilly JF, Chen C, Ricker JL, Secrist JP, Richon VM, Frankel SR, Kantarjian HM. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 2008;111(3):1060–6.CrossRefPubMed Garcia-Manero GYH, Bueso-Ramos C, Ferrajoli A, Cortes J, Wierda WG, Faderl S, Koller C, Morris G, Rosner G, Loboda A, Fantin VR, Randolph SS, Hardwick JS, Reilly JF, Chen C, Ricker JL, Secrist JP, Richon VM, Frankel SR, Kantarjian HM. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 2008;111(3):1060–6.CrossRefPubMed
36.
Zurück zum Zitat Garcia-Manero GAS, Cortes J, Estrov Z, Kantarjian H, Yang H, Newsome WM, Miller Jr WH, Rousseau C, Kalita A, Bonfils C, Dubay M, Patterson TA, Li Z, Besterman JM, Reid G, Laille E, Martell RE, Minden M. Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia. Blood. 2008;112(4):981–9.CrossRefPubMedPubMedCentral Garcia-Manero GAS, Cortes J, Estrov Z, Kantarjian H, Yang H, Newsome WM, Miller Jr WH, Rousseau C, Kalita A, Bonfils C, Dubay M, Patterson TA, Li Z, Besterman JM, Reid G, Laille E, Martell RE, Minden M. Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia. Blood. 2008;112(4):981–9.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Gojo IJA, Trepel JB, Sparreboom A, Figg WD, Rollins S, Tidwell ML, Greer J, Chung EJ, Lee MJ, Gore SD, Sausville EA, Zwiebel J, Karp JE. Phase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemias. Blood. 2007;109(7):2781–90.PubMedPubMedCentral Gojo IJA, Trepel JB, Sparreboom A, Figg WD, Rollins S, Tidwell ML, Greer J, Chung EJ, Lee MJ, Gore SD, Sausville EA, Zwiebel J, Karp JE. Phase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemias. Blood. 2007;109(7):2781–90.PubMedPubMedCentral
38.
Zurück zum Zitat Tan PWA, Mithraprabhu S, Cummings N, Liu HB, Perugini M, Reed K, Avery S, Patil S, Walker P, Mollee P, Grigg A, D’Andrea R, Dear A, Spencer A. Dual epigenetic targeting with panobinostat and azacitidine in acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood Cancer J. 2014;4:e170.CrossRefPubMedPubMedCentral Tan PWA, Mithraprabhu S, Cummings N, Liu HB, Perugini M, Reed K, Avery S, Patil S, Walker P, Mollee P, Grigg A, D’Andrea R, Dear A, Spencer A. Dual epigenetic targeting with panobinostat and azacitidine in acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood Cancer J. 2014;4:e170.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Ocio EMHP, Olave MT, Castro N, Pérez-Simón JA, Brunet S, Oriol A, Mateo M, Sanz MÁ, López J, Montesinos P, Chillón MC, Prieto-Conde MI, Díez-Campelo M, González M, Vidriales MB, Mateos MV, San Miguel JF, PETHEMA Group. Panobinostat as part of induction and maintenance for elderly patients with newly diagnosed acute myeloid leukemia: phase Ib/II panobidara study. Haematologica. 2015;100(10):1294–300.CrossRefPubMedPubMedCentral Ocio EMHP, Olave MT, Castro N, Pérez-Simón JA, Brunet S, Oriol A, Mateo M, Sanz MÁ, López J, Montesinos P, Chillón MC, Prieto-Conde MI, Díez-Campelo M, González M, Vidriales MB, Mateos MV, San Miguel JF, PETHEMA Group. Panobinostat as part of induction and maintenance for elderly patients with newly diagnosed acute myeloid leukemia: phase Ib/II panobidara study. Haematologica. 2015;100(10):1294–300.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Garcia-Manero GTF, Bekele NB, Yang H, Ravandi F, Jabbour E, Borthakur G, Kadia TM, Konopleva MY, Faderl S, Cortes JE, Brandt M, Hu Y, McCue D, Newsome WM, Pierce SR, de Lima M, Kantarjian HM. 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(18):2204–10.CrossRefPubMedPubMedCentral Garcia-Manero GTF, Bekele NB, Yang H, Ravandi F, Jabbour E, Borthakur G, Kadia TM, Konopleva MY, Faderl S, Cortes JE, Brandt M, Hu Y, McCue D, Newsome WM, Pierce SR, de Lima M, Kantarjian HM. 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(18):2204–10.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Garcia-Manero GOM, Pagel JM, Radich JP, Fang M, Rizzieri DA, Marcucci G, Strickland SA, Litzow M, Savoie ML, Medeiros BC, Sekeres MA, Lin TL, Uy GL, Powell BL, Kolitz JE, Larson RA, Stone RM, Claxton DF, Essell J, Luger S, Mohan SR, Moseley A, Appelbaum FR, Erba HP. SWOG S1203: a randomized phase III study of standard cytarabine plus daunorubicin (7+3) therapy versus idarubicin with high dose cytarabine (IA) with or without vorinostat (IA+V) in younger patients with previously untreated acute myeloid leukemia (AML). Blood. 2016;128(22):901. Garcia-Manero GOM, Pagel JM, Radich JP, Fang M, Rizzieri DA, Marcucci G, Strickland SA, Litzow M, Savoie ML, Medeiros BC, Sekeres MA, Lin TL, Uy GL, Powell BL, Kolitz JE, Larson RA, Stone RM, Claxton DF, Essell J, Luger S, Mohan SR, Moseley A, Appelbaum FR, Erba HP. SWOG S1203: a randomized phase III study of standard cytarabine plus daunorubicin (7+3) therapy versus idarubicin with high dose cytarabine (IA) with or without vorinostat (IA+V) in younger patients with previously untreated acute myeloid leukemia (AML). Blood. 2016;128(22):901.
42.
Zurück zum Zitat Kirschbaum MGI, Goldberg SL, Bredeson C, Kujawski LA, Yang A, Marks P, Frankel P, Sun X, Tosolini A, Eid JE, Lubiniecki GM, Issa JP. A phase 1 clinical trial of vorinostat in combination with decitabine in patients with acute myeloid leukaemia or myelodysplastic syndrome. Br J Haematol. 2014;167(2):185–93.CrossRefPubMed Kirschbaum MGI, Goldberg SL, Bredeson C, Kujawski LA, Yang A, Marks P, Frankel P, Sun X, Tosolini A, Eid JE, Lubiniecki GM, Issa JP. A phase 1 clinical trial of vorinostat in combination with decitabine in patients with acute myeloid leukaemia or myelodysplastic syndrome. Br J Haematol. 2014;167(2):185–93.CrossRefPubMed
43.
Zurück zum Zitat Mims ASKR, Garzon R, Walker AR, Devine SM, Walsh KJ, Vasu S, Whitman SP, Marcucci G, Heerema NA, Caligiuri MA, Bloomfield CD, Byrd JC, Piekarz R, Grever MR, Blum W. A novel regimen for acute myeloid leukemia with MLL partial tandem duplication: results of a phase 1 study NCI 8485. Blood. 2016;128(22):900. Mims ASKR, Garzon R, Walker AR, Devine SM, Walsh KJ, Vasu S, Whitman SP, Marcucci G, Heerema NA, Caligiuri MA, Bloomfield CD, Byrd JC, Piekarz R, Grever MR, Blum W. A novel regimen for acute myeloid leukemia with MLL partial tandem duplication: results of a phase 1 study NCI 8485. Blood. 2016;128(22):900.
44.
Zurück zum Zitat Sekeres MAOM, List AF, Odenike O, Stone RM, Gore SD, Litzow MR, Buckstein R, Fang M, Roulston D, Bloomfield CD, Zhang Y, Velasco MR, Gaur R, Atallah E, Attar EC, Appelbaum FR, Erba HP. Additional analyses of a randomized phase II study of azacitidine combined with lenalidomide or with vorinostat vs. azacitidine monotherapy in higher-risk myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML): North American Intergroup Study SWOG S1117. Blood. 2015;126(23):908. Sekeres MAOM, List AF, Odenike O, Stone RM, Gore SD, Litzow MR, Buckstein R, Fang M, Roulston D, Bloomfield CD, Zhang Y, Velasco MR, Gaur R, Atallah E, Attar EC, Appelbaum FR, Erba HP. Additional analyses of a randomized phase II study of azacitidine combined with lenalidomide or with vorinostat vs. azacitidine monotherapy in higher-risk myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML): North American Intergroup Study SWOG S1117. Blood. 2015;126(23):908.
45.
Zurück zum Zitat Prebet TSZ, Figueroa ME, Ketterling R, Melnick A, Greenberg PL, Herman J, Juckett M, Smith MR, Malick L, Paietta E, Czader M, Litzow M, Gabrilove J, Erba HP, Gore SD, Tallman MS. 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(12):1242–8.CrossRefPubMedPubMedCentral Prebet TSZ, Figueroa ME, Ketterling R, Melnick A, Greenberg PL, Herman J, Juckett M, Smith MR, Malick L, Paietta E, Czader M, Litzow M, Gabrilove J, Erba HP, Gore SD, Tallman MS. 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(12):1242–8.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Garcia-Manero G AE, Khaled S, Arellano M, Patnaik M, Esquibel V, Wood K, Medeiros B. Updated results from a phase 2 study of pracinostat (P) in combination With azacitidine (AZA) in elderly patients with acute myeloid leukemia (AML). In: European Hematology Association (EHA). vol. Abstract P568. Vienna, Italy; 2015. Garcia-Manero G AE, Khaled S, Arellano M, Patnaik M, Esquibel V, Wood K, Medeiros B. Updated results from a phase 2 study of pracinostat (P) in combination With azacitidine (AZA) in elderly patients with acute myeloid leukemia (AML). In: European Hematology Association (EHA). vol. Abstract P568. Vienna, Italy; 2015.
47.
Zurück zum Zitat Berthon CRE, Thomas X, Vey N, Gomez-Roca C, Yee K, Taussig DC, Rezai K, Roumier C, Herait P, Kahatt C, Quesnel B, Michallet M, Recher C, Lokiec F, Preudhomme C, Dombret H. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 2016;3(4):e186–95.CrossRefPubMed Berthon CRE, Thomas X, Vey N, Gomez-Roca C, Yee K, Taussig DC, Rezai K, Roumier C, Herait P, Kahatt C, Quesnel B, Michallet M, Recher C, Lokiec F, Preudhomme C, Dombret H. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 2016;3(4):e186–95.CrossRefPubMed
48.
Zurück zum Zitat Krivtsov AVAS. MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 2007;7(11):823–33.CrossRefPubMed Krivtsov AVAS. MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 2007;7(11):823–33.CrossRefPubMed
49.
Zurück zum Zitat Stein EMG-MG, Rizzieri DA, Tibes R, Berdeja JG, Jongen-Lavrencic M, Altman JK, Dohner H, Thomson B, Blakemore SJ, Daigle S, Fine G, Waters NJ, Krivstov AV, Koche R, Armstrong SA, Ho PT, Lowenberg B, Tallman MS. A phase 1 study of the DOT1L inhibitor, pinometostat (EPZ-5676), in adults with relapsed or refractory leukemia: safety, clinical activity, exposure and target inhibition. Blood. 2015;126(23):2547. Stein EMG-MG, Rizzieri DA, Tibes R, Berdeja JG, Jongen-Lavrencic M, Altman JK, Dohner H, Thomson B, Blakemore SJ, Daigle S, Fine G, Waters NJ, Krivstov AV, Koche R, Armstrong SA, Ho PT, Lowenberg B, Tallman MS. A phase 1 study of the DOT1L inhibitor, pinometostat (EPZ-5676), in adults with relapsed or refractory leukemia: safety, clinical activity, exposure and target inhibition. Blood. 2015;126(23):2547.
50.
Zurück zum Zitat Shukla NWC, O’Brien MM, Silverman LB, Brown P, Cooper TM, Thomson B, Blakemore SJ, Daigle S, Suttle B, Waters NJ, Krivstov AV, Armstrong SA, Ho PT, Gore L. Final report of phase 1 study of the DOT1L inhibitor, pinometostat (EPZ-5676), in children with relapsed or refractory MLL-r acute leukemia. Blood. 2016;128(22):2780. Shukla NWC, O’Brien MM, Silverman LB, Brown P, Cooper TM, Thomson B, Blakemore SJ, Daigle S, Suttle B, Waters NJ, Krivstov AV, Armstrong SA, Ho PT, Gore L. Final report of phase 1 study of the DOT1L inhibitor, pinometostat (EPZ-5676), in children with relapsed or refractory MLL-r acute leukemia. Blood. 2016;128(22):2780.
51.
Zurück zum Zitat Kühn MWSE, Feng Z, Sinha A, Chen CW, Deshpande AJ, Cusan M, Farnoud N, Mupo A, Grove C, Koche R, Bradner JE, de Stanchina E, Vassiliou GS, Hoshii T, Armstrong SA. Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia. Cancer Discov. 2016;6(10):1166–81.CrossRefPubMed Kühn MWSE, Feng Z, Sinha A, Chen CW, Deshpande AJ, Cusan M, Farnoud N, Mupo A, Grove C, Koche R, Bradner JE, de Stanchina E, Vassiliou GS, Hoshii T, Armstrong SA. Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia. Cancer Discov. 2016;6(10):1166–81.CrossRefPubMed
52.
Zurück zum Zitat Feng ZYY, Zhou C, Chen F, Wu F, Wei L, Liu W, Dong S, Redell M, Mo Q, Song Y. Pharmacological inhibition of LSD1 for the treatment of MLL-rearranged leukemia. J Hematol Oncol. 2016;9(1):24.CrossRefPubMedPubMedCentral Feng ZYY, Zhou C, Chen F, Wu F, Wei L, Liu W, Dong S, Redell M, Mo Q, Song Y. Pharmacological inhibition of LSD1 for the treatment of MLL-rearranged leukemia. J Hematol Oncol. 2016;9(1):24.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Song YWF, Wu J. Targeting histone methylation for cancer therapy: enzymes, inhibitors, biological activity and perspectives. J Hematol Oncol. 2016;9(1):49.CrossRefPubMedPubMedCentral Song YWF, Wu J. Targeting histone methylation for cancer therapy: enzymes, inhibitors, biological activity and perspectives. J Hematol Oncol. 2016;9(1):49.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Kottaridis PDGR, Frew ME, Harrison G, Langabeer SE, Belton AA, Walker H, Wheatley K, Bowen DT, Burnett AK, Goldstone AH, Linch DC. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9.CrossRefPubMed Kottaridis PDGR, Frew ME, Harrison G, Langabeer SE, Belton AA, Walker H, Wheatley K, Bowen DT, Burnett AK, Goldstone AH, Linch DC. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9.CrossRefPubMed
55.
Zurück zum Zitat Yamamoto YKH, Nakano Y, Suzuki R, Kodera Y, Miyawaki S, Asou N, Kuriyama K, Yagasaki F, Shimazaki C, Akiyama H, Saito K, Nishimura M, Motoji T, Shinagawa K, Takeshita A, Saito H, Ueda R, Ohno R, Naoe T. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97(8):2434–9.CrossRefPubMed Yamamoto YKH, Nakano Y, Suzuki R, Kodera Y, Miyawaki S, Asou N, Kuriyama K, Yagasaki F, Shimazaki C, Akiyama H, Saito K, Nishimura M, Motoji T, Shinagawa K, Takeshita A, Saito H, Ueda R, Ohno R, Naoe T. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97(8):2434–9.CrossRefPubMed
56.
Zurück zum Zitat Serve HKU, Wagner R, Sauerland MC, Heinecke A, Brunnberg U, Schaich M, Ottmann O, Duyster J, Wandt H, Fischer T, Giagounidis A, Neubauer A, Reichle A, Aulitzky W, Noppeney R, Blau I, Kunzmann V, Stuhlmann R, Krämer A, Kreuzer KA, Brandts C, Steffen B, Thiede C, Müller-Tidow C, Ehninger G, Berdel WE. 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(25):3110–8.CrossRefPubMed Serve HKU, Wagner R, Sauerland MC, Heinecke A, Brunnberg U, Schaich M, Ottmann O, Duyster J, Wandt H, Fischer T, Giagounidis A, Neubauer A, Reichle A, Aulitzky W, Noppeney R, Blau I, Kunzmann V, Stuhlmann R, Krämer A, Kreuzer KA, Brandts C, Steffen B, Thiede C, Müller-Tidow C, Ehninger G, Berdel WE. 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(25):3110–8.CrossRefPubMed
57.
Zurück zum Zitat DA Macdonald AS, Brandwein J, Kamel-Reid S, Eisenhauer EA, Couban S, Caplan S, Foo A, Walsh W, Leber B. A phase I/II study of sorafenib in combination with low dose cytarabine in elderly patients with acute myeloid leukemia or high-risk myelodysplastic syndrome from the National Cancer Institute of Canada Clinical Trials Group: trial IND.186. Leuk Lymphoma. 2013;54(4):760–6.CrossRefPubMed DA Macdonald AS, Brandwein J, Kamel-Reid S, Eisenhauer EA, Couban S, Caplan S, Foo A, Walsh W, Leber B. A phase I/II study of sorafenib in combination with low dose cytarabine in elderly patients with acute myeloid leukemia or high-risk myelodysplastic syndrome from the National Cancer Institute of Canada Clinical Trials Group: trial IND.186. Leuk Lymphoma. 2013;54(4):760–6.CrossRefPubMed
58.
Zurück zum Zitat Ohanian MG-MG, Jabbour EJ, Daver N, Borthakur G, Kadia TM, Brandt M, Pierce S, Burger J, Richie MA, Patel K, Cortes JE, Kantarjian H, Ravandi F. Combination of sorafenib and 5-azacytidine in older patients with untreated acute myeloid leukemia with FLT3-ITD mutation. Blood. 2016;128(22):1611. Ohanian MG-MG, Jabbour EJ, Daver N, Borthakur G, Kadia TM, Brandt M, Pierce S, Burger J, Richie MA, Patel K, Cortes JE, Kantarjian H, Ravandi F. Combination of sorafenib and 5-azacytidine in older patients with untreated acute myeloid leukemia with FLT3-ITD mutation. Blood. 2016;128(22):1611.
59.
Zurück zum Zitat Röllig CSH, Hüttmann A, Noppeney R, Müller-Tidow C, Krug U, Baldus CD, Brandts CH, Kunzmann V, Einsele H, Krämer A, Schäfer-Eckart K, Neubauer A, Burchert A, Giagounidis A, Krause SW, Mackensen A, Aulitzky W, Herbst R, Hänel M, Kiani A, Frickhofen N, Kullmer J, Kaiser U, Link H, Geer T, Reichle A, Junghanß C, Repp R, Heits F, Dürk H, Hase J, Klut IM, Illmer T, Bornhäuser M, Schaich M, Parmentier S, Görner M, Thiede C, von Bonin M, Schetelig J, Kramer M, Berdel WE, Ehninger G, Study Alliance Leukaemia. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16(16):1691–9.CrossRefPubMed Röllig CSH, Hüttmann A, Noppeney R, Müller-Tidow C, Krug U, Baldus CD, Brandts CH, Kunzmann V, Einsele H, Krämer A, Schäfer-Eckart K, Neubauer A, Burchert A, Giagounidis A, Krause SW, Mackensen A, Aulitzky W, Herbst R, Hänel M, Kiani A, Frickhofen N, Kullmer J, Kaiser U, Link H, Geer T, Reichle A, Junghanß C, Repp R, Heits F, Dürk H, Hase J, Klut IM, Illmer T, Bornhäuser M, Schaich M, Parmentier S, Görner M, Thiede C, von Bonin M, Schetelig J, Kramer M, Berdel WE, Ehninger G, Study Alliance Leukaemia. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16(16):1691–9.CrossRefPubMed
60.
Zurück zum Zitat Ravandi FAM, Grunwald MR, Rudek MA, Rajkhowa T, Richie MA, Pierce S, Daver N, Garcia-Manero G, Faderl S, Nazha A, Konopleva M, Borthakur G, Burger J, Kadia T, Dellasala S, Andreeff M, Cortes J, Kantarjian H, Levis M. Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood. 2013;121(23):4655–62.CrossRefPubMedPubMedCentral Ravandi FAM, Grunwald MR, Rudek MA, Rajkhowa T, Richie MA, Pierce S, Daver N, Garcia-Manero G, Faderl S, Nazha A, Konopleva M, Borthakur G, Burger J, Kadia T, Dellasala S, Andreeff M, Cortes J, Kantarjian H, Levis M. Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood. 2013;121(23):4655–62.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Chen YBLS, Lane AA, Connolly C, Del Rio C, Valles B, Curtis M, Ballen K, Cutler C, Dey BR, El-Jawahri A, Fathi AT, Ho VT, Joyce A, McAfee S, Rudek M, Rajkhowa T, Verselis S, Antin JH, Spitzer TR, Levis M, Soiffer R. Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(12):2042–8.CrossRefPubMedPubMedCentral Chen YBLS, Lane AA, Connolly C, Del Rio C, Valles B, Curtis M, Ballen K, Cutler C, Dey BR, El-Jawahri A, Fathi AT, Ho VT, Joyce A, McAfee S, Rudek M, Rajkhowa T, Verselis S, Antin JH, Spitzer TR, Levis M, Soiffer R. Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(12):2042–8.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Stone RMFT, Paquette R, Schiller G, Schiffer CA, Ehninger G, Cortes J, Kantarjian HM, DeAngelo DJ, Huntsman-Labed A, Dutreix C, del Corral A, Giles F. Phase IB study of the FLT3 kinase inhibitor midostaurin with chemotherapy in younger newly diagnosed adult patients with acute myeloid leukemia. Leukemia. 2012;26(9):2061–8.CrossRefPubMedPubMedCentral Stone RMFT, Paquette R, Schiller G, Schiffer CA, Ehninger G, Cortes J, Kantarjian HM, DeAngelo DJ, Huntsman-Labed A, Dutreix C, del Corral A, Giles F. Phase IB study of the FLT3 kinase inhibitor midostaurin with chemotherapy in younger newly diagnosed adult patients with acute myeloid leukemia. Leukemia. 2012;26(9):2061–8.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Stone RMMS, Sanford BL, Geyer S, Bloomfield CD, Dohner K, Thiede C, Marcucci G, Lo-Coco F, Klisovic RB, Wei A, Sierra J, Sanz MA, Brandwein JM, de Witte T, Niederwieser D, Appelbaum FR, Medeiros BC, Tallman MS, Krauter J, Schlenk RF, Ganser A, Serve H, Ehninger G, Amadori S, Larson RA, Dohner H. The multi-kinase inhibitor midostaurin (M) prolongs survival compared with placebo (P) in combination with daunorubicin (D)/cytarabine (C) induction (ind), high-dose C consolidation (consol), and as maintenance (maint) therapy in newly diagnosed acute myeloid leukemia (AML) patients (pts) age 18-60 with FLT3 mutations (muts): an international prospective randomized (rand) P-controlled double-blind trial (CALGB 10603/RATIFY [Alliance]). Blood. 2015;126(23):6. Stone RMMS, Sanford BL, Geyer S, Bloomfield CD, Dohner K, Thiede C, Marcucci G, Lo-Coco F, Klisovic RB, Wei A, Sierra J, Sanz MA, Brandwein JM, de Witte T, Niederwieser D, Appelbaum FR, Medeiros BC, Tallman MS, Krauter J, Schlenk RF, Ganser A, Serve H, Ehninger G, Amadori S, Larson RA, Dohner H. The multi-kinase inhibitor midostaurin (M) prolongs survival compared with placebo (P) in combination with daunorubicin (D)/cytarabine (C) induction (ind), high-dose C consolidation (consol), and as maintenance (maint) therapy in newly diagnosed acute myeloid leukemia (AML) patients (pts) age 18-60 with FLT3 mutations (muts): an international prospective randomized (rand) P-controlled double-blind trial (CALGB 10603/RATIFY [Alliance]). Blood. 2015;126(23):6.
64.
Zurück zum Zitat Strati PKH, Ravandi F, Nazha A, Borthakur G, Daver N, Kadia T, Estrov Z, Garcia-Manero G, Konopleva M, Rajkhowa T, Durand M, Andreeff M, Levis M, Cortes J. Phase I/II trial of the combination of midostaurin (PKC412) and 5-azacytidine for patients with acute myeloid leukemia and myelodysplastic syndrome. Am J Hematol. 2015;90(4):276–81.CrossRefPubMedPubMedCentral Strati PKH, Ravandi F, Nazha A, Borthakur G, Daver N, Kadia T, Estrov Z, Garcia-Manero G, Konopleva M, Rajkhowa T, Durand M, Andreeff M, Levis M, Cortes J. Phase I/II trial of the combination of midostaurin (PKC412) and 5-azacytidine for patients with acute myeloid leukemia and myelodysplastic syndrome. Am J Hematol. 2015;90(4):276–81.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Cortes JEKH, Foran JM, Ghirdaladze D, Zodelava M, Borthakur G, Gammon G, Trone D, Armstrong RC, James J, Levis M. 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(29):3681–7.CrossRefPubMedPubMedCentral Cortes JEKH, Foran JM, Ghirdaladze D, Zodelava M, Borthakur G, Gammon G, Trone D, Armstrong RC, James J, Levis M. 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(29):3681–7.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Levis MJPA, Dombret H, Döhner M, Steffen B, Rousselot P, Martinelli G, Estey EH, Burnett AK, Gammon G, Trone D, Leo E, Cortes JE. 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. Blood. 2012;120(21):673. Levis MJPA, Dombret H, Döhner M, Steffen B, Rousselot P, Martinelli G, Estey EH, Burnett AK, Gammon G, Trone D, Leo E, Cortes JE. 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. Blood. 2012;120(21):673.
67.
Zurück zum Zitat Cortes JEPA, Dombret H, Kayser S, Steffen B, Rousselot P, Martinelli G, Estey EH, Burnett AK, Gammon G, Trone D, Leo E, Levis MJ. Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients ≥ 60 years of age with FLT3 ITD positive or negative relapsed/refractory acute myeloid leukemia. Blood. 2012;120(21):48. Cortes JEPA, Dombret H, Kayser S, Steffen B, Rousselot P, Martinelli G, Estey EH, Burnett AK, Gammon G, Trone D, Leo E, Levis MJ. Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients ≥ 60 years of age with FLT3 ITD positive or negative relapsed/refractory acute myeloid leukemia. Blood. 2012;120(21):48.
68.
Zurück zum Zitat Schiller GJ TM, Goldberg SL, Perl AE, Marie JP, Martinelli G, Larson RA, Russell N, Trone D, Gammon G, Levis MJ, Cortes JE. Final results of a randomized phase 2 study showing the clinical benefit of quizartinib (AC220) in patients with FLT3-ITD positive relapsed or refractory acute myeloid leukemia. J Clin Oncol. 2014;suppl; abstract 7100. Schiller GJ TM, Goldberg SL, Perl AE, Marie JP, Martinelli G, Larson RA, Russell N, Trone D, Gammon G, Levis MJ, Cortes JE. Final results of a randomized phase 2 study showing the clinical benefit of quizartinib (AC220) in patients with FLT3-ITD positive relapsed or refractory acute myeloid leukemia. J Clin Oncol. 2014;suppl; abstract 7100.
69.
Zurück zum Zitat Abdelall WKH, Borthakur G, Garcia-Manero G, Patel KP, Jabbour EJ, Daver NG, Kadia T, Gborogen RA, Konopleva M, Ravandi F, Andreef M, Cortes JE. The combination of quizartinib with azacitidine or low dose cytarabine is highly active in patients (Pts) with FLT3-ITD mutated myeloid leukemias: interim report of a phase I/II trial. Blood. 2016;128(22):1642. Abdelall WKH, Borthakur G, Garcia-Manero G, Patel KP, Jabbour EJ, Daver NG, Kadia T, Gborogen RA, Konopleva M, Ravandi F, Andreef M, Cortes JE. The combination of quizartinib with azacitidine or low dose cytarabine is highly active in patients (Pts) with FLT3-ITD mutated myeloid leukemias: interim report of a phase I/II trial. Blood. 2016;128(22):1642.
70.
Zurück zum Zitat Alvarado YKH, Luthra R, Ravandi F, Borthakur G, Garcia-Manero G, Konopleva M, Estrov Z, Andreeff M, Cortes JE. Treatment with FLT3 inhibitor in patients with FLT3-mutated acute myeloid leukemia is associated with development of secondary FLT3-tyrosine kinase domain mutations. Cancer. 2014;120(14):2142–9.CrossRefPubMedPubMedCentral Alvarado YKH, Luthra R, Ravandi F, Borthakur G, Garcia-Manero G, Konopleva M, Estrov Z, Andreeff M, Cortes JE. Treatment with FLT3 inhibitor in patients with FLT3-mutated acute myeloid leukemia is associated with development of secondary FLT3-tyrosine kinase domain mutations. Cancer. 2014;120(14):2142–9.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Randhawa JKKH, Borthakur G, Thompson PA, Konopleva M, Daver N, Pemmaraju N, Jabbour E, Kadia TM, Estrov Z, Ramachandran A, Paradela J, Andreef M, Levis M, Ravandi F, Cortes JE. Results of a phase II study of crenolanib in relapsed/refractory acute myeloid leukemia patients (Pts) with activating FLT3 mutations. Blood. 2014;124(21):389. Randhawa JKKH, Borthakur G, Thompson PA, Konopleva M, Daver N, Pemmaraju N, Jabbour E, Kadia TM, Estrov Z, Ramachandran A, Paradela J, Andreef M, Levis M, Ravandi F, Cortes JE. Results of a phase II study of crenolanib in relapsed/refractory acute myeloid leukemia patients (Pts) with activating FLT3 mutations. Blood. 2014;124(21):389.
72.
Zurück zum Zitat Wang ESSR, Tallman MS, Walter RB, Eckardt JR, Collins R. Crenolanib, a type I FLT3 TKI, can be safely combined with cytarabine and anthracycline induction chemotherapy and results in high response rates in patients with newly diagnosed FLT3 mutant acute myeloid leukemia (AML). Blood. 2016;128(22):1071. Wang ESSR, Tallman MS, Walter RB, Eckardt JR, Collins R. Crenolanib, a type I FLT3 TKI, can be safely combined with cytarabine and anthracycline induction chemotherapy and results in high response rates in patients with newly diagnosed FLT3 mutant acute myeloid leukemia (AML). Blood. 2016;128(22):1071.
73.
Zurück zum Zitat Ohanian MKH, Borthakur G, Kadia TM, Konopleva TM, Garcia-Manero G, Estrov Z, Ferrajoli A, Takahashi K, Jabbour EJ, Daver N, Kornblau SM, Wierda WG, Burger JA, Naqvi K, Benton CB, Bose P, Eckardt JR, Ravandi F, Cortes JE. Efficacy of a type I FLT3 inhibitor, crenolanib, with idarubicin and high-dose ara-C in multiply relapsed/refractory FLT3+ AML. Blood. 2016;128(22):2744. Ohanian MKH, Borthakur G, Kadia TM, Konopleva TM, Garcia-Manero G, Estrov Z, Ferrajoli A, Takahashi K, Jabbour EJ, Daver N, Kornblau SM, Wierda WG, Burger JA, Naqvi K, Benton CB, Bose P, Eckardt JR, Ravandi F, Cortes JE. Efficacy of a type I FLT3 inhibitor, crenolanib, with idarubicin and high-dose ara-C in multiply relapsed/refractory FLT3+ AML. Blood. 2016;128(22):2744.
74.
Zurück zum Zitat Perl AEAJ, Cortes JE, Smith CC, Litzow M, Baer MR, Claxton DF, Erba HP, Gill SC, Goldberg SL, Jurcic JG, Larson RA, Liu C, Ritchie EK, Schiller GJ, Spira AI, Strickland SA, Tibes R, Ustun C, Wang ES, Stuart RK, Röllig C, Neubauer A, Martinelli G, Bahceci E, Levis MJ. Final results of the chrysalis trial: a first-in-human phase 1/2 dose-escalation, dose-expansion study of gilteritinib (ASP2215) in patients with relapsed/refractory acute myeloid leukemia (R/R AML). Blood. 2016;128(22):1069. Perl AEAJ, Cortes JE, Smith CC, Litzow M, Baer MR, Claxton DF, Erba HP, Gill SC, Goldberg SL, Jurcic JG, Larson RA, Liu C, Ritchie EK, Schiller GJ, Spira AI, Strickland SA, Tibes R, Ustun C, Wang ES, Stuart RK, Röllig C, Neubauer A, Martinelli G, Bahceci E, Levis MJ. Final results of the chrysalis trial: a first-in-human phase 1/2 dose-escalation, dose-expansion study of gilteritinib (ASP2215) in patients with relapsed/refractory acute myeloid leukemia (R/R AML). Blood. 2016;128(22):1069.
75.
Zurück zum Zitat Bixby DLSA, Fathi AT, Kovacsovics TJ, Levy MY, Erba HP, Lancet JE, Jillella AP, Ravandi F, Walter RB, Stein EM, Faderl S, DeAngelo DJ, Ho P, O’Meara MM, Voellinger JL, Advani A. Vadastuximab talirine monotherapy in older patients with treatment naive CD33-positive acute myeloid leukemia (AML). Blood. 2016;128(22):590. Bixby DLSA, Fathi AT, Kovacsovics TJ, Levy MY, Erba HP, Lancet JE, Jillella AP, Ravandi F, Walter RB, Stein EM, Faderl S, DeAngelo DJ, Ho P, O’Meara MM, Voellinger JL, Advani A. Vadastuximab talirine monotherapy in older patients with treatment naive CD33-positive acute myeloid leukemia (AML). Blood. 2016;128(22):590.
76.
Zurück zum Zitat Fathi ATEH, Lancet JE, Stein EM, Ravandi F, Faderl S, Walter RB, Advani A, DeAngelo DJ, Kovacsovics TJ, Jillella AP, Bixby DL, Levy MY, O’Meara MM, Ho P, Stein AS. Vadastuximab talirine plus hypomethylating agents: a well-tolerated regimen with high remission rate in frontline older patients with acute myeloid leukemia (AML). Blood. 2016;128(22):591. Fathi ATEH, Lancet JE, Stein EM, Ravandi F, Faderl S, Walter RB, Advani A, DeAngelo DJ, Kovacsovics TJ, Jillella AP, Bixby DL, Levy MY, O’Meara MM, Ho P, Stein AS. Vadastuximab talirine plus hypomethylating agents: a well-tolerated regimen with high remission rate in frontline older patients with acute myeloid leukemia (AML). Blood. 2016;128(22):591.
77.
Zurück zum Zitat Erba HPLM, Vasu S, Stein AS, Fathi AT, Maris MB, Advani AS, Faderl S, Smith SE, Wood BL, Walter RB, Yang J, Donnellan WB, Feldman EJ, Voellinger JL, Ravandi F. A phase 1b study of vadastuximab talirine in combination with 7+3 induction therapy for patients with newly diagnosed acute myeloid leukemia (AML). Blood. 2016;128(22):211. Erba HPLM, Vasu S, Stein AS, Fathi AT, Maris MB, Advani AS, Faderl S, Smith SE, Wood BL, Walter RB, Yang J, Donnellan WB, Feldman EJ, Voellinger JL, Ravandi F. A phase 1b study of vadastuximab talirine in combination with 7+3 induction therapy for patients with newly diagnosed acute myeloid leukemia (AML). Blood. 2016;128(22):211.
78.
Zurück zum Zitat Döhner HLM, Fiedler W, Fouillard L, Haaland A, Brandwein JM, Lepretre S, Reman O, Turlure P, Ottmann OG, Müller-Tidow C, Krämer A, Raffoux E, Döhner K, Schlenk RF, Voss F, Taube T, Fritsch H, Maertens J. Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood. 2014;124(9):1426–33.CrossRefPubMedPubMedCentral Döhner HLM, Fiedler W, Fouillard L, Haaland A, Brandwein JM, Lepretre S, Reman O, Turlure P, Ottmann OG, Müller-Tidow C, Krämer A, Raffoux E, Döhner K, Schlenk RF, Voss F, Taube T, Fritsch H, Maertens J. Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood. 2014;124(9):1426–33.CrossRefPubMedPubMedCentral
79.
80.
Zurück zum Zitat Pan RHL, Benito JM, Bucci D, Han L, Borthakur G, Cortes J, DeAngelo DJ, Debose L, Mu H, Döhner H, Gaidzik VI, Galinsky I, Golfman LS, Haferlach T, Harutyunyan KG, Hu J, Leverson JD, Marcucci G, Müschen M, Newman R, Park E, Ruvolo PP, Ruvolo V, Ryan J, Schindela S, Zweidler-McKay P, Stone RM, Kantarjian H, Andreeff M, Konopleva M, Letai AG. Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia. Cancer Discov. 2014;4(3):362–75.CrossRefPubMed Pan RHL, Benito JM, Bucci D, Han L, Borthakur G, Cortes J, DeAngelo DJ, Debose L, Mu H, Döhner H, Gaidzik VI, Galinsky I, Golfman LS, Haferlach T, Harutyunyan KG, Hu J, Leverson JD, Marcucci G, Müschen M, Newman R, Park E, Ruvolo PP, Ruvolo V, Ryan J, Schindela S, Zweidler-McKay P, Stone RM, Kantarjian H, Andreeff M, Konopleva M, Letai AG. Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia. Cancer Discov. 2014;4(3):362–75.CrossRefPubMed
81.
Zurück zum Zitat Konopleva MPD, Potluri J, Chyla B, Hogdal L, Busman T, McKeegan E, Salem AH, Zhu M, Ricker JL, Blum W, DiNardo CD, Kadia T, Dunbar M, Kirby R, Falotico N, Leverson J, Humerickhouse R, Mabry M, Stone R, Kantarjian H, Letai A. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6(10):1106–17.CrossRefPubMed Konopleva MPD, Potluri J, Chyla B, Hogdal L, Busman T, McKeegan E, Salem AH, Zhu M, Ricker JL, Blum W, DiNardo CD, Kadia T, Dunbar M, Kirby R, Falotico N, Leverson J, Humerickhouse R, Mabry M, Stone R, Kantarjian H, Letai A. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6(10):1106–17.CrossRefPubMed
82.
Zurück zum Zitat Lin TL SS, Fiedler W, Walter RB, Hou JZ, Roboz GJ, Enjeti A, Fakhoui KM, Darden DE, Dunbar M, Zhu M, Hayslip JW, Wei AH. Phase Ib/2 study of venetoclax with low-dose cytarabine in treatment-naive patients age ≥ 65 with acute myelogenous leukemia. J Clin Oncol. 2016;suppl; abstract 7007. Lin TL SS, Fiedler W, Walter RB, Hou JZ, Roboz GJ, Enjeti A, Fakhoui KM, Darden DE, Dunbar M, Zhu M, Hayslip JW, Wei AH. Phase Ib/2 study of venetoclax with low-dose cytarabine in treatment-naive patients age ≥ 65 with acute myelogenous leukemia. J Clin Oncol. 2016;suppl; abstract 7007.
83.
Zurück zum Zitat Pollyea DA DC, Thirman MJ, Letai A, Wei AH, Jonas BA, Arellano ML, Frattini MG, Kantarjian HM, Chyla B, Zhu M, Potluri J, Humerickhouse R, Mabry MH, Konopleva M, Pratz KW. Results of a phase 1b study of venetoclax plus decitabine or azacitidine in untreated acute myeloid leukemia patients ≥ 65 years ineligible for standard induction therapy. J Clin Oncol. 2016;suppl; abstract 7009. Pollyea DA DC, Thirman MJ, Letai A, Wei AH, Jonas BA, Arellano ML, Frattini MG, Kantarjian HM, Chyla B, Zhu M, Potluri J, Humerickhouse R, Mabry MH, Konopleva M, Pratz KW. Results of a phase 1b study of venetoclax plus decitabine or azacitidine in untreated acute myeloid leukemia patients ≥ 65 years ineligible for standard induction therapy. J Clin Oncol. 2016;suppl; abstract 7009.
84.
Zurück zum Zitat Löwenberg BMG, Ossenkoppele GJ, Burnett AK, Zachée P, Dührsen U, Dierickx D, Müller-Tidow C, Sonneveld P, Krug U, Bone E, Flores N, Richardson AF, Hooftman L, Jenkins C, Zweegman S, Davies F. Phase I/II clinical study of tosedostat, an inhibitor of aminopeptidases, in patients with acute myeloid leukemia and myelodysplasia. J Clin Oncol. 2010;28(28):4333–8.CrossRefPubMed Löwenberg BMG, Ossenkoppele GJ, Burnett AK, Zachée P, Dührsen U, Dierickx D, Müller-Tidow C, Sonneveld P, Krug U, Bone E, Flores N, Richardson AF, Hooftman L, Jenkins C, Zweegman S, Davies F. Phase I/II clinical study of tosedostat, an inhibitor of aminopeptidases, in patients with acute myeloid leukemia and myelodysplasia. J Clin Oncol. 2010;28(28):4333–8.CrossRefPubMed
85.
Zurück zum Zitat Cortes JFE, Yee K, Rizzieri D, Advani AS, Charman A, Spruyt R, Toal M, Kantarjian H. Two dosing regimens of tosedostat in elderly patients with relapsed or refractory acute myeloid leukaemia (OPAL): a randomised open-label phase 2 study. Lancet Oncol. 2013;14(4):354–62.CrossRefPubMed Cortes JFE, Yee K, Rizzieri D, Advani AS, Charman A, Spruyt R, Toal M, Kantarjian H. Two dosing regimens of tosedostat in elderly patients with relapsed or refractory acute myeloid leukaemia (OPAL): a randomised open-label phase 2 study. Lancet Oncol. 2013;14(4):354–62.CrossRefPubMed
86.
Zurück zum Zitat DiNardo CDKH, Ravandi F, Konopleva M, Kadia TM, Borthakur G, Daver NG, Durand M, Rivera S, Quintás-Cardama A, Garcia-Manero G, Cortes JE. A phase I/II study of cytarabine or azacitidine in combination with tosedostat in older patients with AML or high-risk MDS. Blood. 2013;122(21):2698. DiNardo CDKH, Ravandi F, Konopleva M, Kadia TM, Borthakur G, Daver NG, Durand M, Rivera S, Quintás-Cardama A, Garcia-Manero G, Cortes JE. A phase I/II study of cytarabine or azacitidine in combination with tosedostat in older patients with AML or high-risk MDS. Blood. 2013;122(21):2698.
87.
Zurück zum Zitat Mawad RBP, Hendrie P, Scott B, Wood BL, Dean C, Sandhu V, Deeg HJ, Walter R, Wang L, Myint H, Singer JW, Estey E, Pagel JM. Phase II study of tosedostat with cytarabine or decitabine in newly diagnosed older patients with acute myeloid leukaemia or high-risk MDS. Br J Haematol. 2016;172(2):238–45.CrossRefPubMed Mawad RBP, Hendrie P, Scott B, Wood BL, Dean C, Sandhu V, Deeg HJ, Walter R, Wang L, Myint H, Singer JW, Estey E, Pagel JM. Phase II study of tosedostat with cytarabine or decitabine in newly diagnosed older patients with acute myeloid leukaemia or high-risk MDS. Br J Haematol. 2016;172(2):238–45.CrossRefPubMed
88.
Zurück zum Zitat Novotny-Diermayr VHS, Goh KC, Cheong A, Ong LC, Hentze H, Pasha MK, Jayaraman R, Ethirajulu K, Wood JM. 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(5):e69.CrossRefPubMedPubMedCentral Novotny-Diermayr VHS, Goh KC, Cheong A, Ong LC, Hentze H, Pasha MK, Jayaraman R, Ethirajulu K, Wood JM. 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(5):e69.CrossRefPubMedPubMedCentral
Metadaten
Titel
Emerging therapies for acute myeloid leukemia
verfasst von
Caner Saygin
Hetty E. Carraway
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2017
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-017-0463-6

Weitere Artikel der Ausgabe 1/2017

Journal of Hematology & Oncology 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.