novel therapies for children with acute myeloid leukaemia306969/moore... · 2019. 10. 9. · moore...

39
Moore et al. Novel therapies for childhood AML 1 Novel therapies for children with acute myeloid leukaemia 1 2 Authors: Andrew S. Moore PhD 1,2,3 , Pamela R. Kearns PhD 4,5 , Steven Knapper DM 6 , 3 Andrew D.J. Pearson MD 3,5 , C. Michel Zwaan PhD 5,7 4 5 Author Affiliations: 1 Queensland Children’s Medical Research Institute, The University of 6 Queensland, Brisbane, Australia; 2 Children’s Health Queensland Hospital and Health 7 Service, Brisbane, Australia; 3 Divisions of Clinical Studies and Therapeutics, The Institute of 8 Cancer Research and Royal Marsden, Sutton, UK; 4 School of Cancer Sciences, University of 9 Birmingham, Birmingham, UK; 5 Innovative Therapies for Children with Cancer (ITCC) 10 European Consortium; 6 Department of Haematology, School of Medicine, Cardiff University, 11 Cardiff, UK; 7 Pediatric Oncology/Hematology, Erasmus MC/Sophia Children’s Hospital, 12 Rotterdam, The Netherlands. 13 Running title: Novel therapies for childhood AML. 14 Funding: ASM is supported by the National Health and Medical Research Council 15 (Australia) and the Children’s Health Foundation Queensland. ADJP is supported by Cancer 16 Research UK (programme grant C1178/A10294). 17 18 Corresponding author: Dr A.S. Moore, Queensland Children’s Medical Research Institute, 19 Royal Children’s Hospital, Herston Rd, Herston, Qld, 4029, Australia. Ph: +67 7 3636 3981, 20 Fax: +61 7 3636 5578, Email: [email protected] 21 22 Word count: 6,285 (including headings and parentheses; abstract word count = 163) 23 24 References: 125 25

Upload: others

Post on 01-Feb-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

  • Moore et al. Novel therapies for childhood AML

    1

    Novel therapies for children with acute myeloid leukaemia 1 2

    Authors: Andrew S. Moore PhD1,2,3, Pamela R. Kearns PhD4,5, Steven Knapper DM6, 3

    Andrew D.J. Pearson MD3,5, C. Michel Zwaan PhD5,7 4

    5

    Author Affiliations: 1Queensland Children’s Medical Research Institute, The University of 6

    Queensland, Brisbane, Australia; 2Children’s Health Queensland Hospital and Health 7

    Service, Brisbane, Australia; 3Divisions of Clinical Studies and Therapeutics, The Institute of 8

    Cancer Research and Royal Marsden, Sutton, UK; 4School of Cancer Sciences, University of 9

    Birmingham, Birmingham, UK; 5Innovative Therapies for Children with Cancer (ITCC) 10

    European Consortium; 6Department of Haematology, School of Medicine, Cardiff University, 11

    Cardiff, UK; 7Pediatric Oncology/Hematology, Erasmus MC/Sophia Children’s Hospital, 12

    Rotterdam, The Netherlands. 13

    Running title: Novel therapies for childhood AML. 14

    Funding: ASM is supported by the National Health and Medical Research Council 15

    (Australia) and the Children’s Health Foundation Queensland. ADJP is supported by Cancer 16

    Research UK (programme grant C1178/A10294). 17

    18

    Corresponding author: Dr A.S. Moore, Queensland Children’s Medical Research Institute, 19

    Royal Children’s Hospital, Herston Rd, Herston, Qld, 4029, Australia. Ph: +67 7 3636 3981, 20

    Fax: +61 7 3636 5578, Email: [email protected] 21

    22

    Word count: 6,285 (including headings and parentheses; abstract word count = 163) 23

    24 References: 125 25

  • Moore et al. Novel therapies for childhood AML

    2

    Abstract 26

    Significant improvements in survival for children with acute myeloid leukaemia (AML) have 27

    been made over the past three decades, with overall survival rates now approximately 60-28

    70%. However, these gains can be largely attributed to more intensive use of conventional 29

    cytotoxics made possible by advances in supportive care, and although over 90% of children 30

    achieve remission with frontline therapy, approximately one third in current protocols 31

    relapse. Furthermore, late effects of therapy cause significant morbidity for many survivors. 32

    Novel therapies are therefore desperately needed. Early phase paediatric trials of several new 33

    agents such as clofarabine, sorafenib and gemtuzumab ozogamicin have shown encouraging 34

    results in recent years. Due to the relatively low incidence of AML in childhood, the success 35

    of paediatric early-phase clinical trials is largely dependent upon collaborative clinical trial 36

    design by international cooperative study groups. Successfully incorporating novel therapies 37

    into frontline therapy remains a challenge, but the potential for significant improvement in 38

    the duration and quality of survival for children with AML is high. 39

    40

    Keywords 41

    Acute myeloid leukaemia, AML, children, novel therapeutics, inhibitors, chemotherapy 42

    43

    44

  • Moore et al. Novel therapies for childhood AML

    3

    Introduction 45

    Acute myeloid leukemia (AML) is a heterogeneous class of leukaemias with an age-related 46

    incidence.1 AML is relatively rare in children, accounting for 15-20% of paediatric 47

    leukaemias, but causes a disproportionate number of childhood cancer deaths. For children 48

    less than 15 years of age overall survival (OS) rates are now approximately 60-70%.2-5 49

    Although there has been a steady and gradual improvement in survival for younger adults, it 50

    is clear that progress has plateaued for children, with survival rates being substantially 51

    inferior to those being achieved for acute lymphoblastic leukaemia (ALL), where OS exceeds 52

    80%.4,6 53

    54

    Conventional AML therapy is essentially the same for adults and children and based on 55

    intensive use of cytarabine or other nucleoside analogs and anthracyclines. Etoposide is 56

    frequently used in paediatric induction regimens, although there is no good evidence it is of 57

    benefit in adult studies and when combined with cytarabine and daunorubicin in children, 58

    etoposide provided no advantage compared to thioguanine with cytarabine and 59

    daunorubicin.2,7 Although chemotherapy will induce complete remission (CR) in 60

    approximately 90% of children, approximately one third relapse.4,5 Relapsed AML is 61

    associated with high morbidity and mortality, with allogeneic haematopoietic stem cell 62

    transplantation (HSCT) generally regarded as the most effective anti-leukaemic therapy 63

    available.6 Most re-induction regimens, at least in children, involve high-dose cytarabine in 64

    combination with other agents such as anthracyclines. Addition of liposomal daunorubicin to 65

    the commonly utilized FLAG regimen (fludarabine, high-dose cytarabine and G-CSF) has 66

    been shown to improve the early response rates to chemotherapy from 58% to 68% (P = 67

    0.047).8 Not surprisingly, the likelihood of achieving a second CR decreases dramatically 68

  • Moore et al. Novel therapies for childhood AML

    4

    with each failed therapeutic attempt, with only one third of children with relapsed AML 69

    becoming long-term survivors.8,9 70

    71

    Although the past 30 years has seen major improvements in overall survival for childhood 72

    AML, additional gains are unlikely to be achieved by simply intensifying conventional 73

    cytotoxic chemotherapy further.6 Chemotherapy such as etoposide and anthracyclines is 74

    limited not only by acute toxicity, but also late effects such as increased risks of secondary 75

    malignancy and cardiotoxicity.10,11 Such late effects are of particular concern in paediatrics 76

    since treatment occurs during periods of growth and development, and the duration of 77

    survivorship is much greater than adults. Less toxic and more effective therapies for AML in 78

    children, and adults, are therefore urgently needed. The ever-expanding array of molecular 79

    abnormalities associated with AML promises to further refine current risk-stratified treatment 80

    and facilitate individualised therapy with novel therapeutics specifically targeting these 81

    leukaemogenic abnormalities.5 Such an approach should improve outcomes for those children 82

    who have historically done poorly and, potentially, permit de-intensification of therapy for 83

    those children with low-risk disease, as has been achieved with paediatric ALL. 84

    85

    Several early-phase clinical trials of novel therapies have been completed over the past 86

    decade and an encouraging number of studies are in progress (Table I). A major limitation for 87

    progress in childhood AML, however, is that paediatric studies with novel agents usually 88

    don’t commence until preliminary toxicity and efficacy profiles have been established in 89

    adults. Whilst this may be justifiable from a patient-safety perspective, it slows the process of 90

    paediatric drug development. Furthermore, it is increasingly recognized that AML biology is 91

    at least partially different in children compared to adults, so extrapolating anticipated efficacy 92

    from adult studies to children can be challenging. For example, ‘epigenetic’ mutations of 93

  • Moore et al. Novel therapies for childhood AML

    5

    DNMT3A, IDH1/2, TET2 and NPM1 all occur much less frequently in children than in 94

    adults.12-16 95

    Novel classes of therapeutics investigated in childhood AML in recent years include purine 96

    nucleoside analogs, small molecule kinase inhibitors and immunomodulatory therapies. Here 97

    we review the results of these trials and highlight some of the more promising agents 98

    emerging from adult trials. 99

    100

    Novel purine nucleoside analogs 101

    The cytidine analog and DNA polymerase inhibitor cytarabine forms the backbone of 102

    conventional AML therapy in both children and adults. It is not surprising that novel 103

    cytarabine derivatives have delivered some promising results. Cladribine and fludarabine 104

    were the first two such derivatives developed. Unlike cytarabine, they inhibit both DNA 105

    polymerase and ribonucleotide reductase, thereby depleting deoxynucleotide pools.17 106

    Cladribine has shown encouraging results in combination with cytarabine18, topotecan19 and 107

    idarubicin.20 Interestingly, the FAB M5 subtype of AML appears more responsive to 108

    cladribine than other morphological subtypes.21 Fludarabine, in combination with cytarabine 109

    ± an anthracycline, has become a widely used re-induction regimen for children with relapsed 110

    AML.8,9,22,23 The dose-limiting neurotoxicity of cladribine and fludarabine however, 111

    prompted the development of clofarabine. After efficacy was demonstrated for 112

    relapsed/refractory ALL, in December 2004 clofarabine became the first anti-leukaemic drug 113

    approved for use in childhood prior to adult use approval.17 The efficacy and potential role of 114

    clofarabine in paediatric AML is discussed in detail below. 115

    116

  • Moore et al. Novel therapies for childhood AML

    6

    Clofarabine 117

    Clofarabine is a structural hybrid of cladribine and fludarabine, designed to have improve 118

    efficacy, through reduced deamination by adenosine deaminase and improved stability, whilst 119

    avoiding the formation of toxic metabolites such as 2-F adenine caused by glycosidic bond 120

    cleavage of fludarabine.17 Clofarabine is converted intracellularly to clofarabine 121

    monophosphate by deoxycytidine kinase (dCK), then to clofarabine triphosphate by 122

    additional kinases. The triphosphate form of clofarabine impairs DNA synthesis and repair 123

    via inhibition of both ribonucleotide reductase and DNA polymerase and induces apoptosis 124

    via mitochondrial pathways.24 As a potent ribonucleotide reductase inhibitor, clofarabine 125

    increases dCK activity, thus potentiating its own activation and theoretically that of other 126

    drugs such as cytarabine.24 The maximum tolerated dose (MTD) as a single-agent is 52 127

    mg/m2/day for 5 days in children and 40mg/m2/day for 5 days in adults with hematologic 128

    malignancies, with reversible hepatotoxicity and skin rash being the main non-129

    haematological dose limiting toxicities (DLTs).25 Although the liver and skin toxicities are 130

    reversible when clofarabine is given as a single agent, it should be noted that a potential 131

    interaction with intrathecal methotrexate exists, with at least one case report describing fatal 132

    skin and hepatoxicity in a child with a CNS-relapse of pre-B ALL given clofarabine 52 133

    mg/m2/day for 5 days, together with triple intrathecal therapy (hydrocortisone, methotrexate 134

    and cytarabine).26 135

    136

    A phase II study of clofarabine as a single agent in children with relapsed/refractory AML 137

    had an overall response rate (ORR) of 26%, comprising one CR with incomplete platelet 138

    recovery (CRp) and 10 partial responses (PR).25 Although most responses were partial, the 139

    trial population was heavily pretreated, with patients having received a median of two prior 140

    treatment regimens (range one to five). Furthermore, six of 28 (21%) patients refractory to 141

  • Moore et al. Novel therapies for childhood AML

    7

    prior therapy responded and approximately one third of clofarabine treated patients 142

    proceeded to HSCT.25 Subsequent interpretation of the HSCT data is problematic however, 143

    considering that most patients went to HSCT in PR, rather than CR. The toxicity profile was 144

    reported as being expected for the patient population, with frequent (≥ 15%) grade 3 adverse 145

    events being febrile neutropaenia, catheter-related infection, epistaxis, hypotension, nausea, 146

    fever, elevated transaminases and hypokalaemia.25 147

    148

    Clofarabine has also been trialed in combination with conventional and targeted therapeutic 149

    agents. In a phase I study of the multi-kinase/FLT3 inhibitor sorafenib in combination with 150

    clofarabine and cytarabine, responses were seen in both FLT3-wild type (-WT) and FLT3-151

    ITD+ patients.27 This trial is discussed further below. A phase I trial of clofarabine in 152

    combination with etoposide and cyclophosphamide for children with relapsed and refractory 153

    acute leukemias resulted in an ORR of 55% (9/20 CR + 2/20 CRp) for ALL patients and 154

    100% (1/5 CR + 4/5 CRp) for AML patients.28 When given for five consecutive days during 155

    induction and four consecutive days in consolidation for up to eight cycles (median time 156

    between cycles 34 days), the recommended phase II doses (RP2D) of clofarabine, 157

    cyclophosphamide and etoposide were 40, 440 and 100 mg/m2 respectively. The toxicity 158

    profile was similar to that seen in the monotherapy phase II study of clofarabine.28 159

    160

    A phase I study of clofarabine in combination with liposomal daunorubicin for children with 161

    relapsed/refractory AML was recently completed. Patients received clofarabine on days 1-5 162

    (30 mg/m2, escalated to 40 mg/m2), and liposomal daunorubicin (60 mg/m2) days 1, 3 and 5. 163

    Nine patients were enrolled, with the most common severe adverse event being febrile 164

    neutropaenia, but no DLTs observed. One third of patients achieved CR and proceeded to 165

    HSCT. The 40 mg/m2 clofarabine cohort is being extended to accrue more efficacy data.29 166

  • Moore et al. Novel therapies for childhood AML

    8

    167

    There is intense ongoing interest in clofarabine for AML in adults and children, with no 168

    fewer than 35 open studies of the drug in AML currently registered on ClinicalTrials.gov. A 169

    major challenge, however, remains in defining how best to use clofarabine in both upfront 170

    and second-line AML treatment regimens. Results of randomised studies in adults are 171

    expected in the near future, including the NCRI AML16 trial for adults over 60 years of age, 172

    which randomised newly-diagnosed patients to induction with either daunorubicin and 173

    cytarabine or daunorubicin and clofarabine (ISRCTN 11036523). The current NCRI AML17 174

    trial for younger adults with AML is randomising poor risk patients (defined by a 175

    statistically-derived risk score based on presenting disease characteristics and remission 176

    status after course I chemotherapy), to either daunorubicin plus clofarabine or FLAG-Ida 177

    prior to HSCT (ISRCTN55675535). For childhood AML, a randomised phase II study 178

    sponsored by St Jude Children’s Research Hospital (SJCRH) is ongoing, comparing end-of-179

    induction MRD for children treated with clofarabine and cytarabine to conventional ADE 180

    therapy (cytarabine, daunorubicin, etoposide; NCT00703820). 181

    182

    FLT3 inhibition 183

    Internal tandem duplication (ITD) or tyrosine kinase domain (TKD) mutations of the fms-like 184

    tyrosine kinase 3 (FLT3) gene occur frequently in AML, resulting in constitutive FLT3 185

    signaling and stimulation of leukaemic proliferation.30 The incidence of FLT3-ITD in AML 186

    increases with age, occurring in approximately 23% of adults and 12% of children overall 187

    with de novo AML, whilst the incidence of FLT3-TKD is relatively constant across all age 188

    groups at approximately 7%.31,32 Although FLT3-TKD mutations do not appear to impact on 189

    prognosis, the presence of FLT3-ITD has consistently been associated with inferior outcome, 190

    principally as a result of increased relapse rate.33 High FLT3-ITD/FLT3-WT allelic ratios 191

  • Moore et al. Novel therapies for childhood AML

    9

    (AR) carry a particularly poor prognosis, with survival rates of less than 20% in both adults 192

    and children.31,34,35 Furthermore, ex vivo studies have demonstrated increased dependence of 193

    high AR FLT3-ITD+ AML blasts to FLT3 signaling, consistent with the concept of 194

    “oncogene addiction”.36 The identification of secondary FLT3-TKD mutations in patients 195

    treated with FLT3 inhibitors is also evidence of clonal selection and cellular dependency on 196

    constitutive FLT3 signaling.37-39 197

    198

    Small molecule inhibition of FLT3 kinase has therefore been vigorously pursued as a 199

    therapeutic strategy over the past decade and detailed critiques of each individual FLT3 200

    inhibitor developed to date have recently been published.33,40 Initial “first generation” FLT3 201

    inhibitors, such as CEP-701 (lestaurtinib) and PKC412 (midostaurin), were non-selective 202

    compounds already in early clinical development and subsequently identified as having 203

    potent anti-FLT3 activity. These compounds have undergone extensive preclinical and 204

    clinical evaluation with major phase III trials ongoing. So-called “second-generation” FLT3 205

    inhibitors such as AC220 (quizartinib), designed with increased potency and selectivity for 206

    FLT3, are currently being evaluated in early-phase clinical trials. 207

    208

    Preclinical studies have demonstrated that sustained inhibition of FLT3 phosphorylation to 209

    less than 15% of baseline is required to achieve cytotoxicity.41,42 Furthermore, several 210

    pharmacokinetic (PK) and pharmacodynamic (PD) studies from clinical trials have 211

    consistently confirmed that clinical responses are unlikely in patients who do not achieve 212

    adequate FLT3 inhibition.41-45 213

    214

    Finally, although newer more potent and selective FLT3 inhibitors such as AC220 are 215

    showing encouraging results in single-agent trials, it appears that FLT3 inhibitors are likely to 216

  • Moore et al. Novel therapies for childhood AML

    10

    be of most benefit when used in combination with conventional chemotherapy.33 It is worth 217

    noting that myelosuppression from conventional chemotherapy itself has been shown to 218

    increase the production of FLT3 ligand (FL) which can decrease the efficacy of a number of 219

    FLT3 inhibitors, at least in vitro.46 These observations highlight the need for effective PD 220

    biomarker assays to monitor how effectively FLT3 is being targeted. Assays such as the 221

    plasma inhibitory activity (PIA) assay have been used to demonstrate that effective FLT3 222

    inhibition can still be achieved clinically however, despite elevations in FL (discussed in 223

    more detail below).47 224

    225

    Sorafenib 226

    The multi-kinase inhibitor, sorafenib, has potent activity against FLT3 and increased activity 227

    against FLT3-ITD+ AML cells compared to FLT3-WT cells.48 Although sorafenib was 228

    assessed by the Pediatric Preclinical Testing Program (PTPP), only one AML cell line was 229

    tested.49 In the PTPP screen, the FLT3-WT cell line Kasumi-1 had an in vitro IC50 of 0.02 230

    M.49 Pre-clinical studies of sorafenib and another multi-kinase inhibitor, sunitinib, at 231

    SJCRH examined a broader panel of AML cell lines and demonstrated similar in vitro 232

    sensitivity of Kasumi-1 (sorafenib IC50 0.04 M), but markedly increased sensitivity of the 233

    FLT3-ITD+ paediatric cell line MV4-11 (sorafenib IC50 0.002 M). In vitro activity of 234

    sorafenib against primary paediatric AML samples (4/6 FLT3-WT, 2/6 unknown FLT3 235

    status) was also demonstrated.50 Increased in vitro sensitivity of FLT3-mutated, primary 236

    paediatric AML samples has also been demonstrated for SU11657, a compound similar to 237

    sunitinib with anti-FLT3 activity51 Unlike sorafenib, sunitinib is no longer being evaluated as 238

    a FLT3 inhibitor in AML due to poor tolerability at doses required to inhibit FLT3.33 239

    240

  • Moore et al. Novel therapies for childhood AML

    11

    Several clinical trials of sorafenib in adult AML have been reported33, with a recent phase I/II 241

    study in combination with cytarabine and idarubicin demonstrating overall CR rates of 93% 242

    (14/15, plus 1 with CRp) in FLT3-ITD+ patients and 66% (24/36 plus 3 with CRp) in FLT3-243

    WT patients.52 In the phase II arm of the study, sorafenib was given at a dose of 400 mg 244

    twice daily for up to 28 days during induction and consolidation (both with concurrent 245

    cytarabine and idarubicin), then continued as maintenance therapy for up to one year.52 In 246

    contrast, a placebo-controlled, randomized phase II study of sorafenib combined with 247

    standard 7+3 induction chemotherapy and intermediate-dose cytarabine consolidation then 248

    continued for 1 year as maintenance in 197 older patients (> 60 years) failed to show any 249

    difference in CR rates, EFS, OS for FLT3-WT or FLT3-ITD+ patients, although the small 250

    number of FLT3-ITD+ patients (n = 28; 14.2%) may have limited the power of subset 251

    analysis. Despite being reportedly well tolerated, there was a trend towards slower leucocyte 252

    and platelet recovery in the sorafenib arm during the induction cycles.53 253

    254

    Encouraging results from a paediatric phase I study of sorafenib in combination with 255

    cytarabine and clofarabine for children with relapsed/refractory acute leukaemia were 256

    recently published. Sorafenib was given as a single agent on days 1 to 7 and then 257

    concurrently with clofarabine (40 mg/m2, or 20 mg/m2 if the patient had HSCT within 6 258

    months or fungal infection within 1 month) and cytarabine (1 g/m2) on days 8 to 12. If 259

    tolerated, sorafenib alone was continued until day 28. Repeated courses of sorafenib, 260

    clofarabine and cytarabine, maintenance therapy with single-agent sorafenib, or 261

    transplantation were administered according to clinical judgment.27 Eleven of the 12 patients 262

    (aged 6-17 years) had AML, including 3 who had previously undergone allogeneic HSCT (2 263

    FLT3-ITD+, 1 FLT3-WT). Of these 11 AML patients, 5 were FLT3-ITD+ and all achieved 264

    morphological CR on day 22 (3 CR + 2 CRp) in combination with cytarabine and 265

  • Moore et al. Novel therapies for childhood AML

    12

    clofarabine, including both patients who had previous allogeneic HSCT.27 Of the 6 FLT3-WT 266

    AML patients, 3 achieved morphological CR and one a PR. Of note, the initial seven-day 267

    course of single-agent sorafenib reduced bone marrow blast percentages in 10 patients 268

    (median 66%, range 9-95%), including all FLT3-ITD+ patients. Ten patients had PD studies 269

    performed (flow cytometric analysis of phosphorylated AKT, 4E-BP1 and S6RP), with 270

    decreases in percentage of positive cells and mean fluorescence intensity unrelated to FLT3-271

    mutational status.27 Six patients proceeded to HSCT.27 The most common toxicity associated 272

    with sorafenib was skin toxicity, with all 12 patients experiencing some degree of hand-foot 273

    skin reaction and/or rash. Skin toxicity was dose-limiting at 200 mg/m2 but not 150 mg/m2, 274

    which was the final recommended dose of sorafenib.27 The hand-foot skin (HFS) reaction to 275

    sorafenib is classically characterised by tender, scaling skin with erythematous halos on areas 276

    of pressure or skin creases which may blister and can progress to hyperkeratosis and impaired 277

    movement and function.54 Consensus guidelines for the management of sorafenib and 278

    sunitinib induced HFS reactions have been developed and include avoidance of skin trauma, 279

    liberal use of emollients and dose reduction/cessation of sorafenib.54 The high incidence of 280

    skin toxicity in this paediatric study compared to previous single agent adult trials was 281

    attributed the concurrent use of cytarabine and clofarabine, as well as the higher 282

    concentration of the active metabolite sorafenib N-oxide observed in the paediatric trial.27 283

    284

    The current Children’s Oncology Group phase III trial (AAML1031, NCT01371981) is using 285

    a FLT3-ITD/FLT3-WT allelic ratio >0.4 (high-AR) to non-randomly stratify children with 286

    FLT3-ITD+ AML to receive sorafenib and HSCT in first remission, if an appropriate donor is 287

    available. Although the HSCT data for adults is inconclusive2, matched related donor 288

    allogeneic HSCT has been demonstrated to be of benefit for FLT3-ITD+ children with a high-289

    AR.31 The dismal prognosis for children with high-AR FLT3-ITD+ AML (3 year DFS/OS ≤ 290

  • Moore et al. Novel therapies for childhood AML

    13

    20% ) clearly warrants novel treatment approaches however, the number of children with 291

    high-AR disease transplanted on the CCG-2941 and CCG-2961 trials was small (total FLT3-292

    ITD+ = 11; high-AR = 6), and all high-AR patients on the current COG AAML1031 trial will 293

    receive both sorafenib and allogeneic HSCT, so it may be difficult to determine the extent to 294

    which sorafenib further improves survival when compared to this historical control. 295

    296

    Paediatric trials of other FLT3 inhibitors, including AC220, CEP-701 and PKC412, are 297

    currently in progress (Table I). An emerging issue with a number of FLT3 inhibitors, 298

    including midostaurin and the more selective compounds AC220 and sorafenib, is resistance 299

    mediated by secondary FLT3-TKD mutations.37-39,55,56 Given the biologically similar nature 300

    of FLT3-ITD+ AML in children and adults, similar resistance patterns could be expected 301

    although the relatively small number of children with FLT3-ITD+ AML may make this a rare 302

    occurrence. Finally, the benefit of “maintenance” FLT3 inhibition (e.g. post-HSCT) is yet to 303

    be proven, however it is plausible that such strategies with compounds like AC220 or 304

    sorafenib may promote acquired resistance, mediated by secondary FLT3-TKD mutations. 305

    Despite the compelling preclinical data supporting FLT3 inhibition as a therapeutic strategy 306

    in FLT3-mutated AML, until therapeutic benefit is proven from ongoing clinical trials in 307

    adults and children, up-front use of FLT3 inhibitors for childhood AML should be limited to 308

    clinical trials. However, for children with relapsed or refractory FLT3-ITD+ AML, limited 309

    therapeutic options (e.g. from anthracycline cardiotoxicity) and no access to a clinical trial, 310

    FLT3 inhibition could be considered to help achieve disease control, e.g., prior to HSCT or 311

    possibly even as a palliative strategy. 312

    313

  • Moore et al. Novel therapies for childhood AML

    14

    Immunotherapy 314

    Gemtuzumab ozogamicin 315

    Gemtuzumab ozogamicin (GO), a humanized anti-CD33 monoclonal antibody conjugated to 316

    the cytotoxic compound N-acetyl-γ-calicheamicin dimethylhydrazine, has been extensively 317

    investigated in adults and children over the past decade. Therapeutic targeting of CD33 has a 318

    sound rationale in AML, since approximately 80% of AML cases express CD33.57 High 319

    CD33 expression in childhood AML is associated with adverse disease characteristics such as 320

    FLT3-ITD and independently predicts poor outcome.58 Initial experience with GO 321

    monotherapy in children with relapsed/refractory AML demonstrated response rates of 53% 322

    (8/15 patients), with 6 patients proceeding to HSCT.59 Subsequent trials, including GO 323

    combined with chemotherapy and HSCT have demonstrated its safety.60-63 Monotherapy 324

    studies and case reports of GO in relapsed/refractory childhood AML have resulted in a 325

    number clinically meaningful responses, whilst phase II studies in combination with 326

    chemotherapy have shown similar results to historical controls. The recently reported multi-327

    centre AML02 phase III trial included a randomization of GO (3 mg/m2) during induction II 328

    chemotherapy with cytarabine, daunorubicin and etoposide.64 Although the specific effects of 329

    GO versus no GO were not reported, GO in combination with ADE resulted in reductions in 330

    minimal residual disease (MRD) in 93% (27/29) of patients who had responded poorly to 331

    induction I, including all 8 patients with MRD > 25% after induction I. Of the 8 patients with 332

    MRD > 25%, half became MRD-negative with ADE+GO. Of the remaining 21 patients, who 333

    all had MRD > 1% after induction I, the ADE+GO combination reduced MRD in 19 (90%), 334

    with 9 patients becoming MRD-negative (43%).64 Paediatric phase III studies of GO in 335

    combination with conventional chemotherapy are ongoing. 336

    337

    The potential for veno-occlusive disease (VOD) of the liver following GO has been of 338

  • Moore et al. Novel therapies for childhood AML

    15

    concern in adult patients, particularly in those patients proceeding to HSCT within 3.5 339

    months of receiving 6 or 9 mg/m2 GO.65 The recently reported UK NCRI AML15 trial (1,113 340

    patients) administered a lower GO dose (3 mg/m2) in adults and failed to show an increased 341

    risk of hepatic toxicity, even when administered within 120 days of HSCT66 The successor 342

    trial, AML17 is randomizing adult patients to 3 vs. 6 mg/m2. In pediatric studies liver toxicity 343

    has been less of an issue, and GO seems better tolerated in children.67 344

    345

    Other dosing strategies of GO have also been examined, including fractionated therapy, 346

    facilitating cumulative doses of up to 27 mg/m2 (ref 68-70). A phase I study in combination 347

    with busulfan and cyclophosphamide HSCT conditioning for 12 children with CD33+ AML 348

    (median age 3 years, range 1-17) failed to identify any GO-associated DLTs at 3.0, 4.5, 6.0 or 349

    7.5 mg/m2 and had no 100-day transplant-related mortality.61 350

    351

    In June 2010 the United States Food and Drug Administration (FDA) withdrew marketing 352

    approval for GO, based on lack of benefit in relapsed/refractory AML and increased 353

    induction mortality in adults. It has been highlighted, however, that the increased induction 354

    mortality associated with GO was relative to an unusually low mortality in the control arm of 355

    one of the studies the FDA based their decision on.66 Furthermore, subsequent data from 356

    1,113 newly diagnosed patients treated on the UK NCRI AML15 trial have demonstrated a 357

    significant benefit of GO for patients with core binding factor (CBF) AML when given as a 358

    single dose of 3 g/m2 during courses one and three.66 The biological explanation for this is 359

    somewhat unclear, since responses to GO were not related to blast CD33 expression.66 360

    Interestingly, children with CBF AML on the NOPHO-AML 2004 trial accounted for 35% 361

    (N = 17) of relapses, independent of whether or not GO (5 mg/m2 x 2 doses) was given as 362

    post-consolidation therapy.71 Definitive results from larger paediatric trials of GO are 363

  • Moore et al. Novel therapies for childhood AML

    16

    awaited. The recently reported French trial ALFA-0701 confirmed the survival advantage for 364

    adults with de novo AML and favourable cytogenetics, but also for those with intermediate-365

    risk cytogenetics. A notable feature of the ALFA-0701 study was the delivery of fractionated, 366

    higher cumulative doses of GO (3 g/m2 days 1, 4 and 7 during induction and in two 367

    consolidation cycles).70 On the basis of these adult studies with fractionated dosing, further 368

    evaluation of GO in children is planned. 369

    370

    Stem cell targeting 371

    Bortezomib 372

    The proteasome inhibitor, bortezomib, was initially approved for use in multiple myeloma. 373

    Pre-clinical studies of proteasomal inhibition in combination with idarubicin in AML 374

    demonstrated down-regulation of NF-κB and preferential cell death in leukaemic stem cells 375

    but not normal haemopoietic stem cells (HSCs).72 A single-agent phase I study in children 376

    demonstrated acceptable toxicity, with a RP2D of 1.3 mg/m2 per dose when administered 377

    twice weekly for 2 weeks.73 Although NF-κB inhibition was observed in 2 of 5 evaluated 378

    patients, there were no objective clinical responses. Phase I studies of bortezomib (twice 379

    weekly for two weeks) in combination with chemotherapy in adult AML74 and paediatric 380

    ALL75 were also well tolerated, with predictable, chemotherapy-related toxicity. In the adult 381

    phase I combination study, nine patients were less than 60 years of age with relapsed AML, 382

    with the remaining 22 patients being over 60 years with untreated AML.74 Overall, 7/9 (78%) 383

    of relapsed patients and 15/22 (68%) untreated older patients achieved CR (including 3 384

    CRp).74 In addition to administering Sorafenib to children with high-AR FLT3-ITD+ AML, 385

    the current Children’s Oncology Group phase III trial (AAML1031, NCT01371981) is 386

  • Moore et al. Novel therapies for childhood AML

    17

    randomising patients with de novo AML (including those with low-AR FLT3-ITD) to receive 387

    conventional chemotherapy with or without Bortezomib for all courses. 388

    389

    Novel agents with promising data in adults 390

    Aurora kinase inhibition 391

    Aurora kinases are a family of serine/threonine kinases with critical roles in mitosis.76 Aurora 392

    A kinase plays a key role in centrosome maturation and mitotic spindle assembly whilst 393

    Aurora B kinase regulates the mitotic checkpoint, forms part of the chromosomal passenger 394

    complex (with inner centromere protein, Borealin and Survivin) and is required for final 395

    cytokinesis.76 Although therapeutic success in solid tumours has been limited, Aurora kinase 396

    inhibitors have shown encouraging responses in leukaemia, particularly AML and 397

    Philadelphia-positive leukaemias.77 A recent phase I/II trial of the selective Aurora B 398

    inhibitor, AZD1152 (barasertib) had an overall response rate of 25%.78 The toxicity profile, 399

    including mucositis and myelosuppression, was reported as being manageable in an older 400

    population. FLT3 kinase is a secondary target of AZD115279 and although the FLT3 status of 401

    patients on the trial was not reported, it is of interest that a number of responses occurred 402

    below the MTD and 55% of patients on the trial had cytogenetically normal AML (CN-403

    AML). Given the high incidence of FLT3 mutations in CN-AML, it is possible that the anti-404

    FLT3 activity of AZD1152 may have contributed to efficacy in patients with FLT3 405

    mutations. A phase II/III study of AZD1152, alone and in combination with low-dose 406

    cytarabine in older adults (> 60 years), is currently ongoing (NCT00952588). A recent 407

    preclinical study has demonstrated that paediatric AML cell lines are sensitive to Aurora B 408

    knockdown by shRNA and small molecule inhibition with AZD1152.80 One potential 409

    limitation of the AZD1152 compound is its delivery via a 7 day continuous IV infusion. 410

  • Moore et al. Novel therapies for childhood AML

    18

    The selective Aurora A inhibitor MLN8237 (alisertib), has shown response rates of 17% in 411

    an adult phase II trial 81 and preclinical activity in paediatric leukaemia.82 Of note, a recent 412

    preclinical study in acute megakaryoblastic leukaemia (AMKL) demonstrated that Aurora A 413

    inhibition with MLN8237 could induce polyploidization in both a Down Syndrome (DS) 414

    AMKL cell line (CMK) and primary blasts from children with non-DS-AMKL. MLN8237 415

    also had in vivo activity against a non-DS-AMKL model.83 A paediatric phase II trial of 416

    MLN8237 in children with relapsed/refractory solid tumours and leukaemia is currently 417

    ongoing (NCT01154816, COG-ADVL0921). Similarly, the Aurora kinase inhibitor AT9283, 418

    which also has potent activity against ABL, JAK2 and FLT3 kinases, has shown encouraging 419

    responses in adult leukaemia trials and is currently being assessed in a paediatric phase I 420

    study (EudraCT No. 2009-016952-36; NCT01431664). 421

    422

    MEK inhibition 423

    Mutations of N- and K-RAS in AML are examples of class I mutations, conferring a 424

    proliferative/survival stimulus.84 A recent comprehensive pre-clinical predictive biomarker 425

    analysis of 218 solid tumor and 81 haematological cancer cell lines revealed AML and CML 426

    cell lines as being particularly sensitive to the MEK inhibitor, GSK1120212.85 Amongst solid 427

    tumour cell lines, RAF/RAS mutations were predictive of sensitivity.85 In a panel of 12 AML 428

    cell lines, single-nanomolar IC50’s were reported for all 6 RAS-mutant (N- or K-RAS) cell 429

    lines, as well as 2 RAS-wt cell lines. The paediatric cell line Kasumi-1 (RAS-wt) was 430

    resistant, although other paediatric cell lines, THP-1 (N-RAS mutant) and MV4-11 (FLT3-431

    ITD+, RAS-wt), were sensitive.85 A phase I trial of GSK1120212 was reported at the 2010 432

    ASH meeting, with 12 of 14 patients treated having AML (including 2 transformed from 433

    MDS).86 Preliminary anti-leukaemic activity was reported, with one patient achieving CR. A 434

    phase I/II study of GSK1120212 in AML is ongoing (NCT00920140). 435

  • Moore et al. Novel therapies for childhood AML

    19

    436

    Aminopeptidase inhibition 437

    Aminopeptidases play a key role in removing amino acids from cellular peptides, a process 438

    required for protein regulation and recycling. Aminopeptidase inhibition depletes intracellular 439

    amino acids required for new protein synthesis.87 AML cells have been shown to be sensitive 440

    to aminopeptidase inhibition, undergoing apoptosis, whilst normal bone marrow cells are less 441

    susceptible.88 The aminopeptidase inhibitor Tosedostat has shown promising activity in phase 442

    I/II trials of older adults with predominantly relapsed/refractory AML, with single-agent 443

    response rates of up to 27%.89,90 Tosedostat appears well tolerated with thrombocytopaenia 444

    being the most common adverse event reported. 445

    446

    c-KIT inhibition 447

    Activating c-KIT mutations are common in paediatric and adult AML, particularly in CBF 448

    AML (incidence 21-54.5% with inv(16) and 17-46.8%% with t(8;21)).91-95 Although c-KIT 449

    mutations confer an increased risk of relapse in adults with CBF AML93, this does not appear 450

    to be the case in children.94,95 Murine models have demonstrated c-KIT mutations cooperate 451

    with AML1-ETO to cause aggressive AML in vivo, but are responsive to c-KIT inhibition 452

    with dasatinib.96 Dasatanib has also been shown to have single-agent activity against the 453

    t(8;21) paediatric cell line, Kasumi-1, which also harbours an N822K point mutation in the 454

    activation loop of c-KIT.97,98 Results of several ongoing clinical trials in AML, particularly 455

    those enriched with CBF patients, are eagerly awaited. 456

    457

    CXCR4 antagonism 458

    The critical role of the bone marrow micro-environment, or “niche” in regulating normal and 459

    malignant haematopoiesis is becoming increasingly recognised. The CXCR4 antagonist, 460

  • Moore et al. Novel therapies for childhood AML

    20

    plerixafor, has demonstrated efficacy in the mobilization of normal HSCs from the marrow 461

    for use in autologous transplantation in non-Hodgkin lymphoma and multiple myeloma.99,100 462

    Plerixafor has also been demonstrated to be a feasible and effective second-line strategy in 463

    mobilizing HSCs in children.101,102 Based on pre-clinical data demonstrating AML blasts 464

    could be mobilized into the peripheral blood, a recent phase I/II study demonstrated the 465

    feasibility of combining plerixafor with cytotoxic chemotherapy in adults with 466

    relapsed/refractory AML. When used in combination with mitoxantrone, etoposide and 467

    cytarabine, plerixafor increased mobilization of AML blasts into the peripheral circulation.103 468

    Overall CR rates of 46% were achieved and no dose-limiting toxicities were observed. 469

    470

    Epigenetic therapies 471

    Genes regulating DNA methylation and demethylation such as DNMT3A, IDH1/2 and TET2 472

    are frequently mutated in adult AML and have prognostic significance.104-107 Consequently, 473

    targeting epigenetic processes has become an attractive therapeutic strategy in adult AML. 474

    The DNA methyltransferase inhibitors decitabine and azacitidine are currently being 475

    evaluated in clinical trials of adults with AML and paediatric evaluation programs are being 476

    designed. A recent analysis of 46 patients treated on two trials of decitabine has shown an 477

    improved response rate in patients with DNMT3A mutations compared to those with wild-478

    type DNMT3A (CR rate 75% (6/8) vs. 34% (13/38) respectively, P=0.05).108 Studies of 479

    azacitidine alone and in sequential combination with lenalidomide have shown responses of 480

    approximately 50% in older adults with newly-diagnosed AML compared to those with 481

    relapsed disease.109,110 482

    483

    Another focus of epigenetic therapy has been histone deacetylase (HDAC) inhibition with 484

    compounds such as valproic acid, vorinostat and panabinostat. Widely used as an anti-485

  • Moore et al. Novel therapies for childhood AML

    21

    convulsant, valproic acid was first demonstrated to inhibit HDAC and cause partial 486

    differentiation of the paediatric AML cell line Kasumi-1 over a decade ago.111 Preclinical 487

    studies, including paediatric models, have also demonstrated single cytotoxic synergy with 488

    cytarabine and clofarabine.112,113 Clinical responses to valproic acid alone or in combination 489

    with cytarabine in adults with AML however, have been variable.114-116 It should be noted 490

    that when combined with decitabine, valproic acid has been associated with 491

    encephalopathy.117 492

    493

    Although a single-agent randomised phase II trial of vorinostat in adults with relapsed or 494

    high-risk untreated AML showed only minimal responses (1 CR from 37 patients)118 a recent 495

    phase II trial of vorinostat in combination with idarubicin and cytarabine (IA) in patients with 496

    AML and no CBF abnormalities resulted in an EFS of 47 weeks (range, 3 to 134 weeks) and 497

    a CR rate of 76%. Although not randomised, these responses compared favourably to 498

    historical data, the combination was reported as not excessively toxic compared to IA alone 499

    and all 11 FLT3-ITD+ patients responded (10 CR, 1 CRp).119 As discussed above, the relative 500

    rarity of mutations in genes regulating DNA methylation in childhood AML may limit the 501

    broader applicability of these agents for children, particularly if responses are confirmed to be 502

    associated with mutational status. 503

    504

    Although adult clinical trials have only recently been initiated, it is worth noting one potential 505

    epigenetic therapy that may be of significant importance to childhood leukemias with MLL 506

    gene rearrangements, given their relatively high incidence (AML 18%, ALL 8.3%).4 The 507

    histone H3 methyltransferase, DOT1L, plays a key role in MLL-fusion mediated 508

    leukemogenesis (reviewed in Ref 120). A small molecule inhibitor of DOT1L, EPZ004777, 509

    was recently shown to have in vivo preclinical efficacy against MLL-rearranged 510

  • Moore et al. Novel therapies for childhood AML

    22

    leukemia.121,122 A phase I trial of an optimized compound, EPZ-5676, opened in September 511

    2012 for adults patients with advanced hematological malignancies, including MLL-512

    rearranged AML and ALL (NCT01684150). 513

    514

    Finding and Hitting the Right Target in AML: Predictive and 515

    Pharmacodynamic Biomarkers 516

    Of critical importance for the rational and efficient development of targeted agents in cancer 517

    and leukaemia is the use of validated predictive and PD biomarkers. Predictive biomarkers 518

    are those that identify which patients are most likely to derive benefit from a given targeted 519

    agent, whilst PD biomarkers are needed to verify whether or not the desired molecular target 520

    is being modulated, and establish how target modulation relates to dose, toxicity and 521

    response. This information, together with careful examination of any resistance mechanisms 522

    or treatment failures, not only decreases the attrition rate of targeted therapeutics, but also 523

    improves the ongoing pre-clinical development of new agents. 524

    525

    FLT3 inhibitors provide arguably the best example of how robust predictive and PD 526

    biomarkers have been successfully applied in AML. It is well established that patients 527

    without activating mutations of FLT3 are less likely to benefit from FLT3 inhibition.33 Even 528

    with potent, second generation FLT3 inhibitors such as AC220, response rates in FLT3-WT 529

    patients are lower than in FLT3-ITD+ patients (34 vs. 44%).123 FLT3 mutational status 530

    therefore serves as a robust predictive biomarker for personalized AML therapy. 531

    Furthermore, results from trials of CEP-701 clearly demonstrate the importance of combining 532

    a predictive biomarker with a robust PD assay, since adequate FLT3 inhibition correlates 533

    with outcome for FLT3-mutated patients treated with CEP-701.45,47 An important PD assay 534

    developed for FLT3 inhibitors is the PIA assay.42 This assay involves incubation of a FLT3-535

  • Moore et al. Novel therapies for childhood AML

    23

    ITD+ cell line in the plasma of patients treated with FLT3 inhibitors. The degree of FLT3 536

    inhibition seen ex vivo for a patient at a given time-point during or after therapy is normalized 537

    to the amount of FLT3 phosphorylation observed when cells are incubated with that patient’s 538

    pre-treatment plasma. FLT3 ligand (FL) levels and protein binding can impact on the efficacy 539

    of FLT3 inhibition.46 The PIA assay is therefore superior to simply calculating the free drug 540

    concentration, since the plasma sample used to treat the cell line ex vivo contains not only 541

    free drug, but also the patient’s actual FL and plasma proteins at that time-point. Using the 542

    PIA assay, it has recently been demonstrated that when adequate FLT3 inhibition is achieved 543

    in combination with chemotherapy, relapse rates are lower and overall survival can be 544

    improved.47 545

    546

    The relatively small circulating blood volume and need for general anaesthetic when 547

    performing bone marrow aspiration can limit the number and nature of PD assays undertaken 548

    in paediatric leukaemia trials. Assays such as the PIA assay have been successfully adapted 549

    for use in children, such that smaller blood volumes are required, permitting use in phase I 550

    paediatric trials.124 Furthermore, the PIA assay has been successfully adapted and validated 551

    for therapeutic targets other than FLT3, with inhibition of phosphorylated histone H3 being 552

    used as a PD biomarker of Aurora kinase inhibition in a paediatric trial of AT9283 (EudraCT 553

    No. 2009-016952-36, NCT01431664).124 554

    555

    Future Directions and Trial Design 556

    Although the prognosis for children with AML has improved over recent decades, such 557

    improvements are largely attributable to more intensive use of cytotoxic chemotherapy and 558

    better supportive care.6 Although a number of novel therapeutic agents have been recently 559

    developed and show promising efficacy in adult AML, significant issues still exist for 560

  • Moore et al. Novel therapies for childhood AML

    24

    executing early phase clinical trials of such drugs in children. Often, patients may progress 561

    rapidly between screening and commencing a novel therapy, by which time they may become 562

    ineligible with poor performance status or complicating conditions such as uncontrolled, 563

    invasive infection. Furthermore, patients relapsing soon after allogeneic HSCT may have 564

    poor organ function deeming them ineligible. One possible solution is to treat patients with 565

    primary refractory or first-relapsed AML with targeted novel agents in a “window” period 566

    prior to allogeneic HSCT. Several factors need to be considered for this approach though, 567

    including the likelihood of achieving CR with second or third line chemotherapy, the 568

    potential of the novel agent causing organ toxicity which could compromise the success of 569

    subsequent HSCT and the time required to screen, enroll and treat patients with the novel 570

    agent prior to HSCT. 571

    572

    Accelerating drug development for childhood AML will rely heavily on effective 573

    international collaboration, particularly if enrolment on targeted therapeutic trials is to be 574

    restricted to the relatively small number of patients with predictive biomarkers of response. 575

    Groups such as the Innovative Therapies for Children with Cancer (ITCC) European 576

    Consortium and the Therapeutic Advances in Childhood Leukaemia and Lymphoma (TACL) 577

    consortium have an existing focus on early-phase trials of novel agents for childhood 578

    leukaemia. Although legislative, regulatory and pharmaceutical supply barriers prevent the 579

    enrolment of children from outside Europe or North America on certain trials, attempts to 580

    facilitate truly international early-phase studies are ongoing. Novel approaches to early-phase 581

    trial design are also critical in accelerating drug development, particularly in children. 582

    Important strategies include phase I trials with expansion cohorts at the RP2D enrolling 583

    patients with predictive biomarkers, and adaptive randomised trials incorporating a number of 584

    new agents with the aim of “picking-the-winner” or “dropping-the-loser”.125 Such approaches 585

  • Moore et al. Novel therapies for childhood AML

    25

    hold promise of reducing the number of large phase III trials needed and improving the 586

    likelihood of beneficial treatments being identified for malignancies like AML with 587

    increasingly heterogeneous biology. The important question of how much can be extrapolated 588

    from adult biology and trial data remains difficult to answer, hence defining the minimum 589

    dataset in children is an ongoing challenge. 590

    591

    Although significant challenges remain for novel drug development in paediatric oncology, 592

    particularly AML, the encouraging results of initial paediatric trials of therapies such as 593

    clofarabine and sorafenib, together with the large number of new agents in the pipeline is 594

    encouraging. Although survival rates in childhood AML are significantly better than adults 595

    with the disease, outcomes for children with AML are dramatically inferior compared with 596

    paediatric ALL.4 Improving outcomes in AML with cytotoxic chemotherapy alone is highly 597

    unlikely, given the current intensity of conventional therapy. Continued efforts to find 598

    effective novel therapies are therefore of critical importance if survival is to be improved and 599

    long-term morbidity reduced. 600

    601

  • Moore et al. Novel therapies for childhood AML

    26

    Acknowledgements 602

    ASM is supported by the National Health and Medical Research Council (Australia) and the 603

    Children’s Health Foundation Queensland. ADJP is supported by Cancer Research UK 604

    (programme grant C1178/A10294). ASM and ADJP acknowledge NHS funding to the NIHR 605

    Biomedical Research Centre at The Institute of Cancer Research and the Royal Marsden 606

    NHS Foundation Trust. 607

    608

    Conflicts of Interest 609

    ASM and ADJP are past and present employees respectively of The Institute of Cancer 610

    Research, which has a commercial interest in drug development programmes (see 611

    www.icr.ac.uk), and are subject to a “Rewards to Inventors Scheme” which may reward 612

    contributors to a programme that is subsequently licensed. ASM has received competitive 613

    research funding from Pfizer Inc., by way of a Pfizer Australia Cancer Research Grant. 614

    615

  • Moore et al. Novel therapies for childhood AML

    27

    References 616

    1. Dores GM, Devesa SS, Curtis RE, Linet MS, Morton LM. Acute leukemia incidence 617

    and patient survival among children and adults in the United States, 2001-2007. Blood 618

    2012; 119(1): 34-43. 619

    620

    2. Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid 621

    leukemia. J Clin Oncol 2011; 29(5): 487-94. 622

    623

    3. Gibson BE, Webb DK, Howman AJ, De Graaf SS, Harrison CJ, Wheatley K. Results 624

    of a randomized trial in children with Acute Myeloid Leukaemia: Medical Research 625

    Council AML12 trial. Br J Haematol 2011; 155(3): 366-376. 626

    627

    4. Pui CH, Carroll WL, Meshinchi S, Arceci RJ. Biology, Risk Stratification, and 628

    Therapy of Pediatric Acute Leukemias: An Update. J Clin Oncol 2011. 629

    630

    5. Creutzig U, van dH-EMM, Gibson B, Dworzak MN, Adachi S, de BE et al. Diagnosis 631

    and management of acute myeloid leukemia in children and adolescents: 632

    recommendations from an international expert panel, on behalf of the AML 633

    Committee of the International BFM Study Group. Blood 2012. 634

    635

    6. Rubnitz JE, Gibson B, Smith FO. Acute myeloid leukemia. Pediatr Clin North Am 636

    2008; 55(1): 21-51, ix. 637

    638

    7. Stevens RF, Hann IM, Wheatley K, Gray RG. Marked improvements in outcome with 639

    chemotherapy alone in paediatric acute myeloid leukemia: results of the United 640

    Kingdom Medical Research Council's 10th AML trial. MRC Childhood Leukaemia 641

    Working Party. Br J Haematol 1998; 101(1): 130-40. 642

    643

    8. Kaspers GJ, Zimmermann M, Reinhardt D, Gibson BE, Tamminga RY, Aleinikova O 644

    et al. Improved outcome in pediatric relapsed acute myeloid leukemia: results of a 645

    randomized trial on liposomal daunorubicin by the International BFM Study Group. J 646

    Clin Oncol. 2013; 31(5): 599-607. 647

    648

    9. Gorman MF, Ji L, Ko RH, Barnette P, Bostrom B, Hutchinson R et al. Outcome for 649

    children treated for relapsed or refractory acute myelogenous leukemia (rAML): a 650

    Therapeutic Advances in Childhood Leukemia (TACL) Consortium study. Pediatr 651

    Blood Cancer 2010; 55(3): 421-9. 652

    653

    10. Meadows AT, Friedman DL, Neglia JP, Mertens AC, Donaldson SS, Stovall M et al. 654

    Second neoplasms in survivors of childhood cancer: findings from the Childhood 655

    Cancer Survivor Study cohort. J Clin Oncol 2009; 27(14): 2356-62. 656

    657

    11. Mulrooney DA, Yeazel MW, Kawashima T, Mertens AC, Mitby P, Stovall M et al. 658

    Cardiac outcomes in a cohort of adult survivors of childhood and adolescent cancer: 659

    retrospective analysis of the Childhood Cancer Survivor Study cohort. BMJ 2009; 660

    339: b4606. 661

    662

  • Moore et al. Novel therapies for childhood AML

    28

    12. Ho PA, Alonzo TA, Kopecky KJ, Miller KL, Kuhn J, Zeng R et al. Molecular 663

    alterations of the IDH1 gene in AML: a Children's Oncology Group and Southwest 664

    Oncology Group study. Leukemia 2010; 24(5): 909-13. 665

    666

    13. Ho PA, Kutny MA, Alonzo TA, Gerbing RB, Joaquin J, Raimondi SC et al. 667

    Leukemic mutations in the methylation-associated genes DNMT3A and IDH2 are rare 668

    events in pediatric AML: a report from the Children's Oncology Group. Pediatr Blood 669

    Cancer 2011; 57(2): 204-9. 670

    671

    14. Damm F, Thol F, Hollink I, Zimmermann M, Reinhardt K, van den Heuvel-Eibrink 672

    MM et al. Prevalence and prognostic value of IDH1 and IDH2 mutations in childhood 673

    AML: a study of the AML-BFM and DCOG study groups. Leukemia 2011; 25(11): 674

    1704–1710. 675

    676

    15. Langemeijer SM, Jansen JH, Hooijer J, van Hoogen P, Stevens-Linders E, Massop M 677

    et al. TET2 mutations in childhood leukemia. Leukemia 2011; 25(1): 189-92. 678

    679

    16. Juhl-Christensen C, Ommen HB, Aggerholm A, Lausen B, Kjeldsen E, Hasle H et al. 680

    Genetic and epigenetic similarities and differences between childhood and adult 681

    AML. Pediatr Blood Cancer 2012; 58(4): 525-31. 682

    683

    17. Pui CH, Jeha S, Kirkpatrick P. Clofarabine. Nat Rev Drug Discov 2005; 4(5): 369-70. 684

    685

    18. Rubnitz JE, Crews KR, Pounds S, Yang S, Campana D, Gandhi VV et al. 686

    Combination of cladribine and cytarabine is effective for childhood acute myeloid 687

    leukemia: results of the St Jude AML97 trial. Leukemia 2009; 23(8): 1410-6. 688

    689

    19. Inaba H, Stewart CF, Crews KR, Yang S, Pounds S, Pui CH et al. Combination of 690

    cladribine plus topotecan for recurrent or refractory pediatric acute myeloid leukemia. 691

    Cancer 2010; 116(1): 98-105. 692

    693

    20. Chaleff S, Hurwitz CA, Chang M, Dahl G, Alonzo TA, Weinstein H. Phase II study 694

    of 2-chlorodeoxyadenosine plus idarubicin for children with acute myeloid leukaemia 695

    in first relapse: a paediatric oncology group study. Br J Haematol 2012; 156(5): 649-696

    55. 697

    698

    21. Krance RA, Hurwitz CA, Head DR, Raimondi SC, Behm FG, Crews KR et al. 699

    Experience with 2-chlorodeoxyadenosine in previously untreated children with newly 700

    diagnosed acute myeloid leukemia and myelodysplastic diseases. J Clin Oncol 2001; 701

    19(11): 2804-11. 702

    703

    22. Abrahamsson J, Clausen N, Gustafsson G, Hovi L, Jonmundsson G, Zeller B et al. 704

    Improved outcome after relapse in children with acute myeloid leukaemia. Br J 705

    Haematol 2007; 136(2): 229-236. 706

    707

    23. Fleischhack G, Hasan C, Graf N, Mann G, Bode U. IDA-FLAG (idarubicin, 708

    fludarabine, cytarabine, G-CSF), an effective remission-induction therapy for poor-709

    prognosis AML of childhood prior to allogeneic or autologous bone marrow 710

    transplantation: experiences of a phase II trial. Br J Haematol 1998; 102(3): 647-55. 711

    712

  • Moore et al. Novel therapies for childhood AML

    29

    24. Bonate PL, Arthaud L, Cantrell WR, Jr., Stephenson K, Secrist JA, 3rd, Weitman S. 713

    Discovery and development of clofarabine: a nucleoside analogue for treating cancer. 714

    Nature reviews. Drug discovery 2006; 5(10): 855-63. 715

    716

    25. Jeha S, Razzouk B, Rytting M, Rheingold S, Albano E, Kadota R et al. Phase II study 717

    of clofarabine in pediatric patients with refractory or relapsed acute myeloid 718

    leukemia. J Clin Oncol 2009; 27(26): 4392-7. 719

    720

    26. Johnston DL, Mandel KM. Fatal skin and liver toxicity in a patient treated with 721

    clofarabine. Pediatr Blood Cancer 2008; 50(5): 1082. 722

    723

    27. Inaba H, Rubnitz JE, Coustan-Smith E, Li L, Furmanski BD, Mascara GP et al. Phase 724

    I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib 725

    in combination with clofarabine and cytarabine in pediatric relapsed/refractory 726

    leukemia. J Clin Oncol 2011; 29(24): 3293-300. 727

    728

    28. Hijiya N, Gaynon P, Barry E, Silverman L, Thomson B, Chu R et al. A multi-center 729

    phase I study of clofarabine, etoposide and cyclophosphamide in combination in 730

    pediatric patients with refractory or relapsed acute leukemia. Leukemia 2009; 23(12): 731

    2259-64. 732

    733

    29. Kearns P, Graham NJ, Cummins M, Gibson B, Grainger JD, Keenan R et al. Phase I 734

    study of clofarabine and liposomal daunorubicin in childhood acute myeloid 735

    leukemia. J Clin Oncol 2011; 29: (suppl; abstr 9521). 736

    737

    30. Meshinchi S, Appelbaum FR. Structural and functional alterations of FLT3 in acute 738

    myeloid leukemia. Clin Cancer Res 2009; 15(13): 4263-9. 739

    740

    31. Meshinchi S, Alonzo TA, Stirewalt DL, Zwaan M, Zimmerman M, Reinhardt D et al. 741

    Clinical implications of FLT3 mutations in pediatric AML. Blood 2006; 108(12): 742

    3654-61. 743

    744

    32. Levis M, Small D. FLT3: ITDoes matter in leukemia. Leukemia 2003; 17(9): 1738-745

    52. 746

    747

    33. Knapper S. The clinical development of FLT3 inhibitors in acute myeloid leukemia. 748

    Expert Opin Investig Drugs 2011; 20(10): 1377-95. 749

    750

    34. Whitman SP, Archer KJ, Feng L, Baldus C, Becknell B, Carlson BD et al. Absence of 751

    the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia 752

    with normal cytogenetics and the internal tandem duplication of FLT3: a cancer and 753

    leukemia group B study. Cancer Res 2001; 61(19): 7233-9. 754

    755

    35. Gale RE, Green C, Allen C, Mead AJ, Burnett AK, Hills RK et al. The impact of 756

    FLT3 internal tandem duplication mutant level, number, size, and interaction with 757

    NPM1 mutations in a large cohort of young adult patients with acute myeloid 758

    leukemia. Blood 2008; 111(5): 2776-84. 759

    760

  • Moore et al. Novel therapies for childhood AML

    30

    36. Pratz KW, Sato T, Murphy KM, Stine A, Rajkhowa T, Levis M. FLT3-mutant allelic 761

    burden and clinical status are predictive of response to FLT3 inhibitors in AML. 762

    Blood 2010; 115(7): 1425-32. 763

    764

    37. Heidel F, Solem FK, Breitenbuecher F, Lipka DB, Kasper S, Thiede MH et al. 765

    Clinical resistance to the kinase inhibitor PKC412 in acute myeloid leukemia by 766

    mutation of Asn-676 in the FLT3 tyrosine kinase domain. Blood 2006; 107(1): 293-767

    300. 768

    769

    38. Man CH, Fung TK, Ho C, Han HH, Chow HC, Ma AC et al. Sorafenib treatment of 770

    FLT3-ITD(+) acute myeloid leukemia: favorable initial outcome and mechanisms of 771

    subsequent nonresponsiveness associated with the emergence of a D835 mutation. 772

    Blood 2012; 119(22): 5133-43. 773

    774

    39. Smith CC, Wang Q, Chin CS, Salerno S, Damon LE, Levis MJ et al. Validation of 775

    ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. 776

    Nature 2012; 485(7397): 260-3. 777

    778

    40. Swords R, Freeman C, Giles F. Targeting the FMS-like tyrosine kinase 3 in acute 779

    myeloid leukemia. Leukemia 2012; 26(10): 2176-85. 780

    781

    41. Smith BD, Levis M, Beran M, Giles F, Kantarjian H, Berg K et al. Single-agent CEP-782

    701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with 783

    relapsed or refractory acute myeloid leukemia. Blood 2004; 103(10): 3669-76. 784

    785

    42. Levis M, Brown P, Smith BD, Stine A, Pham R, Stone R et al. Plasma inhibitory 786

    activity (PIA): a pharmacodynamic assay reveals insights into the basis for cytotoxic 787

    response to FLT3 inhibitors. Blood 2006; 108(10): 3477-83. 788

    789

    43. Knapper S, Burnett AK, Littlewood T, Kell WJ, Agrawal S, Chopra R et al. A phase 2 790

    trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older 791

    patients with acute myeloid leukemia not considered fit for intensive chemotherapy. 792

    Blood 2006; 108(10): 3262-70. 793

    794

    44. Pratz K, Cortes J, Roboz G, Rao N, Arowojolu O, Stine A et al. A pharmacodynamic 795

    study of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical 796

    response. Blood 2009; 113(17): 3938-46. 797

    798

    45. Levis M, Ravandi F, Wang ES, Baer MR, Perl A, Coutre S et al. Results from a 799

    randomized trial of salvage chemotherapy followed by lestaurtinib for patients with 800

    FLT3 mutant AML in first relapse. Blood 2011. 801

    802

    46. Sato T, Yang X, Knapper S, White P, Smith BD, Galkin S et al. FLT3 ligand impedes 803

    the efficacy of FLT3 inhibitors in vitro and in vivo. Blood 2011; 117(12): 3286-93. 804

    805

    47. Knapper S, White P, Levis MJ, Hills RK, Russell NH, Burnett A. The Efficacy of the 806

    FLT3 Inhibitor Lestaurtinib in AML Depends on Adequate Plasma Inhibitory Activity 807

    (PIA), and Is Unaffected by Rising FLT Ligand Levels: An Update of the NCRI 808

    AML15 & 17 Trials. ASH Annual Meeting Abstracts 2011: 421a. 809

    810

  • Moore et al. Novel therapies for childhood AML

    31

    48. Zhang W, Konopleva M, Shi YX, McQueen T, Harris D, Ling X et al. Mutant FLT3: 811

    a direct target of sorafenib in acute myelogenous leukemia. J Natl Cancer Inst 2008; 812

    100(3): 184-98. 813

    814

    49. Keir ST, Maris JM, Lock R, Kolb EA, Gorlick R, Carol H et al. Initial testing (stage 815

    1) of the multi-targeted kinase inhibitor sorafenib by the pediatric preclinical testing 816

    program. Pediatr Blood Cancer 2010; 55(6): 1126-33. 817

    818

    50. Hu S, Niu H, Minkin P, Orwick S, Shimada A, Inaba H et al. Comparison of 819

    antitumor effects of multitargeted tyrosine kinase inhibitors in acute myelogenous 820

    leukemia. Mol Cancer Ther 2008; 7(5): 1110-20. 821

    822

    51. Goemans BF, Zwaan CM, Cloos J, de Lange D, Loonen AH, Reinhardt D et al. FLT3 823

    and KIT mutated pediatric acute myeloid leukemia (AML) samples are sensitive in 824

    vitro to the tyrosine kinase inhibitor SU11657. Leuk Res 2010; 34(10): 1302-7. 825

    826

    52. Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY et al. 827

    Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in 828

    younger patients with acute myeloid leukemia. J Clin Oncol 2010; 28(11): 1856-62. 829

    830

    53. Serve H, Wagner R, Sauerland C, Brunnberg U, Krug U, Schaich M et al. Sorafenib 831

    In Combination with Standard Induction and Consolidation Therapy In Elderly AML 832

    Patients: Results From a Randomized, Placebo-Controlled Phase II Trial. ASH Annual 833

    Meeting Abstracts 2010; 116(21): 333-. 834

    835

    54. Lacouture ME, Wu S, Robert C, Atkins MB, Kong HH, Guitart J et al. Evolving 836

    strategies for the management of hand-foot skin reaction associated with the 837

    multitargeted kinase inhibitors sorafenib and sunitinib. Oncologist 2008; 13(9): 1001-838

    11. 839

    840

    55. Moore AS, Faisal A, Gonzalez de Castro D, Bavetsias V, Sun C, Atrash B et al. 841

    Selective FLT3 inhibition of FLT3-ITD+ acute myeloid leukaemia resulting in 842

    secondary D835Y mutation: a model for emerging clinical resistance patterns. 843

    Leukemia 2012; 26(7): 1462-70. 844

    845

    56. Williams AB, Nguyen B, Li L, Brown P, Levis M, Leahy D et al. Mutations of 846

    FLT3/ITD confer resistance to multiple tyrosine kinase inhibitors. Leukemia 2012. 847

    848

    57. Bain BJ. Leukaemia diagnosis, 4 edn Wiley-Blackwell: Chichester, 2010. 849

    850

    58. Pollard JA, Alonzo TA, Loken M, Gerbing RB, Ho PA, Bernstein ID et al. 851

    Correlation of CD33 expression level with disease characteristics and response to 852

    gemtuzumab ozogamicin containing chemotherapy in childhood AML. Blood 2012; 853

    119(16): 3705-11. 854

    855

    59. Zwaan CM, Reinhardt D, Corbacioglu S, van Wering ER, Bokkerink JP, Tissing WJ 856

    et al. Gemtuzumab ozogamicin: first clinical experiences in children with 857

    relapsed/refractory acute myeloid leukemia treated on compassionate-use basis. Blood 858

    2003; 101(10): 3868-71. 859

    860

  • Moore et al. Novel therapies for childhood AML

    32

    60. Cooper TM, Franklin J, Gerbing RB, Alonzo TA, Hurwitz C, Raimondi SC et al. 861

    AAML03P1, a pilot study of the safety of gemtuzumab ozogamicin in combination 862

    with chemotherapy for newly diagnosed childhood acute myeloid leukemia: A report 863

    from the children's oncology group. Cancer 2011. 864

    865

    61. Satwani P, Bhatia M, Garvin JH, Jr., George D, Dela Cruz F, Le Gall J et al. A Phase 866

    I Study of Gemtuzumab Ozogamicin (GO) in Combination with Busulfan and 867

    Cyclophosphamide (Bu/Cy) and Allogeneic Stem Cell Transplantation in Children 868

    with Poor-Risk CD33(+) AML: A New Targeted Immunochemotherapy 869

    Myeloablative Conditioning (MAC) Regimen. Biol Blood Marrow Transplant 2011. 870

    871

    62. Zwaan CM, Reinhardt D, Zimmerman M, Hasle H, Stary J, Stark B et al. Salvage 872

    treatment for children with refractory first or second relapse of acute myeloid 873

    leukaemia with gemtuzumab ozogamicin: results of a phase II study. Br J Haematol 874

    2010; 148(5): 768-76. 875

    876

    63. Aplenc R, Alonzo TA, Gerbing RB, Lange BJ, Hurwitz CA, Wells RJ et al. Safety 877

    and efficacy of gemtuzumab ozogamicin in combination with chemotherapy for 878

    pediatric acute myeloid leukemia: a report from the Children's Oncology Group. J 879

    Clin Oncol 2008; 26(14): 2390-3295. 880

    881

    64. Rubnitz JE, Inaba H, Dahl G, Ribeiro RC, Bowman WP, Taub J et al. Minimal 882

    residual disease-directed therapy for childhood acute myeloid leukaemia: results of 883

    the AML02 multicentre trial. Lancet Oncol 2010; 11(6): 543-52. 884

    885

    65. Wadleigh M, Richardson PG, Zahrieh D, Lee SJ, Cutler C, Ho V et al. Prior 886

    gemtuzumab ozogamicin exposure significantly increases the risk of veno-occlusive 887

    disease in patients who undergo myeloablative allogeneic stem cell transplantation. 888

    Blood 2003; 102(5): 1578-82. 889

    890

    66. Burnett AK, Hills RK, Milligan D, Kjeldsen L, Kell J, Russell NH et al. Identification 891

    of patients with acute myeloblastic leukemia who benefit from the addition of 892

    gemtuzumab ozogamicin: results of the MRC AML15 trial. J Clin Oncol 2011; 29(4): 893

    369-77. 894

    895

    67. Cooper TM, Franklin J, Gerbing RB, Alonzo TA, Hurwitz C, Raimondi SC et al. 896

    AAML03P1, a pilot study of the safety of gemtuzumab ozogamicin in combination 897

    with chemotherapy for newly diagnosed childhood acute myeloid leukemia: a report 898

    from the Children's Oncology Group. Cancer 2012; 118(3): 761-9. 899

    900

    68. Jager E, van der Velden VH, te Marvelde JG, Walter RB, Agur Z, Vainstein V. 901

    Targeted drug delivery by gemtuzumab ozogamicin: mechanism-based mathematical 902

    model for treatment strategy improvement and therapy individualization. PLoS One 903

    2011; 6(9): e24265. 904

    905

    69. Malfuson JV, Konopacki J, Thepenier C, Eddou H, Foissaud V, de Revel T. 906

    Fractionated doses of gemtuzumab ozogamicin combined with 3 + 7 induction 907

    chemotherapy as salvage treatment for young patients with acute myeloid leukemia in 908

    first relapse. Ann Hematol 2012; 91(12): 1871-7. 909

    910

  • Moore et al. Novel therapies for childhood AML

    33

    70. Castaigne S, Pautas C, Terre C, Raffoux E, Bordessoule D, Bastie JN et al. Effect of 911

    gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid 912

    leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet 2012; 913

    379(9825): 1508-16. 914

    915

    71. Hasle H, Abrahamsson J, Forestier E, Ha SY, Heldrup J, Jahnukainen K et al. 916

    Gemtuzumab ozogamicin as postconsolidation therapy does not prevent relapse in 917

    children with AML: results from NOPHO-AML 2004. Blood 2012; 120(5): 978-84. 918

    919

    72. Guzman ML, Swiderski CF, Howard DS, Grimes BA, Rossi RM, Szilvassy SJ et al. 920

    Preferential induction of apoptosis for primary human leukemic stem cells. Proc Natl 921

    Acad Sci U S A 2002; 99(25): 16220-5. 922

    923

    73. Horton TM, Pati D, Plon SE, Thompson PA, Bomgaars LR, Adamson PC et al. A 924

    phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with 925

    refractory leukemia: a Children's Oncology Group study. Clin Cancer Res 2007; 926

    13(5): 1516-22. 927

    928

    74. Attar EC, De Angelo DJ, Supko JG, D'Amato F, Zahrieh D, Sirulnik A et al. Phase I 929

    and pharmacokinetic study of bortezomib in combination with idarubicin and 930

    cytarabine in patients with acute myelogenous leukemia. Clin Cancer Res 2008; 931

    14(5): 1446-54. 932

    933

    75. Messinger Y, Gaynon P, Raetz E, Hutchinson R, Dubois S, Glade-Bender J et al. 934

    Phase I study of bortezomib combined with chemotherapy in children with relapsed 935

    childhood acute lymphoblastic leukemia (ALL): a report from the therapeutic 936

    advances in childhood leukemia (TACL) consortium. Pediatr Blood Cancer 2010; 937

    55(2): 254-9. 938

    939

    76. Carmena M, Earnshaw WC. The cellular geography of aurora kinases. Nat Rev Mol 940

    Cell Biol 2003; 4(11): 842-54. 941

    942

    77. Moore AS, Blagg J, Linardopoulos S, Pearson AD. Aurora kinase inhibitors: novel 943

    small molecules with promising activity in acute myeloid and Philadelphia-positive 944

    leukemias. Leukemia 2010; 24(4): 671-8. 945

    946

    78. Lowenberg B, Muus P, Ossenkoppele G, Rousselot P, Cahn JY, Ifrah N et al. Phase 947

    I/II study to assess the safety, efficacy, and pharmacokinetics of barasertib 948

    (AZD1152) in patients with advanced acute myeloid leukemia. Blood 2011; 118(23): 949

    6030-6. 950

    951

    79. Grundy M, Seedhouse C, Shang S, Richardson J, Russell N, Pallis M. The FLT3 952

    internal tandem duplication mutation is a secondary target of the aurora B kinase 953

    inhibitor AZD1152-HQPA in acute myelogenous leukemia cells. Mol Cancer Ther 954

    2010; 9(3): 661-72. 955

    956

    80. Hartsink-Segers SA, Zwaan CM, Exalto C, Luijendijk MW, Calvert VS, Petricoin EF 957

    et al. Aurora kinases in childhood acute leukemia: the promise of aurora B as 958

    therapeutic target. Leukemia 2012 Sep 3, doi: 10.1038/leu.2012.256. [Epub ahead of 959

    print] 960

  • Moore et al. Novel therapies for childhood AML

    34

    961

    81. Goldberg SL, Fenaux P, Craig MD, Gyan E, Lister J, Kassis J et al. Phase 2 Study of 962

    MLN8237, An Investigational Aurora A Kinase (AAK) Inhibitor In Patients with 963

    Acute Myelogenous Leukemia (AML) or Myelodysplastic Syndromes (MDS). ASH 964

    Annual Meeting Abstracts 2010; 116(21): 3273-. 965

    966

    82. Maris JM, Morton CL, Gorlick R, Kolb EA, Lock R, Carol H et al. Initial testing of 967

    the aurora kinase A inhibitor MLN8237 by the Pediatric Preclinical Testing Program 968

    (PPTP). Pediatr Blood Cancer 2010; 55(1): 26-34. 969

    970

    83. Wen Q, Goldenson B, Silver SJ, Schenone M, Dancik V, Huang Z et al. Identification 971

    of regulators of polyploidization presents therapeutic targets for treatment of AMKL. 972

    Cell 2012; 150(3): 575-89. 973

    974

    84. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood 975

    2002; 100(5): 1532-42. 976

    977

    85. Jing J, Greshock J, Holbrook JD, Gilmartin AG, Zhang X, McNeil E et al. 978

    Comprehensive Predictive Biomarker Analysis for MEK Inhibitor GSK1120212. Mol 979

    Cancer Ther 2011. 980

    981

    86. Borthakur G, Foran JM, Kadia T, Jabbour E, Wissel P, Cox D et al. GSK1120212, a 982

    MEK1/MEK2 Inhibitor, Demonstrates Acceptable Tolerability and Preliminary 983

    Activity In a Dose Rising Trial In Subjects with AML and Other Hematologic 984

    Malignancies. ASH Annual Meeting Abstracts 2010; 116(21): 3281-. 985

    986

    87. Krige D, Needham LA, Bawden LJ, Flores N, Farmer H, Miles LE et al. CHR-2797: 987

    an antiproliferative aminopeptidase inhibitor that leads to amino acid deprivation in 988

    human leukemic cells. Cancer research 2008; 68(16): 6669-79. 989

    990

    88. Jenkins C, Hewamana S, Krige D, Pepper C, Burnett A. Aminopeptidase inhibition by 991

    the novel agent CHR-2797 (tosedostat) for the therapy of acute myeloid leukemia. 992

    Leuk Res 2011; 35(5): 677-81. 993

    994

    89. Lowenberg B, Morgan G, Ossenkoppele GJ, Burnett AK, Zachee P, Duhrsen U et al. 995

    Phase I/II clinical study of Tosedostat, an inhibitor of aminopeptidases, in patients 996

    with acute myeloid leukemia and myelodysplasia. J Clin Oncol 2010; 28(28): 4333-8. 997

    998

    90. Mathisen MS, Ravandi F. Efficacy of tosedostat, a novel, oral agent for elderly 999

    patients with relapsed or refractory acute myeloid leukemia: a review of the Phase II 1000

    OPAL trial. Future oncology 2012; 8(4): 351-7. 1001

    1002

    91. Beghini A, Ripamonti CB, Cairoli R, Cazzaniga G, Colapietro P, Elice F et al. KIT 1003

    activating mutations: incidence in adult and pediatric acute myeloid leukemia, and 1004

    identification of an internal tandem duplication. Haematologica 2004; 89(8): 920-5. 1005

    1006

    92. Balgobind BV, Hollink IH, Arentsen-Peters ST, Zimmermann M, Harbott J, Beverloo 1007

    HB et al. Integrative analysis of type-I and type-II aberrations underscores the genetic 1008

    heterogeneity of pediatric acute myeloid leukemia. Haematologica 2011; 96(10): 1009

    1478-87. 1010

  • Moore et al. Novel therapies for childhood AML

    35

    1011

    93. Paschka P, Marcucci G, Ruppert AS, Mrozek K, Chen H, Kittles RA et al. Adverse 1012

    prognostic significance of KIT mutations in adult acute myeloid leukemia with 1013

    inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol 2006; 1014

    24(24): 3904-11. 1015

    1016

    94. Goemans BF, Zwaan CM, Miller M, Zimmermann M, Harlow A, Meshinchi S et al. 1017

    Mutations in KIT and RAS are frequent events in pediatric core-binding factor acute 1018

    myeloid leukemia. Leukemia 2005; 19: 1536-42. 1019

    1020

    95. Pollard JA, Alonzo TA, Gerbing RB, Ho PA, Zeng R, Ravindranath Y et al. 1021

    Prevalence and prognostic significance of KIT mutations in pediatric patients with 1022

    core binding factor AML enrolled on serial pediatric cooperative trials for de novo 1023

    AML. Blood 2010; 115: 2372-9. 1024

    1025

    96. Wang YY, Zhao LJ, Wu CF, Liu P, Shi L, Liang Y et al. C-KIT mutation cooperates 1026

    with full-length AML1-ETO to induce acute myeloid leukemia in mice. Proc Natl 1027

    Acad Sci U S A 2011; 108(6): 2450-5. 1028

    1029

    97. Mpakou VE, Kontsioti F, Papageorgiou S, Spathis A, Kottaridi C, Girkas K et al. 1030

    Dasatinib inhibits proliferation and induces apoptosis in the KASUMI-1 cell line 1031

    bearing the t(8;21)(q22;q22) and the N822K c-kit mutation. Leuk Res 2012. 1032

    1033

    98. Kolb EA, Gorlick R, Houghton PJ, Morton CL, Lock RB, Tajbakhsh M et al. Initial 1034

    testing of dasatinib by the pediatric preclinical testing program. Pediatr Blood Cancer 1035

    2008; 50(6): 1198-206. 1036

    1037

    99. DiPersio JF, Micallef IN, Stiff PJ, Bolwell BJ, Maziarz RT, Jacobsen E et al. Phase 1038

    III prospective randomized double-blind placebo-controlled trial of plerixafor plus 1039

    granulocyte colony-stimulating factor compared with placebo plus granulocyte 1040

    colony-stimulating factor for autologous stem-cell mobilization and transplantation 1041

    for patients with non-Hodgkin's lymphoma. J Clin Oncol 2009; 27(28): 4767-73. 1042

    1043

    100. DiPersio JF, Stadtmauer EA, Nademanee A, Micallef IN, Stiff PJ, Kaufman JL et al. 1044

    Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem 1045

    cells for autologous stem cell transplantation in patients with multiple myeloma. 1046

    Blood 2009; 113(23): 5720-6. 1047

    1048

    101. Aabideen K, Anoop P, Ethell ME, Potter MN. The feasibility of plerixafor as a 1049

    second-line stem cell mobilizing agent in children. J Pediatr Hematol Oncol 2011; 1050

    33(1): 65-7. 1051

    1052

    102. Hubel K, Fresen MM, Salwender H, Basara N, Beier R, Theurich S et al. Plerixafor 1053

    with and without chemotherapy in poor mobilizers: results from the German 1054

    compassionate use program. Bone Marrow Transplant 2011; 46(8): 1045-52. 1055

    1056

    103. Uy GL, Rettig MP, Motabi IH, McFarland K, Trinkaus KM, Hladnik LM et al. A 1057

    phase I/II study of chemosensitization with the CXCR4 antagonist plerixafor in 1058

    relapsed or refractory acute myeloid leukemia. Blood 2012. 1059

    1060

  • Moore et al. Novel therapies for childhood AML

    36

    104. Ley TJ, Ding L, Walter MJ, McLellan MD, Lamprecht T, Larson DE et al. DNMT3A 1061

    mutations in acute myeloid leukemia. N Engl J Med 2010; 363(25): 2424-33. 1062

    1063

    105. Delhommeau F, Dupont S, Della Valle V, James C, Trannoy S, Masse A et al. 1064

    Mutation in TET2 in myeloid cancers. N Engl J Med 2009; 360(22): 2289-301. 1065

    1066

    106. Paschka P, Schlenk RF, Gaidzik VI, Habdank M, Kronke J, Bullinger L et al. IDH1 1067

    and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and 1068

    confer adverse prognosis in cytogenetically normal acute myeloid leukemia with 1069

    NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol 2010; 1070

    28(22): 3636-43. 1071

    1072

    107. Weissmann S, Alpermann T, Grossmann V, Kowarsch A, Nadarajah N, Eder C et al. 1073

    Landscape of TET2 mutations in acute myeloid leukemia. Leukemia 2012; 26(5): 1074

    934-42. 1075

    1076

    108. Metzeler KH, Walker A, Geyer S, Garzon R, Klisovic RB, Bloomfield CD et al. 1077

    DNMT3A mutations and response to the hypomethylating agent decitabine in acute 1078

    myeloid leukemia. Leukemia 2012; 26(5): 1106-7. 1079

    1080

    109. Al-Ali HK, Jaekel N, Junghanss C, Maschmeyer G, Krahl R, Cross M et al. 1081

    Azacitidine in patients with acute myeloid leukemia medically unfit for or resistant to 1082

    chemotherapy: a multicenter phase I/II study. Leuk Lymphoma 2012; 53(1): 110-7. 1083

    1084

    110. Pollyea DA, Kohrt HE, Gallegos L, Figueroa ME, Abdel-Wahab O, Zhang B et al. 1085

    Safety, efficacy and biological predictors of response to sequential azacitidine and 1086

    lenalidomide for elderly patients with acute myeloid leukemia. Leukemia 2012; 26(5): 1087

    893-901. 1088

    1089

    111. Gottlicher M, Minucci S, Zhu P, Kramer OH, Schimpf A, Giavara S et al. Valproic 1090

    acid defines a novel class of HDAC inhibitors inducing differentiation of transformed 1091

    cells. EMBO J 2001; 20(24): 6969-78. 1092

    1093

    112. Xie C, Edwards H, Lograsso SB, Buck SA, Matherly LH, Taub JW et al. Valproic 1094

    acid synergistically enhances the cytotoxicity of clofarabine in pediatric acute 1095

    myeloid leukemia cells. Pediatr Blood Cancer 2012; 59(7): 1245-51. 1096

    1097

    113. Xie C, Edwards H, Xu X, Zhou H, Buck SA, Stout ML et al. Mechanisms of 1098

    synergistic antileukemic interactions between valproic acid and cytarabine in pediatric 1099

    acute myeloid leukemia. Clin Cancer Res 2010; 16(22): 5499-510. 1100

    1101

    114. Corsetti MT, Salvi F, Perticone S, Baraldi A, De Paoli L, Gatto S et al. Hematologic 1102

    improvement and response in elderly AML/RAEB patients treated with valproic acid 1103

    and low-dose Ara-C. Leuk Res 2011; 35(8): 991-7. 1104

    1105

    115. Lane S, Gill D, McMillan NA, Saunders N, Murphy R, Spurr T et al. Valproic acid 1106

    combined with cytosine arabinoside in elderly patients with acute myeloid leukemia 1107

    has in vitro but limited clinical activity. Leuk Lymphoma 2012; 53(6): 1077-83. 1108

    1109

  • Moore et al. Novel therapies for childhood AML

    37

    116. Kuendgen A, Schmid M, Schlenk R, Knipp S, Hildebrandt B, Steidl C et al. The 1110

    histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination 1111

    with all-trans retinoic acid in patients with acute myeloid leukemia. Cancer 2006; 1112

    106(1): 112-9. 1113

    1114

    117. Blum W, Klisovic RB, Hackanson B, Liu Z, Liu S, Devine H et al. Phase I study of 1115

    decitabine alone or in combination with valproic acid in acute myeloid leukemia. J 1116

    Clin Oncol 2007; 25(25): 3884-91. 1117

    1118

    118. Schaefer EW, Loaiza-Bonilla A, Juckett M, DiPersio JF, Roy V, Slack J et al. A 1119

    phase 2 study of vorinostat in acute myeloid leukemia. Haematologica 2009; 94(10): 1120

    1375-82. 1121

    1122

    119. Garcia-Manero G, Tambaro FP, Bekele NB, Yang H, Ravandi F, Jabbour E et al. 1123

    Phase II trial of vorinostat with idarubicin and cytarabine for patients with newly 1124

    diagnosed acute myelogenous leukemia or myelodysplastic syndrome. J Clin Oncol 1125

    2012; 30(18): 2204-10. 1126

    1127

    120. Bernt KM, Armstrong SA. Targeting epigenetic programs in MLL-rearranged 1128

    leukemias. Hematology Am Soc Hematol Educ Program 2011: 354-60. 1129

    1130

    121. Daigle SR, Olhava EJ, Therkelsen CA, Majer CR, Sneeringer CJ, Song J et al. 1131

    Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L 1132

    inhibitor. Cancer Cell 2011; 20(1): 53-65. 1133

    1134

    122. Chen L, Deshpande AJ, Banka D, Bernt KM, Dias S, Bus