Skip to main content
Log in

Molecularly Targeted Therapies for Pediatric Acute Myeloid Leukemia

Progress to Date

  • Leading Article
  • Published:
Pediatric Drugs Aims and scope Submit manuscript

Abstract

While acute myeloid leukemia (AML) is significantly less common than acute lymphoblastic leukemia (ALL) in childhood, it is significantly more deadly with only half as many children likely to be cured with standard therapy. In addition, the typical treatment for AML is among the most toxic of treatments for pediatric cancer; it includes intensive multiagent chemotherapy and, often, hematopoietic stem cell transplantation. Given the poor prognosis of pediatric AML and the significant toxicity of standard AML therapy, novel therapies are needed. Improved understanding of the molecular and cellular biology of leukemia has facilitated the development of molecularly targeted therapies.

In this article, we review progress to date with agents that are showing promise in the treatment of pediatric AML including targeted immunoconjugates, inhibitors of signaling molecules (e.g. FMS-like tyrosine kinase 3 [FLT3], farnesyltransferase, and mammalian target of rapamycin [mTOR]), agents that target epigenetic regulation of gene expression (DNA methyltransferase inhibitors and histone deacetylase inhibitors), and proteasome inhibitors. For the specific agents in each of these classes, we summarize the published preclinical data and the clinical trials that have been completed, are in progress, or are being planned for children with AML. Finally, we discuss potential challenges to the success of molecularly targeted therapy including demonstrating adequate targeting of leukemia stem cells, developing synergistic and tolerable combinations of agents, and designing adequately powered clinical trials to test efficacy in molecularly defined subsets of patients.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Notes

  1. The use of trade names is for product identification purposes only and does not imply endorsement.

References

  1. Woods WG. Curing childhood acute myeloid leukemia (AML) at the half-way point: promises to keep and miles to go before we sleep. Pediatr Blood Cancer 2006; 46(5): 565–9

    Article  PubMed  Google Scholar 

  2. Pui CH, Evans WE. Treatment of acute lymphoblastic leukemia. N Engl J Med 2006; 354: 166–78

    Article  PubMed  CAS  Google Scholar 

  3. Pui CH, Relling MV, Downing JR. Acute lymphoblastic leukemia. N Engl J Med 2004; 350: 1535–48

    Article  PubMed  CAS  Google Scholar 

  4. Pui CH, Sandlund JT, Pei D, et al. Improved outcome for children with acute lymphoblastic leukemia: results of Total Therapy Study XIIIB at St Jude Children’s Research Hospital. Blood 2004; 104: 2690–6

    Article  PubMed  CAS  Google Scholar 

  5. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. Blood 2005; 106(4): 1154–63

    Article  PubMed  CAS  Google Scholar 

  6. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997; 3(7): 730–7

    Article  PubMed  CAS  Google Scholar 

  7. Hehlmann R, Hochhaus A, Baccarani M. Chronic myeloid leukemia. Lancet 2007; 370: 342–50

    Article  PubMed  CAS  Google Scholar 

  8. Michor F, Hughes TP, Iwasa Y, et al. Dynamics of chronic myeloid leukaemia. Nature 2005; 435: 1267–70

    Article  PubMed  CAS  Google Scholar 

  9. Graham SM, Jorgensen HG, Allan E, et al. Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood 2002; 99: 319–25

    Article  PubMed  CAS  Google Scholar 

  10. Tallman MS. Acute promyelocytic leukemia as a paradigm for targeted therapy. Semin Hematol 2004; 41 (2 Suppl. 4): 27–32

    Article  PubMed  CAS  Google Scholar 

  11. Grimwade D, Enver T. Acute promyelocytic leukemia: where does it stem from? Leukemia 2004; 18: 375–84

    Article  PubMed  CAS  Google Scholar 

  12. Freeman SD, Kelm S, Barber EK, et al. Characterization of CD33 as a new member of the sialoadhesin family of cellular interaction molecules. Blood 1995; 85: 2005–12

    PubMed  CAS  Google Scholar 

  13. Drexler HG. Classification of acute myeloid leukemias: a comparison of FAB and immunophenotyping. Leukemia 1987; 1: 697–705

    PubMed  CAS  Google Scholar 

  14. Brashem-Stein C, Flowers DA, Smith FO, et al. Ontogeny of hematopoietic stem cell development: reciprocal expression of CD33 and a novel molecule by maturing myeloid and erythroid progenitors. Blood 1993; 82: 792–9

    PubMed  CAS  Google Scholar 

  15. Sievers EL, Larson RA, Stadtmauer EA, et al. Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. J Clin Oncol 2001; 19: 3244–54

    PubMed  CAS  Google Scholar 

  16. Larson RA, Sievers EL, Stadtmauer EA, et al. Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer 2005; 104: 1442–52

    Article  PubMed  CAS  Google Scholar 

  17. Arceci RJ, Sande J, Lange B, et al. Safety and efficacy of gemtuzumab ozogamicin in pediatric patients with advanced CD33+ acute myeloid leukemia. Blood 2005; 106: 1183–8

    Article  PubMed  CAS  Google Scholar 

  18. Walter RB, Gooley TA, van der Velden VH, et al. CD33 expression and P-glycoprotein-mediated drug efflux inversely correlate and predict clinical out come in patients with acute myeloid leukemia treated with gemtuzumab ozogamicin monotherapy. Blood 2007; 109: 4168–70

    Article  PubMed  CAS  Google Scholar 

  19. Linenberger ML, Hong T, Flowers D, et al. Multidrug-resistance phenotype and clinical responses to gemtuzumab ozogamicin. Blood 2001; 98: 988–94

    Article  PubMed  CAS  Google Scholar 

  20. Matsui H, Takeshita A, Naito K, et al. Reduced effect of gemtuzumab ozogamicin (CMA-676) on P-glycoprotein and/or CD34-positive leukemia cells and its restoration by multidrug resistance modifiers. Leukemia 2002; 16: 813–9

    Article  PubMed  CAS  Google Scholar 

  21. Walter RB, Raden BW, Hong TC, et al. Multidrug resistance protein attenuates gemtuzumab ozogamicin-induced cytotoxicity in acute myeloid leukemia cells. Blood 2003; 102: 1466–73

    Article  PubMed  CAS  Google Scholar 

  22. Walter RB, Raden BW, Cronk MR, et al. The peripheral benzodiazepine receptor ligand PK11195 overcomes different resistance mechanisms to sensitize AML cells to gemtuzumab ozogamicin. Blood 2004; 103: 4276–84

    Article  PubMed  CAS  Google Scholar 

  23. Becton D, Dahl GV, Ravindranath Y, et al. Randomized use of cyclosporin A (CsA) to modulate P-glycoprotein in children with AML in remission: Pediatric Oncology Group Study 9421. Blood 2006; 107: 1315–24

    Article  PubMed  CAS  Google Scholar 

  24. Florian S, Sonneck K, Hauswirth AW, et al. Detection of molecular targets on the surface of CD34+/CD38− stem cells in various myeloid malignancies. Leuk Lymphoma 2006; 47: 207–22

    Article  PubMed  CAS  Google Scholar 

  25. Bernstein ID, Singer JW, Smith FO, et al. Differences in the frequency of normal and clonal precursors of colony-forming cells in chronic myelogenous leukemia and acute myelogenous leukemia. Blood 1992; 79: 1811–6

    PubMed  CAS  Google Scholar 

  26. Hauswirth AW, Florian S, Printz D, et al. Expression of the target receptor CD33 in CD34+/CD38−/CD123+ AML stem cells. Eur J Clin Invest 2007; 37: 73–82

    Article  PubMed  CAS  Google Scholar 

  27. Pearce DJ, Taussig DC, Bonnet D. Implications of the expression of myeloid markers on normal and leukemic stem cells. Cell Cycle 2006; 5: 271–3

    Article  PubMed  CAS  Google Scholar 

  28. Lo-Coco F, Cimino G, Breccia M, et al. Gemtuzumab ozogamicin (Mylotarg) as a single agent for molecularly relapsed acute promyelocytic leukemia. Blood 2004; 104: 1995–9

    Article  PubMed  CAS  Google Scholar 

  29. Estey EH, Giles FJ, Beran M, et al. Experience with gemtuzumab ozogamycin (‘mylotarg’) and all-trans retinoic acid in untreated acute promyelocytic leukemia. Blood 2002; 99: 4222–4

    Article  PubMed  CAS  Google Scholar 

  30. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med 2001; 344: 1031–7

    Article  PubMed  CAS  Google Scholar 

  31. Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 2002; 347: 472–80

    Article  PubMed  CAS  Google Scholar 

  32. Cools J, DeAngelo DJ, Gotlib J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N Engl J Med 2003; 348: 1201–14

    Article  PubMed  CAS  Google Scholar 

  33. Sattler M, Scheijen B, Weisberg E, et al. Mutated tyrosine kinases as therapeutic targets in myeloid leukemias. Adv Exp Med Biol 2003; 532: 121–40

    Article  PubMed  CAS  Google Scholar 

  34. Brown P, Small D. FLT3 inhibitors: a paradigm for the development of targeted therapeutics for paediatric cancer. Eur J Cancer 2004; 40: 707–21

    Article  PubMed  CAS  Google Scholar 

  35. Levis M, Small D. FLT3 tyrosine kinase inhibitors. Int J Hematol 2005; 82: 100–7

    Article  PubMed  CAS  Google Scholar 

  36. Carow CE, Levenstein M, Kaufmann SH, et al. Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias. Blood 1996; 87: 1089–96

    PubMed  CAS  Google Scholar 

  37. Kiyoi H, Towatari M, Yokota S, et al. Internal tandem duplication of the FLT3 gene is a novel modality of elongation mutation which causes constitutive activation of the product. Leukemia 1998; 12: 1333–7

    Article  PubMed  CAS  Google Scholar 

  38. Meshinchi S, Woods WG, Stirewalt DL, et al. Prevalence and prognostic significance of Flt3 internal tandem duplication in pediatric acute myeloid leukemia. Blood 2001; 97: 89–94

    Article  PubMed  CAS  Google Scholar 

  39. Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 2001; 97: 2434–9

    Article  PubMed  CAS  Google Scholar 

  40. Iwai T, Yokota S, Nakao M, et al. Internal tandem duplication of the FLT3 gene and clinical evaluation in childhood acute myeloid leukemia: the Children’s Cancer and Leukemia Study Group, Japan. Leukemia 1999; 13: 38–43

    Article  PubMed  CAS  Google Scholar 

  41. Kondo M, Horibe K, Takahashi Y, et al. Prognostic value of internal tandem duplication of the FLT3 gene in childhood acute myelogenous leukemia. Med Pediatr Oncol 1999; 33: 525–9

    Article  PubMed  CAS  Google Scholar 

  42. Meshinchi S, Alonzo TA, Stirewalt DL, et al. Clinical implications of FLT3 mutations in pediatric AML. Blood 2006; 108: 3654–61

    Article  PubMed  CAS  Google Scholar 

  43. Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 2002; 99: 4326–35

    Article  PubMed  CAS  Google Scholar 

  44. Whitman SP, Archer KJ, Feng L, et al. Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a cancer and leukemia group B study. Cancer Res 2001; 61: 7233–9

    PubMed  CAS  Google Scholar 

  45. Levis M, Allebach J, Tse KF, et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood 2002; 99: 3885–91

    Article  PubMed  CAS  Google Scholar 

  46. Brown P, Meshinchi S, Levis M, et al. Pediatric AML primary samples with FLT3/ITD mutations are preferentially killed by FLT3 inhibition. Blood 2004; 104: 1841–9

    Article  PubMed  CAS  Google Scholar 

  47. Smith BD, Levis M, Beran M, et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 2004; 103: 3669–76

    Article  PubMed  CAS  Google Scholar 

  48. Knapper S, Burnett AK, Littlewood T, et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 2006; 108: 3262–70

    Article  PubMed  CAS  Google Scholar 

  49. Levis M, Smith BD, Beran M, et al. A randomized, open-label study of lestaurtinib (CEP-701), an oral FLT3 inhibitor, administered in sequence with chemotherapy in patients with relapsed AML harboring FLT3 activating mutations: clinical response correlates with successful FLT3 inhibition [abstract]. Blood 2005; 106: 403a

    Article  Google Scholar 

  50. Brown P, Levis M, Mclntyre E, et al. Combinations of the FLT3 inhibitor CEP-701 and chemotherapy synergistically kill infant and childhood MLL-rearranged ALL cells in a sequence-dependent manner. Leukemia 2006; 20: 1368–76

    Article  PubMed  CAS  Google Scholar 

  51. Levis M, Pham R, Smith BD, et al. In vitro studies of a FLT3 inhibitor combined with chemotherapy: sequence of administration is important to achieve synergistic cytotoxic effects. Blood 2004; 104: 1145–50

    Article  PubMed  CAS  Google Scholar 

  52. Kottaridis PD, Gale RE, Langabeer SE, et al. Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors. Blood 2002; 100: 2393–8

    Article  PubMed  CAS  Google Scholar 

  53. Levis M, Murphy KM, Pham R, et al. Internal tandem duplications of the FLT3 gene are present in leukemia stem cells. Blood 2005; 106: 673–80

    Article  PubMed  CAS  Google Scholar 

  54. Pollard JA, Alonzo TA, Gerbing RB, et al. FLT3 internal tandem duplication in CD34+/CD33− precursors predicts poor outcome in acute myeloid leukemia. Blood 2006; 108: 2764–9

    Article  PubMed  CAS  Google Scholar 

  55. Farr C, Gill R, Katz F, et al. Analysis of ras gene mutations in childhood myeloid leukaemia. Br J Haematol 1991; 77: 323–7

    Article  PubMed  CAS  Google Scholar 

  56. MacKenzie KL, Dolnikov A, Millington M, et al. Mutant N-ras induces myeloproliferative disorders and apoptosis in bone marrow repopulated mice. Blood 1999; 93: 2043–56

    PubMed  CAS  Google Scholar 

  57. Loh ML, Vattikuti S, Schubbert S, et al. Mutations in PTPN11 implicate the SHP-2 phosphatase in leukemogenesis. Blood 2004; 103: 2325–31

    Article  PubMed  CAS  Google Scholar 

  58. Emanuel PD, Snyder RC, Wiley T, et al. Inhibition of juvenile myelomonocytic leukemia cell growth in vitro by farnesyltransferase inhibitors. Blood 2000; 95: 639–45

    PubMed  CAS  Google Scholar 

  59. Emanuel PD. Juvenile myelomonocytic leukemia. Curr Hematol Rep 2004; 3: 203–9

    PubMed  Google Scholar 

  60. Khosravi-Far R, Cox AD, Kato K, et al. Protein prenylation: key to ras function and cancer intervention? Cell Growth Differ 1992; 3: 461–9

    PubMed  CAS  Google Scholar 

  61. Harousseau JL, Lancet JE, Reiffers J, et al. A phase 2 study of the oral farnesyltransferase inhibitor tipifarnib in patients with refractory or relapsed acute myeloid leukemia. Blood 2007; 109: 5151–6

    Article  PubMed  CAS  Google Scholar 

  62. Lancet JE, Gojo I, Gotlib J, et al. A phase 2 study of the farnesyltransferase inhibitor tipifarnib in poor-risk and elderly patients with previously untreated acute myelogenous leukemia. Blood 2007; 109: 1387–94

    Article  PubMed  CAS  Google Scholar 

  63. Zimmerman TM, Harlin H, Odenike OM, et al. Dose-ranging pharmacodynamic study of tipifarnib (Rl 15777) in patients with relapsed and refractory hematologic malignancies. J Clin Oncol 2004; 22: 4816–22

    Article  PubMed  CAS  Google Scholar 

  64. Cortes J, Albitar M, Thomas D, et al. Efficacy of the farnesyl transferase inhibitor R115777 in chronic myeloid leukemia and other hématologie malignancies. Blood 2003; 101: 1692–7

    Article  PubMed  CAS  Google Scholar 

  65. Karp JE, Lancet JE, Kaufmann SH, et al. Clinical and biologic activity of the farnesyltransferase inhibitor R115777 in adults with refractory and relapsed acute leukemias: a phase 1 clinical-laboratory correlative trial. Blood 2001; 97: 3361–9

    Article  PubMed  CAS  Google Scholar 

  66. Widemann BC, Fox E, Goodspeed W, et al. Phase I trial of the farnesyltransferase inhibitor (FTI) R115777 in children with refractory leukemias [abstract]. Am Soc Clin Oncol 2003; 22: 3250a

    Google Scholar 

  67. Herman JG, Civin CI, Issa JP, et al. Distinct patterns of inactivation of p15INK4B and pl6INK4A characterize the major types of hematological malignancies. Cancer Res 1997; 57: 837–41

    PubMed  CAS  Google Scholar 

  68. Seedhouse CH, Das-Gupta EP, Russell NH. Methylation of the hMLH1 promoter and its association with microsatellite instability in acute myeloid leukemia. Leukemia 2003; 17: 83–8

    Article  PubMed  CAS  Google Scholar 

  69. Gore SD. Combination therapy with DNA methyltransferase inhibitors in hematologic malignancies. Nat Clin Pract Oncol 2005; 2Suppl. 1: S30–5

    Article  PubMed  CAS  Google Scholar 

  70. Friedman AD. Leukemogenesis by CBF oncoproteins. Leukemia 1999; 13: 1932–42

    Article  PubMed  CAS  Google Scholar 

  71. Tenen DG, Hromas R, Licht JD, et al. Transcription factors, normal myeloid development, and leukemia. Blood 1997; 90: 489–519

    PubMed  CAS  Google Scholar 

  72. Liu S, Shen T, Huynh L, et al. Interplay of RUNX1/MTG8 and DNA methyltransferase 1 in acute myeloid leukemia. Cancer Res. 2005; 65: 1277–84

    Article  PubMed  CAS  Google Scholar 

  73. Liu S, Klisovic RB, Vukosavljevic T, et al. Targeting AML1/ETO-HDAC repressor complex: a novel mechanism for valproic acid mediated gene expression and cellular differentiation in AMLl/ETO-positive acute myeloid leukemia cells. J Pharmacol Exp Ther 2007; 321: 953–60

    Article  PubMed  CAS  Google Scholar 

  74. Klisovic MI, Maghraby EA, Parthun MR, et al. Depsipeptide (FR 901228) promotes histone acetylation, gene transcription, apoptosis and its activity is enhanced by DNA methyltransferase inhibitors in AML1/ETO-positive leukemic cells. Leukemia 2003; 17: 350–8

    Article  PubMed  CAS  Google Scholar 

  75. Grignani F, De Matteis S, Nervi C, et al. Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature 1998; 391: 815–8

    Article  PubMed  CAS  Google Scholar 

  76. Lin RJ, Nagy L, Inoue S, et al. Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature 1998; 391: 811–4

    Article  PubMed  CAS  Google Scholar 

  77. Di Croce L, Raker VA, Corsaro M, et al. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science 2002; 295: 1079–82

    Article  PubMed  Google Scholar 

  78. Whitman SP, Liu S, Vukosavljevic T, et al. The MLL partial tandem duplication: evidence for recessive gain-of-function in acute myeloid leukemia identifies a novel patient subgroup for molecular-targeted therapy. Blood 2005; 106: 345–52

    Article  PubMed  CAS  Google Scholar 

  79. Dorrance AM, Liu S, Yuan W, et al. Mll partial tandem duplication induces aberrant Hox expression in vivo via specific epigenetic alterations. J Clin Invest 2006; 116: 2707–16

    Article  PubMed  CAS  Google Scholar 

  80. So CW, Lin M, Ayton PM, et al. Dimerization contributes to oncogenic activation of MLL chimeras in acute leukemias. Cancer Cell 2003; 4: 99–110

    Article  PubMed  CAS  Google Scholar 

  81. Gore SD, Baylin S, Sugar E, et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res 2006; 66: 6361–9

    Article  PubMed  CAS  Google Scholar 

  82. Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B, et al. Phase 1/2 study of the combination of 5-aza-2′-deoxycytidine with valproic acid in patients with leukemia. Blood 2006; 108: 3271–9

    Article  PubMed  CAS  Google Scholar 

  83. Pyatt DW, Stillman WS, Yang Y, et al. An essential role for NF-kappaB in human CD34(+) bone marrow cell survival. Blood 1999; 93: 3302–8

    PubMed  CAS  Google Scholar 

  84. Guzman ML, Neering SJ, Upchurch D, et al. Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 2001; 98: 2301–7

    Article  PubMed  CAS  Google Scholar 

  85. Guzman ML, Swiderski CF, Howard DS, et al. Preferential induction of apoptosis for primary human leukemic stem cells. Proc Natl Acad Sci U S A 2002; 99: 16220–5

    Article  PubMed  CAS  Google Scholar 

  86. Cortes J, Thomas D, Koller C, et al. Phase I study of bortezomib in refractory or relapsed acute leukemias. Clin Cancer Res 2004; 10: 3371–6

    Article  PubMed  CAS  Google Scholar 

  87. Orlowski RZ, Voorhees PM, Garcia RA, et al. Phase 1 trial of the proteasome inhibitor bortezomib and pegylated liposomal doxorubicin in patients with advanced hematologic malignancies. Blood 2005; 105: 3058–65

    Article  PubMed  CAS  Google Scholar 

  88. Horton TM, Pati D, Plon SE, et al. A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: a Children’s Oncology Group study. Clin Cancer Res 2007; 13: 1516–22

    Article  PubMed  CAS  Google Scholar 

  89. Xu Q, Simpson SE, Scialla TJ, et al. Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood 2003; 102: 972–80

    Article  PubMed  CAS  Google Scholar 

  90. Min YH, Eom JI, Cheong JW, et al. Constitutive phosphorylation of Akt/PKB protein in acute myeloid leukemia: its significance as a prognostic variable. Leukemia 2003; 17: 995–7

    Article  PubMed  CAS  Google Scholar 

  91. Recher C, Beyne-Rauzy O, Demur C, et al. Antileukemic activity of rapamycin in acute myeloid leukemia. Blood 2005; 105: 2527–34

    Article  PubMed  CAS  Google Scholar 

  92. Yee KW, Zeng Z, Konopleva M, et al. Phase I/II study of the mammalian target of rapamycin inhibitor everolimus (RAD001) in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res 2006; 12: 5165–73

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

The authors have no conflicts of interest that are directly relevant to the content of this review. This work was supported by grants from the NCI (K23 CA111728, P.B.), Damon Runyon-Lilly Clinical Intestigator Award (P.B.) and Children’s Cancer Foundation (P.B.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrick Brown.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Brown, P., Smith, F.O. Molecularly Targeted Therapies for Pediatric Acute Myeloid Leukemia. Pediatr-Drugs 10, 85–92 (2008). https://doi.org/10.2165/00148581-200810020-00003

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00148581-200810020-00003

Keywords

Navigation