当前位置: 首页 > 期刊 > 《血液学杂志》 > 2005年第16期 > 正文
编号:11175986
Drug therapy for acute myeloid leukemia
http://www.100md.com 《血液学杂志》
     the Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago IL

    the Division of Hematology/Oncology, Brigham and Women's Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA

    the Department of Hematology and Bone Marrow Transplantation, Rambam Medical Center, Technion Israel Institute of Technology, Haifa, Israel.

    Abstract

    Although improvement in outcomes has occurred in younger adults with acute myeloid leukemia (AML) during the past 4 decades, progress in older adults has been much less conspicuous, if at all. Approximately 50% to 75% of adults with AML achieve complete remission (CR) with cytarabine and an anthracycline such as daunorubicin or idarubicin or the anthracenedione mitoxantrone. However, only approximately 20% to 30% of the patients enjoy long-term disease survival. Various postremission strategies have been explored to eliminate minimal residual disease. The optimal dose, schedule, and number of cycles of postremission chemotherapy for most patients are not known. A variety of prognostic factors can predict outcome and include the karyotype of the leukemic cells and the presence of transmembrane transporter proteins, which extrude certain chemotherapy agents from the cell and confer multidrug resistance and mutations in or over expressions of specific genes such as WT1, CeBPA, BAX and the ratio of BCL2 to BAX, BAALC, eVI1, KIT, and FLT3. Most recently, insights into the molecular pathogenesis of AML have led to the development of more specific targeted agents and have ushered in an exciting new era of antileukemia therapy. Such agents include the immunoconjugate gemtuzumab ozogamicin, multidrug resistance inhibitors, farnesyl transferase inhibitors, histone deacetylase and proteosome inhibitors, antiangiogenesis agents, Fms-like tyrosine kinase 3 (FLT3) inhibitors, and apoptosis inhibitors.

    Introduction

    Acute myeloid leukemia (AML) is a heterogeneous group of diseases characterized by uncontrolled proliferation of clonal neoplastic hematopoietic precursor cells and impaired production of normal hematopoiesis leading to neutropenia, anemia, and thrombocytopenia.1 If untreated, patients die of infection or bleeding usually in a matter of weeks. Some older adults may have a slower progressive clinical course. An estimated 10 600 new cases occurred in the United States in 2002 and 7400 patients died of the disease.2 The overall incidence is 3.4 cases per 100 000 population; 1.2 cases per 100 000 population at age 30 and more than 20 cases per 100 000 population at age 80 years.2 The median age is 20 years and has been increasing over the past decade.

    Historically, the diagnosis and response to therapy were established from morphology and cytochemistry. Although morphology remains the initial diagnostic tool for any patient with acute leukemia, the past decade has witnessed increased reliance on cell-surface antigen expression by immunophenotyping, usually carried out by flow cytometry, as well as cytogenetic and molecular markers.

    During the past 4 decades, many studies have investigated a wide variety of cytotoxic antileukemic agents. Most recently, insights into the molecular pathogenesis of AML have led to the development of the more specific targeted therapy. This review focuses on current and evolving drug therapy in the treatment of adults with AML.

    Current treatment results

    Approximately 50% to 75% of adults with AML achieve complete remission (CR) with the deoxycytidine analog cytarabine and an anthracycline antibiotic, such as daunorubicin or idarubicin, or the anthracenedione mitoxantrone, which inhibit the enzyme topoisomerase IIa. However, only 20% to 30% of patients enjoy long-term disease-free survival (DFS). The majority of patients die of their disease, primarily because of persistent or relapsed AML. In an eastern Cooperative Oncology Group (eCOG) analysis of the outcome of approximately 3000 patients with previously untreated AML entered on 5 successive clinical trials with cytarabine and daunorubicin for induction and with increasingly more intensive postremission therapy, 62% achieved CR, but 76% relapsed or died.3,4 The 5-year overall survival (OS) rate among 2000 patients younger than 55 years has improved from 11% in the 1970s to 37% in the 1990s. (Figure 1A-C). In contrast, among 1000 patients age 55 years and older, progress over the past 3 decades has been very modest.5

    Prognostic factors

    The outcome for adults with AML depends on a variety of factors, including age of the patient, intensity of postremission therapy, and biologic characteristics of the disease, the most important of which are the cytogenetics at presentation.4,6-8 The karyotype of the leukemic cells can distinguish 3 groups with either favorable, intermediate, or poor prognostic risk6-8 (Figure 2). Other factors include the presence of transmembrane transporter proteins, which extrude certain chemotherapy agents from the cell and confer multidrug resistance,9 and mutations in or overexpression of specific genes such as WT1,10,11 CeBPA,12 BAX and the ratio of BCL2 to BAX,13 BAALC,14 eVI1,15 KIT,16 and FLT3.17,18

    Induction chemotherapy

    Development of a standard regimen

    During the past 35 years, a series of studies has established an induction regimen that has become a standard of care for patients not participating on a clinical trial. A widely used combination for induction is the cell cycle–specific agent cytarabine 100 mg/m2 by continuous infusion for 7 days and the non–cell-cycle–specific anthracycline antibiotic daunorubicin 45 to 60 mg/m2/d intravenously for 3 days.19-21 To improve the CR rate, studies have tested alternative and higher doses of anthracyclines or the anthracenediones,22-33 higher doses of cytarabine,34-37 new agents combined with cytarabine and daunorubicin such as etoposide, the purine analog fludarabine or the camptothecin topotecan,38-40 or sequential standard therapy followed by high doses of cytarabine.41-44 Despite theoretic advantages,45 none of these approaches is definitively better than the standard regimen (Table 1).

    Hematopoietic growth factors

    Multiple studies have established the safety of hematopoietic growth factors when administered in induction and consolidation. Growth factors have a role in the supportive care of AML when given after induction therapy to reduce the period of neutropenia.46,47 Recently, there has been renewed interest in the use of growth factors as priming agents, to move leukemia cells into a phase of the cell cycle, which might render them more susceptible to cytotoxic chemotherapy. However, benefits of such a strategy have not been definitively established. One large study using granulocyte colony-stimulating factor (G-CSF) showed no effect on CR rate but an improvement in DFS.48 Another study, also using G-CSF, reported the opposite, an improvement in CR rate but no effect on DFS.47 A study with granulocyte-macrophage colony-stimulating factor (GM-CSF) showed no effect on either CR rate or DFS.29

    Postremission therapy

    Various strategies have been explored to eliminate minimal residual disease not apparent in the bone marrow of patients in CR which could contribute to relapse. Such strategies have included intensive consolidation therapy, high-dose chemotherapy, or chemoradiotherapy with either allogeneic or autologous hematopoietic stem-cell transplantation (HSCT), or low-dose maintenance therapy.

    Intensive consolidation chemotherapy

    Retrospective analyses of cooperative group studies and a prospective randomized trial show that increasing the intensity of postremission therapy is beneficial in younger but not older adults.3,4,49 Several studies have evaluated the role of intensive postremission consolidation with high-dose (3 gm/m2/dose) cytarabine (HiDAC). A prospective study by the Cancer and Leukemia Group B (CALGB) demonstrated that 4 courses of HiDAC are significantly better than 4 courses of intermediate- (400 mg/m2/dose) or standard-dose cytarabine (100 mg/m2/dose), confirming a dose-response effect in younger patients and a benefit in patients with good-risk cytogenetics.50 Cerebellar dysfunction, particularly in older adults and in those with hepatic or renal dysfunction, is an important toxicity.

    General conclusions about postremission consolidation chemotherapy

    Although postremission therapy is a sine qua non for curing AML, fundamental issues remain unresolved (Table 1). The optimal dose, schedule, and number of cycles of consolidation chemotherapy for most patients with AML who achieve CR have not been established. In younger patients, cycles of intensive consolidation chemotherapy, often with, but not limited to, high-doses of cytarabine, prolong DFS and OS.

    Prospective studies of intensive postremission chemotherapy, allogeneic hematopoietic stem-cell transplantation, and autologous hematopoietic stem-cell transplantation

    Hematopoietic stem-cell transplantation refers to the administration of very intensive chemotherapy with or without radiation and infusion of previously collected hematopoietic stem cells harvested from either the patient (autologous), or a human leukocyte antigen (HLA)–matched donor (allogeneic). Autologous HSCT is limited by the lack of the immunologic reaction referred to as graft-versus-leukemia (GVL) effect present in patients undergoing allogeneic HSCT, in which the donated allogeneic cells recognize the recipient's leukemic cells as foreign. Furthermore, there is a theoretic risk of infusion of occult residual leukemic cells.51 To decrease toxicities associated with such intensive doses of chemotherapy, lower doses have been explored, relying more on the immunologically mediated GVL effect to eradicate the disease.52 Although the benefits of this strategy of nonmyeloablative HSCT is currently under investigation,52,53 its ultimate role may be in older adults in first CR who are unable to tolerate standard conditioning regimens. Whether such a regimen can reduce the toxicity associated with matched unrelated donor HSCT is currently being explored.54 A potential limitation is the 3 to 9 months required for the immunologic GVL effect to develop in the presence of rapidly proliferating leukemia cells.

    Allogeneic HSCT for patients with AML in first CR is associated with the lowest relapse rate and provides the best antileukemic potential, but it is associated consistently with a higher risk of treatment-related mortality than either autologous HSCT or consolidation chemotherapy.55-58 As a result, the benefit of a lower relapse rate is offset by a higher treatment-related mortality. The decision to recommend allogeneic transplantation in first CR must be made on a patient-by-patient basis, evaluating the risks and patient age, type of donor match, and disease prognosis. However, it is reasonable to consider such a strategy in patients with poor-risk cytogenetics and some younger patients with intermediate-risk cytogenetics who are otherwise suitable candidates.

    Current HSCT techniques have changed significantly, and the transplant-related mortality with contemporary autologous HSCT is 0% to 3% compared with 14% to 18% in published studies, a fact that, historically, has obscured the potential benefit of the lower relapse rate. Furthermore, current understanding of the pathophysiology of AML mandates that therapeutic decisions be made, in part, on cytogenetic and molecular prognostic factors at diagnosis. This has led to subgroup analyses of small patient cohorts that limit confidence in the results.

    Maintenance therapy

    Historic and contemporary studies using various maintenance regimens consistently show a benefit in DFS but not OS.49,59,60 Further studies are needed to establish a definitive role for this therapeutic modality in AML.

    Treatment for acute promyelocytic leukemia

    Acute promyelocytic leukemia (APL) is treated differently from all other subtypes of AML and has become the most curable in adults. This subtype is associated with the t(15;17) translocation which results in a fusion transcript, the promyelocytic leukemia–retinoic acid receptor (PML-RAR), derived from the juxtaposition of the PML gene on chromosome 15 and the retinoic acid receptor gene on chromosome 17.61 The vitamin A derivative, all-trans retinoic acid (ATRA), has the remarkable ability to induce differentiation of leukemic promyelocytes in patients with APL, resulting in high CR rates.62-67 Therefore, APL is the first subtype of AML treated with an agent targeted to a specific genetic mutation. The benefits of ATRA in APL were identified as part of Chinese herbal medicine treatments rather than as a conscious effort to target the molecular abnormality in APL. Two prospective randomized trials demonstrate that ATRA with chemotherapy significantly improves DFS and OS, and approximately 70% of patients are cured of APL.65-67 The concepts that cytarabine may not be required in the treatment of a subtype of AML and that maintenance appears advantageous represent departures from conventional management.65-69

    Treatment of older adults

    The treatment of older adults (ages 55-60 years) with AML is very disappointing with modest, if any, improvement in OS over the past 4 decades.5,70 Older patients do not tolerate intensive chemotherapy as well as younger patients, particularly if they have preexisting comorbidities.71 In addition, older adults frequently have leukemia cells with poor-prognosis karyotypes9 and frequently express the multidrug resistance marker P-glycoprotein, rendering their cells more resistant to chemotherapy.9 It is possible that multidrug resistance marker expression identifies an immature stem-cell–like leukemia. Historic studies using attenuated doses of chemotherapy or delaying intensive therapy until disease progression led to poor results.72,73 Contemporary studies in older patients have established that approximately half can achieve CR.29,31 The major problem is that older adults have a high rate of relapse, and OS rates are less than 10%.5 No clearly effective postremission therapy has been established. Considering that AML occurs much more frequently in older patients,2 this represents the most important challenge in drug therapy for AML. The discouraging overall outcome in older adults has spurred major efforts in the development of new targeted drug therapies.

    Treatment of patients with relapsed or refractory AML

    The treatment of adults with relapsed or refractory AML is also unsatisfactory. The most important factor predicting response to reinduction chemotherapy appears to be the duration of first CR.74 High-dose cytarabine has been an effective reinduction regimen.75 However, the addition of other agents to HiDAC does not improve outcome.75 Allogeneic HSCT may provide the highest likelihood of cure, presumably related to the generation of an immunologically mediated graft-versus leukemia reaction not present with salvage chemotherapy, preferably after induction of a second CR.76-79 However, there is a lack of prospective data to definitively demonstrate a superior outcome with such a strategy. If a suitable histocompatible sibling donor is not available, transplantation from an alternative donor such as a matched unrelated donor80 or a haploidentical stem-cell donor81,82 can be considered. However, given the risks of reactivation of cytomegalovirus (CMV) and other opportunistic infections, this strategy is still investigational. A small number of patients in second CR also may be cured with autologous HSCT.83 Standard chemotherapy alone is rarely curative in patients who have relapsed. Most of these patients, as well as older adults who have not relapsed but who have a poor prognosis, are candidates for novel investigational approaches (Table 2). Much of the search for new agents focuses on biologically targeted strategies that may be effective without the severe myeloablative toxicity that, historically, has been the hallmark of drug therapy for AML.

    Insights into novel therapy based on genetic mechanisms of disease

    There is more known about the genetic basis of AML than perhaps any other human cancer. More than 100 different mutations and/or gene rearrangements have been identified in AML. Because AML is a sporadic disease, only rarely occurring as a heritable trait, many of the initial insights were derived from cloning of acquired recurring chromosomal translocations from bone marrow of affected individuals. More recently, a spectrum of subcytogenetic point mutations and gene rearrangements have been identified through characterization of candidate loci, and it is likely that many more will be identified.84,85

    Mutations that activate signal transduction cascades in AML

    a therapeutic perspective, these mutations can be subdivided into several subgroups. For example, a majority of patients with AML have mutations that result in constitutive activation of signal transduction pathways that confer proliferative and/or survival advantage to hematopoietic progenitors (Table 3). These include mutations that constitutively activate receptor tyrosine kinases, such as FLT3 or C-KIT receptor tyrosine kinase (C-KIT), oncogenic mutations in RAS, or activating mutations in protein tyrosine phosphatases such as PTPN11 (SHP2). These appear to segregate as a complementation group, in that only very rarely are any 2 of these observed in the same patients with AML. Thus, as discussed under "New agents directed at molecular and other specific targets," small molecule inhibitors of these pathways such as FLT3 or C-KIT inhibitors, or inhibitors of RAS through farnesyl transferase inhibition, are attractive as potential therapeutic agents. These mutations collectively account for approximately 50% of patients with AML, but it is plausible that the high-throughput genomic approaches to gene discovery will ultimately identify activating mutations in signal transduction pathways in all patients with AML. In addition, the high frequency of mutations in signal transduction pathways suggests that small molecule inhibitors of common downstream effectors shared by all mutations may also be effective (eg, MeK inhibitors to impair oncogenic RAS signals, or mTOR with rapamycin as a target for PI3K/AKT activation86). Such agents could be used alone or in combination with inhibitors of upstream effectors such as FLT3.

    Mutations in genes that result in dysregulated gene transcription

    A second broad complementation group of mutations and gene rearrangements is defined by loss-of-function mutation in transcription factors and transcriptional coactivators that are important for normal hematopoietic development. These mutations also rarely if ever are present in the same patient with leukemia, appear to confer the property of impaired hematopoietic differentiation to leukemic blasts, and may contribute to self-renewal potential, a hallmark of all human cancers. Novel therapeutic approaches have focused on compounds that might override the block in differentiation, as exemplified by the paradigm of ATRA therapy for APL. The mechanistic basis for therapeutic efficacy of ATRA in APL is complex, but the phenotypic consequence of ATRA therapy is induction of terminal differentiation and apoptosis of leukemic blasts. A key component of ATRA activity is release of the nuclear corepressor complex, containing histone deacetylase (HDAC), that is aberrantly recruited to promoters by the PML-RAR fusion.87 Aberrant recruitment of nuclear corepressor complexes by leukemogenic fusion proteins appears to be a central theme in AML. For example, RUNX1-eTO and CBF-MYH11 associated with t(8;21) and inv(16), respectively, aberrantly recruit the nuclear corepressor complex to RUNX1 target genes.88-90 Thus, development of small molecule inhibitors of HDAC may be of therapeutic value in induction of differentiation and might be used in combination with agents that target signal transduction pathways. Other loss-of-function mutations that impair hematopoietic differentiation and are potential targets for therapy are listed in Table 3 and may involve the hematopoietic transcription factors RUNX1, GATA-1, CeBPA, and PU.1, as well as mutations that affect components of the transcriptional apparatus itself, such as MLL fusions and fusions involving transcriptional coactivators. In addition, there is evidence for overexpression of certain transcription factors such as eVI1 in AML as a consequence of the t(3;21) translocation,91,92 or through as yet unidentified trans- or cis-acting mutations. Overexpression of eVI1 also results in a myelodysplastic phenotype in murine models of disease, suggesting that it may be a viable target for therapeutic intervention.93

    Finally, there are several mutations that occur in genes that encode proteins involved in nuclear cytoplasmic shuttling. These include direct involvement of components of the nuclear pore, such as the NUP98 gene that is fused through chromosomal translocation to a spectrum of partners, including homeobox-containing (HOX) family members,94 and the NUP214 gene that is fused to DeK oncogene (DeK) in patients with t(6;9) AML. Although the fusion partners must play an important role in disease pathogenesis, it also seems likely that the frequent selection for fusions containing components of the nuclear pore in AML must also connote a functional relevance for the nucleoprorins themselves. Most recently, it has been reported that nucleophosmin (NPMI), a nucleolar protein that is involved in nucleocytoplasmic shuttling, is mutated and mislocalized to the cytoplasm of AML cells in approximately 35% of patients.95 It is not known what the functional consequence of this observation is, but it again suggests that dysregulation of the normal nuclear to cytoplasmic trafficking machinery may be important in the pathogenesis of AML.

    Attention has turned to the self-renewal potential of leukemic cells as a therapeutic target. A small population of blasts has been identified in human AML that has the properties of leukemic stem cells96 and is thought to be responsible for continuous growth and propagation of leukemia blasts and for relapse of disease in patients after remission induction with intensive chemotherapy. Several genes and pathways have been identified that are important for this critical property of self-renewal, including the WNT/-catenin pathway, Notch, Bmi-1, and HOX family members.97-101 Although it is not yet certain how these pathways might be effectively targeted therapeutically, or whether it will be possible to target these pathways without excessive toxicities, these are ultimately the most important target cells to ablate (Figure 3).

    New agents directed at molecular and other specific targets

    Among the most exciting developments has been the remarkable ability to classify subtypes of AML by characterizing the expression profiles of all known genes in the human genome using microarray technology.102-105 The expression profile of clusters of genes will be useful not only to classify patients but also to identify specific molecular targets for therapeutic intervention. Currently, cytogenetics at presentation provides the most important prognostic information in AML. However, 70% of patients are in the intermediate-risk category and, of these, about 70% have a normal karyotype. Therefore, the most common karyotype in AML, normal, is a large, heterogeneous and poorly defined population. expression profiling has already refined the diagnostic classification of leukemias and holds promise for individualized approaches to therapy.

    Some targets to which therapy has already been directed include cell-surface antigens such as CD33, a glycoprotein expressed on most AML blast cells and not on normal hematopoietic stem cells; P-glycoprotein, a mediator of multidrug resistance which acts as an efflux pump to extrude chemotherapy from the cell; enzymes involved in signal transduction such as farnesyl transferase; vascular endothelial growth factor and other angiogenic proteins; enzymes that contribute to the enhanced proliferative and survival capacity of leukemia blasts, such as FLT3 or RAS; enzymes that contribute to impaired hematopoietic differentiation, such as HDAC inhibitors, and the antiapoptotic gene BCL-2. While these new strategies have not yet translated into the kind of clinically meaningful advances as has ATRA in APL, they represent a first wave of targeted therapies in AML and provide a platform for development of more efficacious targeted therapies. In addition, it is likely that none of these will be curative as single agents in treatment of this complex disease, but rather they will need to be used in combination with conventional therapy (as is ATRA) or with other molecularly targeted agents.

    Gemtuzumab ozogamicin

    The first of these new targeted agents to be approved by the US Food and Drug Administration is gemtuzumab ozogamicin. This agent is an immunoconjugate of an anti-CD33 antibody chemically linked to a potent cytotoxic agent, calicheamicin.106 Complete remission by conventional criteria is achieved in approximately 15% of patients with AML in first relapse.107,108 Occasionally, patients have developed a veno-occlusive diseaselike syndrome, and caution is indicated for those patients proceeding to transplantation. Two recent studies suggest that gemtuzumab ozogamicin may be associated with a higher CR rate when administered with intensive chemotherapy, and major cooperative groups are conducting phase-3 studies that incorporate gemtuzumab ozogamicin as part of standard chemotherapy in patients newly diagnosed with AML.109,110 If randomized clinical trials confirm these findings, the standard of care for induction in AML will change for the first time in 30 years. Attempts to offer gemtuzumab ozogamicin to older adults as initial induction therapy have not been encouraging.111,112 Data suggest that the effects of gemtuzumab ozogamicin may be potentiated by the use of a P-glycoprotein inhibitor.113

    Multidrug resistance inhibitors

    Although several agents inhibit P-glycoprotein, with the exception of a Southwest Oncology Group (SWOG) trial testing cyclosporine given with cytarabine and infusional daunorubicin,114 randomized trials of MDR modulators such as PSC-833 have not shown benefit.115,116 It is also possible that expression of multidrug resistance simply identifies an immature stem-cell–like leukemia. More potent modulators such as zosuquidar are currently being studied.117

    Farnesyl transferase inhibitors

    Mutations and dysregulation of Ras have been associated with the development of myeloid leukemias.118 Farnesyl transferase inhibitors (FTIs) interfere with RAS signaling by precluding farnesylation of RAS and transfer to the plasma membrane.119 These agents have activity in refractory AML120 and are modestly active in patients newly diagnosed with AML.121 Responses do not correlate with mutational status of Ras, which suggests that activation of native RAS by upstream effectors (such as mutant FLT3) might also be of therapeutic value. However, in some cases response does not even correlate with the degree of FT inhibition, suggesting that there may be other important targets. Preliminary data reporting a CR rate of 21% in 92 patients newly diagnosed has led to a current major intergroup trial evaluation of the FTI tipifarnib (Zarnestra) in older adults who are unable to tolerate conventional chemotherapy.

    Histone deacetylase and proteosome inhibitors

    Aberrant recruitment of the nuclear corepressor complex by leukemogenic fusion proteins is a recurring theme in AML and may result both in modification of chromatin structure at critical hematopoietic promoters, as well as aberrant acetylation of proteins that regulate cell-cycle progression and other functions.122 HDAC inhibitors induce differentiation of malignant cells and are under active investigation in AML.123,124

    The proteosome inhibitor bortezomib, a newly approved drug for multiple myeloma, has demonstrated preclinical synergistic activity with the histone deacetylase inhibitors, as well as potential single-agent activity in leukemia.125,126 Clinical efficacy in AML is intriguing, although as yet, unproven.

    Antiangiogenesis agents

    The potential role for antiangiogenesis therapy in AML is suggested by evidence that bone marrow biopsies for patients with AML demonstrate increased microvessel density that is associated with a poor prognosis.127 Furthermore, vascular endothelial growth factor (VeGF) plays a role in stimulating growth and proliferation of leukemic cells, and an increased endogenous level of VeGF is also associated with a poor prognosis.128 Receptor tyrosine kinase inhibitors of VeGF are another strategy under active study.129 Preliminary data suggest that SU5416, a small molecule inhibitor of phosphorylation of VeGF receptors 1 and 2, C-KIT, the stem cell factor (SCF) receptor and FLT3, has activity in AML.130 Bevacizumab, an anti-VeGF antibody, has been demonstrated to be safe in AML and is currently undergoing phase-2 studies.131

    FLT3 inhibitors

    Mutations that constitutively activate the FLT3 receptor tyrosine kinase occur in approximately 30% of AML and confer a poor prognosis.17 These observations have fueled the development of FLT3-selective–targeted tyrosine kinase inhibitors with in vitro cytotoxicity to leukemia cells.132-134 There are 4 FLT3 inhibitors that are currently in clinical trials, including PKC-412 (Novartis, Basel, Switzerland), CeP-701 (Cephalon, Frazer, PA), MLN518 (Millennium, San Francisco, CA), and SU11248 (SuGen, New York, NY).133 Although data are still preliminary, several generalizations have emerged. The compounds, which like imatinib mesylate are selective but not completely specific, are well tolerated at doses that achieve inhibition of the target FLT3. each of the inhibitors have activity in patients with relapsed AML with activating mutations, although responses have been quite modest and characterized by transient reduction in peripheral blood blasts, less frequent reduction in bone marrow blasts, and rare hematologic responses. In some cases, patients that lack ITD or activation loop mutations have responded to FLT3 inhibitors, although mutations that activate FLT3 outside of these regions have been identified and may explain response in this subset. These data are perhaps not surprising, in that it would not be expected that a kinase inhibitor as a single agent in this setting would be any more effective than imatinib mesylate in patients experiencing chronic myeloid leukemia (CML) blast crisis. Current clinical trials are focusing on use of FLT3 inhibitors in combination with chemotherapy.

    Activating alleles of KIT occur in approximately 5% of patients with AML, but in contrast with activating juxtamembrane deletions observed in gastrointestinal stromal-cell tumors that are imatinib mesylate sensitive, the most common KIT alleles in AML include KIT D816V or KIT D816Y that are resistant to imatinib mesylate. It has recently been reported that PKC-412, however, is a potent inhibitor of these mutant KIT proteins16 and may have clinical therapeutic value for these patients with AML, as well as in systemic mast-cell disease associated with KIT D816V or KIT D816V.

    Apoptosis inhibitors

    Overexpression of the apoptosis inhibitor protein bcl-2 can render tumor cells resistant to induction of apoptosis by drug therapy. A high level of expression of BCL-2 in AML is associated with a poor prognosis.135 Down-regulation of BCL-2 by antisense oligonucleotides in vitro sensitizes leukemic cells to chemotherapy in AML cell lines.136 A phase-1 trial of bcl-2 antisense oligonucleotide (GNS, oblimersen sodium) showed a response in 8 of 20 patients with relapsed or refractory AML.137 In a subsequent phase-1 trial in untreated older adults, to determine feasibility, the bcl-2 antisense oligonucleotide was administered with chemotherapy.138 Ten (45%) of 22 patients achieved CR without unexpected toxicities. A randomized phase-3 study by the CALGB is now evaluating the role of the BCL-2 antisense oligonucleotide both in induction and in consolidation.

    Summary and future directions

    Consistent incremental progress has occurred in younger adults with AML because increasingly intensive chemotherapy can be administered to patients whose cells are relatively sensitive. Older adults usually cannot tolerate such intensive chemotherapy, and their leukemic cells are inherently more resistant. Many new antileukemic agents with novel mechanisms of action and direction are available to explore.

    Several of the current new agents being investigated are likely to have a role in the future therapy for AML. However, diverse drug-resistance mechanisms are a prominent feature of AML, and none alone appear likely to alter the standard of care and have an effect comparable to that of ATRA in APL. Combinations of several agents, targeting more than one gene mutation or antigenic determinant, may hold greater promise. The future also rests with new techniques such as cDNA microarray.102-105,139 and genome-wide approaches to gene discovery, in combination with high-throughput screens for modulators of validated targets. These technologies will likely provide important insights into the molecular pathogenesis of AML and identify critical genes that may be targeted.

    Acknowledgements

    We thank Dr Charles Schiffer for his review of the manuscript and Ms Kristen Burton for her assistance with manuscript preparation.

    Footnotes

    Footnotes

    Prepublished online as Blood First edition Paper, May 3, 2005; DOI 10.1182/blood-2005-01-0178.

    References

    McCulloch e. Stem cells in normal and leukemic hemopoiesis. Blood. 1983;62: 1-13.

    U.S. Government Statistics. Surveillance, epidemiology, and end-results (SeeR) of the National Cancer Institute; 2002.

    Rowe J, Andersen J, Cassileth P, et al. Clinical trials of adults with acute myelogenous leukemia: experience of the eastern Cooperative Oncology Group. In: Acute leukemia IV: experimental approaches and novel therapies. Berlin, Germany: Springer-Verlag; 1994: 541-546.

    Bennett J, Young M, Andersen J, et al. Long-term survival in acute myeloid leukemia: the eastern Cooperative Oncology Group experience. Cancer. 1997;80: 2205-2209.

    Appelbaum F, Rowe J, Radich J, Dick J. Acute myeloid leukemia. Hematology (Am Soc Hematol educ Program). 2001: 62-86.

    Grimwade D, Walker H, Oliver F, et al. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children's Leukaemia Working Parties. Blood. 1998;92: 2322-2333.

    Byrd J, Mrozek K, Dodge R, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood. 2002; 100: 4325-4336.

    Slovak M, Kopecky K, Cassileth P. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/eastern Cooperative Oncology Group study. Blood. 2000;96: 4075-4083.

    Leith C, Kopecky K, Godwin J, et al. Acute myeloid leukemia in the elderly: assessment of multidrug resistance (MDR1) and cytogenetics distinguishes biologic subgroups with remarkably distinct responses to standard chemotherapy. Blood. 1997;89: 3323-3329.

    Schmid D, Heinze G, Linnerth B, et al. Prognostic significance of WT1 gene expression at diagnosis in adult de novo acute myeloid leukemia. Leukemia. 1997;11: 639-643.

    King-Underwood L, Pritchard-Jones K. Wilms' tumor (WTI) gene mutations occur mainly in acute myeloid leukemia and may confer drug resistance. Blood. 1998;91: 2961-2968.

    Frohling S, Schlenk R, Stolze I, et al. CeBPA mutations in younger patients with acute myeloid leukemia and normal cytogenetics: prognostic relevance and analysis of cooperating mutations. J Clin Oncol. 2004;22: 624-633.

    Kohler T, Schill C, Deininger M, et al. High Bad and Bax mRNA expression correlate with negative outcome in acute myeloid leukemia (AML). Leukemia. 2002;16: 22-29.

    Baldus C, Tanner S, Ruppert A, et al. BAALC expression predicts clinical outcome of de novo acute myeloid leukemia patients with normal cytogenetics: a Cancer and Leukemia Group B study. Blood. 2003;102: 1613-1618.

    Barjesteh S, Van Doom-Khosrovani, erpelinick C, Van Putten W, et al. High eVI1 expression predicts poor survival in acute myeloid leukemia: a study of 319 de novo AML patients. Blood. 2003; 101: 837-845.

    Growney J, Clark J, Adelsperger J, Stone R, et al. Activation mutations of human c-KIT resistant to imatinib are sensitive to the tyrosine kinase inhibitor PKC412. Blood. Prepublished on March 24, 2005, as DOI 10.1182/blood-2004-12-4617.

    Kottaridis P, Gale R, Frew M, et al. The presence of FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98: 1752-1759.

    Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood. 2002; 100: 59-66.

    Dillman R, Davis R, Green M, et al. A comparative study of two different doses of cytarabine for acute myeloid leukemia: A phase III study of Cancer and Leukemia Group B. Blood. 1991;78: 2520-2526.

    Yates J, Glidewell O, Wiernik P, et al. Cytosine arabinoside with daunorubicin or adriamycin for therapy of acute myelocytic leukemia: a CALGB study. Blood. 1982;60: 454-462.

    Rowe J, Tallman M. Intensifying induction therapy in acute myeloid leukemia: has a new standard of care emerged? Blood. 1997;90: 2121-2126.

    Berman e, Heller G, Santorsa J, et al. Results of a randomized trial comparing idarubicin and cytosine arabinoside with daunorubicin and cytosine arabinoside in adult patients with newly diagnosed acute myelogenous leukemia. Blood. 1991;77: 1666-1674.

    Wiernik P, Banks P, Case D Jr, et al. Cytarabine plus idarubicin or daunorubicin as induction and consolidation therapy for previously untreated adult patients with acute myeloid leukemia. Blood. 1992;79: 313-319.

    Vogler W, Velez-Garcia e, Weiner R, et al. A phase III trial comparing idarubicin and daunorubicin in combination with cytarabine in acute myelogenous leukemia: a Southeastern Cancer Study Group study. J Clin Oncol. 1992;10: 1103-1111.

    Mandelli F, Petti M, Ardia A, et al. A randomised clinical trial comparing idarubicin and cytarabine to daunorubicin and cytarabine in the treatment of acute non-lymphoid leukaemia. eur J Cancer. 1991;27: 750-755.

    Arlin Z, Case D, Moore J, et al. Randomized multicenter trial of cytosine arabinoside with mitoxantrone or daunorubicin in previously untreated adult patients with acute nonlymphocytic leukemia (ANLL). Leukemia. 1990;4: 177-183.

    Hansen O, Pedersen-Bjergaard J, ellegaard J, et al. Aclarubicin plus cytosine arabinoside versus daunorubicin plus cytosine arabinoside in previously untreated patients with acute myeloid leukemia: a Danish national phase III trial. The Danish Society of Hematology Study Group on AML, Denmark. Leukemia. 1991;5: 510-516.

    Buchner T, Hiddemann W, Schoch C, Haferlach T, Sauerland M-C, Heinecke A. Acute myeloid leukaemia (AML): treatment of the older patient. Best Pract Res Clin Haematol. 2001;14: 139-151.

    Rowe J, Neuberg D, Friedenberg W, et al. A phase 3 study of three induction regimens and of priming with GM-CSF in older adults with acute myeloid leukemia: a trial by the eastern Cooperative Oncology Group. Blood. 2004;103: 479.

    Lowenberg B, Suciu S, Archimbaud e, et al. Mitoxantrone versus daunorubicin in induction-consolidation chemotherapy: the value of low-dose cytarabine for maintenance of remission, and an assessment of prognostic factors in acute myeloid leukemia in the elderly: final report european Organization for the Research and Treatment of Cancer and the Dutch-Belgian Hemato-Oncology Cooperative HOVON Group. J Clin Oncol. 1998;16: 872-881.

    Goldstone AH, Burnett AK, Wheatley K, Smith AG, Hutchinson RM, Clark Re; Medical Research Council Adult Leukemia Working Party. Attempts to improve treatment outcomes in acute myeloid leukemia (AML) older patients: the results of the United Kingdom Medical Research Council AML11 trial. Blood. 2001;98: 1302-1311.

    Anderson J, Kopecky K, Willman C, et al. Outcome after induction chemotherapy for older patients with acute myeloid leukemia is not improved with mitoxantrone and etoposide compared to cytarabine and daunorubicin: a Southwest Oncology Group study. Blood. 2002; 100: 3869-3876.

    Berman e, Arlin Z, Gaynor J, et al. Comparative trial of cytarabine and thioguanine in combination with amsacrine or daunorubicin in patients with untreated acute nonlymphocytic leukemia: results of the L-16M protocol. Leukemia. 1989;3: 115-121.

    Plunkett W, Liliemark J, Adams T, et al. Saturation of l-beta-D-arabinofuranosylcytosine S'-triphosphate accumulation in leukemia cells during high-dose l-beta-D-arbinofuranosylcytosine therapy. Cancer Res. 1987;47: 3005-3011.

    Bishop J, Matthews J, Young G, et al. A randomized trial of high-dose cytarabine in induction in acute myeloid leukemia. Blood. 1996;87: 1710-1717.

    Weick J, Kopecky K, Appelbaum F, et al. A randomized investigation of high-dose versus standard-dose cytosine arabinoside with daunorubicin in patients with previously untreated acute myeloid leukemia: a Southwest Oncology Group Study. Blood. 1996;88: 2841-2851.

    Wolff S, Herzig R, Fay J, et al. High-dose cytarabine and daunorubicin as consolidation therapy for acute myeloid leukemia in first remission: long-term follow-up and results. J Clin Oncol. 1989;7: 1260-1267.

    Bishop J, Lowenthal R, Joshua D, et al. Australian Leukemia Study Group: etoposide in acute nonlymphocytic leukemia. Blood. 1990;75: 27-32.

    Hann I, Stevens R, Goldstone A, et al. Randomized comparison of DAT versus ADe as induction chemotherapy in children and younger adults with acute myeloid leukemia. Results of the Medical Research Council's 10th AML trial (MRC AML10). Blood. 1997;89: 2311-2318.

    estey e, Thall P, Cortes J, et al. Comparison of idarubicin + ara-C-, fludarabine + ara-C-, and topotecan + ara-C-based regimens in the treatment of newly diagnosed acute myeloid leukemia, refractory anemia with excess blasts in transformation, or refractory anemia with excess blasts. Blood. 2001;98: 3575-3583.

    Mitus A, Miller K, Schenkein D, et al. Improved survival for patients with acute myelogenous leukemia. J Clin Oncol. 1995;13: 560-569.

    Cassileth P, Lee S, Miller K, et al. Feasibility study of adding high-dose cytarabine (HDIC) in induction (IND) and in consolidation (CONS) therapy before autologous stem cell transplant (ASCT) in adult acute myeloid leukemia (AML) . Blood. 1998;92: 362b.

    Petersdorf S, Rankin C, Terebolu H, Head D, Appelbaum F. A phase II study of standard dose daunomycin and cytosine arabinoside (Ara-C) with high-dose Ara-C induction therapy followed by sequential high-dose Ara-C consolidation for adults with previously untreated acute myelogenous leukemia: A Southwest Oncology Group study (SWOG 9500) . Proc Am Soc Clin Oncol. 1998;17: 55.

    Buchner T, Hiddemann W, Wormann B, et al. Double induction strategy for acute myeloid leukemia: the effect of high-dose cytarabine with mitoxantrone instead of standard-dose cytarabine with daunorubicin and 6-thioguanine: a randomized trial by the German AML Cooperative Group. Blood. 1999;93: 4116-4124.

    Berman e, McBride M. Comparative cellular pharmacology of daunorubicin and idarubicin in human multidrug-resistant leukemia cells. Blood. 1992;79: 3267-3273.

    Rowe JM. Treatment of acute myeloid leukemia with cytokines: effect on duration of neutropenia and response to infections. Clin Infect Dis. 1998; 26: 1290-1294.

    Amadori S, Suciu S, Jehn U, et al. Use of glycosylated recombinant human G-CSF (lenograstim) during and/or after induction chemotherapy in patients 61 years of age and older with acute myeloid leukemia: final results of AML13, a randomized phase 3 study of the european Organization for Research and Treatment of Cancer and Grippo Italiano Malattie ematologiche dell Advito (eORTG/GIMeMA) Leukemia groups. Blood. Prepublished on Mary 10, 2005, as DOI 10.1182/blood-2004-09-3728.

    Lowenberg B, van Putten W, Theobald M, et al. effect of priming with granulocyte colony-stimulating factor on the outcome of chemotherapy for acute myeloid leukemia. N engl J Med. 2003;349: 727-729.

    Cassileth P, Lynch e, Hines J, et al. Varying intensity of post-remission therapy in acute myeloid leukemia. Blood. 1992;79: 1924-1930.

    Mayer R, Davis R, Schiffer C, et al. Intensive postremission chemotherapy in adults with acute myeloid leukemia. N engl J Med. 1994;331: 896-903.

    Brenner M, Rill D, Moen R, et al. Gene-marking to trace origin of relapse after autologous bone-marrow transplantation. Lancet. 1993;841: 85-86.

    McSweeney P, Niederwieser D, Shizuru J, et al. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood. 2001;97: 3390-3400.

    Diaconescu R, Flowers C, Storer B, et al. Morbidity and mortality with nonmyeloablative compared to myeloablative conditioning before hematopoietic cell transplantation for HLA matched related donors. Blood. 2004;104: 1550-1558.

    Maris M, Niederwieser D, Sandmaier B, et al. HLA-matched unrelated donor hematopoietic cell transplantation after nonmyeloablative conditioning for patients with hematologic malignancies. Blood. 2003;102: 2021-2030.

    Burnett A, Goldstone A, Stevens R, et al. Randomised comparison of addition of autologous bone-marrow transplantation to intensive chemotherapy for acute myeloid leukaemia in first remission: results of MRC AML 10 trial. UK Medical Research Council Adult and Children's Leukaemia Working Parties. Lancet. 1998;351: 700-708.

    Zittoun R, Mandelli F, Willemze R, et al. Autologous or allogeneic bone marrow transplantation compared to intensive chemotherapy in acute myeloid leukemia. N engl J Med. 1995;332: 217-223.

    Cassileth P, Harrington D, Appelbaum F, et al. Chemotherapy compared with autologous or allogeneic bone marrow transplantation in the management of acute myeloid leukemia in first remission. N engl J Med. 1998;339: 1649-1656.

    Harousseau J-L, Cahn J, Pignon B, et al. Comparison of autologous bone marrow transplantation and intensive therapy as postremission therapy in adult acute myeloid leukemia. Blood. 1997;90: 2978-2986.

    Buchner T, Hiddemann W, Berdei W, et al. 6-thioguanine, cytarabine, and daunorubicin (TAD) and high-dose cytarabine and mitoxantrone (HAM) for induction, TAD for consolidation, and either prolonged maintenance by reduced monthly TAD or TAD-HAM-TAD and one course of intensive consolidation by sequential HAM in adult patients at all ages with de novo acute myeloid leukemia (AML). A randomized trial of the German AML Cooperative Group. J Clin Oncol. 2003;21: 4496-4504.

    Hewlett J, Kopecky K, Head D, et al. A prospective evaluation of the roles of allogeneic marrow transplantation and low-dose monthly maintenance chemotherapy in the treatment of adult acute myelogenous leukemia (AML). A Southwest Oncology Group Study. Leukemia. 1995;9: 562-569.

    Grignani F, Valtieri M, Gabbianelli M, et al. PML/RAR fusion protein expression in normal human hematopoietic progenitors dictates myeloid commitment and the promyelocytic phenotype. Blood. 2000;96: 1531-1537.

    Mandelli F, Diverio D, Avvisati G, et al. Molecular remission in PML/RAR-positive acute promyelocytic leukemia by combined all-trans retinoic acid and idarubicin (AIDA) therapy. Gruppo Italiano-Malattie ematologiche Maligne dell'Adulto and Associazione Italiana di ematologia ed Oncologia Pediatrica Cooperative Groups. Blood. 1997;90: 1014-1021.

    Sanz M, Martin G, Rayon C, et al. A modified AIDA protocol with anthracycline-based consolidation results in high antileukemic efficacy and reduced toxicity in newly diagnosed PML/RARa-positive acute promyelocytic leukemia. Blood. 1999;94: 3015-3021.

    Fenaux P, Le Deley M, Castainge S, et al. effect of all trans-retinoic acid in newly diagnosed acute promyelocytic leukemia: results of a multicenter randomized trial. european APL91 Group. Blood. 1993;82: 3241-3249.

    Tallman M, Andersen J, Schiffer C, et al. All-trans retinoic acid in acute promyelocytic leukemia. N engl J Med. 1997;337: 1021-1028.

    Fenaux P, Chastang C, Chevret S, et al. A randomized comparison of all-trans retinoic acid (ATRA) followed by chemotherapy and ATRA plus chemotherapy and the role of maintenance therapy in newly diagnosed acute promyelocytic leukemia. Blood. 1999;94: 1192-1200.

    Tallman M, Andersen J, Schiffer C, et al. All-trans retinoic acid in acute promyelocytic leukemia: long-term outcome and prognostic factor analysis from the North American Intergroup Protocol. Blood. 2002;100: 4298-4302.

    Sanz M, LoCoco F, Martin G, et al. Definition of relapse risk and role of nonanthracycline drugs for consolidation in patients with acute promyelocytic leukemia: a joint study of the PeTHeMA and GIMeMA cooperative groups. Blood. 2000;96: 1247-1253.

    Sanz M. Risk-adapted treatment of acute promyelocytic leukemia with all-trans retinoic acid and anthracycline monochemotherapy: a multicenter study by the PeTHeMA group. Blood. 2004;103: 1237-1243.

    Baudard M, Beauchamp-Nicoud A, Delmer A, et al. Has the prognosis of adult patients with acute myeloid leukemia improved over the years? A single institution experience of 784 consecutive patients over a 16-year period. Leukemia. 1999; 13: 1481-1490.

    Kahn S, Begg C, Mazza J, Bennett J, Bonner H, Glick J. Full dose versus attenuated dose daunorubicin, cytosine arabinoside, and 6-thioguanine in the treatment of acute nonlymphocytic leukemia in the elderly. J Clin Oncol. 1984;2: 865-870.

    Tilly H, Castaigne S, Bordessoule D, et al. Low-dose cytosine arabinoside treatment for acute nonlymphocytic leukemia in elderly patients. Cancer. 1985;55: 1633-1636.

    Lowenberg B, Zittoun R, Kerkhofs H, et al. On the value of intensive remission-induction chemotherapy in elderly patients of 65+ years with acute myeloid leukemia: a randomized phase III study of the european Organization for Research and Treatment of Cancer Leukemia Group. J Clin Oncol. 1989;7: 1268-1274.

    Kern W, Schoch C, Haferlach T, et al. Multivariate analysis of prognostic factors in patients with refractory and relapsed acute myeloid leukemia undergoing sequential high-dose cytosine arabinoside and mitoxantrone (S-HAM) salvage therapy: relevance of cytogenetic abnormalities. Leukemia. 2000;14: 226-231.

    Karanes C, Kopecky K, Head D, et al. A phase III comparison of high dose ARA-C (HIDAC) versus HIDAC plus mitoxantrone in the treatment of first relapsed or refractory acute myeloid leukemia. Southwest Oncology Group Study. Leuk Res. 1999;23: 787-794.

    Biggs J, Horowitz M, Gale R, et al. Bone marrow transplants may cure patients with acute leukemia near achieving remission with chemotherapy. Blood. 1992;80: 1090-1093.

    Byrne J, Dasgupta e, Pallis M, et al. early allogeneic transplantation for refractory or relapsed acute leukemia following remission induction with FLAG. Leukemia. 1991;13: 786-791.

    Fung H, Stein A, Slovak M, et al. A long-term follow-up report on allogeneic stem cell transplantation for patients with primary refractory acute myelogenous leukemia: impact of cytogenetic characteristics on transplantation outcome. Biol Blood Marrow Transplant. 2005;11: 108-114.

    Wong R, Shahjahah M, Wang X, et al. Prognostic factors for outcomes of patients with refractory or relapsed acute myelogenous leukemia or myelodysplastic syndromes undergoing allogeneic progenitor cell transplantation. Biol Blood Marrow Transplant. 2005;11: 108-114.

    Sierra J, Storer B, Hansen J, et al. Transplantation of marrow cells from unrelated donors for treatment of high-risk acute leukemia: the effect of leukemic burden, donor HLA-matching and marrow cell dose. Blood. 1997;89: 4226-4235.

    Aversa F, Tabilio A, Velardi A, et al. Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype. N engl J Med. 1998; 339: 1186-1193.

    Aversa F, Velardi A, Tabilio A, Reisner Y, Martelli M. Haploidentical stem cell transplantation in leukemia. Blood Rev. 2001;15: 111-119.

    Yeager A, Kaizer H, Santos G, et al. Autologous bone marrow transplantation in patients with acute nonlymphocytic leukemia, using ex vivo marrow treatment with 4-hydroperoxy cyclophosphamide. N engl J Med. 1986;315: 141-147.

    Gilliland DG, Tallman MS. Focus on acute leukemias. Cancer Cell. 2002;1: 417-420.

    Gilliland DG. Molecular genetics of human leukemias: new insights into therapy. Semin Hematol. 2002;39: 6-11.

    Mohi MG, Boulton C, Gu TL, et al. Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci U S A. 2004; 101: 3130-3135.

    Gelmetti V, Zhang J, Fanelli M, Minucci S, Pelicci PG, Lazar MA. Aberrant recruitment of the nuclear receptor corepressor-histone deacetylase complex by the acute myeloid leukemia partner eTO. Mol Cell Biol. 1998;18: 7185-7191.

    Amann JM, Nip J, Strom DK, et al. eTO, a target of t(8;21) in acute leukemia, makes distinct contacts with multiple histone deacetylases and binds mSin3A through its oligomerization domain. Mol Cell Biol. 2001;21: 6470-6483.

    Yang G, Khalaf W, van de Locht L, et al. epigenetic regulation of tumor suppressors in t(8:21)-containing AML. Ann Hematol. 2004;83(Suppl 1): S83.

    Hiebert SW, Reed-Inderbitzin eF, Amann J, Irvin B, Durst K, Linggi B. The t(8;21) fusion protein contacts co-repressors and histone deacetylases to repress the transcription of the p14ARF tumor suppressor. Blood Cells Mol Dis. 2003;30: 177-183.

    Buonamici S, Chakraborty S, Senyuk V, Nucifora G. The role of eVI1 in normal and leukemic cells. Blood Cells Mol Dis. 2003;31: 206-212.

    Mitani K. Molecular mechanisms of leukemogenesis by AML1/eVI1. Oncogene. 2004;23: 4263-4269.

    Buonamici S, Li D, Chi Y, et al. eVI1 induces myelodysplastic syndrome in mice. J Clin Invest. 2004;114: 713-719.

    Slape C, Aplan PD. The role of NUP98 gene fusions in hematologic malignancy. Leuk Lymphoma. 2004;45: 1341-1350.

    Falini B, Mecucci C, Tiacci e, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N engl J Med. 2005; 352: 254-266.

    Bonnet D, Dick Je. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3: 730-737.

    Reya T, Duncan AW, Ailles L, et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature. 2003;423: 409-414.

    Ohishi K, Katayama N, Shiku H, Varnum-Finney B, Bernstein ID. Notch signalling in hematopoiesis. Semin Cell Dev Biol. 2003;14: 143-150.

    Lessard J, Sauvageau G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature. 2003;423: 255-260.

    Kyba M, Perlingeiro RC, Daley GQ. HoxB4 confers definitive lymphoid-myeloid engraftment potential on embryonic stem cell and yolk sac hematopoietic progenitors. Cell. 2002;109: 29-37.

    Antonchuk J, Sauvageau G, Humphries RK. HOXB4-induced expansion of adult hematopoietic stem cells ex vivo. Cell. 2002;109: 39-45.

    Bullinger L, Dohner K, Frohling S, et al. Use of gene-expression profiling to identify prognostic subclasses in adult acute myeloid leukemia. N engl J Med. 2004;350: 1605-1616.

    Valk P, Verhaak R, Beijen M, et al. Prognostically useful gene-expression profiles in acute myeloid leukemia. N engl J Med. 2004;350: 1612-1628.

    Grimwade D, Haferlach T. Gene expression profiling in acute myeloid leukemia. N engl J Med. 2004;350: 1676-1678.

    Ley T, Miux P, Walter M, et al. A pilot study of high-throughput, sequence-based mutational profiling of primary human acute myeloid leukemia cell genomes. Proc Natl Acad Sci U S A. 2003; 100: 14275-14280.

    van der Velden V, te Marvelde J, Hoogeveen P, et al. Targeting of the CD33-calicheamicin immunoconjugate Mylotarg (CMA-676) in acute myeloid leukemia: in vivo and in vitro saturation and internalization by leukemic and normal myeloid cells. Blood. 2001;97: 3197-3204.

    Sievers e, Larson R, Stadtmauer e, et al. efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. J Clin Oncol. 2001;19: 3244-3254.

    Larson R, Sievers e, Stadtmauer e, et al. Use of gemtuzumab ozogamicin (Mylotarg) in 157 patients > 60 years of age with acute myeloid leukemia in first relapse . Blood. 2003;102: 176a.

    Kell W, Burnett A, Chopra R, et al. A feasibility study of simultaneous administration of gemtuzumab ozogamicin with intensive chemotherapy in induction and consolidation in younger patients with acute myeloid leukemia. Blood. 2003;102: 4277-4283.

    De Angelo D, Stone R, Durant S, et al. Gemtuzumab ozogamicin (Mylotarg) in combination with induction chemotherapy for the treatment of patients with de novo acute myeloid leukemia: two age-specific phase 2 trials . Blood. 2003;102: 100a.

    estey e, Thall P, Giles F, et al. Gemtuzumab ozogamicin with or without interleukin II in patients 65 years of ago or older with untreated acute myeloid leukemia and high-risk myelodysplastic syndrome: comparison with idarubicin plus continuous-infusion, high-dose cytosine arabinoside. Blood. 2002;99: 4343-4349.

    Nabhan C, Rundhaugen L, Riley M, et al. Phase II pilot trial of gemtuzumab ozogamicin (GO) as first line therapy in acute myeloid leukemia patients age 65 or older. Leuk Res. 2004;28: 909-919.

    Naito K, Takeshita A, Shigeno K, et al. Calicheamicin-conjugated humanized anti-CD33 monoclonal antibody (gemtuzumab ozogamicin, CMA-676) shows cytocidal effect on CD33-positive leukemia cell lines, but is inactive on P-glycoprotein-expressing sublines. Leukemia. 2000;14: 1436-1443.

    List A, Kopecky K, William C, et al. Benefit of cyclosporine modulation of drug resistance in patients with poor-risk acute myeloid leukemia: a Southwest Oncology Group study. Blood. 2001; 98: 3212-3230.

    Greenberg P, Lee S, Advani J, et al. Randomized trial comparing mitoxantrone, etoposide and cytarabine with or without PSC-833 (Valspodar) in patients with relapsed or refractory acute myeloid leukemia and high-risk myelodysplastic syndrome: eastern Cooperative Oncology Group protocol e2995. J Clin Oncol. 2004;22: 1078-1086.

    Lui Y, Wheatley K, Rees J, Burnett A. Comparison of `sequential' versus `standard' chemotherapy as re-induction treatment, with or without cyclosporine in refractory/relapsed acute myeloid leukemia (AML): results of the UK Medical Research council AML-R trial. Br J Haematol. 2001;113: 713-726.

    Rubin e, de Alwis D, Docliquen I, et al. P-glycoprotein inhibitor, Zosuquidar, 3H4 trihydrochloride (LY335979), administered orally in combination with doxorubicin in patients with advanced malignancies. Clin Cancer Res. 2002;8: 3710-3717.

    Reuter CW, Morgan M, Bergmann L. Targeting the RAS signaling pathway: a rational, mechanism-based treatment in hematological malignancies. Blood. 2000;96: 1655-1669.

    Lancet J, Karp J. Farnesyltransferase inhibitors in hematologic malignancies: new horizons in therapy. Blood. 2003;102: 3880-3889.

    Karp J, Lancet J, Kaufmann S, 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: 3361-3369.

    Lancet J, Gojo I, Gotlib J, et al. Tipifarnib (ZARNeSTRA) in previously untreated poor-risk AML and MDS: interim results of a phase 2 trial . Blood. 2003;102: 176a.

    Strahl B, Allis C. The language of covalent histone modifications. Nature. 2000;403: 41.

    Kosugi H, Towatari M, Hatano S, et al. Histone deacetylase inhibitors are the potent inducer/enhancer of differentiation in acute myeloid leukemia: a new approach to anti-leukemia therapy. Leukemia. 1999;13: 1316-1324.

    Gottlicher M, Minucci S, Zhu P, et al. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. eMBO J. 2001;20: 6969-6978.

    Yu C, Rahmani M, Courad D, Subler M, Dent P, Grant S. The proteosome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood. 2003; 102: 3765-3774.

    Yu C, Rahmani M, Dent P, Grant S. The hierarchical relationship between MAPK signaling and ROS generation in human leukemia cells undergoing apoptosis in response to the proteasome inhibitor Bortezomib. exp Cell Res. 2004;295: 555-566.

    de Bont e, Rosati S, Jacobs S, Kamps U, Vellenga e. Increased bone marrow vascularization in patients with acute myeloid leukemia, a possible role for vascular endothelial growth factor. Br J Haematol. 2001;113: 296-304.

    Schuch G, Machluf M, Bartsch G, et al. In vivo administration of vascular endothelial growth factor (VeGF) and its antagonist, soluble necrophilin-1, predicts a role of VeGF in the progression of acute myeloid leukemia in vivo. Blood. 2002; 100: 4622-4628.

    Giles F, Stopeck A, Silverman L, et al. SU5416, a small molecule tyrosine kinase receptor inhibitor, has biologic activity in patients with refractory acute myeloid leukemia and myelodysplastic syndromes. Blood. 2003;102: 795-801.

    Fiedler W, Mesters R, Tinnefeld H, et al. A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia. Blood. 2003;102: 2763-2767.

    Karp J, Gojo I, Gocke D, et al. Timed-sequential therapy (TST) of relapsed and refractory adult acute myelogenous leukemia (AML) with the anti-vascular endothelial growth factor (VeGF) monoclonal antibody bevacizumab . Blood. 2002;100: 744a.

    Levis M, Allebach J, Tse K-F, et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood. 2002;99: 3885-3891.

    Stirewalt DL, Radich JP. The role of FLT3 in haematopoietic malignancies. Nat Rev Cancer. 2003;3: 650-665.

    Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 2002;100: 1532-1542.

    Del Poeta G, Venditti A, Principe M, et al. Amount of spontaneous apoptosis detected by Bax1 Bcl-2 ratio predicts outcome in acute myeloid leukemia. Blood. 2003;101: 2125-2131.

    Konopleva M, Tari A, estrov Z, et al. Liposomal Bcl-2 antisense oligonucleotides enhance proliferation, sensitize acute myeloid leukemia to cytosine-arabinoside, and induce apoptosis independent of other antiapoptotic proteins. Blood. 2000; 95: 3929-3938.

    Marcucci G, Byrd J, Dai G, et al. Phase 1 and pharmacokinetic studies of G3139, a Bcl-2 anti-sense oligonucleotide, in combination with chemotherapy in refractory or relapsed acute leukemia. Blood. 2003;101: 425-432.

    Marcucci G, Stock W, Zwiebel J, et al. Clinical activity of Genasense (GNS, oblimersen sodium), in combination with daunorubicin and cytarabine: a phase 1 study in previously untreated elderly acute myeloid leukemia (abstr). Blood. 2003: 385a.

    Okutsu J, Tsunoda T, Kaneta Y, et al. Prediction of chemosensitivity for patients with acute myeloid leukemia, according to expression levels of 28 genes selected by genome-wide complementary DNA microarray analysis. Mol Cancer Ther. 2002; 1: 1035-1042.(Martin S. Tallman, D. Gar)