aml in the elderly, a review

Upload: modupeh-betts

Post on 07-Apr-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/6/2019 AML in the Elderly, A Review

    1/19

    Acute myeloid leukaemia in the elderly: a review

    Daniel A. Pollyea,1,2 Holbrook E. Kohrt1,2 and Bruno C. Medeiros1

    1

    Divisions of Hematology, and2

    Oncology, Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA, USA

    Summary

    The majority of patients with acute myeloid leukaemia (AML)

    are elderly. Advancements in supportive care and regimen

    intensification have resulted in improvements in clinical

    outcomes for younger AML patients, but analogous improve-

    ments in older patients have not been realized. While

    outcomes are compromised by increased comorbidities and

    susceptibility to toxicity from therapy, it is now recognized

    that elderly AML represents a biologically distinct disease that

    is more aggressive and less responsive to therapy. Some

    patients tolerate and benefit from intensive remission-induc-

    tion approaches, while others are best managed with less

    aggressive strategies. The challenge is to differentiate these

    groups based on host-related and biological features, in order

    to maximize the therapeutic benefit and minimize toxicity. As

    more is understood about the complicated pathogenesis and

    molecular basis of AML, there are more opportunities to

    develop and test targeted therapies. Elderly patients, with their

    narrow therapeutic window, are well positioned to derive a

    benefit from these novel agents, and therefore, despite a

    difficult past, there are reasons to be optimistic about thefuture of elderly AML.

    Keywords: elderly, acute myeloid leukaemia, gene expression

    profiling, stem cell transplantation, prognosis.

    Acute myeloid leukaemia (AML) is a clonal disorder charac-

    terized by arrest of differentiation in the myeloid lineage

    coupled with an accumulation of immature progenitors in the

    bone marrow, resulting in hematopoietic failure. AML is the

    most common acute leukaemia in adults, affecting roughly

    three out of 100 000 people in the UK (Cancer Research UK).

    While the prognosis for younger patients with AML has

    improved in recent decades, the same cannot be said for older

    patients, who continue to have a median overall survival (OS)

    on the order of months with few long-term survivors

    (Dombret et al, 2008). AML patients are predominantly

    elderly, (for the purposes of this review, elderly is defined as

    age 60 and older) with a median age at diagnosis of 67

    (National Cancer Institute 19752007). With improvements in

    life expectancy, by 2031 a 38% increase in elderly cases is

    projected (Pinto et al, 2001).

    Older age is an independent adverse prognosticator, asso-

    ciated with a decreased complete response (CR) rate, disease

    free survival (DFS), relapse free survival (RFS) and OS, with

    higher rates of treatment related mortality (TRM), resistant

    disease and relapse compared to equivalently treated younger

    patients (Harousseau, 1998; Milligan et al, 2006; Dohner et al,

    2010). Explanations for these poor outcomes are not intuitive.

    To review this topic, we conducted a literature search of

    publications from 1960 to the present with PubMed and

    Google Scholar using combinations of the following search

    terms: acute myeloid/myelogenous/myeloblastic leukaemia,

    elderly, prognosis, treatment, survival, remission, toxicity,

    transplantation, induction and consolidation, and reviewed

    relevant works identified in the references sections of these

    publications. We analyzed data from clinical trials that were

    designed exclusively for the elderly, and where possible,

    examined the elderly cohorts of all-inclusive AML trials. Herewe discuss the use of prognostic factors and predictive

    modelling to risk stratify elderly AML patients, and how this

    information can be applied to manage patients. We also review

    treatment options for elderly patients, with an emphasis on

    novel therapies.

    Prognostic factors

    Although age is a risk factor for TRM, disease resistance and

    OS, it is not the most important risk factor (Dohner et al,

    2010). While predicting outcomes for a population that

    universally has poor OS is fraught with difficulty, the

    importance of both age-dependent and independent variables

    have been noted, and are discussed here.

    Biological factors versus host factors

    Elderly patients suffer increased toxicity with therapy

    (Coebergh et al, 1998), as pharmacokinetic and pharmacody-

    namic changes that occur with age inhibit drug clearance and

    result in prolonged exposure to chemotherapeutics (Cohen,

    1986). Additionally, a worse performance status (PS), seen

    Correspondence: Daniel A. Pollyea, MD, Department of Internal

    Medicine, Stanford University Cancer Center, 875 Blake Wilbur Dr,

    Stanford, CA 94305 5820, USA. E-mail: [email protected]

    review

    doi:10.1111/j.1365-2141.2010.08470.x 2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542

  • 8/6/2019 AML in the Elderly, A Review

    2/19

    more commonly in the elderly, predicts poor outcomes

    (Appelbaum et al, 2006). Elderly patients are more prone to

    bleeding complications, and are less able than younger patients

    to tolerate infections (Estey et al, 1982; Harousseau, 1998).

    Finally, psychosocial issues such as cognitive decline, the

    availability of a caretaker, reluctance of physicians to treat

    elderly patients, or the proximity of patients residences to

    treatment centers, can all impact outcomes (Gross et al, 2005;

    Deschler et al, 2006).

    While clinical risk factors do not always differ significantly

    between older and younger patients, outcomes typically do,

    suggesting a biological basis for differential responses. Elderly

    AML more commonly evolves from an antecedent haemato-

    logical disorder (AHD) (Heinemann & Jehn, 1991), a feature

    that is independently associated with inferior responses,

    higher TRM, refractory disease and infectious complications

    (Estey et al, 1982; Gajewski et al, 1989; Heinemann & Jehn,

    1991). AML from an AHD is also more commonly associated

    with adverse cytogenetic profiles, defined as a complex

    karyotype (more than three abnormalities), chromosome 5or 7 deletions, 3q abnormalities, and monosomal deletions

    (Appelbaum, 2008; Medeiros et al, 2010). Morphologically,

    elderly blasts have less granulation and fewer Auer rods

    (Hassan & Rees, 1990), and biologically, elderly AML has a

    more immature stem cell-like phenotype (Fialkow et al,

    1981), resulting in increased cytopenias and toxicity with

    treatment (Stephan et al, 1998). Elderly patients have over-

    expression of the multidrug resistance 1 (MDR1) gene (Leith

    et al, 1997, 1999; Appelbaum et al, 2006; Roboz, 2007;

    Sekeres, 2008; Kuendgen & Germing, 2009), which encodes

    an efflux pump, permeability glycoprotein (Pgp), that

    extrudes chemotherapeutics from the cell and increases

    treatment resistance (Leith et al, 1997; van der Kolk et al,

    2002; Larson, 2003; Solary et al, 2003; Mahadevan & List,

    2004; Burnett & Mohite, 2006). MDR1 over-expression

    correlates with a reduced CR rate, OS and DFS, and is

    associated with relapsed and refractory disease, secondary

    AML and adverse cytogenetics (Wood et al, 1994; Willman,

    1996; Leith et al, 1999; Pinto et al, 2001; Baer et al, 2002;

    Larson, 2003; van der Holt et al, 2005; Burnett & Mohite,

    2006; Estey, 2007; Dombret et al, 2008). Finally, gene

    expression profiling in elderly AML has resulted in the

    identification of distinct subgroups that vary by outcome,

    supporting a molecular basis for poor clinical outcomes in

    elderly patients (Wilson et al, 2006; Raponi et al, 2008; deJonge et al, 2009; Rao et al, 2009) (Table I). All of these

    observations suggest elderly AML represents a distinct

    biological entity.

    Parsing out the impact on outcomes from biological versus

    host factors is a difficult exercise, especially because, as

    exemplified by the relationship between adverse cytogenetics

    and poor PS (Burnett et al, 2007), the two are inextricably

    linked. Although it is important to recognize the contribution

    from both, biological features have a greater impact on

    outcomes than host-related risk factors.

    Genetic factors

    Over 50% of elderly AML patients have a cytogenetic

    abnormality (Tiu et al, 2009), and this information is of

    utmost importance for risk stratification. Elderly patients with

    adverse cytogenetics have poor outcomes (Byrd et al, 2002),

    and those with intermediate-risk cytogenetics [defined as

    trisomy 8, normal karyotype (NK), and 11q23 abnormalities]

    have relatively more favourable outcomes (Grimwade et al,

    2001). However, in contrast to younger patients, in this

    population the presence of favourable cytogenetics (defined as

    translocations involving AML-ETO, CBFB-MYH11 or PML-

    RARa) is uncommon (Rowley et al, 1982), and it is unclear

    whether these abnormalities are associated with better out-

    comes, likely to due to underpowered studies (Frohling et al,

    2006; van der Holt et al, 2007). In a large study of the

    age-specific incidence of cytogenetic abnormalities, adverse

    cytogenetic profiles were disproportionately seen in elderly

    patients; furthermore, balanced translocations decreased and

    unbalanced aberrations increased with age (Bacher et al, 2005).Cytogenetic data predicts CR rate, relapse and DFS (Estey,

    2007) independently of age and other potential confounders

    (Hiddemann et al, 1999; Farag et al, 2006), and represents the

    most powerful biological outcomes predictor. Elderly patients

    with complex karyotypes experience minimal benefit from

    induction chemotherapy (Byrd et al, 2002; Knipp et al, 2007;

    Medeiros et al, 2010), possibly because the numerous genetic

    abnormalities result in increased chemotherapy resistance.

    The predictive power of cytogenetics is clear. However, in

    clinical practice, metaphase cytogenetic studies take several

    days. Practitioners are therefore faced with the dilemma of

    whether it is best to delay treatment with the expectation that

    the regimen will be tailored to the cytogenetic findings, or to

    treat immediately, and use this information purely for post-

    induction treatment decisions. Results of several studies report

    a negative impact from delayed treatment in elderly AML

    patients (Lowenberg et al, 1989; Rowe et al, 2004; Knipp et al,

    2007), while others have not shown that delays result in

    adverse outcomes (Sekeres et al, 2009). Further study is

    necessary to resolve this important question.

    The prognostic implications of molecular mutations for risk

    stratification in NK AML are not fully understood. In younger

    NK patients, nucleophosmin (NPM1) mutations, which confer

    a favourable outcome, are common (Becker et al, 2010). In

    older patients, NPM1 mutations were equally prevalent andalso associated with improved CR rates (Scholl et al, 2008) and

    OS compared to NPM1 wild type (WT) patients (Buchner

    et al, 2009; Becker et al, 2010). Furthermore, a gene expression

    profiling study in elderly patients confirmed that those with

    NPM1 mutations clustered together and had superior out-

    comes (Wilson et al, 2006). Up to one-third of younger

    patients have an internal tandem duplication (ITD) in the

    Fms-like tyrosine kinase 3 (FLT3) gene, which when mutated

    confers a worse prognosis regardless of NPM1 status (Kottari-

    dis et al, 2001). Older patients have a decreased, but

    Review

    2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542 525

  • 8/6/2019 AML in the Elderly, A Review

    3/19

    significant, incidence of FLT3 ITD mutations (Beran et al,

    2004), and although they are less predictive in older patients,

    FLT3 mutations are associated with an increased relapse risk

    and shorter OS and DFS (Stirewalt et al, 2001).

    Other prognostic factors

    In elderly patients for whom treatment carries a high risk of

    mortality, understanding which patients are most likely to

    experience favourable outcomes based on baseline assessments

    is essential. Although prognostications based on retrospective

    analyses have been published, few have been prospectively

    validated, making those large studies that have been of

    particular value. A retrospective analysis of over 1000 older

    AML patients who were treated with intensive regimens found

    cytogenetics, age, white blood cell count (WBC), PS andsecondary AML to be multivariate predictors of OS (Wheatley

    et al, 2009). These factors were validated in independent data

    sets of 1400 patients, the majority of whom received intensive

    regimens (Wheatley et al, 2009). In a large study that has not

    been validated, Katarina et aldivided patients into favourable,

    intermediate and high risk groups, using age, PS, cytogenetics,

    AHD, comorbidities and utilization of strict isolation; the

    groups had significantly different CR rates, TRM and OS.

    Rollig et al (2010) subdivided patients with intermediate-risk

    cytogenetics into two groups, good and adverse, and showed

    significant differences in 3-year OS for these cohorts. Other

    models have used combinations of cytogenetics, AHD, lactate

    dehydrogenase (LDH), leukocytosis, PS and comorbidities to

    predict OS in patients who receive induction chemotherapy

    (Ferrara & Mirto, 1996; Stasi et al, 1996; Wahlin et al, 2001;

    Gupta et al, 2005a; Latagliata et al, 2006; Gardin et al, 2007;

    Giles et al, 2007b) (Table II).

    Multivariate predictors of TRM include PS, laboratory

    abnormalities and the presence of infections (Estey, 2007),

    while multivariate predictors for early death include PS, organ

    function, comorbidity indices, beta-2 microglobulin, LDH,

    leukocytosis and thrombocytopenia (Latagliata et al, 2006;

    Tsimberidou et al, 2008; Burnett et al, 2009). Multivariate

    predictors for disease resistance include cytogenetics, beta-2

    microglobulin and PS (Albitar et al, 2007; Estey, 2008), and for

    OS include cytogenetics, age, luekocytosis, LDH, CD34-expression and NPM1 status (Rollig et al, 2010). More blasts

    at diagnosis predicted worse outcomes (Baudard et al, 1994),

    as did CD34-positive disease (Rollig et al, 2010), the presence

    of chromosomal monosomies (Breems et al, 2008; Medeiros

    et al, 2010), decreased expression of CD65s (Paietta et al,

    2003) and increased expression of CD7 (Stasi et al, 1996).

    Interestingly, age is not a multivariate predictor of outcomes in

    several models (Chen et al, 2005; Gupta et al, 2005a; Malfuson

    et al, 2008), suggesting other risk factors may be more

    important for treatment decisions.

    Table I. Selected gene expression profiling studies in elderly patients with AML suggest this represents a unique biological entity and provides a

    molecular explanation for poor outcomes.

    Reference Patients and methods Results Conclusions

    Wilson et al

    (2006)

    N = 170 AML with median age

    65 years; predominately

    intermediate to poor risk

    Unsupervised analysis showed six

    clusters with outcome differences

    Best prognostic group had 78% NPM1

    mutations, high Wilms tumourgene (WT1) over expression

    Group with most favourable

    cytogenetics did not have best

    outcome

    Differences between six groups with

    respect to refractory disease,

    attaining CR and DFS

    These differences were notexplained by age, cytogenetics or

    other factors

    Older patients can be separated into

    different groups that vary by

    outcome, independent of age and

    cytogenetics

    Rao et al

    (2009)

    N = 144 AML 55 years;

    compared to 175 AML patients

    45 years

    Older patients had differential

    activation of signalling pathways

    compared to younger

    Profile in older patients suggests

    decreased sensitivity to

    anthracyclines

    Gene expression profiling data can be

    utilized to tailor treatment regimens

    de Jonge

    et al (2009)

    N = 175 AML with median age

    59 years compared to 175 AML

    with median age 31 years

    Older patients with intermediate or

    poor risk cytogenetics had decreased

    expression of TSG CDKN2A

    Distinct gene expression profile noted

    for older compared to younger

    patients

    Raponi

    et al (2008)

    N = 67 AML 65 years from a

    clinical trial of 158 patients who

    received tipifarnib

    Identified and validated a two-gene

    expression ratio that predicted

    outcomes for newly diagnosed and

    relapsed/refractory patients who

    receive tipifarnib

    Can use gene expression profiling to

    predict patients more likely to

    respond to a given therapy

    NPM1, nucleophosmin gene; CR, complete remission; DFS, disease free survival; TSG, tumour suppressor gene.

    Review

    526 2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542

  • 8/6/2019 AML in the Elderly, A Review

    4/19

    Table II. Predictive models from elderly AML publications.

    Citation Patients and methods

    Multivariate poor prognostic factors forIndependent

    validation?CR OS

    Kantarjian et al

    (2006)

    N = 998 65 years AML/

    high-risk MDS received

    induction; retrospectively

    evaluated

    Age 75 years

    Treatment-related AML

    AHD 6 months

    Treatment outside LAFR

    Adverse cytogenetics

    Leucocytosis

    Anaemia

    Renal insufficiency

    ECOG PS >2

    Age 75 years

    AHD 12 months

    Treatment outside LAFR

    Adverse cytogenetics

    Renal insufficiency

    Elevated LDH

    ECOG PS >2

    No

    Rollig et al

    (2010)

    N = 909 > 60 years AML

    received two courses of

    induction; retrospectively

    evaluated

    Age >65 years

    Adverse cytogenetic

    NPM1-/FLT3 ITD+

    Elevated LDH

    Leucocytosis

    CD34 > 10%

    >10% BM blasts by day 15

    Age >65 years

    Adverse cytogenetics

    NPM1-/FLT3 ITD+

    Elevated LDH

    Leucocytosis

    CD34 > 10%

    No

    Wheatley et al

    (2009)

    N = 1071 older AML

    received induction;

    validated with independent

    data sets of N = 1412

    (N = 1137 intensively-

    treated, N = 275 treated

    with low intensity strategy)

    Not reported Adverse cytogenetics

    Secondary AML

    Age > 65 years

    Leucocytosis

    ECOG PS 2

    Yes

    Gupta et al

    (2005a)

    N = 117 60 years AML

    received induction;

    retrospectively evaluated

    ECOG PS >2

    Leucocytosis

    Elevated LDH

    Adverse cytogenetics

    Leucocytosis

    Adverse cytogenetics

    AHD

    No

    Johnson et al

    (1993)

    N = 104 60 years AML

    received induction;

    prospectively evaluated.

    65% had cytogenetic data.

    Not reported WHO PS 1

    Adverse cytogenetics

    Zubrod PS >1

    Elevated B2M

    Elevated uric acid

    Elevated LDH

    Yes (in validation

    sample)

    Wahlin et al

    (2001)

    N = 211 60 years AML

    most of whom received

    induction; retrospectively

    evaluated

    AHD

    Adverse cytogenetics

    Administration of less

    intensive induction regimens

    Older age

    Adverse cytogenetics

    Administration of less

    intensive induction regimens

    No

    Giles et al

    (2007a)

    N = 177 60 years AML

    received induction; pro

    spectively evaluated HCT-CI

    score

    HCT-CI predicted CR HCT-CI predicted OS Yes

    Stasi et al

    (1996)

    N = 159 60 years AML

    received a variety of

    therapies; retrospectively

    evaluated

    Older age

    Administration of less

    intensive induction regimens

    Decreased CD14 expression

    (aggressively-treated group only)

    Older age

    Administration of less

    intensive induction regimens

    Adverse cytogenetics

    (aggressively-treated group

    only)

    No

    Review

    2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542 527

  • 8/6/2019 AML in the Elderly, A Review

    5/19

    Gene expression profiling in older patients has revealed

    clusters of patient samples that contain both adverse and

    favourable cytogenetics (Wilson et al, 2006), suggesting there

    are important biological prognostic factors that have yet to be

    discovered. In the future, results from rationally designed

    clinical/translational studies will allow clinicians to predictresponders to a given regimen based on molecular data,

    maximizing the treatment effect while minimizing toxicity.

    Treatment

    Treatment of AML in the elderly is challenging; patients have

    varying tolerance for toxicity, treatments are rarely curative,

    and published studies either exclude the elderly or are limited

    by selection bias. Although many treatment options have been

    explored for elderly patients with AML, achievement of CR [or

    CR with incomplete blood count recovery (CRi)] (Cheson

    et al, 2003) remains necessary for long-term disease-free

    survival. In a meta-analysis of over 12 000 elderly AML

    patients, 50% received intensive therapy and had a median OS

    of 30 weeks compared to 12 weeks from lower intensity

    treatment, but this observation is limited by the fact that

    equivalent patients were not directly compared (Deschler et al,

    2006). However, TRM is a significant limitation for intensive

    treatment in elderly patients, with 1040% experiencing this

    outcome compared to 2

    Decreased plateletsIncreased PB blasts

    No

    CR, complete response; OS, overall survival; MDS, myelodysplastic syndrome, AHD, antecedent haematological disorder, LAFR, laminar airflow

    room, ECOG PS, Eastern Cooperative Oncology Group performance status; PB, peripheral blood; LDH, lactate dehydrogenase; BM, bone marrow;

    B2M, beta 2 microglobulin; HCT-CI, haematopoietic cell transplantation comorbidity index; WHO PS, World Health Organization performance

    status.

    Review

    528 2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542

  • 8/6/2019 AML in the Elderly, A Review

    6/19

    versus 35 mg/m2 of daunorubicin (Burnett et al, 2009). In

    contrast, Lowenberg et al(2009) showed that for the subgroup

    of patients 6065 with an excellent PS and more indolent

    disease, 90 mg/m2 of daunorubicin was superior to 45 mg/m2,

    but these results have yet to be independently validated. Very

    high doses of cytarabine (1 g/m2) are contraindicated in older

    AML patients due to a significantly higher rate of cerebellar

    toxicity (Preisler et al, 1987), and there was no benefit from

    400 mg/m2 of cytarabine compared to 200 mg/m2 (Burnett

    et al, 2009), or 200 mg/m2 compared to 100 mg/m2 (Dillman

    et al, 1991).

    Conversely, supported by observations that reduced

    dosages of chemotherapy in older patients have similar

    half-lives to full dosages given to younger patients (Leoni

    et al, 1995), attempts have been made to improve the

    tolerability to induction by reducing chemotherapeutic

    dosages. Yates et al (1982) reported a higher CR rate in

    older patients who received 30 mg/m2

    of daunorubicincompared to those who received 45 mg/m2, while others did

    not observe an advantage from anthracycline dose reduc-

    tions (Kahn et al, 1984).

    New strategies are necessary to improve induction in elderly

    patients. Oral induction regimens are generally well tolerated

    but have not resulted in significant improvements in responses

    (Ruutu et al, 1994). Substituting fludarabine for an anthracy-

    cline (Ferrara et al, 2005) or adding cladrabine (Juliusson et al,

    2003) or all-trans retinoic acid (Schlenk et al, 2004) to

    standard induction may also be reasonable approaches.

    TRM is limiting in older patients treated with induction,

    and most deaths are due to infection (Dombret et al, 1995;

    Rowe et al, 1995; Stone et al, 1995). Using a growth factor

    cytokine to shorten the duration of neutropenia has been

    proposed to improve outcomes for elderly AML patients. In

    randomized trials, growth factors were found to significantly

    decrease the duration of neutropenia by several days (Buchner

    et al, 1991; Dombret et al, 1995; Rowe et al, 1995; Lowenberg

    et al, 1997; Witz et al, 1998). However, there was no impact on

    TRM, infections, time spent in the hospital or most impor-

    tantly, OS (Maslaket al, 1996; Lowenberg et al, 1997; Godwin

    et al, 1998; Witz et al, 1998; Goldstone et al, 2001; Buchner

    et al, 2004; Rowe et al, 2004), although some studies did show

    a benefit for CR rate in the absence of OS (Dombret et al,

    1995; Rowe et al, 1995). Because reduction in febrile neutro-

    penia is an important endpoint, their use has been recom-

    mended, typically after demonstration of bone marrow aplasia,

    for patients older than 55 (Smith et al, 2006).

    Post-remission and stem cell transplantation

    Elderly patients who achieve a CR experience a remission

    duration of 10 months (Schiller, 1996) and have a >75%

    chance of relapse. While these patients may derive benefit from

    post-remission therapies, it is often prohibitive due to

    comorbidities or residual toxicity from induction. The stan-

    dard post-remission regimen includes one to two cycles of

    cytarabine with or without an anthracycline. In the elderly, no

    Fig 1. Treatment algorithm for elderly AML patients. *Intermediate risk group scoring system. Scoring system adapted from Rollig et al (2010).

    Further therapeutic recommendations.

    Review

    2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542 529

  • 8/6/2019 AML in the Elderly, A Review

    7/19

  • 8/6/2019 AML in the Elderly, A Review

    8/19

    Table

    III.Continued.

    Citation

    Patientsand

    methods

    Outcomes

    CR

    TRM

    OS

    Comments

    Gardinetal(2007)

    N

    =416AML

    65years

    randomized

    toinductionwith

    IDAvs.DN

    R

    59%

    IDAvs.54%

    DN

    R

    (P=0

    28)

    Inductiondeath9%

    IDAvs.10%

    DNR(P=0

    87)

    2-yearforbotharms27%;OS

    estimatesweresimilarinboth

    DNRandIDArandomization

    arms(P=0

    37)

    Nodifferencesinoutcomesfor

    IDA

    orDNR

    Pautasetal(2010)

    N

    =468AML5070years

    randomized

    toARA-Cplus

    standarddo

    seIDA,

    highdose

    IDAorDNR80mg/m

    2.

    Those

    whofailedi

    nitialinduction

    receivedsalvagewithARA-C

    andMA

    Afterfirstinduction,7

    0%

    standarddoseIDAvs.67%

    highdoseIDAvs.61%

    DNR

    (P=0

    25)

    Inductiondeaths:6%

    standard

    doseIDAvs.3%

    highdoseIDA

    vs.8%

    DNR(P=0

    24)

    4-year=32%

    standarddoseIDA

    vs.34%

    highdoseIDAvs.23%

    DNR(P=0

    19)

    OverallCRrate(included

    patie

    ntswhofailedinitial

    inductionbutrespondedto

    salva

    ge)favouredstandard

    dose

    IDA,

    butnoOS

    difference

    Schlenketal(2004)

    N

    =242AML

    61years

    randomized

    to

    induction

    ATRA

    52%forATRAvs.39%

    forno

    ATRA(P=0

    05)

    Notreported;haematological

    andnon-haematological

    toxicitysimilarforATRAand

    noATRAarms

    Median11monthsforATRA

    versusmedian7monthsforno

    ATRA(P or = 60

    years with acute myeloid leukemia treated withuniform intensive therapy. Cancer, 103, 2082

    2090.

    Gupta, V., Daly, A., Lipton, J.H., Hasegawa, W.,

    Chun, K., Kamel-Reid, S., Tsang, R., Yi, Q.L.,

    Minden, M., Messner, H. & Kiss, T. (2005b)

    Nonmyeloablative stem cell transplantation for

    myelodysplastic syndrome or acute myeloid leu-

    kemia in patients 60 years or older. Biology of

    Blood and Marrow Transplantation, 11, 764772.

    Guzman, M.L., Rossi, R.M., Neelakantan, S., Li, X.,

    Corbett, C.A., Hassane, D.C., Becker, M.W.,

    Bennett, J.M., Sullivan, E., Lachowicz, J.L.,

    Vaughan, A., Sweeney, C.J., Matthews, W., Car-

    roll, M., Liesveld, J.L., Crooks, P.A. & Jordan,

    C.T. (2007) An orally bioavailable parthenolide

    analog selectively eradicates acute myelogenous

    leukemia stem and progenitor cells. Blood, 110,

    44274435.

    Harousseau, J.L. (1998) Acute myeloid leukemia in

    the elderly. Blood Reviews, 12, 145153.

    Harousseau, J.L., Lancet, J.E., Reiffers, J., Low-

    enberg, B., Thomas, X., Huguet, F., Fenaux, P.,

    Zhang, S., Rackoff, W., De Porre, P. & Stone, R.

    (2007) A phase 2 study of the oral farnesyl-

    transferase inhibitor tipifarnib in patients with

    Review

    2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542 537

  • 8/6/2019 AML in the Elderly, A Review

    15/19

  • 8/6/2019 AML in the Elderly, A Review

    16/19

    Larson, R.A. (2001) Current use and future devel-

    opment of gemtuzumab ozogamicin. Seminars in

    Hematology, 38, 2431.

    Larson, R.A. (2003) Is modulation of multidrug

    resistance a viable strategy for acute myeloid

    leukemia? Leukemia, 17, 488491.

    Larson, R.A., Boogaerts, M., Estey, E., Karanes, C.,

    Stadtmauer, E.A., Sievers, E.L., Mineur, P., Ben-

    nett, J.M., Berger, M.S., Eten, C.B., Munteanu,M., Loken, M.R., Van Dongen, J.J., Bernstein,

    I.D. & Appelbaum, F.R. (2002) Antibody-tar-

    geted chemotherapy of older patients with acute

    myeloid leukemia in first relapse using Mylotarg

    (gemtuzumab ozogamicin). Leukemia, 16, 1627

    1636.

    Larson, R.A., Sievers, E.L., Stadtmauer, E.A., Low-

    enberg, B., Estey, E.H., Dombret, H., Theobald,

    M., Voliotis, D., Bennett, J.M., Richie, M., Leo-

    pold, L.H., Berger, M.S., Sherman, M.L., Loken,

    M.R., van Dongen, J.J., Bernstein, I.D. & Appel-

    baum, F.R. (2005) Final report of the efficacy and

    safety of gemtuzumab ozogamicin (Mylotarg) in

    patients with CD33-positive acute myeloid leu-

    kemia in first recurrence. Cancer, 104, 14421452.

    Latagliata, R., Bongarzoni, V., Carmosino, I., Men-

    garelli, A., Breccia, M., Borza, P.A., DAndrea,

    M., DElia, G.M., Mecarocci, S., Morano, S.G.,

    Petti, M.C., Mandelli, F. & Alimena, G. (2006)

    Acute myelogenous leukemia in elderly patients

    not eligible for intensive chemotherapy: the dark

    side of the moon. Annals of Oncology, 17, 281

    285.

    Laubach, J. & Rao, A.V. (2008) Current and emer-

    ging strategies for the management of acute

    myeloid leukemia in the elderly. The Oncologist,

    13, 10971108.

    Leith, C.P., Kopecky, K.J., Godwin, J., McConnell,

    T., Slovak, M.L., Chen, I.M., Head, D.R., Ap-pelbaum, F.R. & Willman, C.L. (1997) Acute

    myeloid leukemia in the elderly: assessment of

    multidrug resistance (MDR1) and cytogenetics

    distinguishes biologic subgroups with remarkably

    distinct responses to standard chemotherapy. A

    Southwest Oncology Group study. Blood, 89,

    33233329.

    Leith, C.P., Kopecky, K.J., Chen, I.M., Eijdems, L.,

    Slovak, M.L., McConnell, T.S., Head, D.R.,

    Weick, J., Grever, M.R., Appelbaum, F.R. &

    Willman, C.L. (1999) Frequency and clinical

    significance of the expression of the multidrug

    resistance proteins MDR1/P-glycoprotein,

    MRP1, and LRP in acute myeloid leukemia: a

    Southwest Oncology Group Study. Blood, 94,

    10861099.

    Leone, G., Teofili, L., Voso, M.T. & Lubbert, M.

    (2002) DNA methylation and demethylating

    drugs in myelodysplastic syndromes and sec-

    ondary leukemias. Haematologica, 87, 13241341.

    Leoni, F., Ciolli, S., Giuliani, G., Pascarella, A., Ca-

    porale, R., Salti, F., Cervi, L. & Rossi Ferrini, P.

    (1995) Attenuated-dose idarubicin in acute

    myeloid leukaemia of the elderly: pharmacoki-

    netic study and clinical results. British Journal of

    Haematology, 90, 169174.

    Li, L., Giannopoulos, K., Reinhardt, P., Tabarkie-

    wicz, J., Schmitt, A., Greiner, J., Rolinski, J., Hus,

    I., Dmoszynska, A., Wiesneth, M. & Schmitt, M.

    (2006) Immunotherapy for patients with acute

    myeloid leukemia using autologous dendritic

    cells generated from leukemic blasts. Interna-

    tional Journal of Oncology, 28, 855861.

    Lowenberg, B., Zittoun, R., Kerkhofs, H., Jehn, U.,

    Abels, J., Debusscher, L., Cauchie, C., Peeter-mans, M., Solbu, G., Suciu, S. et al(1989) On the

    value of intensive remission-induction che-

    motherapy 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.

    Journal of Clinical Oncology, 7, 12681274.

    Lowenberg, B., Suciu, S., Archimbaud, E., Ossen-

    koppele, G., Verhoef, G.E., Vellenga, E., Wijer-

    mans, P., Berneman, Z., Dekker, A.W.,

    Stryckmans, P., Schouten, H., Jehn, U., Muus, P.,

    Sonneveld, P., Dardenne, M. & Zittoun, R.

    (1997) Use of recombinant GM-CSF during and

    after remission induction chemotherapy in pa-

    tients aged 61 years and older with acute myeloidleukemia: final report of AML-11, a phase III

    randomized study of the Leukemia Cooperative

    Group of European Organisation for the Re-

    search and Treatment of Cancer and the Dutch

    Belgian Hemato-Oncology Cooperative Group.

    Blood, 90, 29522961.

    Lowenberg, B., Ossenkoppele, G.J., van Putten, W.,

    Schouten, H.C., Graux, C., Ferrant, A., Sonne-

    veld, P., Maertens, J., Jongen-Lavrencic, M., von

    Lilienfeld-Toal, M., Biemond, B.J., Vellenga, E.,

    van Marwijk Kooy, M., Verdonck, L.F., Beck, J.,

    Dohner, H., Gratwohl, A., Pabst, T. & Verhoef, G.

    (2009) High-dose daunorubicin in older patients

    with acute myeloid leukemia. New England

    Journal of Medicine, 361, 12351248.Lowenberg, B., Beck, J., Graux, C., van Putten, W.,

    Schouten, H.C., Verdonck, L.F., Ferrant, A.,

    Sonneveld, P., Jongen-Lavrencic, M., von Li-

    lienfeld-Toal, M., Biemond, B.J., Vellenga, E.,

    Breems, D., de Muijnck, H., Schaafsma, R., Ver-

    hoef, G., Dohner, H., Gratwohl, A., Pabst, T.,

    Ossenkoppele, G.J. & Maertens, J. (2010) Gem-

    tuzumab ozogamicin as postremission treatment

    in AML at 60 years of age or more: results of a

    multicenter phase 3 study. Blood, 115, 2586

    2591.

    Mahadevan, D. & List, A.F. (2004) Targeting the

    multidrug resistance-1 transporter in AML: mo-

    lecular regulation and therapeutic strategies.

    Blood, 104, 19401951.

    Malfuson, J.V., Etienne, A., Turlure, P., de Revel, T.,

    Thomas, X., Contentin, N., Terre, C., Rigaudeau,

    S., Bordessoule, D., Vey, N., Gardin, C. &

    Dombret, H. (2008) Risk factors and decision

    criteria for intensive chemotherapy in older pa-

    tients with acute myeloid leukemia. Haematolo-

    gica, 93, 18061813.

    Marcucci, G., Stock, W., Dai, G., Klisovic, R.B., Liu,

    S., Klisovic, M.I., Blum, W., Kefauver, C., Sher,

    D.A., Green, M., Moran, M., Maharry, K., No-

    vick, S., Bloomfield, C.D., Zwiebel, J.A., Larson,

    R.A., Grever, M.R., Chan, K.K. & Byrd, J.C.

    (2005) Phase I study of oblimersen sodium, an

    antisense to Bcl-2, in untreated older patients

    with acute myeloid leukemia: pharmacokinetics,

    pharmacodynamics, and clinical activity. Journal

    of Clinical Oncology, 23, 34043411.

    Maslak, P.G., Weiss, M.A., Berman, E., Yao, T.J.,

    Tyson, D., Golde, D.W. & Scheinberg, D.A.

    (1996) Granulocyte colony-stimulating factorfollowing chemotherapy in elderly patients with

    newly diagnosed acute myelogenous leukemia.

    Leukemia, 10, 3239.

    Mayer, R.J., Davis, R.B., Schiffer, C.A., Berg, D.T.,

    Powell, B.L., Schulman, P., Omura, G.A., Moore,

    J.O., McIntyre, O.R. & Frei, E. 3rd (1994) In-

    tensive postremission chemotherapy in adults

    with acute myeloid leukemia. Cancer and Leu-

    kemia Group B. New England Journal of Medi-

    cine, 331, 896903.

    McClune, B.L., Weisdorf, D.J., Pedersen, T.L., Tunes

    da Silva, G., Tallman, M.S., Sierra, J., Dipersio, J.,

    Keating, A., Gale, R.P., George, B., Gupta, V.,

    Hahn, T., Isola, L., Jagasia, M., Lazarus, H.,

    Marks, D., Maziarz, R., Waller, E.K., Bredeson, C.& Giralt, S. (2010) Effect of age on outcome of

    reduced-intensity hematopoietic cell transplan-

    tation for older patients with acute myeloid leu-

    kemia in first complete remission or with

    myelodysplastic syndrome. Journal of Clinical

    Oncology, 28, 18781887.

    Medeiros, B.C., Othus, M., Fang, M., Roulston, D. &

    Appelbaum, F.R. (2010) Prognostic impact of

    monosomal karyotype in young adult and elderly

    acute myeloid leukemia: the Southwest Oncology

    Group experience. Blood, 116, 22242228.

    Milligan, D.W., Grimwade, D., Cullis, J.O., Bond, L.,

    Swirsky, D., Craddock, C., Kell, J., Homewood, J.,

    Campbell, K., McGinley, S., Wheatley, K. &

    Jackson, G. (2006) Guidelines on the manage-ment of acute myeloid leukaemia in adults.

    British Journal of Haematology, 135, 450474.

    Mohty, M., de Lavallade, H., Ladaique, P., Faucher,

    C., Vey, N., Coso, D., Stoppa, A.M., Gastaut, J.A.

    & Blaise, D. (2005) The role of reduced intensity

    conditioning allogeneic stem cell transplantation

    in patients with acute myeloid leukemia: a donor

    vs no donor comparison. Leukemia, 19, 916920.

    Moore, J., Seiter, K., Kolitz, J., Stock, W., Giles, F.,

    Kalaycio, M., Zenk, D. & Marcucci, G. (2006) A

    Phase II study of Bcl-2 antisense (oblimersen

    sodium) combined with gemtuzumab ozogami-

    cin in older patients with acute myeloid leukemia

    in first relapse. Leukemia Research, 30, 777783.

    Morgan, W.F. & Sowa, M.B. (2007) Non-targeted

    bystander effects induced by ionizing radiation.

    Mutation Research, 616, 159164.

    Nabhan, C., Rundhaugen, L.M., Riley, M.B., Rade-

    maker, A., Boehlke, L., Jatoi, M. & Tallman, M.S.

    (2005) Phase II pilot trial of gemtuzumab ozo-

    gamicin (GO) as first line therapy in acute

    myeloid leukemia patients age 65 or older. Leu-

    kemia Research, 29, 5357.

    Nand, S., Godwin, J., Smith, S., Barton, K., Mi-

    chaelis, L., Alkan, S., Veerappan, R., Rychlik, K.,

    Germano, E. & Stiff, P. (2008) Hydroxyurea,

    Review

    2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542 539

  • 8/6/2019 AML in the Elderly, A Review

    17/19

    azacitidine and gemtuzumab ozogamicin therapy

    in patients with previously untreated non-M3

    acute myeloid leukemia and high-risk myelo-

    dysplastic syndromes in the elderly: results from a

    pilot trial. Leukemia and Lymphoma, 49, 2141

    2147.

    OFarrell, A.M., Foran, J.M., Fiedler, W., Serve, H.,

    Paquette, R.L., Cooper, M.A., Yuen, H.A., Louie,

    S.G., Kim, H., Nicholas, S., Heinrich, M.C.,Berdel, W.E., Bello, C., Jacobs, M., Scigalla, P.,

    Manning, W.C., Kelsey, S. & Cherrington, J.M.

    (2003) An innovative phase I clinical study de-

    monstrates inhibition of FLT3 phosphorylation

    by SU11248 in acute myeloid leukemia patients.

    Clinical Cancer Research, 9, 54655476.

    OFarrell, A.M., Yuen, H.A., Smolich, B., Hannah,

    A.L., Louie, S.G., Hong, W., Stopeck, A.T., Sil-

    verman, L.R., Lancet, J.E., Karp, J.E., Albitar, M.,

    Cherrington, J.M. & Giles, F.J. (2004) Effects of

    SU5416, a small molecule tyrosine kinase re-

    ceptor inhibitor, on FLT3 expression and phos-

    phorylation in patients with refractory acute

    myeloid leukemia. Leukemia Research, 28, 679

    689.Paietta, E., Neuberg, D., Bennett, J.M., Dewald, G.,

    Rowe, J.M., Cassileth, P.A., Cripe, L., Tallman,

    M.S. & Wiernik, P.H. (2003) Low expression of

    the myeloid differentiation antigen CD65s, a

    feature of poorly differentiated AML in older

    adults: study of 711 patients enrolled in ECOG

    trials. Leukemia, 17, 15441550.

    Pautas, C., Merabet, F., Thomas, X., Raffoux, E.,

    Gardin, C., Corm, S., Bourhis, J.H., Reman, O.,

    Turlure, P., Contentin, N., de Revel, T., Rousse-

    lot, P., Preudhomme, C., Bordessoule, D., Fe-

    naux, P., Terre, C., Michallet, M., Dombret, H.,

    Chevret, S. & Castaigne, S. (2010) Randomized

    study of intensified anthracycline doses for in-

    duction and recombinant interleukin-2 formaintenance in patients with acute myeloid leu-

    kemia age 50 to 70 years: results of the ALFA-

    9801 study. Journal of Clinical Oncology, 28, 808

    814.

    Piccaluga, P.P., Martinelli, G., Rondoni, M., Mala-

    gola, M., Gaitani, S., Visani, G. & Baccarani, M.

    (2004) First experience with gemtuzumab ozo-

    gamicin plus cytarabine as continuous infusion

    for elderly acute myeloid leukaemia patients.

    Leukemia Research, 28, 987990.

    Pinto, A., Zagonel, V. & Ferrara, F. (2001) Acute

    myeloid leukemia in the elderly: biology and

    therapeutic strategies. Critical Reviews in Oncol-

    ogy/Hematology, 39, 275287.

    Preisler, H.D., Raza, A., Barcos, M., Azarnia, N.,

    Larson, R., Walker, I., Browman, M., Grunwald,

    H., DArrigo, P., Doeblin, T. et al (1987) High-

    dose cytosine arabinoside as the initial treatment

    of poor-risk patients with acute nonlymphocytic

    leukemia: a Leukemia Intergroup Study. Journal

    of Clinical Oncology, 5, 7582.

    Pulsoni, A., Pagano, L., Latagliata, R., Casini, M.,

    Cerri, R., Crugnola, M., De Paoli, L., Di Bona, E.,

    Invernizzi, R., Marmont, F., Petti, M.C., Rigolin,

    G., Ronco, F., Spadano, A., Tosti, M.E., Visani,

    G., Mele, A. & Mandelli, F. (2004) Survival of

    elderly patients with acute myeloid leukemia.

    Haematologica, 89, 296302.

    Rao, A.V., Valk, P.J., Metzeler, K.H., Acharya, C.R.,

    Tuchman, S.A., Stevenson, M.M., Rizzieri, D.A.,

    Delwel, R., Buske, C., Bohlander, S.K., Potti, A. &

    Lowenberg, B. (2009) Age-specific differences in

    oncogenic pathway dysregulation and anthracy-

    cline sensitivity in patients with acute myeloid

    leukemia. Journal of Clinical Oncology, 27, 55805586.

    Raponi, M., Lancet, J.E., Fan, H., Dossey, L., Lee, G.,

    Gojo, I., Feldman, E.J., Gotlib, J., Morris, L.E.,

    Greenberg, P.L., Wright, J.J., Harousseau, J.L.,

    Lowenberg, B., Stone, R.M., De Porre, P., Wang,

    Y. & Karp, J.E. (2008) A 2-gene classifier for

    predicting response to the farnesyltransferase in-

    hibitor tipifarnib in acute myeloid leukemia.

    Blood, 111, 25892596.

    Recher, C., Beyne-Rauzy, O., Demur, C., Chicanne,

    G., Dos Santos, C., Mas, V.M., Benzaquen, D.,

    Laurent, G., Huguet, F. & Payrastre, B. (2005)

    Antileukemic activity of rapamycin in acute

    myeloid leukemia. Blood, 105, 25272534.

    Rees, J.K., Gray, R.G. & Wheatley, K. (1996) Doseintensification in acute myeloid leukaemia:

    greater effectiveness at lower cost. Principal re-

    port of the Medical Research Councils AML9

    study. MRC Leukaemia in Adults Working Party.

    British Journal of Haematology, 94, 8998.

    Rezvani, K., Yong, A.S., Mielke, S., Savani, B.N.,

    Musse, L., Superata, J., Jafarpour, B., Boss, C. &

    Barrett, A.J. (2008) Leukemia-associated antigen-

    specific T-cell responses following combined PR1

    and WT1 peptide vaccination in patients with

    myeloid malignancies. Blood, 111, 236242.

    Roboz, G.J. (2007) Treatment of acute myeloid

    leukemia in older patients. Expert Review of An-

    ticancer Therapy, 7, 285295.

    Roboz, G.J., Giles, F.J., Ritchie, E.K., Allen-Bard, S.,Curcio, T.J., Wilkes, M.A., Park, S.L., Kantarjian,

    H.M., Faderl, S., Ravandi, F., Kelner, M.J. &

    Feldman, E.J. (2007) Phase I/II study of con-

    tinuous-infusion troxacitabine in refractory acute

    myeloid leukemia. Journal of Clinical Oncology,

    25, 1015.

    Roboz, G.J., Ritchie, E.K., Curcio, T., Provenzano,

    J., Carlin, R., Samuel, M., Wittenberg, B., Ma-

    zumdar, M., Christos, P.J., Mathew, S., Allen-

    Bard, S. & Feldman, E.J. (2008) Arsenic trioxide

    and low-dose cytarabine in older patients with

    untreated acute myeloid leukemia, excluding

    acute promyelocytic leukemia. Cancer, 113,

    25042511.

    Rollig, C., Thiede, C., Gramatzki, M., Aulitzky, W.,

    Bodenstein, H., Bornhauser, M., Platzbecker, U.,

    Stuhlmann, R., Schuler, U., Soucek, S., Kramer,

    M., Mohr, B., Oelschlaegel, U., Stolzel, F., von

    Bonin, M., Wermke, M., Wandt, H., Ehninger, G.

    & Schaich, M. (2010) A novel prognostic model

    in elderly patients with acute myeloid leukemia

    results of 909 patients entered into the pro-

    spective AML96 trial. Blood, 116, 971978.

    Rowe, J.M., Andersen, J.W., Mazza, J.J., Bennett,

    J.M., Paietta, E., Hayes, F.A., Oette, D., Cassileth,

    P.A., Stadtmauer, E.A. & Wiernik, P.H. (1995) A

    randomized placebo-controlled phase III study of

    granulocyte-macrophage colony-stimulating fac-

    tor in adult patients (> 55 to 70 years of age) with

    acute myelogenous leukemia: a study of the

    Eastern Cooperative Oncology Group (E1490).

    Blood, 86, 457462.

    Rowe, J.M., Neuberg, D., Friedenberg, W., Bennett,

    J.M., Paietta, E., Makary, A.Z., Liesveld, J.L.,

    Abboud, C.N., Dewald, G., Hayes, F.A., Tallman,M.S. & Wiernik, P.H. (2004) 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, 103, 479485.

    Rowley, J.D., Alimena, G., Garson, O.M., Hage-

    meijer, A., Mitelman, F. & Prigogina, E.L. (1982)

    A collaborative study of the relationship of the

    morphological type of acute nonlymphocytic

    leukemia with patient age and karyotype. Blood,

    59, 10131022.

    Ruutu, T., Almqvist, A., Hallman, H., Honkanen, T.,

    Jarvenpaa, E., Jarventie, G., Koistinen, P., Koi-

    vunen, E., Lahtinen, R., Lehtinen, M. et al(1994)

    Oral induction and consolidation of acute mye-loid leukemia with etoposide, 6-thioguanine, and

    idarubicin (ETI) in elderly patients: a rando-

    mized comparison with 5-day TAD. Finnish

    Leukemia Group. Leukemia, 8, 1115.

    Scatena, C.D., Kumer, J.L., Arbitrario, J.P., Howlett,

    A.R., Hawtin, R.E., Fox, J.A. & Silverman, J.A.

    (2010) Voreloxin, a first-in-class anticancer qui-

    nolone derivative, acts synergistically with cytar-

    abine in vitro and induces bone marrow aplasia

    in vivo. Cancer Chemotherapy and Pharmacology,

    66, 881888.

    Schiller, G.J. (1996) Postremission therapy of acute

    myeloid leukemia in older adults. Leukemia,

    10(Suppl 1), S18S20.

    Schiller, G.J., OBrien, S.M., Pigneux, A., Deangelo,D.J., Vey, N., Kell, J., Solomon, S., Stuart, R.K.,

    Karsten, V., Cahill, A.L., Albitar, M.X. & Giles,

    F.J. (2010) Single-agent laromustine, a novel al-

    kylating agent, has significant activity in older

    patients with previously untreated poor-risk

    acute myeloid leukemia. Journal of Clinical On-

    cology, 28, 815821.

    Schlenk, R.F., Frohling, S., Hartmann, F., Fischer,

    J.T., Glasmacher, A., del Valle, F., Grimminger,

    W., Gotze, K., Waterhouse, C., Schoch, R., Pralle,

    H., Mergenthaler, H.G., Hensel, M., Koller, E.,

    Kirchen, H., Preiss, J., Salwender, H., Bie-

    dermann, H.G., Kremers, S., Griesinger, F.,

    Benner, A., Addamo, B., Dohner, K., Haas, R. &

    Dohner, H. (2004) Phase III study of all-trans

    retinoic acid in previously untreated patients 61

    years or older with acute myeloid leukemia.

    Leukemia, 18, 17981803.

    Scholl, S., Theuer, C., Scheble, V., Kunert, C., Heller,

    A., Mugge, L.O., Fricke, H.J., Hoffken, K. &

    Wedding, U. (2008) Clinical impact of nucleo-

    phosmin mutations and Flt3 internal tandem

    duplications in patients older than 60 yr with

    acute myeloid leukaemia. European Journal of

    Haematology, 80, 208215.

    Review

    540 2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542

  • 8/6/2019 AML in the Elderly, A Review

    18/19

  • 8/6/2019 AML in the Elderly, A Review

    19/19

    tifies novel biologic clusters for risk classification

    and outcome prediction. Blood, 108, 685696.

    Witz, F., Sadoun, A., Perrin, M.C., Berthou, C.,

    Briere, J., Cahn, J.Y., Lioure, B., Witz, B., Fran-

    cois, S., Desablens, B., Pignon, B., Le Prise, P.Y.,

    Audhuy, B., Caillot, D., Casassus, P., Delain, M.,

    Christian, B., Tellier, Z., Polin, V., Hurteloup, P.

    & Harousseau, J.L. (1998) A placebo-controlled

    study of recombinant human granulocyte-mac-rophage colony-stimulating factor administered

    during and after induction treatment for de novo

    acute myelogenous leukemia in elderly patients.

    Groupe Ouest Est Leucemies Aigues Myeloblas-

    tiques (GOELAM). Blood, 91, 27222730.

    Wood, P., Burgess, R., MacGregor, A. & Yin, J.A.

    (1994) P-glycoprotein expression on acute mye-

    loid leukaemia blast cells at diagnosis predicts

    response to chemotherapy and survival. British

    Journal of Haematology, 87, 509514.

    Yates, J., Glidewell, O., Wiernik, P., Cooper, M.R.,

    Steinberg, D., Dosik, H., Levy, R., Hoagland, C.,

    Henry, P., Gottlieb, A., Cornell, C., Berenberg, J.,

    Hutchison, J.L., Raich, P., Nissen, N., Ellison,R.R., Frelick, R., James, G.W., Falkson, G., Silver,

    R.T., Haurani, F., Green, M., Henderson, E.,

    Leone, L. & Holland, J.F. (1982) Cytosine arabi-

    noside with daunorubicin or adriamycin for

    therapy of acute myelocytic leukemia: a CALGB

    study. Blood, 60, 454462.

    Zein, N., Sinha, A.M., McGahren, W.J. & Ellestad,

    G.A. (1988) Calicheamicin gamma 1I: an anti-

    tumor antibiotic that cleaves double-stranded

    DNA site specifically. Science, 240, 11981201.

    Zeleny, J. (2008) Obama Weighs Quick Undoing of

    Bush Policy, pp. A17. New York Times, New

    York.Zhu, X., Ma, Y. & Liu, D. (2010) Novel agents and

    regimens for acute myeloid leukemia: 2009 ASH

    annual meeting highlights. Journal of Hematology

    and Oncology, 3, 17.

    Review

    542 2011 Blackwell Publishing Ltd, British Journal of Haematology, 152, 524542