Sunteți pe pagina 1din 20

Acute Myeloid Leukemia

Richard M. Stone, Margaret R. O’Donnell, and Mikkael A. Sekeres

Advances in our understanding of the patho- tyrosine kinase, activated via mutation in 30% of
physiology of acute myeloid leukemia (AML) patients.
have not yet led to major improvements in In Section II Dr. Margaret O’Donnell dis-
disease-free and overall survival of adults with cusses the role of stem cell transplantation in
this disease. Only about one-third of those AML. Several advances including expanded
between ages 18–60 who are diagnosed with AML donor pools, the movement toward peripheral
can be cured; disease-free survival is rare and blood stem cell collection, newer immunosup-
current therapy devastating in older adults. In pressive drugs and antifungals, and particularly
this chapter, challenges in the management of the advent of nonmyeloablative transplant have
the adult with AML are discussed, including made the allogeneic option more viable. The
ongoing questions concerning the optimal subset-specific role for high-dose chemotherapy
choice of induction and postremission therapy with autologous stem cell support and/or for
such as the rationale for and role of allogeneic allogeneic transplant in AML patients in first
and autologous stem cell transplantation in a remission is outlined. Although preconceived
variety of settings, the special considerations notions about the role of transplant abound, the
pertaining to the older patient, and the develop- clinical data supporting a risk-adapted approach
ment of new, so-called targeted therapies. are covered. Finally, guidance concerning the
In Section I, Dr. Richard Stone reviews state- use of nonmyeloablative or reduced-intensity
of the-art therapy in AML in the era of change allogeneic transplantation is provided.
from a morphological to a genetically based In Section III Dr. Mikkael Sekeres reviews the
classification system. Questions being addressed approach to the older patient with AML. Unique
in ongoing randomized cooperative group trials biological and therapeutic considerations make
include anthracycline dose during induction, the AML in this age group a vastly different disease
efficacy of drug-resistance modulators, and the than that in younger adults. The outcome data,
utility of pro-apoptotic agents such as the anti- including the role of specific anthracylines,
bcl-2 antisense oligonucloetide. Developmental hematopoietic growth factors, and drug-resis-
therapeutics in AML include drug resistance tance modulators, are summarized. Communicat-
modulation, anti-angiogenic strategies, immuno- ing with older adults with AML and their families
therapy, and signal transduction-active agents, regarding selection of the optimal treatment
particularly the farnesyl transferase inhibitors as strategy, often a stark choice between induction
well as those molecules that inhibit the FLT3 chemotherapy and palliative care, is covered.

I. AML: CURRENT LANDSCAPE ing of the genetic underpinnings of leukemia is begin-


AND FUTURE DIRECTIONS ning to lead to a wide array of so-called targeted thera-
pies, many of which are in clinical development.
Richard M. Stone, MD* Despite current optimism, most patients with AML
will die of their disease. The basic therapeutic approach
Acute myeloid leukemia (AML) represents a group of to patients with AML has changed little over the last 20
clonal hematopoietic stem cell disorders in which both years. Nonetheless, before beginning to introduce novel
failure to differentiate and overproliferation in the stem therapies in the clinic, a thorough understanding of the
cell compartment result in accumulation of non-func-
tional cells termed myeloblasts. While the specific cause
for this biological abnormality in any individual pa- * Dana-Farber Cancer Institute, 44 Binney Street, Room D-840,
tient is usually unknown, the burgeoning understand- Boston MA 02115-6084

98 American Society of Hematology


current approach to treatment is required. The evolu- classically involves separate treatment phases. The first
tion of the classification system in AML from mor- consists of induction chemotherapy in which the goal
phology to cytogenetic/genetic-based reflects the rec- of myelosuppressive chemotherapy is to “empty” the
ognition of the importance of subtype-specific biology.1 bone marrow of all hematopoietic elements (both be-
The two major prognostic factors in newly diagnosed nign and malignant) and to allow repopulation of the
AML, patient age and chromosome status, form the marrow with normal cells, thereby yielding remission
basis of important treatment decisions. Recently, sev- (< 5% marrow blasts). The primacy of the standard
eral molecularly based prognostic factors, such as the regimen of 3 days of an anthracycline and 7 days of
adverse impact of an FLT3 tyrosine kinase gene length cytarabine has not been definitely altered despite clini-
(or ITD) mutation (repeat of 3–30 amino acids in the cal trials that have substituted alternative anthracyclines
juxtamembrane region,2 leading to constitutive activa- such as idarubicin, added or substituted high-dose
tion) and duplication of the MLL gene on the long arm cytarabine, or added etoposide. Recent trials by the
of chromosome 113 have been described; however, the Cancer and Leukemia Group B (CALGB) have em-
impact of such findings on treatment decisions remains ployed high doses of daunorubicin and etoposide.4 While
unclear. Patients with treatment-related AML require tolerable, whether such higher doses offer disease-free
special consideration because, in the absence of bal- or overall survival benefits remains to be proven by
anced translocations known to carry a favorable prog- ongoing clinical trials (Table 1). Given the high (75%–
nostic impact, such patients fare poorly with standard 80%) complete remission rate typically achieved in
approaches. The management of AML in older adults, younger adults with standard chemotherapy, an ex-
who have a highly inferior prognosis and an increased tremely large trial or a highly effective agent would be
rate of treatment morbidity and mortality, is highlighted required to show superiority over conventional chemo-
in Section III. therapy. Most of the trials that have reported an im-
The approach to adults aged 18–60 years with AML provement with a new chemotherapeutic agent or with

Table 1. Selected clinical trials in acute myeloid leukemia (AML).

US Intergroup APL: ATRA/daunorubicin/ara-C induction, followed by As203 + daunorubicin/ATRA x 2 vs daunorubicin/


ATRA x 2 consolidation, followed by ATRA/6MP/methotrexate vs ATRA x 1 yr for maintenance.
AML (age > 70 years and not a candidate for chemotherapy): Oral tipifarnib (R115777) at 2 doses and 2
schedules
CALGB AML (age > 60): dauno/ara-C vs dauno/ara-C/oblimersen
AML (age < 60): dauno/ara-C/etoposide, followed by post-chemotherapy/PBSCT, followed by IL-2 vs observation
ECOG AML (age > 60): dauno/ara-C ± MDR modulator (LY335979)
AML (age < 60): daunorubicin (45 mg/m2 x 3 d)/ara-C vs daunorubicin (90 mg/m2 x 3 d)/ara-C, followed by
± gemtuzumab ozogamicin (GO) prior to autoPBSCT (if no sib donor)
SWOG AML (age > 55): continuous infusion daunorubicin/ara-C ± cyclosporine A followed by assignment to mini-allo BMT
(if HLA-matched sibling donor)
AML (age < 55): daunorubicin/ara-C vs daunorubicin/ara-C + GO
EORTC AML (age > 60): ida/ara-C vs ida/ara-C/GO
AML (age < 60): ida/ara-C vs ida/high dose ara-C, followed by intensive consolidation/allogeneic BMT,
followed by IL-2
HOVON AML (age > 60): dauno (45 mg/m2)/ara-C vs dauno (90 mg/ m2)/ara-C f/b intermediate dose ara-C,
followed by GO x 4 vs observation
AML (age < 60): ida/ara-C ± G-CSF vs ida/high-dose ara-C ± G-CSF, followed by mitoxantrone/etoposide
(good risk) or Mito/etop vs autoBMT or alloBMT (if sibling donor)
MRC APL: ATRA/dauno/high-dose ara-C/6-thioguanine (MRC) vs ATRA/ida (Spanish), followed by MRC or Spanish
chemo ± GO
AML (age > 60): intensive chemo (dauno/ara-c at various doses) vs non-intensive chemo (hydroxyurea/
low dose ara-C ± ATRA)
AML (age < 60): dauno/high dose ara-C/6-thioguanine vs fludarabine/ara-C/G-CSF/ida +/- GO, followed by
allo BMT (if sibling; and will be nonmyeloablative if > 50, ‘full’ if under 35 years old)

Abbreviations: APL, acute promyelocytic leukemia; ATRA, all-trans retinoic acid; ara-C, cytosine arabinoside; 6MP, 6-mercapto-
purine; AML, acute myeloid leukemia; PBSCT, peripheral blood stem cell transplantation; IL, interleukin; MDR, multidrug resistance;
BMT, bone marrow transplantation; G-CSF, granulocyte colony-stimulating factor

Hematology 2004 99
a higher dose of a standard drug during induction have response (CR) has proven useful. Few patients who
documented a disease-free survival benefit, implying achieve polymerase chain reaction (PCR)-negative sta-
that such remissions yield a lower disease burden. How- tus after postremission chemotherapy relapse; whereas
ever, the inability to translate such benefits into overall those with persistently detectable PML-RARα fusion
survival improvements suggests a limited ability to af- transcripts have a 25% risk of relapse,10 yet whether
fect a major difference in the depth of remission. such relapses can be prevented with additional therapy
It is definitely clear that once remission is achieved, remains unclear. While the cure rate in APL with “stan-
additional therapy is required to reduce the undetect- dard” therapy is favorable (60%–70%), the problems
able burden of leukemic cells to a level low enough of secondary myelodysplasia11 and late central nervous
that long-term disease-free survival (i.e., cure) might system (CNS) relapses8 have recently been described.
be possible. The most effective anti-leukemic approach In summary, the young adult who presents with
is allogeneic stem cell transplantation. However, this AML should have studies performed on leukemic cells
technique carries a high degree of initial mortality and leading to morphologic and cytogenetic classification.
a significant degree of long-term morbidity in the form Those with non-APL AML should receive induction
of chronic graft-versus-host disease (GVHD), tending therapy with 3 days of an anthracyline and 7 days of
to offset the low likelihood of disease relapse. Chemo- infusional cytarabine followed by risk-adapted post-
therapy-based approaches with or without autologous remission therapy [intensive chemotherapy for those
stem cell rescue can be performed relatively safely, but with inv 16 or t(8;21)], allogeneic transplant for those
there remains a high chance for disease recurrence. Some with high-risk cytogenetics, and either intensive che-
of the patients who relapse after chemotherapy or even motherapy, high-dose chemotherapy with autologous
high-dose chemotherapy with autologous stem cell res- stem cell rescue or sibling-matched allogeneic trans-
cue can be salvaged with an allogeneic transplant per- plant for the remainder. That a “standard approach” to
formed in early relapse or second remission. The role the treatment of AML in the 18–60 year old patient can
of autologous and allogeneic (both standard and reduced be described is still compatible with the notion that all
intensity) strategies in the post-remission management of should be referred for a clinical trial. Many questions
AML patients with various risks of disease relapse based need to be answered, even for those with so-called ‘fa-
on karyotype at diagnosis is discussed in Section II. vorable’ prognoses, that usually involve the addition of
Specific therapies have proven benefit for small a new agent onto a backbone of “standard” therapy.
subsets of patients defined by recurring cytogenetic ab- The ideal candidates for investigational therapy, many
normalities. Post-remission chemotherapy with high- of whom are described subsequently, with a single agent
dose cytarabine is generally accepted as the best ap- are those likely to fare poorly with induction chemo-
proach for the 15% of patients with favorable progno- therapy, including older adults and those with disease
sis chromosome abnormalities [e.g., t(8:21); or relapse less than a year from diagnosis. While many
inv(16)].5 While the optimal number of such cycles re- different chemotherapy regimens exist for AML in re-
mains to be defined, at least three are probably required.6 lapse, the choice of regimen is less important than the
Whether the high-dose cytarabine or repetitive cycles duration of first remission. Those who relapse more
of intensive chemotherapy is the critical factor remains than one year after diagnosis have good chance to achieve
debatable; however, a cure rate of 60%–70% with che- a second remission after administration of the original
motherapy suggests that the more risky strategy of al- induction regimen, or at least one of similar intensity.
logeneic transplant should be reserved for early relapse However a second remission is achieved, the only post-
or second complete remission in this subset of patients. remission therapy with significant utility is allogeneic
Patients who present with acute promyelocytic leuke- transplant if possible, or high-dose chemotherapy with
mia (APL; another 8%–12% of patients) should be autologous stem cell rescue.
treated with all-trans retinoic acid (ATRA) and an
anthracycline in induction.7,8 Although many still em- Developmental Therapeutics in AML
ploy cytarabine in the induction and postremission man- The increased understanding of the pathophysiology of
agement of APL patients, its use is probably not neces- AML has led to the development of a host of new so-
sary.7 ATRA/anthracycline-based postremission therapy called targeted therapies. The success of imatinib in the
should be augmented with a year of maintenance therapy, treatment of chronic myeloid leukemia (CML) and other
not generally thought effective in other AML subtypes, diseases pathophysiologically based on the constitutive
with ATRA, probably in combination with oral anti- activation of a tyrosine kinase that is inhibited by the
metabolites.9 APL is the one subtype of APL in which drug has spurred the search for similarly effective agents
molecular monitoring after achievement of complete in AML. Table 2 lists some of the newer therapies ac-

100 American Society of Hematology


Table 2. Categories of novel therapies for acute myeloid with untreated first relapse who had, in most cases, a
leukemia (AML).
first remission of 6 months or longer.13 Disappointment
with this drug due to its low single-agent efficacy in
Drug-resistance modifiers treatment of patients with refractory AML as well as its
Proteosome inhibitors association with hepatic veno-occlusive disease when
Pro-apoptotic approaches administered proximal to or following a bone marrow
Signal transduction inhibitors transplant14 has dampened enthusiasm. Nonetheless,
“RAS”-targeted (e.g., farnesyl transferase inhibitors) based on the apparent safety of combining this agent
Tyrosine kinase targeted (e.g., FLT3, c-kit) with chemotherapy in newly diagnosed patients,15 im-
Downstream signal inhibitors portant studies are now underway to determine if
Immunotherapeutic approaches gemtuzumab ozogamicin combined with induction or
Antigens known consolidation chemotherapy, or as a single agent in the
anti-CD33 post-remission setting in older adults might lead to a
anti-GM-CSF receptor better outcome (Table 1).
Antigens unknown
stimulate immune system (IL-2, GM-CSF) Agents Not Expected to Be Effective as Single Drugs
present tumor antigens effectively
dendritic cell fusion
Drug-resistant modulators
The notion that blasts from AML patients, and particu-
transfer hematopoietic growth factor genes
larly from older individuals, are likely to express genes
capable of mediating drug resistance, most notably the
Abbreviations: IL, interleukin; GM-CSF, granulocyte-macroph-
age colony-stimulating factor GP170 drug efflux pump, has prompted clinical trials
with so-called reversing agents that inhibit this func-
tion. Since many of these agents also inhibit the me-
tabolism of chemotherapeutic drugs like anthracyclines,
cording to their proposed mechanism of action. Whether pharmacokinetic considerations are important. Most of
any of these will prove to alter the natural history of the trials that have compared chemotherapy with or
AML when used either alone or in combination with without a drug-resistant modulator (DRM) have shown
each other or with standard chemotherapy remains to no benefit or have been terminated early due to excess
be determined. That there are so many therapies in de- toxicity.16 However, a randomized clinical trial involv-
velopment is certainly positive; the challenge posed by ing cyclosporine A as a reversing agent has been posi-
this surfeit in a relatively rare disease for which fairly tive,17 prompting the Southwest Oncology Group to treat
effective therapy already exists remains daunting. Im- older adults with chemotherapy (using a novel regimen
munological therapies in AML based on enhancement including continuous infusion daunorubicin) with and
of the host immunity or elegant strategies of making without cyclosporine A. Secondly, trials with newer
AML cells “visible” to the immune system are men- DRMs that do not have pharmacological effects on che-
tioned in Section II. motherapy are also underway (Table 1).
Two agents have recently been approved for use in A problem with these drugs as well as with many
AML: arsenic trioxide12 and gemtuzumab ozogamicin.13 novel approaches is that the mechanism of resistance is
Their place in the therapeutic armamentarium is now likely to be pleiotropic. The inability to initiate cell
being better defined. Both could be considered models death in response to chemotherapy is another potential
for other new therapies, because they were shown to be mechanism of resistance. Overexpression of the bcl-2
effective (in highly varying degrees) in relapsed pa- anti-apoptotic gene has been associated with poor prog-
tients. Post-approval studies are being done to define nosis in AML patients. Preclinical data documenting
their role in newly diagnosed patients to increase their synergism between chemotherapy and the antisense 18-
overall impact. Arsenic trioxide leads to a four-log re- mer oligonucleotide oblimersen (G3139) have prompted
duction in the acute promyelocytic leukemia disease Phase I clinical trials documenting the safety of induc-
burden in heavily pre-treated relapsed patients12 and is tion chemotherapy with this agent.18 The CALGB has
now being evaluated to consolidate first remissions begun a randomized a trial in older adults that com-
(Table 1). Gemtuzumab ozogamicin is an anti-CD33 pares chemotherapy alone to chemotherapy adminis-
immunotoxin conjugate that was approved based on a tered with oblimersen (Table 1). Synergistic antileuke-
30% response rate (half of whom were patients who mic activity between chemotherapy agents and the
never fully recovered their platelet count) in patients proteosome inhibitor bortezomib19 has spurred the clini-

Hematology 2004 101


cal development of this agent in conjunction with che- these differentiating agents in combination with agents
motherapy in AML. that inhibit mitogenic signals.
The use of hematopoietic growth factors (HGFs)
in AML has received much attention. Once concerns Signal Transduction Inhibition
diminished that growth factors would enhance disease The mechanism by which cells receive and transmit
resistance via their known ability to stimulate leukemia mitogenic signals is complex and involves the coordi-
cells to enter S-phase, a host of clinical trials examined nated action of many different proteins, commonly rep-
the ability of HGFs to shorten the period of neutrope- resented by so-called signal transduction cascades. In-
nia and reduce infectious mortality. While the duration sofar as such cascades might be overactive in leukemia
of neutropenia was reduced by several days when G- cells compared to normal hematopoietic cells, a thera-
CSF or granulocyte-macrophage colony-stimulating peutic opportunity is provided. Moreover, an activat-
factor (GM-CSF) was used in the postchemotherapy ing mutation in one of the proteins responsible for trans-
period, the inability of these agents to alter the nadir, mitting proliferative signals in AML defines a poten-
allay mucositis, or decrease the death rate diminished tial target. Mutations in one of the RAS family pro-
their utility. Perhaps more interesting is the use of these teins, 21-KD guanine-nucleotide binding proteins, have
agents before or during chemotherapy in an effort to been described in 10%–50% of AML patients. Such
increase the S-phase fraction of cells, thereby using them activating mutations are thought to allow autonomous
as chemosensitizing agents. Although most of the trials growth. RAS proteins require several post-translational
using HGFs in this fashion have been disappointing, a modification steps including addition of a farnesyl lipid
recent randomized study in which G-CSF was used be- moiety that allows translocation to the plasma mem-
fore and during chemotherapy, yielding a disease free brane and activation. Farnesyl transferase inhibitors
survival benefit,20 has prompted renewed interest in this (FTIs) have been shown to inhibit the growth of mu-
strategy. At the present time the use of G- or GM-CSF tant RAS-transformed cell lines. The FTI in furthest
can be considered optional using supportive care guide- development in AML, tipifarnib (R115777), was asso-
lines for neutropenic fever while their use as chemo- ciated with responses in advanced-stage patients in a
therapy-enhancing agents needs to be explored further Phase I trial.22 Despite disappointing results in an inter-
before routine use is warranted. national Phase II trial in refractory/relapsed patients
(although RAS mutations were not required), the drug
Agents That Promote Differentiation has been administered to over 100 chemotherapy-naïve
Almost a quarter century has elapsed since clinical tri- AML patients. A preliminary report documented a 20%
als suggested that low-dose cytarabine’s effect in AML complete remission rate in this group of poor progno-
and myelodysplasia resulted from hematopoietic cell sis, largely older patients who received this oral agent
differentiation. In the ensuing years, the enthusiasm for as their initial therapy.23 These exciting results, if con-
low-dose cytarabine as an effective therapy that induces firmed, might provide a new and less toxic therapy com-
maturation of AML cells has waned. However, an in- pared to chemotherapy for older adults with AML. FTIs
creased understanding of how the transcription of dif- are also being evaluated in younger patients in earlier
ferentiation-associated genes is regulated has prompted disease states and in combination with chemotherapy.
the exploration of new and potentially effective thera- An active area of current research in AML is the devel-
pies. Among the many biochemical events that must opment of agents known as FLT3 inhibitors. FLT3 is a
occur as a cell changes from a stem cell to a mature transmembrane tyrosine kinase. Approximately 30% of
functional blood cell are removal of methyl groups from AML patients can be shown to have an activating mu-
DNA bases and addition of acetyl groups to the histone tation that generally carries a poor prognosis.2 The ac-
DNA protein coat. Such effects change the conforma- tivating mutation may be a duplication of between 3
tion of the DNA and allow transcription of certain genes. and 60 amino acids in the juxtamembrane region or,
The DNA hypomethylating agent azacitidine21 was re- less commonly, a point mutation in the activation loop.2
cently approved by the US Food and Drug Administra- Activating mutations of the FLT3 tyrosine kinase trans-
tion (FDA) for use in all subtypes of myelodysplastic form leukemic cell lines into growth factor indepen-
syndrome. Many clinical trials are now underway with dence and cause a fatal myeloproliferative syndrome in
so-called histone deacetylase inhibitors, although out- a murine bone marrow transplant model. Small mol-
come data are sparse. Given the general hypothesis that ecules capable of inhibiting FLT3 enzyme activity can
AML results from a combination of over proliferation selectively kill such transformed cell lines and improve
of the stem cell compartment as well as failure to dif- survival in the murine model.2 Thus, the preclinical
ferentiate, it is rational to consider the future use of data supporting the development of these drugs as thera-

102 American Society of Hematology


Table 3. FLT3 inhibitors in clinical development (modified from Wadleigh et al29).

Tyrosine Receptor
Kinase Inhibitor Clinical Trials/
Inhibitor Class Activity† FLT3 IC50 ‡ Comments Toxicity
PKC-412 Benzoylstaurosporine PKC Phase II: AML with/without FLT3-ITD Nausea,
PDGFR In FLT 3 mut pts (n = 20), 35% emesis, fatigue
KDR significant reduction in blast
KIT count25
FLT3 528 nM
ABL
CEP-701 Indolocarbazole FLT3 2–3 nM Phase II: AML with FLT3-ITD Nausea,
TRKA Several pts had reduced blast emesis, fatigue
KDR counts, autophosphorylation
PKC inhibited26
PDGFR
EGFR
MLN-518 Piperazinyl quinazoline KIT Phase I: AML/MDS with/without Generalized
PDGFR FLT3-ITD weakness,
FLT3 170–220 nM Phase II: AML with FLT3-ITD fatigue, nausea
FMS A few pts with FLT3 ITD treated and vomiting
at higher doses had biological
response 27
SU5416 Indolinone FLT3 250 nM Phase II: Refractory Fatigue,
KDR AML/MDS/MPD/MM nausea, sepsis
KIT Phase II: Refractory AML (c-KIT and bone pain
positive). No responses in
FLT3 ITD pts.24
† Receptor inhibitor activity in descending order of potency
‡ FLT3 autophosphorylation in vitro 1
Abbreviations: AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; MPD, myeloproliferative disease; MM, multiple
myeloma

peutic agents in AML is at least as strong as that used to summarized in Table 3. Biological responses, demon-
support clinical trials with imatinib in patients with strated by a major reduction in peripheral blast count,
CML. There are two major differences between imat- have occurred with each of these drugs. However, they
inib’s development and that of the FLT3 inhibitors: 1) have minimal ability to reduce the bone marrow blast
CML in chronic phase is probably based solely on the count, and therefore, complete remissions have occurred
activation of bcr-abl, whereas AML is almost certainly in 1/42 reported patients. Moreover, the duration of
a “multi-hit” disease; 2) Multiple agents and drug com- response has been brief. Although much more work
panies are developing FLT3 inhibitors. However, just with these oral agents needs to be done, initial impres-
as was the case for imatinib, these drugs have a spec- sions suggest that (1) the multiplicity of genetic lesions
trum of activity beyond FLT3 inhibition alone. For in the typical AML cell may be problematic for expect-
example, PKC-412 (N-benzoylstaurosporine) inhibits ing these agents to work alone; (2) pharmacokinetic
FLT3 as well as protein kinase C and the vascular en- issues require prolonged therapeutic drug levels and the
dothelial growth factor receptor. Such a wide spec- ability to get to the target leukemia progenitor cell; and
trum of activity could have positive or negative conse- (3) the contribution of alternative enzymes and path-
quences. SU5416, which inhibits FLT3 reasonably po- ways are important.28
tently, was developed as a c-kit inhibitor. The drug dem- Nonetheless, further development of these drugs at
onstrated modest activity in AML, but no activity was different doses and schedules and in combination with
observed in the 7 patients who retrospectively were other signal transduction inhibitors and/or chemotherapy
found to have an activating mutation of FLT3 in their is warranted. Other tyrosine kinase inhibitors, includ-
myeloblasts.24 Minor responses have been observed in ing those that inhibit the vascular endothelial growth
patients without known FLT3 mutations who have re- factor receptor, are also in clinical trials in AML.29
ceived PKC-412. Available clinical data from early tri-
als with the three drugs specifically developed as FLT3
inhibitors, PKC-412,25 CEP-701,26 and MLN-518,27 are

Hematology 2004 103


II. THE ROLE OF AUTOLOGOUS AND ALLOGENEIC median of 28 days versus 47 days for platelets, respec-
(“FULL” AND “MINI”) STEM CELL tively), which decreases the risk for bacterial infections
TRANSPLANTATION IN AML in the early post-transplant period.1 Despite initial con-
cerns that a 10-fold increase in the number of CD3+
Margaret R. O’Donnell, MD* cells in the PBSC graft would lead to higher incidence
of severe GVHD, this has not been demonstrated in
Hematopoietic cell transplantation (HCT) is an effec- clinical trials. Several studies have shown no signifi-
tive therapy for AML. Obstacles to broad applicability cant difference in the incidence of acute GVHD; how-
of HCT therapy for the majority of patients with AML ever, higher rates of late onset chronic GVHD (beyond
have included inability to control leukemia with pri- 6 months) have been reported.2 In both a large random-
mary induction therapy, lack of suitable hematopoietic ized trial and a retrospective review of International
cell donors, toxicities of HCT conditioning regimens Bone Marrow Transplant Registry (IBMTR) data, PBSC
and long-term complications of the transplant proce- grafts were associated with improved relapse-free sur-
dure. While all antileukemia therapies are complicated vival in patients with leukemia beyond first remission
by the problems of chemotherapy-related toxicities and (CR1).1,3,4 In addition to producing a HPC product that
disease relapse, allogeneic HCT also exposes patients results in more rapid engraftment, G-CSF exposure also
to the risks of potential failure of engraftment; organ shifts donor T cells to Type 2 cytokine secretion
toxicities caused by GVHD and prolonged immuno- (interleukin [IL]-4 and IL-10) and downregulates Type
suppression with its attendant risks of post-HCT infec- 1 (IL-2 and interferon γ ) cytokine production, phe-
tious complications. nomena which may be associated with less severe acute
In the last decade, several developments have made GVHD.
allogeneic HCT a more “user-friendly” treatment mo- Improvements in post-transplant supportive care
dality. Expansion of the pool of donors in national and and the development of newer immunosuppressive
international registries as well as the establishment of agents have also had an impact on transplant-related
cord blood banks has vastly increased the likelihood of toxicities. More refined PCR-based screening for cy-
success in identifying a suitable HLA match for pa- tomegalovirus reactivation allows pre-emptive therapy
tients who lack matched family donors, although cord tailored to viral load. Newer less toxic antifungal agents
blood HCT is still utilized primarily for pediatric re- such as voriconazole and caspofungin have decreased
cipients. The refinement of tissue typing using molecu- early mycotic infections and make it feasible to pro-
lar probes permits better matching of unrelated donor- vide long-term fungal prophylaxis in patients at high
recipient pairs, thereby decreasing the risks of both graft risk due to chronic GVHD. Newer immunosuppressive
rejection and GVHD. agents, such as sirolimus and mycophenolate mofetil
There has also been a recent change in the type of (MMF) are being incorporated into GVHD prophylac-
hematopoietic progenitor cell (HPC) product used for tic regimens to decrease the incidence of acute and chronic
graft reconstitution. Autologous transplant procedures GVHD in high-risk populations including unrelated do-
had transitioned from marrow to cytokine-primed (G- nor recipients and older (> 50 years) patients.5,6
CSF and/or GM-CSF) peripheral blood stem cells However, the change that may have the greatest
(PBSC) for ease of collection and rapid engraftment by impact on AML treatment is the advent of reduced in-
the early 1990s. However, expectation of higher rates tensity or nonmyeloablative HCT. Reduced-intensity
of GVHD due to the 10-fold increase in T cells in PBSC HCT relies upon the graft-versus-leukemia effect of
products delayed their use in allogeneic settings until the allograft rather than the direct tumoricidal activity
the latter-half of the 1990s. In 2003, National Marrow of the conditioning regimen. Because of the paucity of
Donor Program (NMDP) statistics showed 60% of un- any direct antileukemic effect of the conditioning regi-
related donor products collected for transplants for AML mens, the truly nonmyeloablative (NMABT) regimens
were PBSC. Collection via apheresis is physically easier can only be used in patients with low volumes of dis-
for many donors. The 4- to 5-fold higher number of ease, whereas the reduced-intensity conditioning regi-
CD34+ cells in a PBSC product facilitates more rapid mens, which usually contain fludarabine and some alky-
engraftment in comparison with a marrow product (me- lating agent, do have direct antileukemia activity and
dian of 17 days versus 24 days for granulocytes and can be used with more extensive disease. The shortened
duration of cytopenias and the minimal mucosal toxic-
ity of the newer reduced-intensity conditioning regi-
* City of Hope National Medical Center, 1500 East Duarte mens provide a reasonably safe transplant option for
Road, Duarte CA 91010 patients two decades older than the population that was

104 American Society of Hematology


treated with traditional fully ablative high-dose chemo- salvage therapy for patients in second remission. To
radiotherapy regimens. minimize both the total body leukemic burden and the
contamination of the PBSC product, current practice is
Choosing the Type and Timing of HCT to administer one to two cycles of a high-dose
(To Transplant or Not to Transplant) cytarabine-based consolidation to achieve “in vivo purg-
The possible need for HCT as a component of future ing” prior to collection of G-CSF–mobilized PBSC.
therapy should be acknowledged at diagnosis in AML. To assure that an adequate hematopoietic progenitor
For all patients under age 56 who do not have signifi- cell product of at least 5 × 106 CD34+ cells/kg recipient
cant co-morbid conditions, HLA typing should be part body weight can be collected, it is recommended that
of the patient’s initial evaluation, and family typing no more than 2 cycles of consolidation occur before
should be initiated when the cytogenetic information is PBSC collection. While the nonhematologic toxicities
available. Only a minority of patients will have a fa- of the conditioning regimens used for autologous HCT
vorable karotype that would obviate consideration of are significantly greater than those seen with high-dose
allogeneic HCT either as part of consolidation or for cytarabine consolidation, the hematologic recovery is
salvage of resistant disease. For patients with poor-risk quite rapid (8–11 days) following infusion of autolo-
cytogenetics, antecedent myelodysplasia or therapy- gous G-CSF–mobilized PBSC. The combined treat-
related AML, an unrelated donor search should be ment-related mortality (TRM) for one cycle of high-
promptly instituted in patients lacking a family donor. dose cytarabine-based consolidation followed by 1200
Early donor identification allows optimal timing for cGy fractionated total body irradiation (FTBI), etopo-
HCT as consolidation or salvage therapy. In older pa- side and cyclophosphamide (Cy) and autologous HCT
tients (56–70 years), however, it is reasonable to delay at our institution ranged between 3% and 6% in three
HLA typing of family members until remission is sequential trials inclusive of patients up to age 60. At-
achieved and the patient has recovered a good perfor- trition between consolidation and autologous transplant
mance status since there is substantial attrition in the ranged from 18% to 24% due to relapse, inadequate
rank of candidates for the investigational use of reduced- HPC collection or treatment-related toxicities such as
intensity or nonmyeloablative HCT as consolidation due invasive fungal infection and cytarabine-induced neuro-
to failure to achieve a remission or decline of func- toxicity, making this a reasonable consolidation option
tional status as a consequence of induction therapy. for most patients up to age 60.7
The decision to proceed to either autologous or allo- Over the last 15 years, the upper age limit consid-
geneic HCT is strongly influenced by the following ered to be acceptable for a fully ablative allogeneic HCT
factors: expectations of outcome with the prevailing has advanced from 40 to “robust” 60 year olds (perhaps
non-HCT conventional therapy, the effectiveness of sal- in response to the maturing age of the transplanters them-
vage options for disease relapse, the toxicities of trans- selves as much as to improvements in supportive care
plant including long-term complication of infertility and GVHD prophylaxis).8 If an HLA-matched donor
and second malignancy, and the individual patient’s co- has been identified while the patient was receiving in-
morbid conditions. The patient’s cytogenetic risk group duction therapy, there is no advantage to administering
and time to achieve remission are currently the major postremission chemotherapy prior to allogeneic HCT.9
disease-specific factors to be considered in opting for GVHD and transplant-related toxicity remain the ma-
HCT as consolidation therapy, whereas donor availability jor obstacles to successful outcome with allogeneic HCT.
and the impact of recipient performance status on the The introduction of two new immunomodulatory drugs
ability to tolerate the rigors of even a reduced-intensity for GVHD prophylaxis may have an important impact
HCT are important factors to consider at relapse. While on both these problems. Investigators from the Dana-
there are several studies with long-term outcome data Farber recently reported a Phase II trial in which
comparing myeloablative conditioning regimens fol- sirolimus was substituted for methotrexate (MTX) in a
lowed by either autologous or allogeneic HCT, the fol- tacrolimus-based GVHD prophylaxis regimen for PBSC
low-up data on reduced intensity HCT is much shorter HCT utilizing a fully myeloablative conditioning regi-
and the patient populations are not comparable. men.5 Engraftment was prompt and only 3/30 (10%)
patients developed acute GVHD (all Grade II) com-
Comparisons of Autologous Transplantation, pared to the expected incidence of 35%–40% acute
Allogeneic and Reduced-Intensity HCT GVHD in sibling-donor HCT with tacrolimus/MTX.
Autologous transplantation has been used primarily as TRM was only 6% at 1 year compared to 25%–30% in
a component of consolidation following initial remis- most traditional allogeneic studies. Chronic GVHD de-
sion; less commonly, it can be used as a component of veloped in 40% of the study patients. Toxicities in-

Hematology 2004 105


cluded significant hyperlipidemia, thrombotic micro- still being performed for AML patients beyond CR1.
angiopathy and a 10% incidence of veno-occlusive dis- The foremost advantage of the nonmyeloablative
ease primarily in patients with recent exposure to or reduced-intensity HCT is the reduction of TRM. Mu-
gemtuzumab ozogamicin. Other studies of the substitu- cositis in the immediate post-HCT period is much re-
tion of mycophenolate mofetil (MMF) for MTX in duced due to the low antiproliferative activity of the
GVHD prophylaxis regimens have demonstrated de- conditioning regimen and the substitution of MMF for
creased regimen-related toxicities of severe mucositis and MTX in the GVHD prophylaxis regimen. The reduc-
interstitial pneumonias, as well as more rapid engraft- tion in mucosal disruption led to a decreased incidence
ment compared to a MTX-containing regimen (14–16 of acute (12%) and chronic (19%) GVHD for reduced-
days versus 19–22 days for neutrophil engraftment).10,11 intensity fludarabine/melphalan versus 36% acute and
Improvements in molecular matching for unrelated 40% chronic GVHD for patients receiving ablative sib-
donor-recipient pairs have significantly improved out- ling HCT in an MD Anderson Cancer Center study.11
comes for this group, although there continues to be an Infectious complications are also reduced, with a 9%
approximately 10% higher incidence of TRM when sib- incidence of bacteremia at day 100 for nonmyeloablative
ling allogeneic HCT is compared to allogeneic HCT conditioning versus 27% for standard HCT and a dif-
from molecularly matched unrelated donors for patients ference in day 100 survivals of 93% versus 81%, re-
in CR1. Because of the perception of higher TRM, the spectively, in a case-matched control series from Se-
profile of patients who undergo unrelated donor HCT attle.12 However, other investigators have reported higher
in CR1 is skewed toward those with poorer risk cytoge- rates of CMV reactivation before day 100 and persis-
netics, prolonged time to achieve a remission, or a his- tent risk of bacteremia and fungal infections late post-
tory of antecedent myelodysplasia or therapy-related HCT with nonmyeloablative regimens because of the
AML. The majority of unrelated donor transplants are prolonged immunosuppressive effects of fludarabine

Table 4. Treatment outcomes and toxicity for autologous and allogeneic hematopoietic cell transplantation (HCT) for
consolidation or salvage therapy.

Reduced-Intensity
Autologous HCT Allo HCT (Sib) AlloHCT/MUD AlloHCT + MUD
CONSOLIDATION (CR1)
Cytogenetic risk
t(15;17) No role No role No role No role
t(8;21)inv(16) DFS 60%–80%* DFS 65%
TRM 4%–8% TRM 18% = No Role No role No role
Intermediate DFS 42%–55% DFS 48%–62% Insufficient data for
TRM 4%–6% TRM 16%–20% nonmyeloablative
Poor DFS 18%–25% DFS 35%–45% 5-yr DFS 30%–40% Reduced intensity DFS 50%
TRM 4%–8% TRM 18%–20% TRM 30% for older AML CR1 at 2 yr
SALVAGE
CR2 DFS 30% overall DFS 40% Pediatric 40% 2-yr DFS 40%–50%reduced
DFS 60%–80% Adult 5-yr DFS 30% intensity
for t(15;17) TRM 30%
Relapse Not an option unless DFS 20%–30% Pediatric DFS 20% 2-yr DFS 10%–30%
“back-up” product from Adult 5-yr DFS depending on the volume
CR1 available 10%–15% of residual disease
Induction Failure No Role DFS 30%–40% (3 yrs) DFS 20%–30%** 1-yr DFS 15%–30% (sibling)
(20% for untreated
secondary AML)

* Because of high salvage rate and long-term sequelae, many would reserve autologous HCT for relapse for patients with
favorable cytogenetics.
** Most of these patients represent patients with high grade myelodysplastic syndrome (MDS) in which an unrelated donor search
was initiated prior to leukemic evolution.
*** DFS interval will be 5 years unless otherwise noted
Abbreviations: AML, acute myeloid leukemia; DFS, disease-free survival; TRM, treatment-related mortality; HCT, hematopoietic cell
transplantation; MUD, matched unrelated donor; allo, allogeneic; CR, complete response

106 American Society of Hematology


even in the absence of GVHD. a molecularly negative product can be collected.17 How-
Table 4 provides a schematic of current thinking ever, for patients with persistence of the PML/RAR gene
on the role of the various types of HCT in the therapy product, allogeneic transplantation should be consid-
of AML. A caveat for interpretation of all consolida- ered since the relapse rate following infusion of a mo-
tion trials in which HCT is a component is the exist- lecularly positive product exceeds 85%.
ence of selection bias. There have been several large Relapse free-survivals of 60%–83% have been re-
national or co-operative group trials conducted between ported for patients with t(8;21) or inv(16) using intent-
1986 and 1995 that tried to answer the question of the to-treat analysis from the start of high-dose cytarabine
role of transplantation in consolidation in younger pa- consolidation through autologous HCT with relapse rates
tients with AML.13-15 Patients with sibling donors were of 15%–20% post-HCT and TRM of 4%–8% over-
assigned to the allogeneic marrow transplantation and all.18,19 While these disease-free survival (DFS) rates
the remainder were randomized, between a variety of are higher than the DFS seen with multiple cycles of
consolidation (2–4 cycles) chemotherapy versus autolo- high-dose cytarabine-based consolidation and the time
gous marrow transplant after 1–4 cycles of consolida- to completion of treatment may be shorter, long-term
tion. The conclusions ranged from no survival advan- toxicities associated with autologous HCT need to be
tage to either form of HCT compared with consolida- factored into the decision to pursue autologous HCT in
tion chemotherapy in the initial analysis of the US Na- individual patients. Many with expertise in AML would
tional Trial involving the Eastern Cooperative Oncol- reserve autologous HCT for relapse in patients with
ogy Group (ECOG), Southwest Oncology Group good risk cytogenetics. In patients who opt for conven-
(SWOG) and CALGB to a significant difference in dis- tional chemotherapy, one should consider cryopreserving
ease-free survival for both allogeneic (55%) and au- autologous PBSC as a “back-up” for salvage therapy in
tologous HCT (48%) compared to chemotherapy (30%) the event of relapse in patients who lack a histocompat-
in the European Organisation for Research and Treat- ible sibling. Autologous HCT can provide long-term
ment of Cancer (EORTC) trial (albeit with no overall salvage for 40% of patients who achieve a second re-
survival advantage as a high percentage [58%] of che- mission overall; factors that correlate with better out-
motherapy relapses were salvaged with transplant). come include cytogenetics and duration of first remis-
When the US National Trial was reanalyzed using sion.20 Sibling allogeneic HCT in CR1 in the group with
cytogentic risk groupings there were clear differences favorable cytogenetics had only 62% DFS with a 17%
in favor of HCT (allo and auto) for favorable risk and TRM in the EORTC/GIMEMA (Gruppo Italiano
for allogeneic in patients with poor risk cytogenetics.15 Malattie Ematologiche dell’ Adulto) trial based on in-
In all these trials as well as the MRC10 trial, there was tent-to-treat from time of remission that compared au-
considerable patient attrition from achieving remission tologous to allogeneic transplant during the time pe-
to initiating any form of consolidation.16 Fully one third riod of 1993–1999 (Figure 1A). The high TRM asso-
of younger (< 55 yr) patients received no documented ciated with allogeneic HCT indicates that allogeneic
form of consolidation; 50%–80% of patients actually HCT has no advantage over conventional chemotherapy
received the designated HCT treatment. While it is ap- or autologous HCT until the toxicity profile improves.19
propriate to compare treatment strategies on an intent-
to-treat basis from the time CR is achieved, the inter- Intermediate cytogenetics
position of multiple nontransplant therapies before HCT The European Bone Marrow Transplant (EBMT) group
may cause high attrition rates that provide a negative reported an overall 2-year leukemia-free survival of
bias, while analyses that start only at time of transplant 49% for 1040 patients who received autologous
may give an overly optimistic prediction of outcome. unpurged marrow transplant during the period of 1986–
1994.21 The TRM was 11%, with a median time to
Consolidation granulocyte and platelet recovery of 29 days and 42
days, respectively. Factors that influenced outcomes
Good-risk cytogenetics were time to achieve remission (1 vs 2 inductions) for
Given the high curability of acute promyelocytic leu- leukemia-free survival and older age (> 35 years) for
kemia (APL) with conventional chemotherapy, there is TRM. Karyotypic analysis was not included in that study.
no role for autologous or allogeneic HCT for patients In the EORTC/GIMEMA study, patients with normal
in CR1 who achieve a molecular remission by the karyotype who underwent autologous HCT had a 48%
completion of consolidation. Several series have dem- DFS with a 5% TRM compared to 45% DFS and 20%
onstrated that autologous HCT can produce durable sec- TRM in patients transplanted from a sibling donor.14 In
ond remissions in 75%–100% of the patients in whom a consortium study that included 128 patients up to age

Hematology 2004 107


Figure 1. Disease-free survival (DFS) from complete response (CR) for four prognostic cytogenetic risk groups—
EORTC/GIMEMA trial comparing sibling allogeneic versus autologous HCT for consolidation.
The cytogenetic risk groups are good risk (1A), normal (1B) and bad/very bad risk (1C). 1D captures the data for patients in whom
cytogenetics were unknown. Reprinted with permission from Blood. 2003;102,1237.

65 receiving autologous HCT in CR1, Linker et al re- Poor-risk cytogenetics


ported a 51% DFS for patients with intermediate risk A donor search for a related or unrelated donor should
cytogenetics, with a combined TRM of 3% for both be initiated as soon as the patient is identified as having
consolidation and transplant.18 Since TRM in alloge- poor-risk cytogenetics as this group may not achieve
neic recipients increases significantly with age, autolo- remission and may require allogeneic HCT for early
gous transplant may offer equivalent DFS with less mor- salvage. Patients with poor-risk cytogenetics are pro-
bidity for older patients (≥ 50 years) than standard portionally underrepresented in autologous transplant
myeloablative allogeneic transplant. Younger (≤ 40 series, accounting for less than 10% of patients because
years) patients with a sibling donor can expect a better remissions are more difficult to achieve and the recov-
relapse-free survival of 62% with a 15% relapse rate ering marrow may have underlying myelodysplasia,
and TRM of 15%–20%. Since TRM is 10%–18% higher which impairs the ability to collect adequate numbers
for unrelated-donor HCT using myeloablative condi- of stem cells with normal proliferative capacity. In most
tioning, a donor search is rarely initiated during CR1 series, the results of autologous HCT for patients in
for intermediate-risk patients in the absence of other this group are poor, with DFS of 20% or less.21 In con-
risk factors such as antecedent MDS. trast, sibling allogeneic transplant for this group results

108 American Society of Hematology


in a 40%–45% DFS for patients ≤45 years of age19 and (2) the likeliness of achieving a remission, which
(Figure 1C). Most unrelated donor transplants in CR1 in turn is based on cytogenetic risk group, duration of
are performed in patients with poor-risk cytogenetics. CR 1 and comorbid conditions. Data from EBMT,
Data from the NMDP demonstrated an overall 40% 5- IBMTR and the NMDP show DFS of 44% with sibling
year relapse-free survival for all AML patients in CR1 allografts and 30% with matched unrelated donor al-
transplanted between 1987 and 2001, which is compa- lografts at 5 years for patients transplanted in second
rable to the 44% reported by the EORTC for sibling remission and DFS of 35%–40% in sibling transplants
HCT of patients with poor-risk karyotypes. and 10% in unrelated donor transplants for patients with
Reported outcomes for reduced intensity transplants induction failures or HCT in relapse.24 It should be noted
in patients with AML in CR1 are based on small that the majority of these patients received marrow rather
samples. The group from the University of Michigan than PBSC. In the Seattle study comparing sibling mar-
treated 21 patients over age 55 (median 61 years) with row versus PBSC in patients with more advanced dis-
MDS or AML who had HLA-matched sibling donors ease, there was a significant difference in DFS (57% vs
with the reduced intensity regimen of fludarabine (Flu) 33%) using PBSC compared to marrow.1 Additional
and busulfan (Bu) with tacrolimus and MMF for GVHD data from the IBMTR also showed an advantage for
prophylaxis.22 Seven of 12 patients who were in CR at PBSC compared to marrow for patients transplanted in
time of transplant remain in remission with a median second remission.3 The reduced intensity regimens have
follow-up of 1 year (range 210–733 days). There were had a tremendous impact on TRM in the setting of sec-
3 relapses and 2 treatment-related deaths. All the pa- ond transplants or secondary MDS/AML following prior
tients with active disease died (5 relapse and 4 TRM) autologous HCT, increasing the DFS from 16% with
with a median survival of 108 days. Overall, the inci- standard Bu plus Cy conditioning to 40% with Flu/Bu
dence of acute GVHD was 38% with a maximum se- or Flu/Melphalan conditioning.25 The recent formation
verity of grade 2. TRM at day 100 for the whole group of the Blood and Marrow Transplant Clinical Trials
was 19% and was only 8% in the patients who did not Network (CTN) to facilitate the conduct of large scale
have active disease at transplant. comparative transplant trials should enable the trans-
The German Cooperative Transplant Study Group plant community to expeditiously define the role of
reported on 113 AML patients treated with Bu/Flu (93 newer reduced-intensity conditioning regimens and
patients) or Flu/TBI (20 patients) with a radiation dose GVHD prophylaxis regimens in exploiting the antileu-
of either 400 or 800 cGy. The reduced intensity regi- kemic effect of donor alloreactivity, minimizing trans-
mens were chosen for these patients because of age > 50, plant-related toxicities and broadening the applicabil-
co-morbid condition or prior HCT; the median age was ity of HCT as a curative treatment strategy for patients
51 and more than half the patients received unrelated with AML.
donor allografts.23 The minority of patients (22%) were
in CR1, and half the patients were in relapse at HCT. III. AML IN OLDER ADULTS: ARE WE LISTENING?
Five patients with relapse did not clear their disease
and 1 did not engraft. Acute GVHD (Grade 2–4) oc- Mikkael A. Sekeres, MD, MS*
curred in 42% of patients and chronic GVHD occurred
in 35% of patients at risk after day 100 with the major- AML is a disease of older adults. In the US, the median
ity of these cases being of limited extent. The 2-year age is 68 years and the age-adjusted population inci-
DFS was 50% for those in CR1, 40% for those in CR2 dence is 17.6 per 100,000 for people 65 years of age or
or 3, and 15% for those with relapsed disease. Causes older, compared with an incidence of 1.8 per 100,000
of death were evenly divided between relapse and treat- for people under the age of 65 years.1 Therefore, of the
ment-related toxicity (29 patients each). In addition to estimated 11,900 new AML diagnoses in the US in
remission status, performance status (≤70%) and do- 2004, over half will affect patients 60 years of age or
nor type (sibling versus unrelated donor) were predic- older, a population considered “elderly” in the leuke-
tive factors for event-free survival. mia literature.2-5 This number will only increase as the
population of US citizens 65 years or older, estimated
Salvage to be approximately 35 million in the year 2000, is
The majority of allogeneic transplants are performed
as salvage for patients who have relapsed after conven-
tional chemotherapy or autologous HCT. The decision * The Cleveland Clinic Foundation, Hematology and Medical
regarding reinduction prior to salvage transplant is in- Oncology, Desk R35, 9500 Euclid Avenue, Cleveland OH
fluenced by (1) the availability of an identified donor 44195

Hematology 2004 109


expected to double by the year 2030. older patients,8,9 though it is not clear that this finding
Older adults with AML, when compared to holds for patients older than 65 years.
younger patients with the same disease, have a poor
prognosis and represent a discrete population in terms Bone marrow biology
of disease biology, treatment-related complications, and In older adults, AML is more likely to arise from a
overall outcome. As a result, older patients require dis- proximal bone marrow stem cell disorder, such as
tinctive management approaches to determine whether myelodysplastic syndrome (MDS), and with leukemia-
standard treatment, investigational treatment, or low- specific abnormalities in more than one hematopoietic
dose therapy or palliative care is most appropriate. Par- cell lineage. This may explain the different disease be-
ticularly in this population, the tradeoff between po- havior in this group, as well as prolonged neutropenia
tential for cure or survival prolongation and quality of following chemotherapy.3 Older adults with AML also
life must be weighed carefully. are more likely to have reduced proliferative capacities
in normal hematopoietic stem cells, which may affect
Disease Biology blood count recovery following intensive chemotherapy.

Cytogenetics Drug-resistance genes


Older adults with AML have a lower incidence of fa- The expression of genes that mediate drug resistance
vorable chromosomal abnormalities [including the core occurs with increased frequency in this age cohort.
binding factor abnormalities, such as t(8;21) or abnor- MDR1, the so-called P-glycoprotein (gp170) chemo-
malities of chromosome 16, or the t(15;17) associated therapy efflux pump, was found in 71% of leukemic
with APL] and a higher incidence of unfavorable ab- blasts in subjects in a SWOG study of AML patients
normalities (including complex cytogenetics or abnor- over the age of 55 years, compared to a prevalence of
malities of chromosomes 5, 7, or 8) compared to 35% in younger AML patients.10 MDR1/P-glycopro-
younger adults with AML (Table 5).6-8 The rare older tein expression is associated with lower complete re-
patient fortunate enough to have good-risk cytogenetic mission (CR) rates and more chemo-resistant disease.
abnormalities may have a survival advantage over other
Prior stem cell insult
Older adults with AML are more likely to have a sec-
Table 5. Characteristics of older and younger adults with ondary leukemia arising from an antecedent MDS or
acute myeloid leukemia (AML).
from prior treatment with chemotherapy or radiation
Older AML Younger AML
therapy for another cancer. Patients with this type of
Characteristic PatientsA PatientsA AML are predisposed to having abnormalities in chro-
Population incidenceB 17.6 1.8 mosome 5 and/or 7.7 Secondary AML (AML that arose
Favorable cytogeneticsC after MDS, myeloproliferative disorders, and therapies
t(8;21) 2% 9% of malignancy) comprises 24%–56% of AML diagno-
inv 16 or t(16;16) 1–3% 10% sis in older patients,10,11 compared to a prevalence of
t(15;17) 4% 6–12%
approximately 8% in younger AML patients in the
Unfavorable cytogeneticsC
-7 8–9% 3%
Medical Research Council (MRC) AML 10 trial.7 AML
+8 6–10% 4% arising from prior bone marrow stem cell disorders or
Complex 18% 7% antecedent hematologic disorders, particularly when the
MDR1 expression 71% 35% process is greater than 10 months in duration prior to
Secondary AML 24–56% 8% the development of AML, is less responsive to chemo-
Treatment-related mortalityD 25–30% 5–10% therapy, resulting in shorter event-free survival, a lower
Complete remissionD 38–62% 65–73% CR rate, and conferring a worse prognosis.12
Long-term survivalC 5–15% 30%
Response to Therapy
A In general, Older AML Patients are defined as ≥ 60 years of Older adults are not as tolerant of or responsive to re-
age; Younger AML Patients as < 60 years of age. mission induction and consolidation chemotherapy com-
B New diagnoses, per 100,000 US citizens per year. Older/ pared to their younger counterparts. High mortality rates
younger division occurs at 65 years. likely result from inherent disease biology, an increased
C Percentages rounded to nearest whole number prevalence of comorbid disease, and a differential me-
D Rates following remission-induction therapy with an tabolism of induction regimen drugs, particularly
anthracycline- or anthracenedione-based regimen.
cytarabine, resulting in supratherapeutic drug levels.4

110 American Society of Hematology


Concern over potential treatment-related toxicities may ments other than the one they chose, despite physi-
result in undertreatment of disease. Paradoxically, ad- cian documentation of alternatives in all cases. Pa-
ministration of full-dose daunorubicin, for example, tients choosing to receive remission induction therapy
may result in a reduction in early deaths by effecting a spent 79% of their days during the first 6 weeks of
more rapid CR. the study period either hospitalized or being seen in
The outcome of older adults with AML is also clinic, compared to 14% of days for patients choos-
worse. Adults under the age of 60 years treated with an ing less aggressive therapy or best supportive care.
induction regimen consisting of an anthracycline com- Thus, treatment decisions should be based on indi-
bined with cytarabine have a 65%–73% chance of at- vidual patient preferences after an informed discus-
taining a complete remission (CR), while those over 60 sion has taken place that incorporates risk estimates
years of age have a 38%–62% chance of a CR.2-4,13-15 modified to a patient’s performance status, comorbid-
Patients who fall into the “very elderly” category (80 ities, and leukemia-specific risk factors, without us-
years or older) can attain a CR with intensive therapy, ing absolute cutoffs for prognostic factors or chrono-
but their chance of doing so is 30%, and only 7% of logical age. For example, an active, “younger” older
treated patients are alive at one year. Similar numbers adult with de novo AML and favorable cytogenetics
hold for patients 70 years of age or older.9 Moreover, should be presented with different prognostic informa-
long-term survival occurs in approximately 30% of tion than a bed-bound septuagenarian with secondary
younger adults (or 45% of those entering a CR), com- disease and complex cytogenetics. Ideally, this discus-
pared to only 5%–15% long-term DFS in adults over sion should include specifics about prognosis and treat-
the age of 60 years.2-4,15 ment-related complications, and the potential impact
therapy will have on a patient’s quality of life. When
Indications for Treatment available and clinically appropriate, older patients
For 85%–95% of older AML patients, any therapy ul- should always be offered the opportunity to partici-
timately will be purely palliative. Treatment options pate in clinical trials for up-front therapy.
range from supportive care (blood and platelet transfu-
sions when needed, antibiotics to treat infections, and Treatment Options
growth factor support) to low-dose chemotherapy (e.g.,
hydroxyurea or low-dose cytarabine) or investigational Remission induction therapy
agents as part of clinical trials, and high-dose chemo- As with younger AML patients, the backbone of remis-
therapy (anthracycline- or anthracenedione-based re- sion induction in older adults consists of an anthracycline
mission induction therapy). Two trials have random- (daunorubicin or idarubicin) or anthracenedione
ized older patients to receive either immediate remis- (mitoxantrone) and cytosine arabinoside (Ara-C), a regi-
sion induction chemotherapy with an anthracycline- men that has not changed in over two decades. Typi-
based regimen versus a less aggressive or palliative ap- cally, daunorubicin is given at a dose of 45 mg/m2/d × 3
proach.16,17 Only one of these studies16 demonstrated a days, or mitoxantrone or idarubicin are given at doses
survival advantage for patients receiving induction che- of 12 mg/m2/d × 3 days, in combination with Ara-C,
motherapy (21 vs 11 weeks, P = 0.015), for a median which is administered as a continuous infusion at 100
survival time only 16 days longer than the median amount or 200 mg/m2/d × 7 days (frequently referred to as 7+3
of time they spent hospitalized to receive therapy. Thus, chemotherapy). While studies have compared different
the decision of whether or not to offer remission induc- anthracyclines and anthracenediones, varied doses and
tion therapy (on the part of physicians) or to receive it schedules, and added additional agents with some im-
(on the part of patients) is not straightforward. provement in CR rates, they have not demonstrated an
In making this decision, older patients overesti- improvement in overall survival (OS) rates (Table
mate the potential benefit they may derive from in- 6).2,5,13,15,19-21 For example, a recent study from the ECOG
tensive chemotherapy and may not recall all treat- randomized older AML patients to remission induction
ment options. One study18 found that 74% of older therapy with either daunorubicin, idarubicin, or
patients estimated their chance to be cured by re- mitoxantrone along with a standard dose of Ara-C.9
mission induction therapy to be ≥ 50%, and almost The outcome was not significantly different, with CR
90% estimated their chance of being alive in one rates of 40%, 43%, and 46% and median survivals of
year to be ≥ 50%. In contradistinction, physicians 7.7, 7.5, and 7.2 months, respectively. Once a decision
caring for these patients estimated the chance of cure has been made to initiate intensive chemotherapy, it
to be ≤ 10% almost 89% of the time. Nearly two- should not be delayed, as this may impact outcome.9,22
thirds of patients did not recall being offered treat-

Hematology 2004 111


Table 6. Selected randomized studies defining remission induction therapy in older adults.

Remission Complete
Goal Induction Agents Remission Overall Survival Comments
Compare Anthracyclines and Anthracenediones
Lowenberg, 1998 2 M (8 mg/m2/d) + 47% 39 weeks (median) Early and post-induction death rates
A (100 mg/m2/d) 9% (5 years) were similar for both arms. Patients
vs. vs. vs. receiving mitoxantrone had a significantly
D (30 mg/m2/d) + 38% 36 weeks (median) higher rate of severe infections (25.1%
A (100 mg/m2/d) P = 0.07 6% (5 years) vs 18.6%) and a trend toward a longer
P = 0.23 duration of aplasia (22 vs 19 days).
AML Collaborative I (8-20 mg/m2/d) + 51% 33.6 weeks (median) Early induction failure tended to be higher
Group, 199819 A (100-200 mg/m2/d) with idarubicin, while late induction failure
vs. vs. vs. was lower. Myelosuppression was
D (45-50 mg/m2/d) + 46% 29.9 weeks (median) greater in patients receiving idarubicin.
A (100-200 mg/m2/d) P = ND P = 0.58
Archimbaud, 199920 I (8 mg/m2/d) + 45% 7 months (median) No difference between groups in degree
A (100 mg/m2/d) + 21% (2 years) of myelosuppression or in early death
E (100 mg/m2/d) rates.
vs. vs. vs.
M (7 mg/m2/d) + 50% 7 months (median)
A (100-200 mg/m2/d)+ P = 0.52 21% (2 years)
E (100 mg/m2/d) P = ND
Vary 7+3 Dose
Buchner, 199721 D (60 mg/m2/d) + 54% 16% (5 years) The 30 mg daunorubicin arm was closed
A (100 mg/m2/d) prematurely due to higher response rates
vs. vs. vs. in the 60 mg arm, resulting in 42 patients
D (30 mg/m2/d) + 42% 10% (5 years) receiving lower dose daunorubicin, and
A (100 mg/m2/d) P = 0.038 P = 0.11 130 patients receiving the higher dose.
Dillman, 199113 D (45 mg/m2/d) + 44% 11.0 weeks (median) This study has short overall survival times
A (100 mg/m2/d) compared to other studies in this patient
vs. vs. vs. population.
D (45 mg/m2/d) + 38% 9.6 weeks (median)
A (200 mg/m2/d) P = 0.68 P = 0.23
Add Agents
Goldstone, 200115 D (50 mg/m2/d) + 62% 12% (5 years) There were no significant differences in
A (100 mg/m2 q 12 hours)+ myelosuppression or other toxicities,
T (100 mg/m2 q 12 hours) neutrophils were slower to recover in the
vs. vs. vs. mitoxantrone arm. Patients receiving ADE
D (50 mg/m2/d) + 50% 8% (5 years) had higher rates of induction death (26%
A (100 mg/m2 q 12 hours) + P = 0.0021 P = 0.021 compared to 16% for DAT and 17% for
E (100 mg/m2/d) MAC)
vs. vs. vs.
M (12 mg/m2/d) + 55% 10% (5 years)
A (100 mg/m2/d P = 0.042 P = 0.1,2 0.23
Baer, 20025 D (60 mg/m2/d) + 46% 7 months (median) Because of concern about excessive
A (100 mg/m2/d)+ mortality on the ADEP arm (25 deaths vs
E (100 mg/m2/d) 12 on the ADE arm), it was closed early to
vs. vs. vs. to further accrual. Survival between the
D (40 mg/m2/d) + 39% 2 months (median) two arms was similar at one year.
A (100 mg/m2/d) +
E (60 mg/m2/d) + P = 0.008 P = 0.48
P (10 mg/kg/d)
Use Hematopoietic Growth Factors
Stone, 1995 3 D (45 mg/m2/d) + 54% 10.8 months Median duration of neutropenia was 15
A (200 mg/m2/d) (median) days in the GM-CSF arm and 17 days in
vs. vs. vs. placebo arm (P = 0.02). The duration of
D (45 mg/m2/d) + 51% 8.4 months hospitalization did not differ between the
A (200 mg/m2/d) + (median) arms; nor did the rates of life-threatening
GM-CSF (5 µg/kg/d) P = 0.61 P = 0.10 infection, or persistent leukemia.
Godwin, 199814 D (45 mg/m2/d) + 50% 9 months The duration of neutropenia was 15%
A (200 mg/m2/d) (median) shorter in the G-CSF arm compared to the
vs. vs. vs. placebo arm (P = 0.14). The duration of
D (45 mg/m2/d) + 41% 6 months hospitalization did not differ between the
A (200 mg/m2/d) + (median) arms; nor did the rates of life-threatening
G-CSF P = 0.89 P = 0.71 infection, or persistent leukemia.
Abbreviations: M, mitoxantrone; A, Ara-C (cytosine arabinoside); D, daunorubicin; I, idarubicin; E, etoposide; T, thioguanine; P, PSC-833;
GM-CSF, granulocyte macrophage colony stimulating factor; G-CSF, granulocyte colony stimulating factor; ND, not done
1 For comparison of DAT to ADE 2 For comparison of DAT to MAC 3 For comparison of ADE to MAC

112 American Society of Hematology


Hematopoietic growth factors Postremission bone marrow transplantation
In the majority of AML patients, death results from An even more aggressive approach than induction
bleeding or infectious complications. This is particu- therapy followed by consolidation consists of bone
larly true in older adults with AML. The utility of he- marrow transplantation. Nonmyeloablative allogeneic
matopoietic growth factors (HGF) for ameliorating the bone marrow transplants take advantage of a graft-ver-
myelosuppressive complications of AML therapy in sus-leukemia effect using a less-intensive preparative
older adults has been studied extensively.3,14 These tri- regimen with lower up-front mortality. The reduced
als were also designed to determine whether or not HGF TRM and ability to perform these transplants in the
had detrimental effects due to inappropriate stimula- outpatient setting make them an appealing option for
tion of leukemic cell proliferation and thus resistance, the older AML patient with few comorbidities and an
or whether they had beneficial effects in “priming” leu- adequate performance status. Preliminary studies that
kemic cells to proliferate prior to the administration of include older AML patients have demonstrated that du-
S-phase specific chemotherapy agents such as Ara-C.9,23 rable complete remissions are attainable with this treat-
With the exception of one ECOG study that demon- ment, though with limited follow-up. One study of 19
strated a CR rate and overall survival benefit in patients patients with myeloid malignancies (17 of whom had
randomized to the GM-CSF arm (compared to patients advanced MDS or AML) and a median age of 64 years
receiving no growth factor support), these trials found (range, 60–70 years) demonstrated a 68% survival at a
that while HGF are safe, reduce the duration of neutro- median follow-up of 825 days following nonmyeloab-
penia (by a range of 2–6 days), and do not support lative transplantation.24 These early data have prompted
leukemia cell proliferation, they also do not reliably cooperative groups to explore the role of bone marrow
improve the CR rate, the length of hospitalization, or transplantation in older AML patients in first CR.
the induction death rate or prolong survival.
Newer approaches
Post-remission chemotherapy Remission induction therapy for older adults with AML
No randomized trial has ever demonstrated that any is no panacea, with median and 5-year survival rates
amount of post-remission therapy in older AML pa- resembling those of patients with advanced lung can-
tients provides better outcomes than no post-remission cer.1 It is thus reasonable to consider investigational
therapy. That being said, the only studies demonstrat- agents for older AML patients as initial therapy, par-
ing that long-term DFS is possible in older AML pa- ticularly those that may be associated with less TRM.
tients have included remission induction and post-re- Potential targets for antileukemia therapy include spe-
mission therapy. It is reasonable, then, to administer cific signaling molecules required for the maintenance
post-remission therapy consisting of a repeat of remis- of the leukemic state, such as tyrosine kinases;
sion induction therapy, single-agent Ara-C, or 2 days overexpression of bcl-2, an anti-apoptosis signal; DNA
of an anthracycline or anthracenedione (the same type methylation, associated with suppression of regulatory
of drug given at the same doses as with remission in- genes and with disease progression; indirect pathways
duction therapy) combined with 5 days of Ara-C, again that maintain leukemogenesis, including angiogenesis
given at the same dose as with remission induction and drug resistance; and investigational agents with
therapy (frequently referred to as post-remission mechanisms of action that differ from anthracyclines,
therapy). There does not appear to be any additional anthracenediones, and Ara-C, such as nucleoside ana-
survival benefit attained from administering more than logs, farnesyl transferase inhibitors (FTIs), and MDR
1–2 cycles of post-remission therapy or in treating older modulators, alone or in combination with standard thera-
AML patients with maintenance therapy. In the Medi- pies.25-29 These approaches are discussed in more detail
cal Research Council (MRC) AML 11 trial, 371 pa- in Section I. The efficacy of the FTI Zarnestra will be
tients who entered a complete remission following examined in newly diagnosed AML patients over the
anthracycline- or anthracenedione-based remission in- age of 70 years in the US cooperative group setting
duction therapy were randomized to receive either 1 (SWOG 0432). One recent trial from the Dutch-Bel-
cycle of daunorubicin, Ara-C, and thioguanine (DAT) con- gian Hemato-Onocology Cooperative Group (HOVON)
solidation therapy, or DAT along with 3 additional cycles randomized predominantly older patients, most of whom
of Ara-C–based consolidation therapy (for a total of 4 had a diagnosis of advanced MDS or AML, to Ara-C
cycles of post-remission therapy).15 Of those randomized and G-CSF or the same regimen plus fludarabine for 2
to the long consolidation course, 61% were able to com- cycles followed by Ara-C and daunorubicin for 1 cycle.30
plete all 4 cycles. Survival was similar at 5 years for pa- This study is typical of many novel combination trials
tients randomized to the short and long consolidation arms. in older AML patients in that, while the CR rate was

Hematology 2004 113


improved in AML patients receiving fludarabine (95% reduced toxicity in newly diagnosed PML/RARα-positive
vs 71%, P = 0.046), survival was not impacted. acute promyelocytic leukemia. Blood. 1999;94:3015-3021.
8. Tallman MS, Nabhan C, Feusner JH, et al. Acute
promyelocytic leukemia: evolving therapeutic strategies.
Supportive/palliative care Blood. 2002;99:759-767.
Intensive chemotherapy provides only marginal, if any, 9. Fenaux P, Chastang C, Chevret S, et al. A randomized
survival benefit to older AML patients, so non-inten- comparison of all transretinoic acid (ATRA) followed by
chemotherapy and ATRA plus chemotherapy and role of
sive (or non-chemotherapy-based) approaches are rea- maintenance therapy in newly diagnosed acute promyelocytic
sonable. We use the phrase aggressive supportive care leukemia. Blood. 1999;94:1192-1200.
to emphasize that symptoms will be treated vigorously 10. Diverio D, Rossi V, Avvisati G, et al. Early detection of
and to distinguish this modality from hospice. Blood relapse by prospective reverse transcriptase-polymerase chain
reaction analysis of the PML/RAR alpha fusion gene in
and platelet transfusions should be administered to al- patients with acute promyelocytic leukemia enrolled in the
leviate symptoms stemming from anemia and thromb- GIMEMA-AIEOP multicenter “AIDA” trial. GIMEMA-
ocytopenia, and antibiotics started when appropriate. AIEOP Multicenter “AIDA” Trial. Blood. 1998;92:784-789.
Low-dose chemotherapy should only be used in the set- 11. Latagliata R, Petti MC, Fenu S, et al. Therapy-related
myelodysplastic syndrome-acute myelogenous leukemia in
ting of leukocytosis and/or associated symptoms. Any
patients treated for acute promyelocytic leukemia: an
recommendations for the institution of neutropenic pre- emerging problem. Blood. 2002;99:822-824.
cautions (i.e., avoiding crowds, refraining from 12. Soignet SL, Frankel SR, Douer D, et al. United States
ingestions of raw foods) must be balanced with the lack multicenter study of arsenic trioxide in relapsed acute
of evidence supporting the benefit of these maneuvers promyelocytic leukemia. J Clin Oncol. 2001;19:3852-3860.
13. Sievers EL, Larson RA, Stadtmauer EA, Stey E, Lowenberg
and the impact such restrictions will have on a patient’s B, Dombret H. Efficacy and safety of gemtuzumab
quality of life. Hospice services should be instituted ozogamicin in patients with CD33-positive acute myeloid
within 6 months of anticipated demise. While some leukemia in first relapse. J Clin Oncol. 2004;22:1087-1094.
hospice organizations prohibit blood product transfu- 14. Wadleigh M, Richardson PG, Zahrich D, et al. Prior
gemtuzumab ozogamicin exposure significantly increases the
sions, we consider these to be palliative in this popula- risk of venoocclusive disease in patients who undergo
tion as they may result in improved quality of life in myeloablative allogeneic stem cell transplantation. Blood.
terminal cancer patient populations. 2003;102:1578-1582.
15. De Angelo DJ, Stone RM, Durrant S, Liu D, Baccarani M,
Schiffer CA, Amrein PC, Sherman ML. Gemtuzumab
REFERENCES ozogamicin (Mylotarg®) in combination with induction
chemotherapy for the treatment of patients with de novo acute
I. AML: Current Landscape and myeloid leukemia: two age-specific phase II trials [abstract].
Future Directions Blood. 2003;102:100a.
1. Vardiman JW, Harris NL, Brunning RD. The World Health 16. Baer MR, George SL, Dodge RK, et al. Phase 3 study of the
Organization (WHO) classification of the myeloid neoplasms. multidrug resistance modulator PSC-833 in previously
Blood. 2002;100:2292-2302. untreated patients 60 years of age and older with acute
2. Gilliland DG, Griffin JD. The role of FLT3 in hematopoiesis myeloid leukemia: Cancer and Leukemia Group B Study
and leukemia. Blood. 2002;100:1332-1342. 9720. Blood. 2002;100:1224-1232.
3. Poppe B, Vandesompele J, Schoch C, et al. Expression 17. List AF, Kopecky KJ, Willman CL, et al. Benefit of
analyses identify MLL as a prominent target of 11q23 cyclosporine modulation of drug resistance in patients with
amplification and support an etiologic role for MLL gain of poor-risk acute myeloid leukemia: a Southwest Oncology
function in myeloid malignancies. Blood. 2004;103:229-235. Group study. Blood. 2001;98:3212-3220.
4. Kolitz JE, George SL, Dodge RK, Hoke E, Hurd DD. Dose 18. Marcucci G, Byrd JC, Guowei D, Klisovic MI, Kourlas PJ.
escalation studies of Ara-C (A), daunorubicin (D) and Phase I and pharmacodynamic studies of G3139, a Bcl-2
etoposide (E) with and without multidrug resistance (MDR) antisense oligonucleotide, in combination with chemotherapy
Modulation with PSC-833 (P in untreated adults with acute in refractory or relapsed acute leukemia. Blood.
myeloid leukemia (AML) [abstract]. Blood. 2003;101:425-432.
2001;98(Suppl):461a. 19. Cortes J, Thomas D, Koller C, Giles F, Estey E, Faderl S.
5. Bloomfield CD, Lawrence D, Byrd JC, et al. Frequency of Phase I study of bortezomib in refractory or relapsed acute
prolonged remission duration after high-dose cytarabine leukemias. Clin Cancer Res. 2004;10:3371-3376.
intensification in acute myeloid leukemia varies by cytoge- 20. Lowenberg B, van Putten W, Theobald M, et al. Effect of
netic subtype. Cancer Res. 1998;58:4173-4179. priming with granulocyte colony-stimulating factor on the
6. Byrd JC, Dodge RK, Carroll A, et al. Patients with outcome of chemotherapy for acute myeloid leukemia. N
t(8;21)(q22;q22) and acute myeloid leukemia have superior Engl J Med. 2003;349:743-752.
failure-free and overall survival when repetitive cycles of 21. Silverman LR, Demalos EP, Peterson BL, et al. Randomized
high-dose cytarabine are administered. J Clin Oncol. 1999; controlled trial of azacitidine inpatients with the
17:3767-3775. myelodysplastic syndrome: a study of the Cancer and
7. Sanz MA, Martin G, Rayon C, et al for the PETHEMA Group. Leukemia Group B. J Clin Oncol. 2002;20:2429-2440.
A modified AIDA protocol with anthracycline-based 22. Karp JE, Lancet JE, Kaufmann SH, et al. Clinical and
consolidation results in high antileukemic efficacy and biological activity of the farnesyltransferase inhibitor

114 American Society of Hematology


R115777 in adults with refractory and relapsed acute tion from related and unrelated donors in older patients with
leukemias: a phase I clinical-laboratory correlative trial. myeloid leukemia. J Clin Oncol. 2003;21:1480-1484.
Blood. 2001;97:3361-3369. 9. Tallman MS, Rowlings PA, Milone J, et al. Effect of post
23. Lancet JE, Gojo I, Gotlib J, et al. Tipifarnib (ZARNESTRA) remission chemotherapy before HLA identical sibling
in previously untreated poor-risk AML and MDS: interim transplantation for acute myelogenous leukemia in first
results of a phase 2 trial [abstract]. Blood. 2003;102:176a. complete remission. Blood. 2000;96:1254-1258.
24. Fiedler W, Mesters R, Tinnefeld H, Loges S. A phase 2 clinical 10. Bornhauser M, Storer B, Slattery JT, et al. Conditioning with
study of SU5416 in patients with refractory acute myeloid fludarabine and targeted busulfan for transplantation of
leukemia. Blood. 2003;102:2763-2767. allogeneic hematopoietic stem cells. Blood. 2003;102:820-
25. Stone RM, DeAngelo DJ, Klimek V, et al. Acute myeloid 826.
leukemia patients with an activating mutation in FLT3 11. Couriel DR, Saliba RM, Giralt S, et al. Acute and chronic
respond to a small molecule FLT3 tyrosine kinase inhibitor, graft-versus-host disease after ablative and nonmyeloablative
PKC412. Blood. First Edition Paper, prepublished online conditioning for allogeneic hematopoietic transplantation.
September 2, 2004; DOI 10.1182/blood-2004-03-0891 Biol Blood Marrow Transplant. 2004;10:178-185.
26. Smith DB, Levis M, Beran M, Giles F, Kantarjian K. Single- 12. Jungbanss C, Marr KA, Carter RA, et al. Incidence and
agent CEP 701, a novel FLT3 inhibitor, shows biologic and outcome of bacterial and fungal infections following
clinical activity in patients with relapsed or refractory acute nonmyeloablative compared with myeloablative allogeneic
myeloid leukemia. Blood. 2004;103:3669-3676. hematopoietic stem cell transplantation: a matched control
27. DeAngelo D, Stone RM, Bruner RJ, et al. Phase I clincal study. Biol Blood Marrow Transplant. 2002;8:512-520.
results with MLN518, a novel FLT3 antagonist: tolerability. 13. Cassileth PA, Harrington DP, Appelbaum FR, et al. Chemo-
Pharmacokinetics and pharmacodynamics [abstract]. Blood. therapy compared with autologous or allogeneic bone
2003;102:65a. marrow transplantation in the management of acute myeloid
28. Wadleigh M, DeangeloDJ, Griffin JD, Stone RM. After leukemia in first remission. N Engl J Med. 1998;339:1649-
chronic myelogenous leukemia: tyrosine kinase inhibitors in 1656.
other hematologic malignancies. Blood. 2004; Epub 10.1182/ 14. Zittoun RA, Mandelli F, Willemze R, De Witte, T, et al.
blood-2003-11-3896. Autologous or allogeneic bone marrow transplantation
29. Karp JE, Gojo I, Pili R, Gocke CD, Greer J, Guo C. Targeting compared with intensive chemotherapy in acute myelogenous
vascular endothelial growth factor for relapsed and refractory leukemia. N Engl J Med. 1995;332:217-223.
adult acute myelogenous leukemias: therapy with sequential 15. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, et al.
1-B-D- arabinofuransylcytosine, mitoxantrone, and Karyotypic analysis predicts outcome of preremission and
bevacizumab. Clin Cancer Res. 2004;10:3577-3585. post remission therapy in adult acute myeloid leukemia: a
Southwest Oncology Group/Eastern Cooperative Oncology
II. The Role of Autologous and Allogeneic (“Full” Group study. Blood. 2000;96:4075-4077.
and “Mini”) Stem Cell Transplantation in AML 16. Burnett AK, Goldstone AH, Stevens RMF, Hann IM, et al.
Randomized comparison of addition of autologous bone-
1. Bensinger WI, Martin PJ, Storer B, et al. Transplantation of
marrow transplantation to intensive chemotherapy for acute
bone marrow as compared with peripheral blood cells from
myeloid leukaemia first remission: results of MRC AML 10
HLA-identical relatives in patients with hematologic cancers.
trial. Lancet. 1998;351:700-708.
N Engl J Med. 2001;344;175-181.
17. Meloni G, Diverio D, Vignette M, et al. Autologous bone
2. Zaucha JM, Gooley T, Bensinger WI, et al. CD34 cell dose in
marrow transplantation for acute promyelocytic leukemia in
granulocyte colony-stimulating factor-mobilized peripheral
second remission: prognostic relevance of pretransplant
blood mononuclear cell grafts affects engraftment kinetics
minimal residual disease assessment by reverse-transcription
and development of extensive chronic graft-versus-host
polymerase chain reaction of the PML/RARα fusion gene.
disease after human leukocyte antigen-identical sibling
Blood. 1997;90:1321-1325.
transplantation. Blood. 2001;98:3221-3227.
18. Linker CA, Ries CA, Damon LE, et al. Autologous stem cell
3. Champlin RE, Schmitz N, Horowitz MM, et al. Blood stem
transplantation for acute myeloid leukemia in first remission.
cells compared with marrow as a source of hematopoietic cells
Biol Blood Marrow Transplant. 2000; 6:50-57.
for allogeneic transplantation. Blood. 2000;95:3702-3709.
19. Suciu S, Mandelli F, de Witte T, et al. Allogeneic compared
4. Schmitz N, Beksac M, Hasenclever D, et al. Transplantation of
with autologous stem cell transplantation in the treatment of
mobilized peripheral blood cells to HLA-identical siblings
patients younger than 46 years with acute myeloid leukemia
with standard-risk leukemia. Blood. 2002:100:761-767.
(AML) in first complete remission (CR1): an intention-to-treat
5. Cutler C, Kim HT, Hochberg E, et al. Sirolimus and
analysis of the EORTC/GIMEMA AML-10 trial. Blood.
tacrolimus without methotrexate as graft-versus-host disease
2003;102:1232-1240.
prophylaxis after matched related donor peripheral blood
20. Linker CA, Damon LE, Ries CA, Navarro WA, Case D, Wolf
stem cell transplantation. Biol Blood Marrow Transplant.
JL. Autologous stem cell transplantation for advanced acute
2004;10:328-336.
myeloid leukemia. Bone Marrow Transplant. 2002;29:297-
6. Michallet M, Bilger K, Garban F, et al. Allogeneic hematopoi-
301.
etic stem-cell transplantation after nonmyeloablative
21. Reiffers J, Labopin M, Sanz M, et al. Autologous blood cell
preparative regimens: impact of pretransplantation and
vs marrow transplantation for acute myeloid leukemia in
posttransplantation factors on outcome. J Clin Oncol.
complete remission: an EBMT retrospective analysis. Bone
2001;19:3340-3349.
Marrow Transplant. 2000;25:1115-1119.
7. Stein AS, O’Donnell MR, Slovak ML, et al. Interleukin-2
22. Levine JE, Uberti JP, Ayash L, et al. Lowered-intensity
after autologous stem-cell transplantation for adult patients
preparative regimen for allogeneic stem cell transplantation
with acute myeloid leukemia in first complete remission. J
delays acute graft-versus-host disease but does not improve
Clin Oncol. 2003;21:615-623.
outcome for advanced hematologic malignancy. Biol Blood
8. Bertz H, Potthoff K, Finke J. Allogeneic stem-cell transplanta-

Hematology 2004 115


Marrow Transplant. 2003;9:189-197. leukemia in the elderly: assessment of multidrug resistance
23. Sayer HG, Kroger M, Veyer J, et al. Reduced intensity (MDR1) and cytogenetics distinguishes biologic subgroups
conditioning for allogeneic hematopoietic stem cell transplan- with remarkably distinct responses to standard chemotherapy.
tation in patients with acute myeloid leukemia: disease status A Southwest Oncology Group study. Blood. 1997;89:3323-
by marrow blasts is the strongest prognostic factor. Bone 3329.
Marrow Transplant. 2003;31:1089-1095. 11. Bauduer F, Ducout L, Dastugue N, Capdupuy C, Renoux M.
24. Michallet M, Thomas X, Vernant JP, et al. Long-term outcome De novo and secondary acute myeloid leukemia in patients
after allogeneic hematopoietic stem cell transplantation for over the age of 65: a review of fifty-six successive and
advanced stage acute myeloblastic leukemia: a retrospective unselected cases from a general hospital. Leuk Lymphoma.
study of 379 patients reported to the Societe Francaise de 1999;35:289-296.
Greffe de Moelle (SFGM). Bone Marrow Transplant. 12. Estey E, Thall P, Beran M, Kantarjian H, Pierce S, Keating M.
2000;26:1157-1163. Effect of diagnosis (refractory anemia with excess blasts,
25. Feinstein LC, Sandmaier BM, Maloney DG, et al. Allografting refractory anemia with excess blasts in transformation, or
after nonmyeloablative conditioning as a treatment after a acute myeloid leukemia [AML]) on outcome of AML-type
failed conventional hematopoietic cell transplant. Biol Blood chemotherapy. Blood. 1997;90:2969-2977.
Marrow Transplant. 2003;9:266–272. 13. Dillman RO, Davis RB, Green MR, et al. A comparative study
of two different doses of cytarabine for acute myeloid
III. AML in Older Adults: Are We Listening? leukemia: a phase III trial of Cancer and Leukemia Group B.
1. Ries L, Eisner M, Kosary C, et al. SEER Cancer Statistics Blood. 1991;78:2520-2526.
Review, 1975-2001, National Cancer Institute. http:// 14. Godwin JE, Kopecky KJ, Head DR, et al. A double-blind
seer.cancer.gov/csr/1975_2001/. Bethesda, MD: National placebo-controlled trial of granulocyte colony-stimulating
Cancer Institute; 2004. factor in elderly patients with previously untreated acute
2. Lowenberg B, Suciu S, Archimbaud E, et al. Mitoxantrone myeloid leukemia: a Southwest oncology group study (9031).
versus daunorubicin in induction-consolidation chemotherapy Blood. 1998;91:3607-3615.
- the value of low-dose cytarabine for maintenance of 15. Goldstone AH, Burnett AK, Wheatley K, Smith AG,
remission, and an assessment of prognostic factors in acute Hutchinson RM, Clark RE. Attempts to improve treatment
myeloid leukemia in the elderly: final report. European outcomes in acute myeloid leukemia (AML) in older patients:
Organization for the Research and Treatment of Cancer and the results of the United Kingdom Medical Research Council
the Dutch-Belgian Hemato-Oncology Cooperative Hovon AML11 trial. Blood. 2001;98:1302-1311.
Group. J Clin Oncol. 1998;16:872-881. 16. Lowenberg B, Zittoun R, Kerkhofs H, et al. On the value of
3. Stone RM, Berg DT, George SL, et al. Granulocyte-macroph- intensive remission-induction chemotherapy in elderly
age colony-stimulating factor after initial chemotherapy for patients of 65+ years with acute myeloid leukemia: a
elderly patients with primary acute myelogenous leukemia. randomized phase III study of the European Organization for
Cancer and Leukemia Group B. N Engl J Med. Research and Treatment of Cancer Leukemia Group. J Clin
1995;332:1671-1677. Oncol. 1989;7:1268-1274.
4. Mayer RJ, Davis RB, Schiffer CA, et al. Intensive 17. Tilly H, Castaigne S, Bordessoule D, et al. Low-dose
postremission chemotherapy in adults with acute myeloid cytarabine versus intensive chemotherapy in the treatment of
leukemia. Cancer and Leukemia Group B. N Engl J Med. acute nonlymphocytic leukemia in the elderly. J Clin Oncol.
1994;331:896-903. 1990;8:272-279.
5. Baer MR, George SL, Dodge RK, et al. Phase 3 study of the 18. Sekeres MA, Stone RM, Zahrieh D, et al. Decision-making
multidrug resistance modulator PSC-833 in previously and quality of life in older adults with acute myeloid leukemia
untreated patients 60 years of age and older with acute or advanced myelodysplastic syndrome. Leukemia.
myeloid leukemia: Cancer and Leukemia Group B Study 2004;18:809-816.
9720. Blood. 2002;100:1224-1232. 19. A systematic collaborative overview of randomized trials
6. Byrd JC, Mrozek K, Dodge RK, et al. Pretreatment cytoge- comparing idarubicin with daunorubicin (or other
netic abnormalities are predictive of induction success, anthracyclines) as induction therapy for acute myeloid
cumulative incidence of relapse and overall survival in adult leukaemia. AML Collaborative Group. Br J Haematol.
patients with de novo acute myeloid leukemia: results from 1998;103:100-109.
CALGB 8461. Blood. 2002;100:4325-4336. 20. Archimbaud E, Jehn U, Thomas X, et al. Multicenter
7. Grimwade D, Walker H, Oliver F, et al. The importance of randomized phase II trial of idarubicin vs mitoxantrone,
diagnostic cytogenetics on outcome in AML: analysis of combined with VP-16 and cytarabine for induction/
1,612 patients entered into the MRC AML 10 trial. The consolidation therapy, followed by a feasibility study of
Medical Research Council Adult and Children’s Leukaemia autologous peripheral blood stem cell transplantation in
Working Parties. Blood. 1998;92:2322-2333. elderly patients with acute myeloid leukemia. Leukemia.
8. Grimwade D, Walker H, Harrison G, et al. The predictive 1999;13:843-849.
value of hierarchical cytogenetic classification in older adults 21. Buchner T, Hiddemann W, Wormann B, et al. Daunorubicin
with acute myeloid leukemia (AML): analysis of 1065 60 instead of 30 mg/sqm improves response and survival in
patients entered into the United Kingdom Medical Research elderly patients with AML [abstract]. Blood. 1997;90:583a.
Council AML11 trial. Blood. 2001;98:1312-1320. 22. Sekeres M, Rybicki L, Sobecks R, et al. The Time Interval
9. Rowe JM, Neuberg D, Friedenberg W, et al. A phase 3 study from Diagnosis of Acute Myeloid Leukemia (AML) in older
of three induction regimens and of priming with GM-CSF in adults to start of remission induction therapy predicts survival
older adults with acute myeloid leukemia: a trial by the [abstract]. Blood. 2003;102:3257a.
Eastern Cooperative Oncology Group. Blood. 2004;103:479- 23. Lowenberg B, van Putten W, Theobald M, et al. Effect of
485. priming with granulocyte colony-stimulating factor on the
10. Leith CP, Kopecky KJ, Godwin J, et al. Acute myeloid outcome of chemotherapy for acute myeloid leukemia. N

116 American Society of Hematology


Engl J Med. 2003;349:743-752. 28. Silverman LR, Demakos EP, Peterson BL, et al. Randomized
24. Bertz H, Potthoff K, Finke J. Allogeneic stem-cell transplanta- controlled trial of azacitidine in patients with the
tion from related and unrelated donors in older patients with myelodysplastic syndrome: a study of the cancer and
myeloid leukemia. J Clin Oncol. 2003;21:1480-1484. leukemia group B. J Clin Oncol. 2002;20:2429-2440.
25. Stone R, Klimek V, DeAngelo D, et al. PKC412, an oral FLT3 29. Smith BD, Levis M, Beran M, et al. Single-agent CEP-701, a
inhibitor, has activity in mutant FLT3 acute myeloid leukemia novel FLT3 inhibitor, shows biologic and clinical activity in
(AML): a phase II clinical trial [abstract]. Blood. patients with relapsed or refractory acute myeloid leukemia.
2002;100:316a. Blood. 2004;103:3669-3676.
26. Karp JE, Lancet JE, Kaufmann SH, et al. Clinical and biologic 30. Ossenkoppele GJ, Graveland WJ, Sonneveld P, et al. The
activity of the farnesyltransferase inhibitor R115777 in adults value of fludarabine in addition to ARA-C and G-CSF in the
with refractory and relapsed acute leukemias: a phase 1 treatment of patients with high-risk myelodysplastic syn-
clinical-laboratory correlative trial. Blood. 2001;97:3361- dromes and AML in elderly patients. Blood. 2004;103:2908-
3369. 2913.
27. Giles FJ. The emerging role of angiogenesis inhibitors in
hematologic malignancies. Oncology (Huntingt). 2002;16:23-
29.

Hematology 2004 117

S-ar putea să vă placă și