You are on page 1of 8

VOLUME 25 䡠 NUMBER 14 䡠 MAY 10 2007

JOURNAL OF CLINICAL ONCOLOGY R E V I E W A R T I C L E

Acute Myeloid Leukemia and Myelodysplastic Syndromes in


Older Patients
Elihu Estey
From the Department of Leukemia, The
A B S T R A C T
University of Texas M.D. Anderson
Cancer Center, Houston, TX. The median age of patients with acute myeloid leukemia (AML) is 65 to 70 years. The majority of
Submitted December 5, 2006; accepted older patients with AML probably do not receive specific treatment, and those who receive
February 8, 2007. standard regimens have a median survival time of less than 1 year. This suggests that, in general,
Author’s disclosures of potential con- older patients should receive investigational therapy; however, factors other than age influence
flicts of interest and author contribu- survival after administration of standard treatment and need to be accounted for when making
tions are found at the end of this treatment recommendations. In some cases where investigational therapy is unavailable, palliative
article.
care may be the best option. Like AML, myelodysplastic syndrome (MDS) is a disease of the
Address reprint requests to Elihu Estey, elderly. It is divided into higher and lower risk groups. The natural history of high-risk MDS (eg,
MD, Department of Leukemia, Unit ⬎ 10% marrow blasts) bears more resemblance to that of AML than to that of an indolent
428, The University of Texas M.D.
disorder; accordingly, similar therapeutic considerations apply. The more benign natural history of
Anderson Cancer Center, 1515
Holcombe Blvd, Box 61, Houston, TX
lower risk MDS leads to consideration of reduction in transfusion needs and improvement in
77030; e-mail: ehestey@mdanderson.org. quality of life as primary goals of therapy. Lenalidomide, azacitidine, and decitabine, each recently
© 2007 by American Society of Clinical
approved by the US Food and Drug Administration, are useful in achieving these objectives.
Oncology
J Clin Oncol 25:1908-1915. © 2007 by American Society of Clinical Oncology
0732-183X/07/2514-1908/$20.00

DOI: 10.1200/JCO.2006.10.2731
(TID) and the probability of therapeutic resistance.
ACUTE MYELOID LEUKEMIA
Resistance means either a transient remission or no
Any discussion of acute myeloid leukemia (AML) in remission, with the inability to maintain a remission
a geriatric population should begin with recognition incompatible with survival. The distinction between
of the difficulty in accurately defining such a popu- TID and resistance is arbitrary, with overlap be-
lation. In particular, it seems that each additional tween the two categories, but often, deaths occur-
year of age above age 5 to 10 years is associated with ring within the first 6 weeks after beginning therapy
a worse outcome.1 Thus, although clinical trials of- are considered TID, with deaths occurring later as-
ten consider patients aged 60 years and older to be cribed to resistance; using this definition, resistance
elderly, there is less difference in outcome between, is the cause of death in 75% of older patients. Of
for example, a 58-year-old patient and a 61-year-old particular note, at any given age, the risk of TID
patient than between the same 61-year-old patient depends on, among other factors, performance sta-
and a 69-year-old patient, although the 58-year-old tus, bilirubin and creatinine (even within the normal
patient would be considered young, and both the range), the presence of infection, albumin, and
61-year-old and 68-year-old patients would be con- beta2-microglobulin.3-5 The major predictor of re-
sidered elderly. However, for practical purposes, age sistance is not age but leukemia-cell cytogenetics,6
ⱖ 60 years is a commonly accepted criterion for with a normal karyotype and the very rare cases with
elderly AML (with 60 years being 5 to 10 years less inv(16) or t (8;21) associated with the best outcome,
than the median age of all AML patents). Impor- abnormalities of chromosomes 5 and/or 7 (⫺5/⫺7)
tantly, the effect of age is not constant throughout a associated with the worst outcome (except when,
patient’s course of treatment. Rather, approximately rarely, present as single abnormalities7), and other
1 year after treatment begins, the effect of age on karyotypes associated with an intermediate out-
prognosis disappears; this reflects the dispropor- come. Furthermore, at any given age, an abnormal
tionate influence of age on early death.2 blood count for at least 1 month before diagnosis of
Clinical trials typically include as eligible all AML (antecedent hematologic disorder [AHD]) or
older patients except those with a poor performance AML arising after chemotherapy for another dis-
status, abnormal organ function, or active infection. ease (secondary AML) confers a worse prognosis,
These exclusions suggest that factors (covariates) usually independent of cytogenetics.8 Leith et al9
other than age affect outcome. Such outcome is a found that presence of a protein (MDR1, also
sum of the probability of treatment-induced death called P-glycoprotein) that promoted extrusion

1908
Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138
AML and MDS in the Elderly

of anthracyclines commonly used to treat AML was associated with not known in adequate detail. Hence, the natural history of AML
a lower complete response (CR) rate, but not relapse-free survival becomes difficult to specify, particularly compared with outcome with
rate, in older patients. In contrast, internal tandem duplications standard anti-AML therapy. In one of the few randomized studies
(ITD) of the FLT3 gene (but seemingly not mutations that do not to address the issue, the European Organisation for Research and
affect the gene’s length) decrease relapse-free survival time but not Treatment of Cancer assigned patients aged more than 65 years to
CR rate; such ITDs are most commonly seem in the normal cyto- immediate 3⫹7 or to observation/supportive care, with use of chem-
genetic group.10 However, although older age is associated with otherapy (hydroxyurea or LDAC) if blood counts worsened or symp-
poorer performance status, ⫺5/⫺7, an AHD, secondary AML, and toms developed.19 Median survival time was 21 weeks in the 3⫹7 arm
MDR1 (but not FLT3 ITDs), the deleterious effect of advancing age and 10 weeks in the observation arm, and the number of days spent in
generally remains after accounting for these covariates.1,3-5 None- hospital was essentially identical. However, the degree to which the
theless, it is important to consider covariates other than age when enrolled patients represented AML in the elderly is in some doubt
deciding whether to recommend standard anti-AML therapy, investi- because the enrolled patients had a performance status of less than 3
gational anti-AML therapy, or no specific anti-AML therapy (ie, sup- and relatively normal organ function, and 50% of the patients ran-
portive care only) for an older patient with untreated AML. This domly assigned to observation had to begin therapy within 1 month,
decision is at the heart of the management of AML in the elderly. suggesting that they were on the verge of progression when randomly
assigned. In contrast, it is commonly observed that AML in older
General Inadequacy of Standard Anti-AML Therapy
patients can have an indolent course for months if not longer. How-
Two types of regimens may be considered standard. The first,
ever, regardless of their relative merits, 3⫹7, LDAC, and supportive
known as 3⫹7, consists of 3 days of an anthracycline (usually dauno-
care only are not appealing approaches for the majority of older
rubicin or idarubicin) and 7 days of cytarabine (ara-C). Table 1 lists
patients with AML.
series reporting outcome with 3⫹7 in older patients. Despite varia-
tions in postremission therapy in these studies and possible differences Most Elderly Patients Are Candidates for
in therapy after relapse, the table indicates that the average older Investigational Therapies
patient with untreated AML can expect to live 8 to 12 months if The data in Table 1 indicate that a principal reason to recom-
administered 3⫹7 as initial therapy.11-15 The second standard regi- mend investigational therapy for older patients with AML is the un-
men omits anthracyclines and reduces the dose of ara-C (low-dose satisfactory outcome with standard therapy. Although scientific
ara-C [LDAC]). In one of the only randomized comparisons of 3⫹7 promise is a frequently cited reason for recommending an investiga-
and LDAC, which involved 87 patients aged more than 65 years, Tilly tional therapy, history suggests that the promise of many therapies is
et al16 found median survival times of 9 months with LDAC and 13 not borne out by clinical trials. Furthermore, investigational therapies
months with 3⫹7; however, patients administered LDAC spent less can prove worse than standard therapies. (An example is a trial of the
time in hospital (median time, 28 v 34 days, respectively) and required MDR1 antagonist PSC-833 in older patients with untreated AML.20)
fewer transfusions (median, seven v 10 transfusions, respectively). If this was not the case, random assignment between standard and
Reported survival data likely overestimate the effectiveness of investigational therapies would be problematic. Nonetheless, I believe
standard therapy in older patients. This overestimation results from that, in the majority of older patients with AML, the results with newer
the tendency to treat only very fit older patients.17,18 Indeed, most therapy cannot be much worse than those obtained with standard or
elderly patients probably do not receive anti-AML therapy, leading to no therapy. The view that elderly patients with AML are typically
the contention that that there is no standard therapy for AML of the candidates for investigational therapy is widespread. For example,
elderly. The characteristics of untreated patients, as a group, are often the National Comprehensive Cancer Network,21 a consortium of

Table 1. Outcomes in Older Patients Administered Anthracycline Plus Cytarabine for AML
Median Probability
Age of Survival of Survival Induction
No. of Patients Time at 2 Years CR Rate Death Rate
Study Group or Institution Patients (years) (months) (%) (%) (%) Comment
Rowe et al11 ECOG 234 ⱖ 56 7-8 ⬇ 20 41 19 Results same with daunorubicin,
idarubicin, or mitoxantrone and ⫾
GM-CSF priming
Goldstone et al12 NCRI (formerly MRC) 1,314 ⱖ 56 ⬇ 12 ⬇ 25 62 16 Results same with 1 or 4 courses
after CR and ⫾ G-CSF starting 8
days after end of induction
Anderson et al13 SWOG 161 ⱖ 56 9 19 43 15 Survival worse with mitoxantrone ⫹
etoposide
Van der Holt et al14 HOVON 211 ⱖ 60 ⬇ 10 ⬇ 25 48 15 Results same ⫾ PSC-833
Estey et al15 M.D. Anderson Cancer 31 ⱖ 65 ⬇ 12 ⬇ 20 48 Not given Cytarabine at 1.5 g/m2 daily ⫻ 3;
Center survival worse with single-agent
gemtuzumab

Abbreviations: AML, acute myeloid leukemia; CR, complete response; ECOG, Eastern Cooperative Oncology Group; GM-CSF, granulocyte-macrophage
colony-stimulating factor; NCRI, National Cancer Research Institute; MRC, Medical Research Council; G-CSF, granulocyte colony-stimulating factor; SWOG,
Southwest Oncology Group; HOVON, Stichting Haemato-Oncologie voor Volwassenen Nederland.

www.jco.org 1909
Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138
Elihu Estey

prominent American cancer centers, explicitly cites clinical trial as the


1.0
preferred option in patients with untreated AML who are aged 60 Total Died
years and olders. Better* 115 77
0.8 + Worse 533 465
Thinking in terms of covariates other than age allows identifica-

Survival Probablity
tion of groups of older patients for whom standard therapy (3⫹7 or ++ +
+
LDAC) might be reasonable. From the preceding discussion, one such 0.6 +
+ +
+
group might be patients with the following characteristics: aged 60 to ++ +++ +++++
69 years, performance status less than 2, normal bilirubin and creati- 0.4 ++
+
nine, neither an AHD nor secondary AML, and no pretreatment +++ ++++ + + ++++ + + +
+++
++ ++
infection. Such patients have had a median survival time of 14 months +++ + +
0.2 ++
after administration of various ara-C– containing regimens at M.D. ++ + ++++ ++
+ +++ + +++ +
Anderson Cancer Center over the past decade, with a 38% actuarial +++
survival rate at 2 years, a 5% TID rate at 6 weeks, and a 70% CR rate
(Table 2). Because investigational therapy can be worse than standard 0 1 2 3 4 5

therapy, some patients in this group might prefer standard therapy. Time (years)
The same might be said, although to a lesser extent, of patients aged 70
to 79 years but with an otherwise favorable configuration of perfor- Fig 1. Survival in two groups of older patients with untreated acute myeloid
mance status, bilirubin/creatinine/infection, and AHD/secondary leukemia (log-rank test, P ⬍ .0001). As described in the text, patients in the better
group are more plausible candidates for standard therapy (eg, 3 days of an
AML (Table 2; the number of such patients aged 80 years and older is anthracycline plus 7 days of cytarabine or low-dose cytarabine) than patients in
too small to warrant comment). However, patients less than age 80 the worse group. (ⴱ) Age ⬍ 88 years, performance status ⬍ 2, no antecedent
hematologic disorder or secondary acute myeloid leukemia, bilirubin/creatinine
years with otherwise favorable features composed only 17% of the ⬍ 2, and no infection or fever of unknown origin.
patients seen during this time. The outcome of the remaining 83% of
patients after ara-C– containing therapy is more problematic (Fig 1).22
For example, those patients aged ⱖ 80 years only had an 11-week
most plausible cause of this discrepancy is patients’ natural tendency
median survival time, with 39% dead within 6 weeks of beginning
to believe what they want to believe, there may also be gaps in com-
therapy (Table 2), which is similar to reports from an earlier period.
munication between physicians and patients.
The dominant effect of performance status is seen in the 21-month
median survival time and 24% 6-week mortality rate in otherwise Role of Cytogenetics
favorable patients who have a performance status more than 1. Pa- Optimally, cytogenetics would inform initial management of
tients with an AHD or secondary AML as their only unfavorable untreated AML. For example, considering the most favorable patients
characteristic still had a median survival time of only 35 weeks, despite in Table 2 (row 1), median survival time was 90 weeks in patients with
only a 9% 6-week TID rate. All of these patients are candidates for a normal karyotype or, more rarely, an inv(16) or t (8;21) karyotype,
investigational therapy. However, if such therapy is unavailable, then with 46% alive at 2 years, versus a median survival time of 40 weeks in
the preferred option in those patients aged ⱖ 80 years or with a patients with other karyotypes, with 20% alive at 2 years (Fig 2).
performance status more than 1 would seem to be supportive care Accordingly, the former might be less receptive than the latter to
only given their high early death rates; in contrast, 3⫹7 or LDAC investigational therapy. However, results of cytogenetic tests are often
might be more plausible in patients with only an AHD or second- unavailable for at least 1 week, necessitating that physicians decide
ary AML. As the number of unfavorable covariates increases, so whether it is riskier to delay initiation of therapy or to administer
should the reluctance to give standard therapy rather than investi- potentially toxic standard therapy that is unlikely to be effective
gational therapy.22,23 given the patient’s cytogenetics (or, in the case of a normal karyo-
Any discussion of choice of therapy must refer to the observa- type, to administer investigational therapy when standard therapy
tions of Sekeres et al24 that 74% of older patients estimated that their might be reasonably successful). The Eastern Cooperative Oncol-
chances of cure with 3⫹7 were at least 50%; in contrast, 85% of their ogy Group reported that the 113 patients aged ⱖ 55 years in whom
physicians estimated this chance to be less than 10%. Although the logistical reasons necessitated a 3- to 5-day delay in beginning

Table 2. Outcome According to Indicated Covariates After Cytarabine-Containing Therapy for AML at M.D. Anderson Cancer Center 1996 to 2006
Median % of Probability
Survival Patients of Survival
Age Performance AHD or Bilirubin/Creatinine No. of Time Dead By CR Rate at 2 Years
(years) Status Secondary AML (mg/dL) Infection/FUO Patients (weeks) Day 42 (%) (%)
60-69 0-1 No ⬍2 No 64 58 5 70 38
70-79 0-1 No ⬍2 No 51 46 2 59 28
ⱖ 80 Any Yes or no Any Yes or no 46 11 39 39 11
60-79 ⬎1 No ⬍2 No 21 21 24 67 5
60-69 0-1 Either or both ⬍2 No 127 35 9 55 18

Abbreviations: AML, acute myeloid leukemia; AHD, antecedent hematologic disorder; FUO, fever of unknown origin; CR, complete response.

1910 JOURNAL OF CLINICAL ONCOLOGY


Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138
AML and MDS in the Elderly

this issue by conducting relatively small randomized trials intended to


1.0
Cytogenetics Total Died select which of several agents to investigate in larger trials.34,35 Third,
Normal, Inv(16), t(8;21) 37 21 new drugs are typically studied in patients with active disease. Given
0.8 + Other 27 21 average CR rates less than 50% in older patients, this approach is
Survival Probablity

necessary. However, because the volume of disease is less in CR than at


0.6 + + diagnosis, new therapies might prove to be more effective in prolong-
ing than in inducing remissions. Furthermore, based on well-known
++
+ ++ + + + + relationships between probability of response to previous new drugs
0.4 ++
+ + (eg, administered at relapse) and beneficial effect of a prior therapy
(eg, as measured by duration of initial CR),36 it is plausible that a new
0.2
drug’s benefit will be more readily demonstrable in patients in CR
+
than in untreated patients. Finally, in none of the studies in Table 3 is
it noted that the patients were consecutive patients, raising the possi-
0 1 2 3 4 5 bility of selection bias, similar to that described for minitransplanta-
Time (years) tion (see next paragraph).
Numerous studies comparing allogeneic stem-cell transplanta-
Fig 2. Influence of cytogenetics on survival in patients in the better group tion (SCT) with standard chemotherapy in patients less than age 60
described in Figure 1 who were aged 60 to 69 years (log-rank test, P ⫽ .02). years have found that the likely greater anti-AML effect of SCT is
counterbalanced by greater TID, resulting on average in equivalent
survival.37-41 Current thinking assigns more of a role to an immuno-
induction therapy had a CR rate of 38% compared with a CR rate of
logically mediated graft-versus-leukemia effect than to high-dose
50% in the 235 similarly aged patients who began the same thera-
chemotherapy as the principal mediator of the anti-AML effect of
pies without such delay.11 M.D. Anderson Cancer Center data
SCT.41 If this is the case, it becomes feasible to use reduced-intensity
(unpublished data) suggest that a 1-week delay in beginning ther-
conditioning (RIC) SCT (minitransplantation) to reduce TID. Estey
apy in older patients with WBC counts less than 50,000/␮L is
et al42 described a systematic attempt to perform RIC-SCT in first CR
associated with decreases in median survival time ranging from 4
in all M.D. Anderson patients aged ⱖ 50 years with an abnormal
to 28 weeks depending on other covariates.
karyotype and a sibling or matched unrelated donor. Matching for
If it does not enter into initial management decisions, cytogenetic
known prognostic factors suggested that the14 patients who received
information should be used in planning postremission therapy. In
transplantation had longer survival time than the 83 patients who did
particular, patients with an abnormal karyotype would generally be
not receive transplantation, but these 14 patients represented only 5%
poorer candidates for ara-C or anthracycline plus ara-C than patients
of all treated patients aged more than 50 years with abnormal karyo-
with a normal karyotype.
types. These results question the general applicability of RIC-SCT and
Investigational Approaches suggest that it should be performed before patients enter first CR to
The first three agents listed in Table 3 (tipifarnib,25 lestaurtinib,26 increase the number of patients who might be candidates given the
and decitabine27) are examples of targeted therapy, although, partic- low CR rates in older patients. For example, older patients might
ularly with tipifarnib and less so with lestaurtinib and decitabine, there initially receive a new agent. While waiting to see this agent’s effect, a
is no direct correlation between response and the pretreatment status donor search might be completed, as might various logistical arrange-
of, or the effect of the drug on, the presumed target. As seen in Table 3, ments. RIC-SCT would be performed once response to the new agent
response rates seem higher when the new agent is not a targeted is known.
therapy but bears more resemblance to a traditional cytotoxic agent Cytotoxic T lymphocytes (CTLs) are proposed mediators of
(eg, clofarabine28 or VNP40101M [Cloretazine; Vion Pharmaceuti- graft-versus-leukemia.43 Molldrem et al44 have identified PR1, an
cals, New Haven, CT]29). Accordingly, it is generally assumed that HLA-A2– binding peptide derived from proteinase-3, as a myeloid
many targeted agents will eventually be combined with cytotoxic leukemia–associated antigen for CTL, finding PR1-specific high-
agents, as has been done with gemtuzumab ozogamicin (GO)30 and avidity CTLs in 11 of 12 patients with CML responding to interferon
oblimersen sodium (Marcucci et al, submitted for publication; Table compared with none of seven patients without a response. They sug-
3. In younger patients with favorable or intermediate karyotypes, gested that failure of myeloid leukemia patients to spontaneously
gemtuzumab ozogamicin plus chemotherapy has been reported to develop an immune response to their disease resulted from overex-
lengthen relapse-free survival time32; whether the same is true in older pression of proteinase-3, leading to apoptosis of PR1-specific high-
patients is unknown.) Although not exhaustive, Table 3 makes several avidity CTLs.45 Clinical responses have followed PR1 vaccination of
other relevant points. First, there is an increasing tendency to report patients with AML and have occurred only in patients in whom there
responses (eg, CR with incomplete platelet recovery) less demanding was an immune response, which was defined as ⱖ two-fold increase in
than CR. Although it is known that achievement of CR prolongs the number of PR1-specific CTLs; in contrast, there was no relation-
survival, the effect of these lesser responses on survival remains to be ship between response and the number of CTLs directed against the
established.33 Second, although patients are more interested in the control antigen PP65.43,46 Immune response was more common in
comparative activity of different drugs than in the activity of a given patients in remission when beginning vaccination, leading to a soon to
drug, particularly with respect to survival, most new drug studies are be initiated trial comparing PR1 vaccination with no further therapy
single-arm studies. The National Cancer Research Institute (formerly in patients who are HLA-A2 positive and have completed three
the Medical Research Council) in the United Kingdom is addressing courses of postremission therapy.

www.jco.org 1911
Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138
Elihu Estey

Table 3. Investigational Therapies in Patients Age ⱖ 60 Years With Untreated AML


Response
Median Age ⬍ CR
Type of Drug Example Study No. of Patients (years) CR Rate (%) Rate (%) Effect on Survival
25
Farnesyltransferase Tipifarnib Lancet et al 158 74 14 9 Median survival time,
inhibitor 5.3 months
26
FLT3 inhibitor Lestaurtinib Knapper et al 27 73 0 11 Not given
(CEP701)
Hypomethylating agent Decitabine (plus Lubbert et al27 29 72 14 17 Median survival time,
all-trans-retinoic 7.5 months
acid
Nucleoside analog Clofarabine Burnett et al28 66 71; included 29 20 Median survival time,
only patients 5 months
aged ⱖ 65
Alkylating agent VNP40101M Karp et al29 45 (no AHD or 72 49 — 22% survival at 1
(Cloretazine) secondary year
AML)
Anti-CD33 antibody Gemtuzumab De Angelo et al30 21 67 43 — 48% of patients alive
attached to toxin ozogamicin with median
(⫹ ara-C) follow-up time of 7
months
Bcl-2 antagonist: Oblimersen sodium Marcucci et al 503 — 48 in patients who — None; 1-year survival
enhances apoptosis (added or not (submitted for received rate: 36% with
added to 3 ⫹ 7) publication) oblimersen oblimersen
sodium; 52 in sodium, 40%
patients who did without oblimersen
not receive sodium
oblimersen
sodium
MDR1 antagonist
PSC-833 Added or not Baer et al20 120 70 39 in patients — Median survival time
added to 3 ⫹ 7 who received of 2 months with
plus etoposide PSC-833; 46 in PSC-833 and 7
patients who months without
did not receive PSC-833; study was
PSC-833 stopped because of
excess early deaths
with PSC-833
Zosuquidar Added or not Cripe et al31 442 67 — — None; median
added to 3 ⫹ 7 survival time of 7.7
with zosuquidar
and 9.4 months
without zosuquidar

Abbreviations: AML, acute myeloid leukemia; CR, complete response; AHD, antecedent hematologic disorder; AML, acute myeloid leukemia; ara-C, cytarabine;
3 ⫹ 7, 3 days of an anthracycline and 7 days of cytarabine.

Clinical Practice Issues


MYELODYSPLASTIC SYNDROME
Several practices that may have little effect on mortality but
considerable effect on patients’ lives should be mentioned. First is Like AML, myelodysplastic syndrome (MDS) is a disease that has an
hospitalization; given the more serious nature of hospital-acquired onset usually after age 60 years. Patients are usually classified as high or
infection compared with community-acquired infection, the use of low risk.
routine hospitalization after completion of a course of therapy should
be questioned. The second practice involves masks and avoidance of High-Risk MDS
crowds. The advice to avoid crowds seems inconsistent with observa- High-risk MDS generally implies International Prognostic Scor-
tions that bacteria and fungi, rather than viruses, are the typical causes ing System (IPSS) scores of intermediate-2 or high.49 All patients with
of infection in AML.47,48 The bacteria are invariably residents of the 10% to 20% marrow blasts will meet these criteria, as will patients with
patients’ own skin, mouth, or intestines. The fungi are similarly found two to three cytopenias and either 5% to 10% blasts and intermediate
on the skin or are airborne. However, because of the organism’s size, it cytogenetics or less than 5% blasts and unfavorable cytogenetics, as
is unclear whether masks will restrict entry of Aspergillus into the nose. defined by the IPSS.49 Life expectancy without treatment is typically
The third practice involves consumption of fresh fruits and vegetables. less than 1.5 to 2 years. Thus, the natural history of high-risk MDS
There is conflicting data as to whether avoidance of these foods lessens bears more resemblance to that of AML than to that of an indolent
the risk of infection. An ongoing M.D. Anderson Cancer Center trial smoldering disease, which is how MDS, at times, is regarded. Further-
suggests that patients who are encouraged to eat fresh fruits and more, patients with AML and patients with high-risk MDS have sim-
vegetables have similar infection rates as patients randomly assigned to ilar outcomes when treated identically, after accounting for the older
not eat them (unpublished data). age of MDS patients and their tendency to have ⫺5/⫺7 and longer

1912 JOURNAL OF CLINICAL ONCOLOGY


Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138
AML and MDS in the Elderly

durations of abnormal blood counts.50 Indeed, the boundary between istered and the interval between courses. Thus, in 50% of patients, at
high-risk MDS and AML is malleable; previously defined by a marrow least three courses are needed to observe response to azacitidine58,59;
blast count of 30%, AML is now diagnosed when more than 20% 25% of responses or best responses were seen between course 5 and
blasts are seen.51 Although there are undoubtedly biologic differ- course 17. A similar phenomenon occurs with decitabine.57 Use of
ences between high-risk MDS and AML, there is also likely to be lower doses (eg, 20 mg/m2 daily for 5 days) facilitates administration
biologic overlap, given the somewhat arbitrary distinction between of more courses at more frequent intervals (eg, every 4 to 5 weeks).
the two entities.52 Using such an approach, Kantarjian et al60 reported a CR rate of 34%
Given the natural history of high-risk MDS, the primary of goal in 95 MDS patients, two thirds of whom were high-risk patients. In
of therapy is to lengthen survival.53 The US Food and Drug Adminis- contrast, the CR rate was 9% in the randomized (higher dose, fewer
tration has recently approved lenalidomide, decitabine, and azaciti- and less frequent courses) trial.57 A similar approach might be taken
dine for treatment of MDS. However, too few patients with high-risk with azacitidine.
MDS have received lenalidomide to draw conclusions.54-56 Approval Another investigational therapy is hypomethylating agents plus
of decitabine in MDS was based on a randomized trial comparing histone deacetylase inhibitors. The effect of azacitidine and decitabine
supportive care plus decitabine (15 mg/m2 every 8 hours for 3 days is believed to be mediated by removal of methyl groups from various
intravenously repeated every 6 weeks) with supportive care alone; tumor suppressor genes, with hypomethylation restoring gene expres-
approximately 70% of patients had high-risk MDS, with 25% of these sion.61 Through a different mechanism, histone deacetylase inhibitors
patients having 20% to 30% blasts.57 Decitabine produced a response are also postulated to reactivate transcription of tumor suppressor
rate (CR ⫹ partial response ⫹ hematologic improvement, as defined genes.62 The possibility of synergy between these two classes of
by List et al54) of 30%, reduced RBC transfusion need, and improved epigenetic-acting drugs has led to their combination (eg, azacitidine
quality of life. An analogous trial randomly assigning patients to either plus valproic acid, decitabine plus suberoylanilide hydroxamic acid)
supportive care or supportive care plus azacitidine (75 mg/m2 subcu- in ongoing clinical trials. Given the overlap between high-risk MDS
taneously on days 1 through 7) prompted approval of azacitidine.58,59 and AML, low-dose decitabine and combination epigenetic therapy
Unlike the decitabine trial, crossover was permitted for patients in the are also plausibly effective in AML.
supportive care group who remained transfusion dependent, devel-
Lower Risk MDS
oped worsening cytopenias, or had increased blasts. Forty-six percent
Lower risk MDS includes patients with IPSS scores of
of the patients for whom IPSS scores were known had high-risk MDS,
intermediate-1 or low.48 The natural history is quite variable, but
and substantial numbers of these patients had AML, using the 20%
median survival times usually range from 2 to 10 years. This more
blast criterion. The CR ⫹ partial response ⫹ hematologic improve-
benign natural history leads to consideration of reduction in RBC
ment rate was 47% in the azacitidine arm compared with 17% in
transfusion need and concomitant improvement in quality of life as
the supportive care arm. Both decitabine and azacitidine produced
primary goals.53 Table 4 presents an approach to management of
statistically significant delays in time to AML or death (eg, median
lower risk MDS in transfusion-dependent patients. Lenalidomide is
time, 12.0 months for decitabine v 6.8 months for supportive care
particularly effective in patients with deletion of the long arm of
in high-risk patients). But neither drug resulted in a more than
chromosome 5 (5q–).55 Sixty-seven percent of 148 patients with this
5-month delay in median time to death; in the decitabine trial, the
abnormality became transfusion independent within a median of 4.6
median time to death was 14.0 months for decitabine compared
weeks (range, 1 to 49 weeks) of beginning lenalidomide 10 mg daily for
with 14.9 months for supportive care, with data not broken down
21 days every 4 weeks, with independence maintained for at least 1
by risk group; and in the azacitidine trial, the median time to death
year in 62% of patients. There was no relationship between the com-
in patients who would probably currently be considered as high
plexity of the 5q karyotype and the likelihood of response, but multi-
risk was 18 months in the azacitidine arm compared with 13
variate analysis indicated that response was less likely in patients with
months in the supportive care arm. Although the ability to cross
thrombocytopenia or who required more than 4 units of RBCs in the
over to azacitidine decreased the difference in survival between
8 weeks before study entry.
supportive care and azacitidine, such a crossover would of course
Preliminary data suggest that lenalidomide may also be effec-
be done in clinical practice. The inability of azacitidine and decit-
tive (but less so) in patients with a normal karyotype.56 However,
abine to have more of an effect on survival may reflect the low CR
rates each drug produced (9% for decitabine and 10% for azaciti-
dine). In general, I believe time to AML or death is considerably less
important than time to death; after all, patients presumably care Table 4. Treatment of Low-Risk MDS
little about whether they die with or without AML, and the defini-
Candidate for
tion of AML is itself arbitrary. EPO Level Immunosuppressive
Given the data, I believe that decitabine, azacitidine (as adminis- Cytogenetics (mU/mL) Therapy Treatment
tered in the randomized trials), and lenalidomide are not satisfactory 5q- Any NA Lenalidomide
options for patients with high-risk MDS. Accordingly, like older pa- Not 5q- ⬍ 500 NA EPO ⫾ G-CSF
tients with AML, older patients with high-risk MDS are primarily Not 5q- ⬎ 500 Yesⴱ ATG
Not 5q- ⬎ 500 Noⴱ Azacitidine, decitabine
candidates for investigational treatments.
Besides the investigational treatments noted earlier and in Table Abbreviations: MDS, myelodysplastic syndrome; EPO, erythropoietin; NA,
not applicable; G-CSF, granulocyte colony-stimulating factor; ATG, antithymo-
3, there are other investigational therapies to consider. The first is cyte globulin.

low-dose decitabine. Two confounding factors in any analysis of the See text.
effects of azacitidine and decitabine are the number of courses admin-

www.jco.org 1913
Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138
Elihu Estey

at present, I recommend erythropoietin (EPO) or darbepoetin


SUMMARY
(⫾ granulocyte colony-stimulating factor) for patients without 5q–
with a serum EPO level of less than 500 mU/mL, primarily because Most patients with untreated AML or high-risk MDS should receive
these drugs have fewer adverse effects than lenalidomide. Such pa- investigational therapy. Covariates other than age affect outcome and
tients have a 25% to 75% response rate, with the specific rate inversely should be used in deciding whether specific patients receive investiga-
related to the transfusion rate (eg, ⬍ 2 v ⱖ 2 units/month).63 tional or standard therapy or supportive care only in cases where
Some patients with MDS respond to immunosuppressive ther- investigational therapy is not feasible. There are several problems with
apy (eg, antithymocyte globulin [ATG]), reflecting the autoimmune the approaches used to conduct trials of new agents. Supportive care
nature of MDS; indeed, there is overlap between aplastic anemia and practices for older patients with AML or high-risk MDS should be
MDS. ATG-responsive patients tend to be positive for HLA-DR15 and questioned. Lenalidomide, azacitidine, and decitabine are important
have an age in years and a duration of RBC transfusion dependence in advances for patients with lower risk MDS.
months summing to less than 72.64
Patients who are not candidates for lenalidomide, EPO, or
AUTHOR’S DISCLOSURES OF POTENTIAL CONFLICTS
ATG are candidates for azacitidine, decitabine, or investigational OF INTEREST
therapies. One example of the latter is the PR1 vaccine discussed in
the section on AML.43 The author indicated no potential conflicts of interest.

study and usefulness as a marker for the detection and older with acute myeloid leukemia: Cancer and
REFERENCES of minimal residual disease. Blood 100:59-66, 2002 Leukemia Group B Study 9720. Blood 100:1224-
11. Rowe JM, Neuberg D, Friedenberg W, et al: A 1232, 2002
1. Estey EH, Thall PF, Cortes JE, et al: Compar- phase 3 study of three induction regimens and of 21. National Comprehensive Cancer Network
ison of idarubicin ⫹ ara-C-, fludarabine ⫹ ara-C-, and priming with GM-CSF in older adults with AML: A AML Panel: AML clinical practice guidelines. J Natl
topotecan ⫹ ara-C-based regimens in treatment of trial by the Eastern Cooperative Oncology Group. Comprehensive Cancer Network 1:520-539, 2003
newly-diagnosed acute myeloid leukemia, refractory Blood 103:479-485, 2004 22. Estey E: General approach to, and perspec-
anemia with excess blasts in transformation, or 12. Goldstone AH, Burnett AK, Wheatley K, et al: tives on clinical research in, older patients with
refractory anemia with excess blasts. Blood 98: Attempts to improve treatment outcomes in AML in newly-diagnosed acute myeloid leukemia. Semin
3575-3583, 2001 older patients: The results of the United Kingdom Hematol 43:89-95, 2006
2. Thall P, Estey E: Graphical methods for Medical Research Council AML11 trial. Blood 98: 23. Estey E: How I treat older patients with AML.
evaluating covariate effects in the Cox model, in 1302-1311, 2001 Blood 96:1670-1673, 2000
Crowley J (ed): Handbook of Statistics in Clinical 13. Anderson JE, Kopecky KJ, Willman CL, et al: 24. Sekeres MA, Stone RM, Zahrieh D, et al:
Oncology. New York, NY, Marcel-Dekker, 2001, Outcome after induction chemotherapy for older Decision-making and quality of life in older adults
pp 411-433 patients with AML is not improved with mitox- with AML or advanced MDS. Leukemia 18:809-816,
3. Estey E, Smith T, Keating M, et al: Prediction antrone and etoposide compared to daunorubicin 2004
of survival during induction therapy in patients with and cytarabine: A Southwest Oncology Group 25. Lancet J, Gojo I, Gotlib J, et al: A phase 2
newly diagnosed AML. Leukemia 3:257-263, 1989 Study. Blood 100:3869-3876, 2002 study of the farnesyl transferase inhibitor tipifarnib
4. Appelbaum F, Gundacker H, Head D, et al: 14. Van der Holt B, Lowenberg B, Burnett AK, et in poor-risk and elderly patients with previously
Aging and acute myeloid leukemia. Blood 107:3481- al: The value of the MDR1 reversal agent PSC-833 in untreated acute myelogenous leukemia. Blood 109:
3485, 2006 addition to daunorubicin and cytarabine in the treat- 1387-1394, 2007
5. Tsimberidou A-M, Kantarjian H, O’Brien S, et ment of elderly patients with previously untreated 26. Knapper S, Burnett A, Littlewood T, et al: A
al: Prognostic significance of beta-2 microglobulin AML in relation to MDR1 status at diagnosis. Blood phase 2 trial of the FLT3 inhibitor lestaurtinib
levels in acute myeloid leukemia. Blood 108:240a, 106:2646-2654, 2005 (CEP701) as first-line treatment for older patients
2006 (abstr 802) 15. Estey EH, Thall PF, Giles FG, et al: Gemtu- with acute myeloid leukemia not considered fit for
6. Grimwade D, Walker H, Harrison G, et al: The zumab ozogamicin with or without interleukin 11 in intensive chemotherapy. Blood 108:3262-3270,
predictive value of hierarchical cytogenetic classifi- patients 65 years of age or older with untreated 2006
cation in older adults with acute myeloid leukemia: AML or high-risk MDS: Comparison with idarubicin 27. Lubbert M, Ruter B, Schmid M, et al: Contin-
Analysis of 1065 patients entered into the United plus high-dose continuous infusion cytosine arabino- ued low-dose decitabine is an active first-line treat-
Kingdom Medical Research Council AML 11 trial. side. Blood 99:4343-4349, 2002 ment of older AML patients: First results of a
Blood 98:1312-1320, 2001 16. Tilly H, Castaigne S, Bordessoule D, et al: Low multicenter phase II study. Blood 106:527a, 2005
7. Estey E, Pierce S, Keating M: Identification of dose cytarabine versus intensive chemotherapy in (abstr 1852)
a group of AML/MDS patients with a relatively the treatment of acute nonlymphocytic leukemia in 28. Burnett A, Baccarani M, Johnson P, et al: A
favorable prognosis who have chromosome 5 the elderly. J Clin Oncol 8:272-279, 1990 phase 2 study of clofarabine monotherapy in pa-
and/or 7 abnormalities. Haematologica 85:246-249, 17. Mengis C, Aebi S, Tobler A, et al: Assessment tients aged 65 years or older with acute myeloid
2000 of differences in patient populations selected for or leukemia for whom standard intensive chemothera-
8. Goldstone AH, Burnett AK, Avivi I, et al: excluded from participation in clinical phase III acute py is not considered suitable. Blood 108:130a, 2006
Secondary AML has a worse outcome than de novo myelogenous leukemia trials. J Clin Oncol 21:3933- (abstr)
AML even taking into account cytogenetics and age: 3939, 2003 29. Karp J, Rizzieri D, Vey N, et al: Cloretazine is
AML 10, 11, 12 MRC trials. Blood 100:88a, 2002 18. Hutchins LF, Unger JM, Crowley JJ, et al: an effective induction therapy in elderly patients
(abstr) Underrepresentation of patients 65 years of age or with poor-risk de novo AML. J Clin Oncol 24:340s,
9. Leith C, Kopecky K, Godwin J, et al: Acute older in cancer-treatment trials. N Engl J Med 341: 2006 (suppl, abstr 6512)
myeloid leukemia in the elderly: Assessment of 2061-2067, 1999 30. De Angelo D, Stone RM, Durant S, et al:
multidrug resistance (MDR1) and cytogenetics dis- 19. Lowenberg B, Zittoun R, Kerkhofs H, et al: On Gemtuzumab ozogamicin (Mylotarg) in combination
tinguishes biologic subgroups with remarkably dis- the value of intensive remission induction therapy in with induction chemotherapy for the treatment of
tinct responses to standard chemotherapy—A elderly patients of 65⫹ years with AML: A random- patients with de novo acute myeloid leukemia: Two
Southwest Oncology Group study. Blood 89:3323- ized phase III study of the European Organization for age-specific phase 2 trials. Blood 102:100a, 2003
3329, 1997 Research and Treatment of Cancer Group. J Clin (abstr)
10. Schnittger S, Schoch C, Dugas M, et al: Oncol 7:1268-1274, 1989 31. Cripe L, Li X, Litzow M, et al: A randomized
Analysis of FLT3 length mutations in 1003 patients 20. Baer MR, George SL, Dodge RK, et al: Phase placebo-controlled, double-blind trial of the MDR
with acute myeloid leukemia: Correlation to cytoge- 3 study of the multidrug resistance modulator PSC- modulator zosuquidar during conventional induction
netics, FAB subtype, and prognosis in the AMLCG 833 in previously untreated patients 60 years of age and post-remission therapy for patients ⬎ 60 years

1914 JOURNAL OF CLINICAL ONCOLOGY


Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138
AML and MDS in the Elderly

of age with newly-diagnosed acute myeloid leuke- 42. Estey E, de Lima M, Tibes R, et al: Prospec- the International Working Group (IWG) response
mia or high-risk myelodysplastic syndrome. Blood tive feasibility analysis of reduced intensity condi- criteria in myelodysplasia. Blood 108:419-425, 2006
108:129, 2006 (abstr 423) tioning regimens for hematopoietic stem cell 54. List A, Kurtin S, Roe DJ, et al: Efficacy of
32. Burnett A, Kell W, Goldstone A, et al: The transplantation (HSCT) in elderly patients with acute lenalidomide in myelodysplastic syndromes. N Engl
addition of gemtuzumab ozogamicin to induction myeloid leukemia and high-risk myelodysplastic syn- J Med 352:549-557, 2005
chemotherapy for AML improves disease-free sur- drome. Blood 109:1395-1400, 2007 55. List A, Dewald G, Bennett J, et al: Lenalido-
vival without extra toxicity: Preliminary results of 43. Molldrem JJ: Vaccination for leukemia. Biol mide in the myelodysplastic syndrome with chromo-
1115 patients in the MRC AML15 trial. Blood 108: Blood Marrow Transplant 12:13-18, 2006 (suppl 1) some 5q deletion. N Engl J Med 355:1456-1465,
8a, 2006 (abstr 13) 44. Molldrem JJ, Lee PP, Wang C, et al: Evidence 2006
33. Estey E, Garcia-Manero G, Giles F, et al: that specific T lymphocytes may participate in the 56. Raza A, Reeves JE, Feldman EJ, et al: Long
Clinical relevance of CRp in untreated AML. Blood elimination of chronic myelogenous leukemia. Nat term clinical benefit of lenalidomide treatment in
106, 2005 (abstr 541) Med 6:1018-1023, 2000 patients with myelodysplastic syndrome without del
34. Estey E: Clinical trials in AML of the elderly: 45. Molldrem JJ, Lee PP, Kant S, et al: Chronic 5q cytogenetic abnormalities. Blood 108:78a, 2006
Should we change our methodology? Leukemia myelogenous leukemia shapes host immunity by (abstr)
18:1772-1774, 2004 selective elimination of high-avidity leukemia- 57. Kantarjian H, Issa JP, Rosenfeld CS, et al:
35. Estey E, Thall P: New designs for phase 2 specific T cells. J Clin Invest 111:639-647, 2003 Decitabine improves patient outcomes in myelodys-
clinical trials. Blood 102:442-448, 2003 46. Qazilbash M, Wieder E, Rios R, et al: Vaccina- plastic syndromes. Cancer 106:1794-1803, 2006
36. Breems D, Van Putten W, Huijgens P, et al: tion with the PR1 leukemia-associated antigen can 58. Silverman LR, Demakos EP, Peterson BL, et
al: Randomized controlled trial of azacitidine in pa-
Prognostic index for adult patients with acute my- induce complete remission in patients with myeloid
tients with the myelodysplastic syndrome: A study
eloid leukemia in first relapse. J Clin Oncol 23:1969- leukemia. Blood 104, 2004 (abstr 259)
of the Cancer and Leukemia Group B. J Clin Oncol
1978, 2005 47. Anderlini P, Luna M, Kantarjian HM, et al:
20:2429-2440, 2002
37. Cassileth PA, Harrington DP, Appelbaum FR, Causes of initial remission induction failure in pa-
59. Silverman LR, McKenzie DR, Peterson BL, et
et al: Chemotherapy compared with autologous or tients with acute myeloid leukemia and myelodys-
al: Further analysis of trials with azacitidine in pa-
allogeneic bone marrow transplantation in the man- plastic syndromes. Leukemia 10:600-608, 1996
tients with myelodysplastic syndrome: Studies
agement of acute myeloid leukemia in first remis- 48. Mattiuzzi G, Alvarado G, Alvarez R, et al:
8421, 8921, and 9221 by the Cancer and Leukemia
sion. N Engl J Med 339:1649-1656, 1998 Causes of death in patients with acute myeloid
Group B. J Clin Oncol 24:3895-3903, 2006
38. Zittoun RA, Mandelli F, Willemze R, et al: leukemia and myelodysplastic syndromes undergo-
60. Kantarjian H, Oki Y, Garcia-Manero G, et al:
Autologous or allogeneic bone marrow transplanta- ing induction chemotherapy. Blood 102:878a, 2003
Results of a randomized study of 3 schedules of
tion compared with intensive chemotherapy in (abstr) low-dose decitabine in higher risk myelodysplastic
AML. N Engl J Med 332:217-223, 1995 49. Greenberg P, Cox C, LeBeau M, et al: Inter- syndrome and chronic myelomonocytic leukemia.
39. Burnett AK, Wheatley K, Goldstone AH, et al: national scoring system for evaluating prognosis in Blood 109:52-57, 2007
The value of allogeneic bone marrow transplant in myelodysplastic syndromes. Blood 89:2079-2088, 61. Egger G, Liang G, Aparicio A, et al: Epigenet-
patients with acute myeloid leukaemia at differing 1997 ics in human disease and prospects for epigenetic
risk of relapse: Results of the UK MRC AML 10 trial. 50. Estey E, Thall P, Beran M, et al: Effect of therapy. Nature 429:457-463, 2004
Br J Haematol 118:385-400, 2002 diagnosis (refractory anemia with excess blasts, 62. Bolden JE, Peart MJ, Johnstone RW: Antican-
40. Jourdan E, Boiron J, Dastugue N, et al: Early refractory anemia with excess blasts in transforma- cer activities of histone deacetylase inhibitors. Nat
allogeneic stem-cell transplantation for young adults tion, or acute myeloid leukemia [AML]) on outcome Rev Drug Discov 5:769-784, 2006
with acute myeloblastic leukemia in first complete of AML-type chemotherapy. Blood 90:2969-2977, 63. Hellstrom-Lindberg E, Gulbrandsen N, Lindberg
remission: An intent-to-treat long-term analysis of 1997 G, et al: A validated decision model for treating the
the BGMT experience. J Clin Oncol 23:7676-7684, 51. Vardiman JW, Harris NL, Brunning RD: The anaemia of myelodysplastic syndromes with erythro-
2005 World Health Organization (WHO) classification of poietin ⫹ granulocyte colony-stimulating factor: Sig-
41. Kolb HJ, Schattenberg A, Goldman JM, et al: the myeloid neoplasms. Blood 100:2292-2302, 2002 nificant effects on quality of life. Br J Haematol
Graft-versus-leukemia effect of donor lymphocyte 52. Albitar M, Manshouri T, Shen Y, et al: Myelo- 120:1037-1046, 2003
transfusions in marrow grafted patients: European dysplastic syndrome is not merely “preleukemia.” 64. Saunthararajah Y, Nakamura R, Wesley R, et
Group for Blood and Marrow Transplantation Work- Blood 100:791-798, 2002 al: A simple method to predict response to immu-
ing Party Chronic Leukemia. Blood 86:2041-2050, 53. Cheson BD, Greenberg PL, Bennett JM, et al: nosuppressive therapy in patients with myelodys-
1995 Clinical application and proposal for modification of plastic syndrome. Blood 102:3025-3027, 2003

■ ■ ■

www.jco.org 1915
Information downloaded from jco.ascopubs.org and provided by at Christian Medical College-Vellore on August 9, 2011 from
Copyright © 2007 American Society of Clinical Oncology. All rights reserved.
220.225.126.138

You might also like